WO2005014775A2 - Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a - Google Patents

Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a Download PDF

Info

Publication number
WO2005014775A2
WO2005014775A2 PCT/US2003/020746 US0320746W WO2005014775A2 WO 2005014775 A2 WO2005014775 A2 WO 2005014775A2 US 0320746 W US0320746 W US 0320746W WO 2005014775 A2 WO2005014775 A2 WO 2005014775A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
recombinant adeno
associated viral
viral vector
nucleic acid
Prior art date
Application number
PCT/US2003/020746
Other languages
French (fr)
Other versions
WO2005014775A3 (en
Inventor
Barry J. Byrne
Cathryn S. Mah
Original Assignee
University Of Florida Research Foundation, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Florida Research Foundation, Inc. filed Critical University Of Florida Research Foundation, Inc.
Priority to AU2003304396A priority Critical patent/AU2003304396A1/en
Publication of WO2005014775A2 publication Critical patent/WO2005014775A2/en
Publication of WO2005014775A3 publication Critical patent/WO2005014775A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates generally to the fields of molecular biology and virology, and in particular, to methods for using recombinant adeno-associated virus (rAAV) compositions that express nucleic acid segments encoding therapeutic antihemophilic factor polypeptides useful in the treatment of complex human disorders, including for example, blood disorders such as hemophilia.
  • rAAV adeno-associated virus
  • methods are provided for preparing rAAV-based vector constructs that deliver one or more therapeutic antihemophilic factor compositions to cells, and in particular, human plasma protein Factor VII for the treatment of hemophilia or Factor VII deficiency.
  • Coagulopathic (bleeding and clotting) disorders encompass a wide range of medical problems that lead to poor blood clotting and continuous bleeding. These disorders can result from de ects in the blood vessels or from abnormalities in the blood itself. The abnormalities may be in blood clotting factors or in platelets. Coagulation, the process that controls bleeding, is a complex multi-component process that involves as many as twenty different plasma proteins, or blood clotting factors.
  • Hemophilia is a bleeding disorder caused by a deficiency in one of the blood clotting factors.
  • Hemophilia A (or "classic" hemophilia) is a deficiency in clotting Factor VIII, and accounts for about 80 percent of all hemophilia cases.
  • Hemophilia A is a hereditary disorder in which tlie clotting ability of the blood is impaired and excessive bleeding results. Small wounds and punctures are usually not a problem. But uncontrolled internal bleeding can result in pain and swelling and permanent damage, especially to joints and muscles. Severity of symptoms can vary, and severe forms become apparent early on. The incidence of hemophilia A is 1 out of 10,000 live male births.
  • hemophilia About 20,000 Americans have hemophilia. Women may have it, but the condition is veiy rare in females. Flemophilia is caused by several different gene abnormalities. The severity of the symptoms of hemophilia A depends on how a particular gene abnormality affects the activity of factor VIII. When the activity is less than 1 percent of normal, episodes of severe bleeding occur and recur for no apparent reason.
  • the symptoms of hemophilia include bruising, spontaneous bleeding, bleeding into joints and associated pain and swelling, gastrointestinal tract and urinary tract hemorrhage, blood in the urine or stool, and prolonged bleeding from cuts, tooth extraction, etc.
  • Factor VII deficiency is an extremely rare disorder that can be inherited or acquired by 5 persons without hemophilia who take Coumadin, a d g used to inhibit blood clotting. In this disorder, bleeding can vary from mild to severe within the same person over time. Bleeding doesn't always correspond with the severity of the deficiency shown in blood tests. A history of bleeding may occur in infancy or childhood. Gastrointestinal and central nervous system bleeding can also occur.
  • Factor VII deficiency occurs in approximately one in 500,000 males and females. Congenital Factor VII deficiency is distinguished from Acquired Factor VII Deficiency that may result from liver disease, vitamin K deficiency, or other malabsorption conditions. Unlike hemophilia, Factor FVII deficiency is not sex-linked. It affects both males and females with equal frequency. It is also autosomal recessive, which means that if the 15 clotting defect is inherited from a parent, the child will be a genetic carrier of the condition, but may or may not have symptoms. Those who have inherited a defective Factor VII gene from only one parent will usually have only moderate levels of tlie factor, but without symptoms.
  • Factor VII deficiency often include bleeding of mucous » 0 membranes, spontaneous nosebleeds, excessive bruising, prolonged menstrual bleeding, and bleeding into joints or muscles.
  • the diagnosis for Factor VII deficiency is usually made by testing for Factor VII in the blood, prolonged prothrombm time, or a normal partial thromboplastin time in combination with decreased Factor VII Assay.
  • Existing treatments for Factor VII deficiency rely on the admimsfration of normal plasma or concentrates containing Factor VII. Severe bleeding is typically treated with fresh frozen plasma or PCCs (Prothrombin complex concentrates). However, because the life span of infused factor VII is very short (2 to 4 hours), patients require treatment every 2 to 6 hours for severe bleeding or surgery.
  • the present invention overcomes these and other limitations inherent in the prior art by providing rAAV compositions that comprise a genetic construct that encodes one or more mammalian therapeutic polypeptides, for use in the amelioration, treatment and/prevention of a variety of bleeding disorders, such as for example, hemophilia and/or diseases or conditions that result from a lack, deficiency in, or absence of sufficient biologicall-active Factor VII peptide or polypeptides in one or more cells of the affected mammal.
  • the invention discloses particular rAAV compositions useful in a variety of therapeutic and diagnostic regimens and in the manufacture of medicaments for treating various mammalian bleeding disorders, including Factor VII deficiency and hemophilia A, in particular.
  • the invention provides compositions and methods for treating or ameliorating Factor VII polypeptide deficiencies in a mammal, and particularly Factor VII deficiency in a human, diagnosed with, at risk for developing, or presenting clinical symptoms of one or more bleeding disorders, such as Factor VII deficiency or hemophilia.
  • the invention provides methods for treating or reducing the severity or extent of Factor V ⁇ polypeptide deficiency in a human manifesting one or more of the disorders linked to a deficiency in Factor VII polypeptide in cells and tissues of a human in need thereof.
  • the method involves administration of at least a first composition that comprises a rAAV-based genetic construct that encodes one or more Factor VII peptides, polypeptides, or proteins in a pharmaceutically-acceptable vehicle to the animal in an amount and for a period of time sufficient to prevent, treat, or ameliorate one or more symptoms of the Factor VII deficiency, defect, disorder, disease, or dysfunction in the animal suspected of suffering from one or more disorders linked to a deficiency in Factor VII.
  • the invention provides for superior advantages over the currently-existing products as it results in long-term expression of the Factor VII protein in the patient from a single, or few doses.
  • Current products are extremely unstable and often in short supply as it is extremely difficult and costly to produce.
  • Delivery of a gene therapy therapeuticum such as recombinant Factor VII via recombinant adeno-associated virus (rAAV)-mediated gene transfer overcomes the need for frequent and costly administration of exogenous Factor VII polypeptides, or administration of plasma, plasma concentrates, or plasma extracts containing the Factor VII polypeptide.
  • the present therapy method provides a vital alternate form of treatment for hemophiliacs which are refractory to current replacement therapies. Additional benefits of the present therapy include the elimination of a need for repeated injections or exogenous Factor VH polypeptide. In fact, recipients of the current therapy would receive multiple benefits including, for example, the increased efficacy of treating the disease than with the currently available modalities, and an overall reduced healthcare cost, as repeated injections of the purified protein are obviated by the gene therapy methods provided herein. Many patients with hemophilia A are not able to control their disease with current replacement therapies and are left with few or no options.
  • This invention provides a cost- effective option, with improved patient compliance, more convenient dosage and administration regimens, and less repeated injections.
  • the recombinant AAV- Factor VII compositions disclosed herein also lower expenses due to far fewer dosings and increased stability and longevity of treatment.
  • a single dose of recombinant AAV has been shown to produce therapeutic proteins over the course of weeks, months, and even years.
  • the invention provides an rAAV vector comprising a polypeptide that comprises at least a first nucleic acid segment that encodes a mammalian Factor VII peptide or polypeptide, and in particular, a biologically-active Factor VII (FVII) polypeptide, or biologically-active fragment thereof, operably linked to at least a first promoter capable of expressing the nucleic acid segment in a suitable host cell transformed with such a vector.
  • FVII biologically-active Factor VII
  • the nucleic acid segment encodes a mammalian, and in particular, a human Factor VII polypeptide, such as for example, one or more of the polypeptides as disclosed in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, and SEQ ID NO: 17.
  • a human Factor VII polypeptide such as for example, one or more of the polypeptides as disclosed in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, and SEQ ID NO: 17.
  • the invention also encompasses treatment modalities involving the use of one or more other
  • rAAV vectors may comprise sequences of murine, porcine, feline, canine, bovine, ovine,
  • the rAAV-Factor VII constructs comprise at least a first genetic sequence that encodes a human Factor VII peptide, polypeptide, or
  • the therapeutic constructs of the invention may encompass nucleic acid segments that encode modified hemophilia-inhibitory polypeptides obtained from any
  • Preferred rAAV vector backbones for the practice of the present invention include,
  • rAAV serotype 1 rAAVl
  • rAAV serotype 2 rAAV2
  • the polynucleotides comprised in the vectors and viral particles of the present invention preferably comprise at least a first constitutive or inducible promoter operably linked to the nucleic acid segments disclosed herein.
  • promoters may be homologous or heterologous promoters, and may be operatively positioned upstream of the nucleic acid segment encoding the therapeutic polypeptide of interest, such that the expression of the segment is under the control of the promoter.
  • the construct may comprise a single promoter, or alternatively, two or more promoters may be used to facilitate expression of the therapeutic gene sequence.
  • Exemplary promoters useful in the practice of the invention include, but are in no way limited to, those promoter sequences that are operable in mammalian, and in particular, human host cells, tissues, and organs, such as for example, a
  • CMV promoter a ⁇ -actin promoter, a hybrid CMV promoter, a hybrid ⁇ -actin promoter, an
  • EF1 promoter a Ula promoter, a Ulb promoter, a Tet-inducible promoter and a VP16-LexA promoter being particularly useful in the practice of the invention.
  • a polynucleotide encoding a therapeutic polypeptide was placed under the
  • CBA chicken ⁇ -actin
  • the polynucleotides comprised in the vectors and viral particles of the present invention may also further optionally comprise one or more native, synthetic, homologous, heterologous, or hybrid enhancer or 5' regulatory elements, for example, a CMV enhancer, a synthetic enhancer, or a liver- or tissue-specific enhancer operably linked to the therapeutic polypeptide-encoding nucleic acid segments disclosed herein.
  • the polynucleotides and nucleic acid segments comprised within the vectors and viral particles of the present invention may also further optionally comprise one or more intron sequences.
  • the polynucleotides comprised in the vectors and viral particles of the present invention may also further optionally comprise one or more native, synthetic, homologous, heterologous, or hybrid post-transcriptional or 3' regulatory elements operably positioned relative to the therapeutic polypeptide-encoding nucleic acid segments disclosed herein to provide greater expression, stability, or translation of the encoded polypeptides.
  • One such example is the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), operably positioned downstream of the therapeutic gene of interest.
  • WPRE woodchuck hepatitis virus post-transcriptional regulatory element
  • the invention concerns administration of one or more biologically-active neovascularization-inhibitory peptides or polypeptides that comprise an at least 20, at least 40, at least 60, at least 80, at least 100, at least 120, at least 140, at least 160, at least 180, at least 200, at least 220, at least 240, at least 260, at least 280, at least 300, or more contiguous amino acid sequence from one or more of the polypeptide sequences disclosed in Section 5.2 hereinbelow and particularly those polypeptides as recited in any one of SEQ ID NO:2, SEQ ID NO:4. SEQ ID NO:6.
  • the invention concerns administration of one or more biologically-active Factor VII polypeptides that are encoded by a nucleic acid segment that comprises at least 30, at least 60, at least 90, at least 120, at least 150, at least 180, at least 210, at least 240, at least 270, at least 300, at least 330, at least 360, at least 390, at least 420, at least 450, at least 480, at least 510, at least 540, at least 570, or at least 600, 700, 800, or 900, or more contiguous nucleic acid residues, up to and including substantially full-length, and full-length sequences from the DNA sequences disclosed in Section 5.2 hereinbelow and particularly those DNA sequences as recited in any one of SEQ ID NO:l,
  • adeno-associated viral vector constructs and polynucleotides of the present invention include those that comprise, consist essentially of, or consist of at least a first nucleic acid segment that encodes a peptide or polypeptide that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the sequence of SEQ ID NO:2, SEQ ID NO:4. SEQ ID NO:6.
  • SEQ ID NO:8 SEQ ID NO:10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO:15, SEQ ID NO:16, or SEQ ID NO: 17, wherein the peptide or polypeptide has Factor VII activity when administered to, and expressed in, a suitable mammalian cell.
  • Exemplary polynucleotides of the present invention also include those sequences that comprise, consist essentially of, or consist of at least a first nucleic acid segment that encodes a polypeptide that is at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about
  • Particularly preferred adeno-associated viral vector constructs and polynucleotides of the present invention include those that comprise, consist essentially of, or consist of at least a first nucleic acid segment that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the sequence of any one of SEQ ID NO:l, SEQ ID NO:3.
  • SEQ ID NO:5 SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13, wherein the segment encodes a peptide or polypeptide that has Factor VII activity when administered to, and expressed in, a suitable mammalian cell.
  • Exemplary polynucleotides of the present invention also include those sequences that comprise, consist essentially of, or consist of at least a first nucleic acid segment that is at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about
  • SEQ ID NO:5 SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13, wherein the peptide or polypeptide encoded by the nucleic acid segment has Factor VII activity when administered to a mammalian eye.
  • rAAV particles and virions that comprise the vectors of the present invention, pluralities of such particles and virions, as well as pharmaceutical compositions and host cells that comprise one or more of the rAAV vectors disclosed herein, such as for example pharmaceutical formulations of the rAAV vectors or virions intended for administration to a mammal through suitable means, such as, by intramuscular, intravenous, or direct injection to selected cells, tissues, or organs of the mammal, for example, to the muscle tissue, the circulatory system, or directly to one or more organs of the selected mammal, such as for example, by direct administration to the liver, or to liver cells.
  • suitable means such as, by intramuscular, intravenous, or direct injection to selected cells, tissues, or organs of the mammal, for example, to the muscle tissue, the circulatory system, or directly to one or more organs of the selected mammal, such as for example, by direct administration to the liver, or to liver cells.
  • compositions will be formulated with pharmaceutically- acceptable excipients, buffers, diluents, adjuvants, or carriers, as described hereinbelow, and may further comprise one or more liposomes, lipids, lipid complexes, microspheres, microparticles, nanospheres, or nanoparticle formulations to facilitate administration to the selected organs, tissues, and cells for which therapy is desired.
  • compositions of the invention also will optionally further comprising at least a first pharmaceutical vehicle, and particularly those formulations that are acceptable for administration to a human through one or more conventional routes of administration, such 5 as for example, oral, nasal, inhalation, trasndermal, intravenous, subcutaneous, or intramuscular administration.
  • compositions of the invention may also further comprise one or more liposomes, lipids, proteins, peptides, polypeptides, nucleic acids, polysaccharides, antibodies, antigens, antigen binding fragments, enzymes, lipid complexes, or at least a first detectable label, marker, or tag.
  • the rAAV particles may be conjugated or 0 otherwise associated with one or more surfaces of a micro- or nanoparticle, such as for example, the nanospheres and microspheres described herein.
  • kits for treating or ameliorating the symptoms of hemophilia, bleeding or clotting disorders, or other condition resulting from defect, deficiency, or dysfunction of the native Factor FII polypeptide in a mammal are also part of the present invention. Such kits are also part of the present invention.
  • !5 typically comprise one or more of the disclosed AAV vector constructs, virion or virus particles, host cells, or therapeutic AAV compositions described herein, and instructions for using the kit. Another important aspect of the present invention concerns methods of use of the
  • Such methods generally involve administration to a mammal.
  • the methods may also provide a defect, dysfunction, or deficiency in the affected mammal.
  • Such symptoms may include,
  • compositions that comprise one or more of
  • compositions comprising such are particularly useful as described herein.
  • the present invention also provides for a host cell that comprises the rAAV-Factor
  • such host cells are mammalian cells, with
  • the host cell may be a human pancreas, kidney, muscle, epithelial, endothelial, perivascular, liver, heart, lung, brain, blood, bone, or nerve cell.
  • the invention also provides methods for delivering therapeutically-effective amounts of a biologically-active Factor VII polypeptide to a mammal in need thereof.
  • Such methods generally comprise at least the step of providing or administering to such a mammal, one or more of the AAV-Factor VII compositions disclosed herein.
  • the method may involve providing to such a mammal, one or more of the rAAV vectors,0 virions, viral particles, host cells, or pharmaceutical compositions as described herein.
  • such providing or such administration will be in an amount and for a time effective to provide a therapeutically-effective amount of one or more of the Factor VII peptides or polypeptides disclosed herein to selected cells, tissues, or organs of the mammal, and in particular, therapeutically-effective levels to the cells, tissues, or organs of 5 the mammal.
  • Such methods may include systemic injection(s) of the therapeuticum, or may even involve direct or indirect administration, injection, or introduction of the therapeutic compositions to particular cells, tissues, or organs of the mammal, such as for example, by direct injection into muscle or liver tissues.
  • the invention also provides methods of treating, ameliorating the symptoms, and
  • the invention provides a method for treating, preventing, or ameliorating the symptoms of a Factor VII protein, peptide, or polypeptide deficiency or dysfunction in a mammal.
  • the method generally involves administering to a mammal in need thereof, one or more of the disclosed rAAV-Factor VII vector compositions disclosed herein, in an amount and for a time sufficient to treat, prevent, or ameliorate the symptoms of the Factor VII deficiency or the bleeding disorder or dysfunction, such as e.g., hemophilia, in the mammal.
  • the mammal is a human that is has, is at risk for developing, or has been diagnosed with one or more diseases, disorders, or dysfunctions that result from the deficiency or lack of one or more Factor VII peptides, polypeptides, or proteins normally present in a healthy subject.
  • compositions of the invention may be administered to the patient in an amount and for a time sufficient to treat or prevent the symptoms of the Factor VII deficiency or dysfunction through a single dose, or by administration of a plurality of doses given over a relatively short, or even relatively long period of therapy.
  • the patient may require only one or two administrations of the disclosed rAAV constructs to achieve relatively short-term, relatively medium-term, or even relatively long-term treatment.
  • one or two administrations of the disclosed compositions may provide sufficient therapeutic levels of the Factor VII composition for a period of several days, several weeks, or several months.
  • three or four administrations of the disclosed compositions may provide sufficient therapeutic levels of the Factor VII composition for a period of several weeks, several months, several years, or even tens of years, up to and including the natural lifetime of the treated mammal.
  • the therapeutic effectiveness of a single administration or of multiple administrations of the disclosed compositions may be provided.
  • administration or of multiple administrations of the disclosed compositions may persist for a
  • compositions may persist for a period of about 41, 42, 43, 44, 45, 46, 47, 48, 49, or
  • compositions contemplate that particular therapeutic regimens involving one or more of the compositions
  • months at least about 4 months, at least about 5 months, at least about 6 months, at least
  • rAAV-Factor VII compositions disclosed herein may be administered by any of the conventional drug delivery methods, such as for example, orally, intranasally,
  • compositions disclosed herein include, but are not limited to, the vascular or circulatory system, the0 pancreas, liver, heart, lung, brain, kidney, joint, bone, neural, and muscles.
  • kits for treating, preventing, or ameliorating the symptoms of a Factor VII protein, peptide, or polypeptide-related deficiency or disorder in a mammal comprising (i) one or more rAAV-Factor VII composition disclosed herein; and (ii) instructions for using the kit in diagnostic, therapeutic, or5 prophylactic treatment regimens.
  • FIG. 1 shows that functional Factor VII can be produced in vitro.
  • the chromogenic Coaset assay was used to determine the levels of functional Factor VII secreted.
  • Factor VIII KO plasma was used as a positive control as these mice have been shown to produce normal levels
  • Adeno-associated virus is a single-stranded DNA parvo virus with a 4.7 kb genome and a particle diameter of approximately 20 nm.
  • the AAV genome is flanked by two identical inverted terminal repeat (ITR) sequences (Lusby et al, 1980). These ITRs provide all the cw-acting sequence required for replication, packaging and integration (Samulski et al, 1989).
  • ITRs inverted terminal repeat
  • There are two large open reading frames (Srivastava et al, 1983).
  • the open reading frame in the right half of the genome encodes 3 overlapping coat proteins (VP1, VP2 and VP3).
  • the open reading frame in the left half encodes 4 regulatory proteins with overlapping sequences which are known as Rep proteins (Rep78, Rep68, Rep52 and j?ej_>40), because frame shift mutations at most locations within the open reading frame inhibit viral DNA replication (Hermonat et al, 1984).
  • the Rep proteins are multi-functional DNA binding proteins.
  • the functions of the Rep proteins in viral DNA replication include helicase activity and a site-specific, strand-specific endonuclease (nicking) activity (Ni et al, 1994).
  • AAV infects a broad spectrum of vertebrates from birds to humans, although in nature specific types are species specific (Berns, 1996). In humans AAV can infect a large variety of cells derived from different tissues.
  • rAAV is the safest of the currently used gene therapy vectors. Because of its propensity to establish latency and because it has not been implicated as a pathogen, AAV has been of considerable interest as a potential vector for human gene therapy (Flotte and Ferkol, 1997; Flotte and Carter, 1995). In general, rAAV vectors are produced by deleting the viral coding sequences and substituting the fransgene of interest under control of a non-AAV promoter between the two AAV inverted terminal repeats (ITRs).
  • ITRs inverted terminal repeats
  • rAAV genomes can be efficiently packaged. Considerations in the development of AAV as a vector have included difficulties in attaining high vector titers and the limited insertional capacity (>5 kb). Although these issues can still be improved, recently developed packaging techniques for high titer and Ad- contamination free vectors, and strategies to overcome the packaging limitation, have dramatically impacted the applications of rAAV (Zolotukhin et al, 1999; Duan et al, 2000;
  • rAAV vectors are remarkably nonimmunogenic with little host response (Jooss et al, 1998; Song et al, 1998).
  • rAAV have mediated long-term fransgene expression in a wide variety of tissues, including muscle (Song et al, 1998; Kessler et al, 1996; Xiao et al, 1996; Clark et al, 1997; Snyder et al, 1997a), lung (Flotte et al, 1993), liver (Snyder et al, 1997b; Xiao et al, 1998; Song et al, 2001a; Xu et al, 2001), brain (Kaplitt et al, 1994) and eye (Flannery et al, 1997).
  • rAAV vectors appear to have significant advantages over other commonly used viral vectors.
  • Six serotypes of AAV have been cloned and sequenced. Of the six AAV serotypes, serotype 2 (AAV2) is the best-characterized and has been predominantly used in gene transfer studies.
  • Membrane-associated heparan sulfate proteoglycan is the primary receptor for AAV
  • integrin are co-receptors for AAV2 (Qing et al, 1999; Summerford et al, 1999). Serotypes 1 and 6 share >99% amino acid homology in their capsid proteins. Sequence analysis supports a recombination event between seroType I and 2. Comparison of the serotype capsid amino acid sequences suggests that serotypes, 1, 2, and 3 share homology across the three capsids in accord with heparan sulfate binding (Summerford and Samulski, 1998). In contrast, AAV type 4 and 5 are the most divergent of the six AAV serotypes, exhibiting only 60% homology to AAV2 or to each other.
  • AAV4 and AAV5 require different sialic acid-containing glycoproteins for binding and transduction of target cells.
  • the different tropisms of AAV serotypes provide opportunities to optimize the transduction efficiency in different target cells.
  • Data showed that of the serotypes, AAV1 mediated the highest fransgene expression in skeletal muscle and murine islets (Chao et ⁇ /., 2000).
  • the AAV life cycle is quite unusual (Berns and Linden, 1995).
  • AAV binds to cells via a heparan sulfate proteoglycan receptor (Summerford and Samulski, 1998). Once attached, AAV
  • a co-receptor which may consist of either the fibroblast
  • FGF-R growth factor receptor
  • v - ⁇ 5 integrin molecule the v - ⁇ 5 integrin molecule
  • UV irradiation or treatment with genotoxic agents will undergo productive replication of AAV
  • AAV Rep protein binding and nicking by the AAV Rep protein, a non-structural protein that is also involved in
  • the AAV genome consists of two 145-nucleotide inverted terminal repeat (ITR)
  • rep and cap Tratschin et al , 1984.
  • the rep gene is transcribed from two promoters, the
  • Rep78 and Rep68 are multifunctional DNA
  • binding proteins which possess helicase activity and site-specific, strand-specific nickase
  • the long Rep proteins are also capable of binding to the chromosomal target
  • Rep78/68 are potent bi-functional transcription regulators that generally activate transcription from AAV promoters during productive infection and repress their transcription during latent infection (Pereira and Muzyczka, 1997; Pereira et al, 1997).
  • the shorter Rep proteins, Rep52 and Rep40 act as modifier proteins for long Rep transcriptional activities, and are required for sequestration of single-stranded AAV genomes into capsids during productive infection.
  • the AAV cap gene is transcribed from the p40 promoter.
  • the 5' end of the mRNA transcript from p40 contains an intron which can utilize either of two downstream splice acceptor sites.
  • the longer of the two processed messages contains an ATG codon which is used in the translation of VP1, the longest of the three AAV capsid proteins.
  • the shorter mRNA can initiate translation using either a non-canonical ACG start codon, which represents the start of
  • VP3 is the shortest and most abundant of the AAV capsid proteins, but all three are required for the production of infectious virions.
  • rAAV RECOMBINANT AAV VECTORS
  • rAAV vectors have been developed by replacement of the viral coding sequences with fransgene of interest (Tratschin et al, 1984; Hermonat and Muzyczka, 1984).
  • the ITR sequences must be retained in rAAV since these serve as origins for viral DNA replication and contain the packaging signals.
  • the maximum packaging capacity of rAAV is approximately 5 kb, including the ITRs, the fransgene, its promoter, and polyadenylation signal
  • rAAV vectors are uniquely suitable for in vivo gene therapy for genetic and metabolic disorders, since they are non-toxic (Flotte et al, 1993; Conrad et al, 1996; Flotte and Carter,
  • differentiated cells such as neurons (Kaplitt et al, 1994; McCown et al, 1996; Peel et al, 1997;
  • AAV adeno-associated virus 2
  • AAV is an ideal vector for viral-based human gene therapy because it has not been associated with any known pathology and post-infection, the viral genome integrates into the human chromosome (Muzyczka, 1992).
  • Recombinant adeno-associated virus (rAAV) vectors have important utility as vehicles for the in vivo delivery of polynucleotides to target host cells (Kessler etal, 1996; Koeberl etal, 1997; Kotin, 1994;
  • rAAV vectors are useful vector for efficient and long-term gene transfer in a variety of mammalian tissues, e.g., lung (Flotte et ah, 1993), muscle (Kessler et al, 1996; Xiao etal, 1996; Clark etal, 1997; Fisher etal, 1997), brain (Kaplitt etal, 1994; Klein etal, 1998) retina (Flannery etal, 1997; Lewin etal, 1998), and liver (Snyder, 1997).
  • mammalian tissues e.g., lung (Flotte et ah, 1993), muscle (Kessler et al, 1996; Xiao etal, 1996; Clark etal, 1997; Fisher etal, 1997), brain (Kaplitt etal, 1994; Klein etal, 1998) retina (Flannery etal, 1997; Lewin etal, 1998), and liver (Snyder, 1997
  • rAAV can evade the immune response of the host by failing to transduce dendritic cells.
  • Clinical trials have been initiated for several important mammalian diseases including hemophilia B, muscular dystrophy and cystic fibrosis (Flotte etal, 1996; Wagner etal, 1998; Flotte and Carter, 1995; Kay et al, 2000).
  • hemophilia B muscular dystrophy
  • cystic fibrosis Flotte etal, 1996; Wagner etal, 1998; Flotte and Carter, 1995; Kay et al, 2000.
  • obstacles in the efficient use of rAAV vectors for a variety of disease models include sub-therapeutic levels of transduction and the ability to target the site(s) of gene transfer.
  • expression vector or construct means any type of genetic construct containing a nucleic acid in which part or all of the nucleic acid encoding sequence is capable of being transcribed. In preferred embodiments, expression only includes transcription of the nucleic acid, for example, to generate a biologically-active Factor VII polypeptide product from a transcribed gene. Particularly useful vectors are contemplated to be those vectors in which the nucleic acid segment to be transcribed is positioned under the transcriptional control of a promoter.
  • a “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene.
  • a recombinant or heterologous promoter is intended to refer to a promoter that is not normally associated with a biologically-active Factor V ⁇ gene in its natural environment.
  • Such promoters may include promoters normally associated with other genes, and/or promoters isolated from any bacterial, viral, eukaryotic, or mammalian cell.
  • promoter that effectively directs the expression of the biologically-active Factor VH-encoding DNA segment in the cell type, organism, or even animal, chosen for expression.
  • the use of promoter and cell type combinations for protein expression is generally known to those of skill in the art of molecular biology, for example, see Sambrook etal. (1989), incorporated herein by reference.
  • the promoters employed may be constitutive, or inducible, and can be used under the appropriate conditions to direct high-level expression of the introduced DNA segment.
  • At least one module in a promoter functions to position the start site for RNA synthesis.
  • TATA box In some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl fransferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation. Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upsfream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
  • the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • individual elements can function either co-operatively or independently to activate transcription.
  • the particular promoter that is employed to control the expression of a nucleic acid is not believed to be critical, so long as it is capable of expressing the biologically-active Factor VII polypeptide-encoding nucleic acid segment in the targeted cell.
  • such a promoter might include either a human or viral promoter, such as a beta- actin, CMV, an HSV promoter, or even a human tissue-specific or otherwise inducible promoter.
  • a human or viral promoter such as a beta- actin, CMV, an HSV promoter, or even a human tissue-specific or otherwise inducible promoter.
  • the chicken beta-actin promoter has been demonstrated to be particularly desirable in some embodiments disclosed herein.
  • the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression of transgenes.
  • Factor VII vectors and compositions of the present invention This list is not intended to be exhaustive of all the possible elements involved in the promotion of fransgene expression, but merely to be exemplary thereof.
  • Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of DNA with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins. The basic distinction between enhancers and promoters is operational.
  • an enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements.
  • a promoter must have one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization. Additionally any promoter/enhancer combination (as per the Eukaryotic Promoter Data Base EPDB) could also be used to drive expression.
  • Use of a T3, T7 or SP6 cytoplasmic expression system is another possible embodiment. Eukaryotic cells can support cytoplasmic transcription from certain bacterial promoters if the appropriate bacterial polymerase is provided, either as part of the delivery complex or as an additional genetic expression construct.
  • NCAM Neural Cell Adhesion Molecule Hirsch et al. , 1990
  • Troponin I (TN I) Yutzey et ⁇ /., 1989
  • Retrovimses Kriegler and Botchan, 1982, 1983; Levinson et al, 1982; Kriegler etal, 1983, 1984a, b, 1988; Bosze et al, 1986; Miksicek et al, 1986; Celander and Haseltine, 1987; Thiesen et al, 1988; Celander et al, 1988; Choi etal, 1988; Reisman and Rotter, 1989
  • MMTV mimmary Glucocorticoids Huang et al. , 1981; Lee et al. , tumor vims
  • Majors and Varmus 1983; Chandler et al, 1983; Lee et al, 1984; Ponta et al, 1985; Sakai et /., 1988
  • engineered and recombinant cells are intended to refer to a cell into which an exogenous DNA segment, such as DNA segment that leads to the transcription of a biologically-active Factor VII polypeptide or a ribozyme specific for such a biologically-active Factor VII polypeptide product, has been introduced. Therefore, engineered cells are distinguishable from naturally occurring cells, which do not contain a recombinantly introduced exogenous DNA segment. Engineered cells are thus cells having DNA segment introduced through the hand of man. To express a biologically-active Factor VII encoding gene in accordance with the
  • an rAAV expression vector that comprises a biologically- active Factor VH polypeptide-encoding nucleic acid segment under the confrol of one or more
  • promoters To bring a sequence "under the control of a promoter, one positions the 5' end of the transcription initiation site of the transcriptional reading frame generally between about
  • promoter stimulates transcription of the DNA and promotes expression of the encoded
  • polypeptide This is the meaning of "recombinant expression” in this context. Particularly,
  • preferred recombinant vector constructs are those that comprise an rAAV vector. Such vectors are described in detail herein.
  • the present invention concerns formulation of one or more of
  • islet cells such as for example, islet cells.
  • nucleic acid segments RNA, DNA or PNA
  • compositions that express one or more of the biologically-active Factor VII therapeutic gene that express one or more of the biologically-active Factor VII therapeutic gene
  • products as disclosed herein may be administered in combination with other agents as well, such as
  • proteins or polypeptides as, e.g., proteins or polypeptides or various pharmaceutically-active agents, including one or
  • compositions may thus be delivered along with various other agents as required in the particular instance.
  • Such compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein.
  • such compositions may further comprise substituted or derivatized RNA, DNA, or PNA compositions.
  • Formulation of pharmaceutically-acceptable excipients and carrier solutions is well- known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular administration and formulation.
  • these formulations may contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 70% or 80% or more of the weight or volume of the total formulation.
  • the amount of active compound(s) in each therapeutically- useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound.
  • Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • compositions disclosed herein either subcutaneously, intraopancreatically, parenterally, intravenously, intramuscularly, intrathecally, or even orally, infraperitoneally, or by nasal inhalation, including those modalities as described in U. S. Patent 5,543,158; U. S. Patent 5,641,515 and U. S. Patent 5,399,363 (each specifically incorporated herein by reference in its entirety).
  • Solutions of the active compounds as freebase or pharmacologically acceptable salts may be prepared in sterile water and may also suitably mixed with one or more surfactants, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U. S. Patent 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., vegetable oils
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion
  • isotonic agents for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the solution may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences” 15th Edition, pages 1035- 1038 and 1570-1580).
  • Some variation in dosage will necessarily occur depending on the condition of the subject being treated.
  • the person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • preparations should meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologies standards.
  • Sterile injectable solutions are prepared by incorporating the active AAV vector- delivered biologically-active Factor VII polypeptide-encoding polynucleotides in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • compositions disclosed herein may also be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the
  • carrier includes any and all solvents, dispersion media, vehicles, and the like.
  • coatings diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents,
  • compositions are pharmaceutically-acceptable.
  • pharmaceutically-acceptable refers to molecular entities and compositions
  • compositions are prepared as mjectables, either as liquid
  • solutions or suspensions solid forms suitable for solution in, or suspension in, liquid prior to
  • injection can also be prepared.
  • the preparation can also be emulsified.
  • the inventors contemplate the use of liposomes, nanocapsules,
  • a microparticle is well
  • microparticles are typically on the order of 5 mm or less in diameter, and they may assume any shape, such as spherical, polygonal, fiber-like, or simply a fractured piece of a larger structure. Nanoparticles are understood to encompass even smaller particles, typically those having diameters in the 5 micron or less range. These particles can be made by a wide variety of known methods such as suspension, emulsion or dispersion polymerization. Ball-milling, or grinding (for instance of a larger piece cooled to low temperature if not brittle at room temperature) can be used to create smaller fragments.
  • Aerosol spraying and solidification by cooling or photo cross-linking can also be employed to prepare microparticles or nanoparticles of preferred sizes and compositions.
  • Ultrasonication for example, may be used to disperse one liquid in another.
  • Many microparticles can be obtained directly from commercial sources (for example, Bangs Laboratories) or simply made by precipitating iron oxides and using those particles without polymers, or co-precipitation them with other solids.
  • combinations of materials may also be used to prepare suitable microspheres.
  • polymers of degradable solids polylactic acid, polyglycolic acid, and their copolymers
  • hydrogels such as dextran or starch or alginate
  • non-degradable hydrogels such as polyhydroxyethyl methacrylate (PHEMA)
  • PHEMA polyhydroxyethyl methacrylate
  • non-degradable solids like polystyrene or hydroxy apatite or plaster of paris
  • magnetic field-responsive compounds e.g., magnetite
  • even finely milled particles of magnetic metals e.g., iron metal
  • Such nanoparticle- and microparticle-associated formulations may be preferred for the introduction of pharmaceutically acceptable formulations of the rAAV-Factor VII vectors disclosed herein.
  • the inventors also contemplate the use of other delivery regimens to improve introduction of the rAAV vectors into suitable animals.
  • Such delivery regimens may include, for example, the use of nanocapsules, liposomes, lipid particles, and the like.
  • the formation and use of liposomes for pharmaceutical administration is generally known to those of skill in the art (see for example, Couvreur et al, 1977; Couvreur, 1988; Lasic, 1998; which describes the use of liposomes and nanocapsules in the targeted antibiotic therapy for intracellular bacterial infections and diseases).
  • liposomes were developed with improved serum stability and circulation half-times (Gabizon and Papahadjopoulos, 1988; Allen and Choun, 1987; U. S. Patent 5,741,516, specifically incorporated herein by reference in its entirety). Further, various methods of liposome and liposome like preparations as potential dmg carriers have been reviewed (Takakura, 1998; Chandran et al, 1997; Margalit, 1995; U. S.
  • Patent 5,567,434 U. S. Patent 5,552,157; U. S. Patent 5,565,213; U. S. Patent 5,738,868 and U. S. Patent 5,795,587, each specifically incorporated herein by reference in its entirety).
  • Exemplary microparticles and nanoparticles, and methods for their synthesis have been described in a variety of patents including, for example, U.S. Patents 6,383,470, 6,346,274, 6,238,294, 6,284,280, 6,331,310, 6,254,890, 6,177,088, 5,972,707, 6,2587,588, and 6,361,994, each specifically incorporated herein by reference in its entirety.
  • Magnetically responsive microparticles are useful in biological techniques requiring the separation of bound from free fractions.
  • Magnetically responsive particles useful in immunoassays, for the separation of cells, as magnetic resonance imaging agents, etc. have been described in U.S. Pat. Nos. 3,215,572, 4,452,773, 4,795,698, 4,770,183, 4,695,392,
  • Such biologically-active magnetic particles may find use in a variety of preparative and
  • HGMS HGMS which uses a magnetic field to separate magnetic particles from suspension.
  • the material of interest may thereby be separated from other materials not bound to the
  • suspendable coated particle refers to a finely divided solid
  • Magnetic encompasses material which may or may not be permanently
  • magnetic which also may be paramagnetic or superparamagnetic but which in all cases exhibits
  • Liposomes have been used effectively to introduce genes, drags (Heath and Martin,
  • Liposomes are formed from phospholipids that are dispersed in an aqueous medium and
  • multilamellar concentric bilayer vesicles also termed multilamellar vesicles
  • MLVs generally have diameters of from 25 nm to 4 ⁇ m. Sonication of MLVs results
  • Liposomes bear resemblance to cellular membranes and are contemplated for use in connection with the present invention as carriers for the peptide compositions. They are widely
  • lipid-soluble substances suitable as both water- and lipid-soluble substances can be entrapped, i.e. in the aqueous spaces and within the bilayer itself, respectively. It is possible that the drug-bearing liposomes may
  • Phospholipids can form a variety of
  • liposomes depend on pH, ionic strength and the presence of divalent cations. Liposomes can
  • phase transition which markedly alters their permeability.
  • the phase fransition involves a
  • soluble proteins such as cytochrome c, bind, deform and penetrate the bilayer, thereby
  • MLVs are moderately efficient at trapping solutes, but SUVs are extremely inefficient. SUVs
  • Polar compounds are released through permeation or when the bilayer is broken, but nonpolar compounds remain affiliated with the bilayer unless it is dismpted by temperature or exposure to
  • lipoproteins Both types show maximum efflux rates at the phase transition temperature.
  • Liposomes interact with cells via four different mechanisms: Endocytosis by phagocytic
  • liposomal contents into the cytoplasm; and by transfer of liposomal lipids to cellular or
  • compositions such as size, fluidity, and surface charge. They may persist in tissues for h or days, depending on their composition, and half lives in the blood range from min to several h. Larger
  • liposomes such as MLVs and LUVs
  • MLVs and LUVs are taken up rapidly by phagocytic cells of the
  • liposomes potential targeting of liposomes to only those organs and tissues accessible to their large size. These include the blood, liver, spleen, bone marrow, and lymphoid organs.
  • Targeting is generally not a limitation in terms of the present invention. However, should specific targeting be desired, methods are available for this to be accomplished.
  • Antibodies may be used to bind to the liposome surface and to direct the antibody and its drag
  • glycoprotein or glycolipid cell-surface components that play a role in cell-cell
  • recognition, interaction and adhesion may also be used as recognition sites as they have
  • the invention provides for pharmaceutically acceptable nanocapsule
  • Nanocapsules can generally entrap compounds in a stable and reproducible way (Henry- Michelland etal, 1987; Quintanar-Guerrero et al, 1998; Douglas et al, 1987).
  • ulfrafine particles sized around 0.1 ⁇ m
  • Biodegradable polyalkyl- cyanoacrylate nanoparticles that meet these requirements are contemplated for use in the present 5 invention.
  • Such particles may be are easily made, as described (Couvreur etal, 1980; Couvreur, 1988; zur Muhlen et al, 1998; Zambaux et al. 1998; Pinto-Alphandry et al, 1995 and U. S. Patent 5,145,684, specifically incorporated herein by reference in its entirety).
  • Patent 5,797,898 ophthalmic formulations (Bourlais et al, 1998), transdermal matrices (U. S. Patent 5,770,219 and U. S. Patent 5,783,208) and feedback-controlled delivery (U. S. Patent 5,697,899), each specifically incorporated herein by reference in its entirety.0 4.8 THERAPEUTIC AND DIAGNOSTIC KITS
  • the invention also encompasses one or more compositions together with one or more pharmaceutically-acceptable excipients, carriers, diluents, adjuvants, and/or other components, as may be employed in the formulation of particular rAAV-polynucleotide delivery
  • kits may comprise one or more of the disclosed microsphere-conjugated rAAV compositions in combination with instructions for using the viral vector in the treatment of such disorders in a mammal, and may typically further include containers prepared for convenient commercial packaging.
  • preferred animals for administration of the pharmaceutical compositions disclosed herein include mammals, and particularly humans. Other preferred animals include murines, bovines, equines, ovines, epines, porcines, canines, felines, and other animals of veterinary significance which may benefit from the therapeutic and/or prophylactic methods disclosed herein.
  • the composition may include partially or significantly purified rAAV compositions, either alone, or in combination with one or more additional active ingredients, which may be obtained from natural or recombinant sources, or which may be obtainable naturally or either chemically synthesized, or alternatively produced in vitro from recombinant host cells expressing DNA segments encoding such additional active ingredients.
  • Therapeutic kits may also be prepared that comprise at least one of the compositions disclosed herein and instructions for using the composition as a therapeutic agent.
  • the container means for such kits may typically comprise at least one vial, test tube, flask, bottle, syringe or other container means, into which the disclosed rAAV composition(s) may be placed, and preferably suitably aliquoted.
  • the kit may also contain a second distinct container means into which this second composition may be placed.
  • the plurality of biologically-active therapeutic polypeptide compositions may be prepared in a single pharmaceutical composition, and may be packaged in a single container means, such as a vial, flask, syringe, bottle, or other suitable single container means.
  • the kits of the present invention will also typically include a means for containing the vial(s) in close confinement for commercial sale, such as, e.g., injection or blow-molded plastic containers into which the desired vial(s) are retained.
  • RESULTS OF NUCLEIC ACID DELIVERY AND DNA TRANSFECTION it may be desirable to use other methods for the transfer of expression constructs into target mammalian cells. Some of these techniques may be successfully adapted for in vivo or ex vivo use, as discussed below. Likewise, in some applications, it may be desirable to transfer a naked DNA expression construct into cells using methods such as particle bombardment. This method depends on the ability to accelerate DNA coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them. Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force.
  • microprojectiles used have consisted of biologically inert substances such as tungsten or gold beads.
  • one or more polynucleotide compositions disclosed herein will be used to transfect an appropriate host cell.
  • Technology for introduction of nucleic acids into cells is well known to those of skill in the art. These include calcium phosphate precipitation (Graham and Van Der Eb, 1973; Chen and Okayama, 1987; Rippe et al, 1990) DEAE-dexfran (Gopal, 1985), elecfroporation (Wong and Neumann, 1982; Fromm et al , 1985; Tur-Kaspa et al. , 1986; Potter et al.
  • nucleic acid sequences means that a particular subject sequence, for example, an nucleotide
  • sequence is substantially complementary to all or a portion of the selected target sequence
  • sequences will be highly complementary to the selected target sequence, and will have no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 base
  • sequences may be exact matches, i.e. be completely complementary
  • nucleotide sequence specifically binds, and therefore have zero mismatches along the
  • cellular mRNA e.g., antisense molecules
  • highly complementary sequences will typically bind
  • polypeptides in a selected cell through the introduction of one or more rAAV- vectored antisense
  • substantially complementary oligonucleotide sequences will typically be greater than about 80 percent complementary (or '% exact-match') to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and will, more preferably be greater than about 85 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds.
  • the oligonucleotide sequences will be greater than about 90 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and may in certain embodiments be greater than about 95 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and even up to and including 96%, 97%, 98%, 99%, and even 100% exact match complementary to all or a portion of the target mRNA to which the designed oligonucleotide specifically binds.
  • Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG).
  • the GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences.
  • the preferred default parameters for the GAP program include:
  • PNA peptide nucleic acids
  • PNA is a DNA mimic in which the nucleobases are attached to a pseudopeptide backbone (Good and Nielsen, 1997). PNAs may be utilized in a number of methods that traditionally have used RNA or DNA.
  • PNA sequences perform better in techniques than the corresponding RNA or DNA sequences and have utilities that are not inherent to RNA or DNA.
  • An excellent review of PNA including methods of making, characteristics of, and methods of using, is provided by Corey (1997) and is incorporated herein by reference.
  • PNA compositions may be used to regulate, alter, decrease, or reduce the translation of the specific mRNA, and thereby alter the level of polypeptide encoded by the targeted mRNA in a host cell to which such PNA compositions have been administered.
  • PNAs have 2-aminoethyl-glycine linkages replacing the normal phosphodiester backbone of DNA (Nielsen et al , 1993 ; Hanvey et al , 1992; Hyrup and Nielsen, 1996; Nielsen,
  • PNAs are neutral molecules
  • PNAs are achiral, which avoids the need to develop a stereoselective synthesis
  • PNA synthesis uses standard Boc (Dueholm et al, 1992) or Fmoc (Bonham et al, 1995) protocols for solid-phase peptide synthesis, although other methods, including a modified Merrifield method, have been
  • PNA monomers or ready-made oligomers are commercially available from PerSeptive
  • modified nucleotide compositions such as, for example, in the generation of the nucleic acid segments that encode either parts of the AAV vector itself, or the promoter, or even the therapeutic gene delivered by
  • Site-specific mutagenesis is a technique useful in the preparation of individual peptides, or biologically functional equivalent polypeptides, through specific mutagenesis of the underlying polynucleotides that encode them.
  • the technique well-known to those of skill in the 0 art, further provides a ready ability to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA.
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a 5 primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Mutations may be employed in a selected polynucleotide sequence to improve, alter, decrease, modify, or change the properties of the polynucleotide itself, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide.
  • the inventors contemplate the mutagenesis of the disclosed polynucleotide sequences to alter the activity or effectiveness of such viral vector constructs in a transformed host cell. Likewise in certain embodiments, the inventors contemplate the mutagenesis of the viral genome itself to facilitate improved infectivity, replication, stability, activity, or viral titers, as well as efficiency of transfection both
  • site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector or melting apart of two strands of a double-stranded vector which includes within its sequence a DNA sequence which encodes the desired polypeptide(s).
  • An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to
  • DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand.
  • E. coli polymerase I Klenow fragment DNA polymerizing enzymes
  • This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected which include recombinant vectors bearing the mutated sequence arrangement.
  • sequence variants of the selected polynucleotide segments using site- directed mutagenesis is provided as a means of producing potentially useful species and is not meant to be limiting, as there are other ways in which sequence variants of polypeptides and the DNA sequences encoding them may be obtained.
  • recombinant vectors encoding a desired polypeptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.
  • mutagenic agents such as hydroxylamine
  • specific details regarding these methods and protocols are found in the teachings of Maloy etal, 1994; Segal, 1976; Prokop and Bajpai, 1991; Kuby, 1994; and Maniatis et al, 1982, each incorporated herein by reference, for that purpose.
  • oligonucleotide directed mutagenesis procedure refers to template-dependent processes and vector-mediated propagation that result in an increase in the concentration of a specific nucleic acid molecule relative to its initial concentration, or in an increase in tlie concentration of a detectable signal, such as amplification.
  • oligonucleotide directed mutagenesis procedure is intended to refer to a process that involves the template-dependent extension of a primer molecule.
  • template dependent process refers to nucleic acid synthesis of an RNA or a DNA molecule wherein the sequence of
  • methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector
  • the nucleic acid may be genomic DNA or fractionated or whole cell
  • RNA Ribonucleic acid
  • it may be desired to convert the RNA to a complementary DNA.
  • the RNA is whole cell RNA and is used directly as the template for amplification.
  • Pairs of primers that selectively hybridize to nucleic acids corresponding to the ribozymes or conserved flanking regions are contacted with the isolated nucleic acid under
  • primers are oligonucleotides from ten to twenty base
  • Primers may be provided in double-
  • nucleic acid:primer complex is contacted with one or more
  • amplification also referred to as "cycles,” are conducted until a sufficient amount of
  • the amplification product is detected.
  • the detection may be
  • PCRTM polymerase chain reaction
  • PCRTM two primer sequences are prepared that are complementary to regions on opposite complementary strands of the marker sequence.
  • An excess of deoxynucleoside triphosphates is added to a reaction mixture along with a DNA polymerase, e.g., Tag polymerase. If the marker sequence is present in a sample, the primers will bind to the marker and the polymerase will cause the primers to be extended along the marker sequence by adding on nucleotides.
  • a DNA polymerase e.g., Tag polymerase
  • a reverse transcriptase PCR amplification procedure may be performed in order to quantify the amount of mRNA amplified.
  • Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et al, 1989.
  • Alternative methods for reverse transcription utilize thermostable, RNA-dependent DNA polymerases. These methods are described in WO 90/07641, filed December 21, 1990, incorporated herein by reference in its entirety.
  • Polymerase chain reaction methodologies are well known in the art.
  • Another method for amplification is the ligase chain reaction (“LCR”), disclosed in Eur. Pat.
  • Q ⁇ R Qbeta Replicase
  • RNA polymerase RNA polymerase
  • An isothermal amplification method in which restriction endonucleases and ligases are used to achieve the amplification of target molecules that contain nucleotide 5'-[alpha-thio]- triphosphates in one strand of a restriction site may also be useful in the amplification of nucleic acids in the present invention.
  • Strand Displacement Amplification (SD A), described in U. S. Patent Nos. 5,455,166,
  • a probe having 3' and 5' sequences of non-specific DNA and a middle sequence of specific RNA is hybridized to DNA that is present in a sample.
  • the reaction is treated with RNase H, and the products of the probe identified as distinctive products that are released after digestion.
  • the original template is annealed to another cycling probe and the reaction is repeated.
  • Still another amplification methods described in Great Britain Patent 2202328, and in Intl. Pat. Appl. Publ. No. PCT/US89/01025, each of which is incorporated herein by reference in its entirety, may be used in accordance with the present invention.
  • "modified" primers are used in a PCR-like, template- and enzyme-dependent synthesis.
  • the primers may be modified by labeling with a capture moiety (e.g. , biotin) and/or a detector moiety (e.g., enzyme).
  • a capture moiety e.g. , biotin
  • a detector moiety e.g., enzyme
  • nucleic acid amplification procedures include transcription-based amplification systems (TAS), including nucleic acid sequence based amplification (NASBA) and 3SR Gingeras et al, PCT Application WO 88/10315, incorporated herein by reference.
  • TAS transcription-based amplification systems
  • NASBA nucleic acid sequence based amplification
  • 3SR Gingeras et al PCT Application WO 88/10315, incorporated herein by reference.
  • the nucleic acids can be prepared for amplification by standard phenol/chloroform extraction, heat denaturation of a clinical sample, treatment with lysis buffer and minispin columns for isolation of DNA and RNA or guanidinium chloride extraction of RNA.
  • amplification techniques involve annealing a primer that has target specific sequences.
  • DNA/RNA hybrids are digested with RNase H while double stranded DNA molecules are heat denatured again.
  • the single stranded DNA is made fully double stranded by addition of second target specific primer, followed by polymerization.
  • the double-stranded DNA molecules are then multiply transcribed by an RNA polymerase such as T7 or SP6.
  • an RNA polymerase such as T7 or SP6.
  • the RNA's are reverse transcribed into single stranded DNA, which is then converted to double-stranded DNA, and then transcribed once again with an RNA polymerase such as T7 or SP6.
  • the resulting products whether truncated or complete, indicate target specific sequences. Davey et al, Eur. Pat. Appl.
  • ssRNA single- stranded RNA
  • dsDNA double-stranded DNA
  • the ssRNA is a template for a first primer oligonucleotide, which is elongated by reverse transcriptase (RNA-dependent DNA polymerase).
  • RNA-dependent DNA polymerase reverse transcriptase
  • the RNA is then removed from the resulting DNA:RNA duplex by the action of ribonuclease H (RNase H, an RNase specific for RNA in duplex with either DNA or RNA).
  • RNase H ribonuclease H
  • the resultant ssDNA is a template for a second primer, which also includes the sequences of an RNase H
  • RNA polymerase promoter (exemplified by T7 RNA polymerase) 5' to its homology to the template.
  • This primer is then extended by DNA polymerase (exemplified by the large "Klenow" fragment of E. coli DNA polymerase I), resulting in a double-stranded DNA (“dsDNA”) molecule, having a sequence identical to that of the original RNA between the primers and having additionally, at one end, a promoter sequence.
  • This promoter sequence can be used by the appropriate RNA polymerase to make many RNA copies of the DNA. These copies can then re-enter the cycle leading to very swift amplification. With proper choice of enzymes, this amplification can be done isothermally without addition of enzymes at each cycle.
  • the starting sequence can be chosen to be in the form of either DNA or RNA.
  • Miller et al, PCT Application WO 89/06700 (incorporated herein by reference in its entirety) disclose a nucleic acid sequence amplification scheme based on the hybridization of a promoter/primer sequence to a target single-stranded DNA ("ssDNA”) followed by transcription of many RNA copies of the sequence. This scheme is not cyclic, i.e., new templates are not produced from the resultant RNA transcripts.
  • Other amplification methods include "RACE” and "one-sided PCR” (Frohman, 1990 incorporated by reference).
  • Methods based on ligation of two (or more) oligonucleotides in the presence of nucleic acid having the sequence of the resulting "di-oligonucleotide", thereby amplifying the di- oligonucleotide may also be used in the amplification step of the present invention. Following any amplification, it may be desirable to separate the amplification product from the template and the excess primer for the purpose of determining whether specific amplification has occurred. In one embodiment, amplification products are separated by agarose, agarose-acrylamide or polyacrylamide gel electrophoresis using standard methods (Sambrook et al. , 1989). Alternatively, chromatographic techniques may be employed to effect separation.
  • Amplification products must be visualized in order to confirm amplification of the marker sequences.
  • One typical visualization method involves staining of a gel with ethidium bromide and visualization under UV light.
  • the amplification products can then be exposed to x-ray film or visualized under the appropriate stimulating spectra, following separation. In one embodiment, visualization is achieved indirectly.
  • a labeled, nucleic acid probe is brought into contact with the amplified marker sequence.
  • the probe preferably is conjugated to a chromophore but may be radiolabeled.
  • the probe is conjugated to a binding partner, such as an antibody or biotin, and the other member of the binding pair carries a detectable moiety.
  • detection is by Southern blotting and hybridization with a labeled probe. The techniques involved in Southern blotting are well known to those of skill in the art and can be found in many standard books on molecular protocols (Sambrook etal, 1989). Briefly, amplification products are separated by gel electrophoresis.
  • the gel is then contacted with a membrane, such as nitrocellulose, permitting transfer of the nucleic acid and non- covalent binding.
  • a membrane such as nitrocellulose
  • the membrane is incubated with a chromophore-conjugated probe that is capable of hybridizing with a target amplification product. Detection is by exposure of the membrane to x-ray film or ion-emitting detection devices.
  • BIOLOGICAL FUNCTIONAL EQUIVALENTS Modification and changes may be made in the stracture of the polynucleotides and polypeptides of the present invention and still obtain a functional molecule that still possesses desirable characteristics. As mentioned above, it is often desirable to introduce one or more mutations into a specific polynucleotide and/or encoded polypeptide sequence. In certain circumstances, the resulting encoded polypeptide sequence is altered by this mutation, or in other cases, the sequence of the polypeptide is unchanged by one or more mutations in the encoding polynucleotide.
  • the amino acid changes may be achieved by changing one or more of the codons of the encoding DNA sequence, according to Table 3.
  • certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the polynucleotide sequences disclosed herein, without appreciable loss of their biological utility or activity.
  • the hydropathic index of amino acids may be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporate herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary stracture of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics (Kyte and Doolittle, 1982), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5). It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e. still obtain a biological functionally equivalent protein. In making such changes,
  • proline (-0.5 ⁇ 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine
  • amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • Exemplary substitutions which take several of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
  • polynucleotides, nucleic acid segments, nucleic acid sequences, and the like include, but are not limited to, DNAs (including and not limited to genomic or extragenomic DNAs), genes, peptide nucleic acids (PNAs) RNAs
  • Expression The combination of intracellular processes, including transcription and translation undergone by a polynucleotide such as a stmctural gene to synthesize the encoded peptide or polypeptide.
  • Promoter a term used to generally describe the region or regions of a nucleic acid sequence that regulates transcription.
  • Regulatory Element a term used to generally describe the region or regions of a nucleic acid sequence that regulates transcription.
  • Structural gene A gene or sequence region that is expressed to produce an encoded peptide or polypeptide. Transformation: A process of introducing an exogenous polynucleotide sequence (e.g.
  • Transfection, elecfroporation, and naked nucleic acid uptake all represent examples of techniques used to transform a host cell with one or more polynucleotides.
  • Transformed cell A host cell whose nucleic acid complement has been altered by the introduction of one or more exogenous polynucleotides into that cell.
  • Transgenic cell Any cell derived or regenerated from a transformed cell or derived from a transgenic cell, or from the progeny or offspring of any generation of such a transformed host cell.
  • a nucleic acid molecule (typically comprised of DNA) capable of replication in a host cell and/or to which another nucleic acid segment can be operatively linked so as to bring about replication of the attached segment.
  • a plasmid, cosmid, or a virus is an exemplary vector.
  • the terms "substantially corresponds to”, “substantially homologous”, or “substantial identity” as used herein denotes a characteristic of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid or amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence.
  • the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity. More preferably still, highly homologous sequences often share greater than at least about 96,
  • the reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome.
  • the reference sequence will typically comprise at least about 18-25 nucleotides, more typically at least about 26 to 35 nucleotides, and even more typically at least about 40, 50, 60, 70, 80, 90, or even 100 or so nucleotides.
  • the extent of percent identity between the two sequences will be at least about 80%, preferably at least about 85%, and more preferably about 90% or 95% or higher, as readily determined by one or more of the sequence comparison algorithms well-known to those of skill in the art, such as e.g., the FASTA program analysis described by Pearson and Lipman (1988).
  • naturally occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including vimses) that can be isolated from a source in nature and which has not been intentionally modified by the hand of man in a laboratory is naturally-occurring.
  • laboratory strains of rodents that may have been selectively bred according to classical genetics are considered naturally occurring animals.
  • a heterologous is defined in relation to a predetermined referenced gene sequence.
  • a heterologous promoter is defined as a promoter which does not naturally occur adjacent to the referenced stmctural gene, but which is positioned by laboratory manipulation.
  • a heterologous gene or nucleic acid segment is defined as a gene or segment that does not naturally occur adjacent to the referenced promoter and/or enhancer elements.
  • Transcriptional regulatory element refers to a polynucleotide sequence that activates transcription alone or in combination with one or more other nucleic acid sequences.
  • a transcriptional regulatory element can, for example, comprise one or more promoters, one or more response elements, one or more negative regulatory elements, and/or one or more enhancers.
  • a "transcription factor recognition site” and a “transcription factor binding site” refer to a polynucleotide sequence(s) or sequence motif(s) which are identified as being sites for the sequence-specific interaction of one or more transcription factors, frequently taking the form of direct protein-DNA binding.
  • transcription factor binding sites can be identified by DNA footprinting, gel mobility shift assays, and the like, and/or can be predicted on the basis of known consensus sequence motifs, or by other methods known to those of skill in the art.
  • the term “operably linked” refers to a linkage of two or more polynucleotides or two or more nucleic acid sequences in a functional relationship.
  • a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the coding sequence.
  • Operably linked means that the DNA sequences being linked are typically contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame.
  • enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not contiguous.
  • Transcriptional unit refers to a polynucleotide sequence that comprises at least a first stmctural gene operably linked to at least a first cw-acting promoter sequence and optionally linked operably to one or more other -acting nucleic acid sequences necessary for efficient transcription of the stmctural gene sequences, and at least a first distal regulatory element as may be required for the appropriate tissue-specific and developmental franscription of the stmctural gene sequence operably positioned under the confrol of the promoter and/or enhancer elements, as well as any additional cis sequences that are necessary for efficient transcription and translation (e.g., polyadenylation site(s), mRNA stability controlling sequence(s), etc.
  • substantially complementary when used to define either amino acid or nucleic acid sequences, means that a particular subject sequence, for example, an oligonucleotide sequence, is substantially complementary to all or a portion of the selected sequence, and thus will specifically bind to a portion of an mRNA encoding the selected sequence.
  • sequences will be highly complementary to the mRNA "target" sequence, and will have no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 base mismatches throughout the complementary portion of the sequence.
  • oligonucleotide sequences will be greater than about 80 percent complementary (or '% exact-match') to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and will, more preferably be greater than about 85 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds.
  • the oligonucleotide sequences will be greater than about 90 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and may in certain embodiments be greater than about 95 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and even up to and including 96%, 97%, 98%, 99%, and even 100% exact match complementary to all or a portion of the target mRNA to which the designed oligonucleotide specifically binds.
  • Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG).
  • the GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences.
  • the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • a unary comparison matrix containing a value of 1 for identities and 0 for non-identities
  • the weighted comparison matrix of Gribskov and Burgess (1986) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap
  • no penalty for end gaps include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • vector refers to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, vims, virion, etc., which is capable of replication when associated with the proper control sequences and/or which can transfer nucleic acid sequences between cells.
  • the term includes cloning and expression vectors, as well as viral vectors.
  • host and “expression host” refer to organisms and/or cells which harbor an exogenous DNA sequence (e.g., via transfection), an expression vector or vehicle, as well as organisms and/or cells that are suitable for use in expressing a recombinant gene or protein.
  • viral replicons and “viral origins of replication” refer to viral DNA sequences that allow for the extrachromosomal replication of a vector in a host cell expressing the appropriate replication factors.
  • vectors which contain either the SV40 or polyoma virus origin of replication replicate to high copy number, while vectors which contain the replicons from bovine papillomaviras or Epstein-Barr virus replicate extrachromosomally at low copy number may be utilized in other embodiments.
  • wild type refers to a gene or gene product which has the characteristics of that gene or gene product when isolated from a naturally occurring source.
  • a wild-type gene is that which is most frequently observed in a population and is thus arbitrarily designed the "normal” or “wild-type” form of the gene.
  • modified or mutant refers to a gene or gene product which displays modifications in sequence and or functional properties (i.e., altered characteristics) when compared to the wild-type gene or gene product. It is noted that naturally-occurring mutants can be isolated; these are identified by the fact that they have altered characteristics when compared to the wild-type gene or gene product.
  • heterologous as it relates to nucleic acid sequences such as coding sequences and control sequences, denotes sequences that are not normally joined together, and/or are not normally associated with a particular cell.
  • a heterologous region of a nucleic acid construct or a vector is a segment of nucleic acid within or attached to another nucleic acid molecule that is not found in association with the other molecule in nature.
  • a heterologous region of a nucleic acid constmct could include a coding sequence flanked by sequences not found in association with the coding sequence in nature.
  • Another example of a heterologous coding sequence is a construct where the coding sequence itself is not found in nature (e.g., synthetic sequences having codons different from the native gene).
  • coding sequence or a sequence which "encodes” a particular
  • antigen is a nucleic acid sequence which is transcribed (in the case of DNA) and translated (in the case of mRNA) into a polypeptide in vitro or in vivo, when placed under the control of appropriate regulatory sequences.
  • the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a franslation stop codon at the 3' (carboxy) terminus.
  • a coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and even synthetic DNA sequences.
  • a transcription termination sequence will usually be located 3' to the coding sequence.
  • control sequences refers collectively to regulatory elements such as promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate recipient cell.
  • “Operably linked” or “operatively linked” refers to an arrangement of elements wherein the components so described are configured so as to perform their usual function. Thus, control sequences operably linked to a coding sequence are capable of effecting the expression of the coding sequence.
  • control sequences need not be contiguous with the coding sequence, so long as they function to direct the expression thereof.
  • intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked” or "operatively linked” to the coding sequence.
  • isolated when used in relation to a nucleic acid, as in "an isolated oligonucleotide” or “isolated polynucleotide” refers to a nucleic acid sequence that is identified and separated from at least one contaminant nucleic acid with which it is ordinarily associated in its natural source.
  • Isolated nucleic acid is such present in a form or setting that is different from that in which it is found in nature.
  • non-isolated nucleic acids are nucleic acids such as DNA and RNA found in the state they exist in nature.
  • a given DNA sequence e.g., a gene
  • RNA sequences such as a specific mRNA sequence encoding a specific protein, are found in the cell as a mixture with numerous other mRNAs which encode a multitude of proteins.
  • the isolated nucleic acid, oligonucleotide, or polynucleotide may be present in single-stranded or double-stranded form.
  • the oligonucleotide or polynucleotide When an isolated nucleic acid, oligonucleotide or polynucleotide is to be utilized to express a protein, the oligonucleotide or polynucleotide will contain at a minimum the sense or coding strand (i.e., the oligonucleotide or polynucleotide may single-stranded), but may contain both the sense and anti-sense strands (i.e., the oligonucleotide or polynucleotide may be double-stranded).
  • the term "purified” or "to purify” refers to the removal of contaminants from a sample.
  • antibodies may be purified by removal of contaminating non-immunoglobulin proteins; they may also purified by the removal of immunoglobulin that does not bind the antigen of interest (e.g., at least a portion of Factor VII).
  • the removal of non-immunoglobulin proteins and/or the removal of immunoglobulins that do not bind the antigen of interest (e.g., at least a portion of Factor VII) results in an increase in the percent of desired antigen-reactive immunoglobulins in the sample.
  • composition comprising a given polynucleotide sequence refers broadly to any composition containing the given polynucleotide sequence.
  • the composition may comprise an aqueous solution.
  • at risk is used in references to individuals who are at risk for experiencing hemorrhagic episodes. In particularly preferred embodiments, the individuals are hemophiliacs with mild, moderate, or severe hemophilia.
  • subject refers to any animal (i.e., vertebrates and invertebrates), while the term “vertebrate subject” refers to any member of the subphylum
  • Chordata any member of this subphylum, including, but not limited to humans and other primates, rodents (e.g., mice, rats, and guinea pigs), lagomorphs (e.g., pikas, hares, rabbits), bovines (e.g., cattle), ovines (e.g., sheep), caprines
  • rodents e.g., mice, rats, and guinea pigs
  • lagomorphs e.g., pikas, hares, rabbits
  • bovines e.g., cattle
  • ovines e.g., sheep
  • domestic cats e.g., goats
  • porcines e.g., swine
  • equines e.g., horses
  • canines e.g., dogs, wolves
  • felines e.g., lions, tigers, cheetahs, domestic cats
  • domestic fowl e.g., chickens, turkeys, ducks, geese, other gallinaceous birds, etc.
  • domestic fowl e.g., chickens, turkeys, ducks, geese, other gallinaceous birds, etc.
  • feral or wild animals including, but not limited to, such animals as ungulates, including members of the Artiodactyla (e.g., antelope, deer, etc.) and the Perissodactyla (e.g., rhinoceros, tapir, horse, zebra, etc.), etc.
  • a "therapeutically effective amount” or “therapeutic effective dose” is an amount or dose of AAV vectors or AAV virions capable of producing sufficient amounts of Factor VII to decrease the time it takes for a subject's blood to clot.
  • Adeno-associated virus 2 (AAV)-based vectors have emerged at the forefront of gene therapy.
  • AAV has been used effectively for the treatment of factor IX (FIX) deficiency in both murine and canine models (Koeberl et al, 1997; Snyder et al, 1997; Chao et al, 1999; Herzog et al, 1999; Hagsfrom et al, 2000), leading to current phase I clinical trials.
  • the present example describes the use of a recombinant AAV-based vector system for the treatment of a clinically relevant murine model of hemophilia A. This study demonstrates expression of Factor VII polypeptide from rAAV vectors expressing the FVII gene, administered either intravenously or intramuscularly.
  • rAAV is most often produced by co-transfection of rAAV vector plasmid and wt AAV helper plasmid into Ad-infected 293 cells (Hermonat and Muzyczka, 1984).
  • Recent improvements in AAV helper design Li et al, 1997) as well as construction of non-infectious mini-Ad plasmid helper (Grimm et al, 1998; Xiao et al, 1998; Salvetti, 1998) have eliminated the need for Ad infection, and made it possible to increase the yield of rAAV up to 10 5 particles per fransfected cell in a cmde lysate.
  • Scalable methods of rAAV production that do not rely on DNA transfection have also been developed (Cbiorini et al, 1995; frioue and Russell, 1998;
  • the conventional protocol for downstream purification of rAAV involves the stepwise precipitation of rAAV using ammonium sulfate, followed by two or preferably, three rounds of CsCl density gradient centrifugation. Each round of CsCl centrifugation involves fractionation of the gradient and probing fractions for rAAV by dot-blot hybridization or by PCRTM analysis.
  • the AAV vectors and rAAV virions of the present invention can be produced using standard methodology known to those of skill in the art.
  • Such methods typically involve one or more steps such as: (a) introducing an AAV vector into an appropriate mammalian host cell; (b) introducing an AAV helper construct into the host cell, where the helper construct includes AAV coding regions capable of being expressed in the host cell to complement AAV helper functions missing from the AAV vector; (c) introducing one or more helper viruses and/or accessory function vectors into the host cell, wherein the helper virus and/or accessory function vectors provide accessory functions capable of supporting efficient recombinant AAV (“rAAV”) virion production in the host cell; and (d) culturing the host cell to produce rAAV virions.
  • steps such as: (a) introducing an AAV vector into an appropriate mammalian host cell; (b) introducing an AAV helper construct into the host cell, where the helper construct includes AAV coding regions capable of being expressed in the host cell to complement AAV helper functions missing from the AAV vector; (c) introducing one or more helper viruses and/or accessory function vector
  • DNA may be delivered to a suitable mammalian host cell as an expression vector construct.
  • Preferred gene therapy vectors of the present invention are generally viral vectors.
  • Adeno-associated virus (AAV) is particularly attractive for gene transfer because it does not induce any pathogenic response and can integrate into the host cellular chromosome (Kotin et al, 1990).
  • TRs The AAV terminal repeats (TRs) are the only essential cw-components for the chromosomal integration (Muzyczka and McLaughin, 1988). These TRs are reported to have promoter activity (Flotte et al, 1993). They may promote efficient gene transfer from the cytoplasm to the nucleus or increase the stability of plasmid DNA and enable longer-lasting gene expression. Studies using recombinant plasmid
  • AAV-based plasmids have been shown to drive higher and longer fransgene expression than the identical plasmids lacking the TRs of AAV in most cell types (Shafron et al, 1998).
  • AAV (Ridgeway, 1988; Hermonat and Muzyczka, 1984) is a paroviras, discovered as a contamination of adenoviral stocks. It is a ubiquitous virus (antibodies are present in 85% of the
  • the virion in using viral delivery systems, one may desire to purify the virion sufficiently to render it essentially free of undesirable contaminants, such as defective interfering viral particles or endotoxins and other pyrogens such that it will not cause any untoward reactions in the cell, animal or individual receiving the vector construct.
  • One preferred means of purifying the vector involves the use of buoyant density gradients, such as cesium chloride gradient centrifugation, heparin affinity chromatography (Clark et al, 1999), or non-ionic iodixinol gradients followed by heparin affinity chromatography (Zolotukhin et al, 1999).
  • the titer of AAV in a given sample may be determined using any one of the methods routinely accepted in the AAV arts. For example, the inventors routinely use the methods of
  • the infectivity of a given AAV sample may be determined using any one of the methods routinely accepted in the AAV arts. For example, the inventors routinely use the methods of Hermonat and Muzyczka (1984) or Clark et al. (1999) to determine the infectivity of a given AAV stock. 5.1.4 MATERIALS AND METHODS
  • Monolayer cells may be maintained at 37°C, 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and antibiotics.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • the albumin promoter may be obtained from K. Zaret (Foxchase, Philadelphia, PA).
  • a truncated hybrid cytomegalovirus immediate-early enhancer chicken b- actin (CBA) promoter may be generated by using _3sj_> 1201 to delete 700 bp from the 39 end of the promoter, according to standard cloning methods.
  • rAAV plasmids pCMVp-/ ⁇ cZ, UF5 (pCMVp-GFP), and UF11 (pCBAp-GFP) have been described previously (Kessler et al., 1996; Klein et al, 1998).
  • rAAV plasmids containing the appropriate DNA sequence encoding the desired mammalian Factor VII polypeptide may be generated by standard cloning methods.
  • HC-SD and pSALC may be cloned from the intron region of the pCI cloning vector (Stratagene, La Jolla, CA).
  • rAAV vectors may be generated, purified, and titered at the University of Florida (Gainesville, FL) Vector Core Laboratory as previously described (Zolotukhin et al, 1999).
  • In vitro fransfections may be performed by the calcium phosphate-DNA coprecipitation method as described by Sambrook et al. (1989).
  • cells may be infected with the Factor VII vectors at the indicated multiplicities of infection (MOIs) and coinfected with wild-type adenovirus type 5 at an MOI of 1.
  • MOIs multiplicities of infection
  • conditioned medium may be assayed for the presence of functional FVII by the Coatest kit assay (Chromogenix, Milan, Italy).
  • HeLa or 293 cells may be infected with the indicated vectors at the indicated MOIs.
  • HeLa cells may also be coinfected with wild-type adenovirus type 5 at an MOI of 1.
  • HeLa cells (48 hr postinfection) and 293 cells (72 hr postinfection) may be subjected to fluorescence-activated cell sorting (FACS) analysis at a suitable facility, such as the University of Florida Interdisciplinary Center for Biotechnology Research (ICBR) Flow Cytometry Core (FACScan; BD Immunocytometry Systems, San Jose, CA).
  • FACS fluorescence-activated cell sorting
  • a suitable model of hemophilia such as for example a mouse FVII knockout [KO] mouse may be utilized in the practice of the present invention.
  • One-day-old FVII KO mice may be administered a suitable concentration of infectious particles (IP) (for example about 2.4 x 10 8 ) via the superficial temporal vein as described by Sands and Barker (1999). Seven-month-old FVII KO mice may also be administered a suitable concentration of IP via intramuscular injection into the gastrocnemius of the hind leg. Beginning, for example, about 4 weeks postinj ection, plasma may then be collected from tail vein bleeds and functional mFVII activity determined by a suitable Factor VII assay, such as the Coatest assay. Plasma samples are obtained by tail bleeding of anesthetized animals followed by the immediate addition of sodium citrate to a final concentration of about 0.38% (wt./vol.).
  • IP infectious particles
  • IMMUNOHISTOCHEMISTRY Tissue samples are fixed in 10% buffered formalin, embedded, and sectioned (4-mm thickness). Sections are immunostained with a suitable anti-mFVII antibody, the mouse-on- mouse (M.O.M.) immunostaining kit (Vector Laboratories, Burlingame, CA), and 3,39- diaminobenzidine (DAB), and then counterstained with Gill's hematoxylin (Vector Laboratories). Photographs are taken with a light microscope (Zeiss, Thomwood, NY), camera (Olympus America, Melville, NY) and MagnaFire digital recording system (Pro
  • Relative transduction efficiency is determined as described by Nakai et al. (2002). Briefly, the number of positively stained cells and total number of cells are counted. Ten fields of approximately 500 nuclei per field are counted for each section.
  • Tris-glycine sodium dodecyl sulfate (SDS)-polyacrylamide minigels Invitrogen, Carlsbad, CA.
  • Western transfer is performed with a Novex Western blot module (Invitrogen) and Hybond ECL nitrocellulose (Amersham Pharmacia Biotech, Piscataway, NJ). Blots are hybridized with a suitable anti-FVII antibody (Bi et al, 1996; Sarkar et al, 2000) (diluted 1 :50) and horseradish peroxidase (HRP)-conjugated secondary antibody (diluted 1:1000) (Amersham Pharmacia Biotech). Hybridization is detected with the ECL Plus Western blotting detection system (Amersham Pharmacia Biotech).
  • FIG. 1 shows that functional Factor VII can be produced in vitro.
  • the chromogenic Coaset assay was used to determine the levels of functional Factor VII secreted.
  • Factor VIII KO plasma was used as a positive control as these mice have been shown to produce normal levels of Factor VII.
  • KLQVGVFRLPLL From Aasru and Prydz , Biochemistry Mosc . , 67 ( l ) : 25-32 , 2002 . )
  • Auricchio, O'Connor, Hildinger and Wilson "A single-step affinity column for purification of serotype-5 based adeno-associated viral vectors," Mol. Ther., 4:372-74, 2001.
  • Baskar Smith, Ciment, Hoffmann, Tucker, Tenney, Colberg-Poley, Nelson and Ghazal, "Developmental analysis of the cytomegalovirus enhancer in transgenic animals," J Virol, 70:3215-26, 1996a.
  • Baskar Smith, Nilaver, Jupp, Hoffmann, Peffer, Tenney, Colberg-Poley, Ghazal and Nelson, "The enhancer domain of the human cytomegalovirus major immediate-early promoter determines cell type-specific expression in transgenic mice," J Virol, 70:3207-14, 1996b.
  • Blacklow, Dolin and Hoggan "Studies of the enhancement of an adenovirus-associated virus by herpes simplex virus," J Gen. Virol, 10:29-36, 1971b. Blanar, Baldwin, Flavell and Sharp, "A gamma-interferon-induced factor that binds the interferon response sequence of the MHC Class I gene, H-2Kb," EMBO J, 8:1139, 1989.
  • Bodine and Ley "An enhancer element lies 3' to the Human A gamma globin gene," EMBO J, 6:2997, 1987. Bonham, Brown, Boyd, Brown, Bruckenstein, Hanvey, Thomson, Pipe, Hassman, Bisi, et al, "An assessment of the antisense properties of RNase H-competen and steric-blocking oligomers," Nucleic Acids Res. , 23 : 1197-203, 1995. Boshart, Weber, Jahn, Dorsch-Hasler, Fleckenstein and Schaffner, "A very strong enhancer is located upstream of an immediate early gene of human cytomegalovirus," Cell, 41:521, 1985.
  • prealbumin gene binds a common factor at one site and a liver-specific factor(s) at two other sites
  • Culotta and Hamer "Fine mapping of a mouse metallothionein gene metal-response element," Mol. Cell. Biol, 9:1376, 1989.
  • Cunningham and Wells "High resolution epitope mapping of hGH-receptor interactions by alanine-scanning mutagenesis," Science, 244:1081-85, 1989.
  • Flotte, Barraza-Ortiz, Solow, Afione, Carter and Guggino "An improved system for packaging recombinant adeno-associated virus vectors capable of in vivo transduction," Gene Ther., 2:29-37, 1995.
  • Flotte, Carter, Conrad, Guggino, Reynolds, Rosenstein, Taylor, Walden and Wetzel "A phase I study of an adeno-associated virus-CFTR gene vector in adult CF patients with mild lung disease," Hum. Gene Ther., 7:1145-59, 1996.
  • Human factor VIII can be packaged and functionally expressed in an adeno-associated virus background: Applicability to haemophilia A gene therapy," Br. J. Haematol, 104:27-36, 1999.
  • B cell stimulatory factor unresponsiveness of X chromosome-linked immunodeficiency B cells," J Exp. Med., 172:1625-31, 1990.
  • subtilis, phage SP01, and phage T are homologous proteins
  • Grifman, Trepel, Speece, Gilbert, Arap, Pasqualini and Weitzman "Incorporation of rumor- targeting peptides into recombinant adeno-associated virus capsids," Mol. Ther., 3:964-75, 2001.
  • Grimm, Kem, Pawlita, Ferrari, Samulski and Kleinschmidt "Tifration of AAV-2 particles via a novel capsid ELISA: packaging of genomes can limit production of recombinant AAV-2," Gene Ther., 6:1322-30, 1999. Grosschedl and Baltimore, "Cell-type specificity of immunoglobulin gene expression is regulated by at least three DNA sequence elements," Cell, 41 :885, 1985.
  • Haslinger and Karin "Upstream promoter element of the human metallothionein-II gene can act like an enhancer element," Proc. Natl. Acad. Sci. USA, 82:8572, 1985.
  • PNA protein nucleic acids
  • the AAV origin binding protein Rep68 is an ATP-dependent site- specific endonuclease with DNA helicase activity," Cell, 61:447-57, 1990.
  • Jakobovits Smith, Jakobovits and Capon, "A discrete element 3' of human immunodeficiency vims 1 (HIV-1) and HIV-2 mRNA initiation sites mediates transcriptional activation by an HIV trans-activator," Mol. Cell. Biol, 8:2555, 1988.
  • Kessler, Podsakoff, Chen, McQuiston, Colosi, Matelis, Kurtzman and Byrne "Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein," Proc. Natl Acad. Sci. USA, 93:14082-87, 1996.
  • Kharkevich, Alyautdin and Filippov “Employment of magnet-susceptible microparticles for the targeting of drugs," J Pharm. Pharmacol, 41:286-88, 1989. Kiledjian, Su and Kadesch, "Identification and characterization of two functional domains within the murine heavy-chain enhancer," Mol. Cell. Biol, 8:145, 1988.
  • Kotin "Prospects for the use of adeno-associated vims as a vector for human gene therapy," Hum. Gene Ther., 5:793-801, 1994. Kotin, Siniscalco, Samulski, Zhu, Hunter, Laughlin, McLaughlin, Muzyczka, Rocchi and Berns, "Site-specific integration by adeno-associated virus,” Proc. Natl. Acad. Sci. USA, 87:2211-15, 1990. Kotin, Menninger, Ward and Bems, “Mapping and direct visualization of a region-specific viral DNA integration site on chromosome 19ql3-qter,” Genomics, 10:831-34, 1991.
  • Bovine papilloma vims contains an activator of gene expression at the distal end of the early transcription unit," Mol. Cell. Biol. 3:1108, 1983.
  • LDL receptor elements is highly conserved in humans and chickens," Arterioscler.
  • Muralidhar, Becerra and Rose "Site-directed mutagenesis of adeno-associated virus type 2 structural protein initiation codons: effects on regulation of synthesis and biological activity," J Virol, 68:170-76, 1994.
  • Muzyczka "Use of adeno-associated virus as a general transduction vector for mammalian cells," Curr. Top. Microbiol Immunol, 158:97-129, 1992.
  • AAV Rep protein acts as both a repressor and an activator to regulate AAV transcription during a productive infection
  • the LDL receptor is the major pathway for ⁇ -VLDL uptake by mouse peritoneal macrophages," Atherosclerosis, 154 : 51 -60, 2001.
  • Senapathy, Tratschin and Carter "Replication of adeno-associated viras DNA. Complementation of naturally occurring rep ' mutants by a wild-type genome or an ori " mutant and correction of terminal palindrome deletions," J. Mol. Biol, 179:1-20, 1984. Senyei, Reich, Gonczy and Widder, "In vivo kinetics of magnetically targeted low-dose doxorabicin,” J Pharm. Set, 70:389-91, 1981.
  • SR ⁇ promoter An efficient and versatile mammalian cDNA expression system composed of the simian viras 40 early promoter and the R-U5 segment of human T-cell leukemia virus Type 1 long terminal repeat," Mol. Cell. Biol, 8:466, 1988.
  • rat albumin promoter Cooperation with upstream elements is required when binding of APF/HNF 1 to the proximal element is partially impaired by mutation or bacterial methylation," Mol. Cell Biol, 9:4759, 1989. Tranche, Rollier, Herbomel, Bach, Cereghini, Weiss and Yaniv, "Anatomy of the rat albumin promoter," Mol. Biol. Med, 7:173, 1990. Trudel and Constantini, "A 3' enhancer contributes to the stage-specific expression of the human ⁇ -globin gene," Genes andDev., 6:954, 1987.
  • Tyndell La Mantia, Thacker, Favaloro and Kamen, "A region of the polyoma vims genome between the replication origin and late protein-coding sequences is required in cis for both early gene expression and viral DNA replication," Nuc. Acids. Res., 9:6231, 1981. Vanbever, Fouchard, Jadoul, De Morre, Preat and Marty, "In vivo noninvasive evaluation of hairless rat skin after high-voltage pulse exposure," Skin Parmacol Appl. Skin Physiol, 11:23-34, 1998.
  • CMV- ⁇ -actin promoter directs higher expression from an adeno-associated viral vector in the liver than the cytomegalovirus or elongation factor l ⁇ promoter and results in therapeutic levels of human factor X in mice," Hum. Gene Ther., 12:563-73, 2001.
  • AAV5 Addeno-associated viras type 5 but not AAV2 binds to the apical surfaces of airway epithelia and facilitates gene transfer
  • compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Abstract

Disclosed are improved recombinant adeno-associated viral (rAAV) vector compositions useful in the delivery of antihemophilic factor polypeptides to selected mammalian host cells. The disclosed rAAV vector compositions preferably comprise one or more polynucleotide sequences that express one or more mammalian Factor VII proteins, polypeptides, peptides, a operably linked to one or more promoter and/or enhancer sequences that are capable of expressing the encoded antihemophilic therapeutics in cells suitably transformed with the disclosed rAAV vector constructs, virions, and viral particles comprising the contructs of interest. These compositions, and methods for their use, including the manufacture of medicaments, have desirable therapeutic and/or prophylactic efficacy in the amelioration, treatment, and/or prevention of a variety of diseases, disorders, and dysfunctions in selected mammals, and in particular, humans diagnosed with Factor VII deficiency and/or hemophilia A.

Description

DESCRIPTION RAAV COMPOSITIONS AND METHODS FOR DELIVERY OF
HUMAN FACTOR VII POLYPEPTIDES AND TREATMENT OF HEMOPHILIA A
1. BACKGROUND OF THE INVENTION The present application claims priority to United States Provisional Application Serial No. 60/392,725, filed June 28, 2002, the entire contents of which is specifically incorporated herein by reference.
1.1 FIELD OF THE INVENTION The present invention relates generally to the fields of molecular biology and virology, and in particular, to methods for using recombinant adeno-associated virus (rAAV) compositions that express nucleic acid segments encoding therapeutic antihemophilic factor polypeptides useful in the treatment of complex human disorders, including for example, blood disorders such as hemophilia. In illustrative embodiments, methods are provided for preparing rAAV-based vector constructs that deliver one or more therapeutic antihemophilic factor compositions to cells, and in particular, human plasma protein Factor VII for the treatment of hemophilia or Factor VII deficiency.
1.2 DESCRIPTION OF RELATED ART
1.2.1 BLEEDING DISORDERS Coagulopathic (bleeding and clotting) disorders encompass a wide range of medical problems that lead to poor blood clotting and continuous bleeding. These disorders can result from de ects in the blood vessels or from abnormalities in the blood itself. The abnormalities may be in blood clotting factors or in platelets. Coagulation, the process that controls bleeding, is a complex multi-component process that involves as many as twenty different plasma proteins, or blood clotting factors.
Normally, a complex chemical process occurs using these clotting factors to form a substance called fibrin that stops bleeding. When certain coagulation factors are deficient or missing, the process doesn't occur normally.
1.2.2 HEMOPHILIA A Hemophilia is a bleeding disorder caused by a deficiency in one of the blood clotting factors. Hemophilia A (or "classic" hemophilia) is a deficiency in clotting Factor VIII, and accounts for about 80 percent of all hemophilia cases. Hemophilia A is a hereditary disorder in which tlie clotting ability of the blood is impaired and excessive bleeding results. Small wounds and punctures are usually not a problem. But uncontrolled internal bleeding can result in pain and swelling and permanent damage, especially to joints and muscles. Severity of symptoms can vary, and severe forms become apparent early on. The incidence of hemophilia A is 1 out of 10,000 live male births. About 20,000 Americans have hemophilia. Women may have it, but the condition is veiy rare in females. Flemophilia is caused by several different gene abnormalities. The severity of the symptoms of hemophilia A depends on how a particular gene abnormality affects the activity of factor VIII. When the activity is less than 1 percent of normal, episodes of severe bleeding occur and recur for no apparent reason. The symptoms of hemophilia include bruising, spontaneous bleeding, bleeding into joints and associated pain and swelling, gastrointestinal tract and urinary tract hemorrhage, blood in the urine or stool, and prolonged bleeding from cuts, tooth extraction, etc. 1.2.3 FACTOR VII (PROCONVERTIN) DEFICIENCY In addition to Factor VIII deficiency, some individuals suffer from a lack or deficiency in Factor VII. This factor is often referred to a "stable" factor or "proconvertin." Factor VII deficiency is an extremely rare disorder that can be inherited or acquired by 5 persons without hemophilia who take Coumadin, a d g used to inhibit blood clotting. In this disorder, bleeding can vary from mild to severe within the same person over time. Bleeding doesn't always correspond with the severity of the deficiency shown in blood tests. A history of bleeding may occur in infancy or childhood. Gastrointestinal and central nervous system bleeding can also occur. 10 Factor VII deficiency occurs in approximately one in 500,000 males and females. Congenital Factor VII deficiency is distinguished from Acquired Factor VII Deficiency that may result from liver disease, vitamin K deficiency, or other malabsorption conditions. Unlike hemophilia, Factor FVII deficiency is not sex-linked. It affects both males and females with equal frequency. It is also autosomal recessive, which means that if the 15 clotting defect is inherited from a parent, the child will be a genetic carrier of the condition, but may or may not have symptoms. Those who have inherited a defective Factor VII gene from only one parent will usually have only moderate levels of tlie factor, but without symptoms. The symptoms of Factor VII deficiency often include bleeding of mucous »0 membranes, spontaneous nosebleeds, excessive bruising, prolonged menstrual bleeding, and bleeding into joints or muscles. The diagnosis for Factor VII deficiency is usually made by testing for Factor VII in the blood, prolonged prothrombm time, or a normal partial thromboplastin time in combination with decreased Factor VII Assay. Existing treatments for Factor VII deficiency rely on the admimsfration of normal plasma or concentrates containing Factor VII. Severe bleeding is typically treated with fresh frozen plasma or PCCs (Prothrombin complex concentrates). However, because the life span of infused factor VII is very short (2 to 4 hours), patients require treatment every 2 to 6 hours for severe bleeding or surgery.
1.2.4 DEFICIENCIES IN THE PRIOR ART Current treatment methods for factor VII deficiency and hemophilia A are costly, in high demand, and typically most often involve replacement therapy using plasma, plasma-derived or recombinant Factor VII or VIII. Such therapy is limited by the production of Factor VII or VIII and the short half-life of these Factors in vitro. In addition, patients treated via replacement therapy often build resistance to treatment due to an immune response to the recombinant factor. What is lacking in the prior art is treatment of Factor VH deficiency and hemophilia A by administration of therapeutically-effective amounts of human plasma protein Factor VII. What is also lacking in the prior art are methods for long-term therapy of patients who are either unresponsive or refractive to current therapy regimens.
2.0 SUMMARY OF THE INVENTION The present invention overcomes these and other limitations inherent in the prior art by providing rAAV compositions that comprise a genetic construct that encodes one or more mammalian therapeutic polypeptides, for use in the amelioration, treatment and/prevention of a variety of bleeding disorders, such as for example, hemophilia and/or diseases or conditions that result from a lack, deficiency in, or absence of sufficient biologicall-active Factor VII peptide or polypeptides in one or more cells of the affected mammal. In illustrative embodiments, the invention discloses particular rAAV compositions useful in a variety of therapeutic and diagnostic regimens and in the manufacture of medicaments for treating various mammalian bleeding disorders, including Factor VII deficiency and hemophilia A, in particular. The invention provides compositions and methods for treating or ameliorating Factor VII polypeptide deficiencies in a mammal, and particularly Factor VII deficiency in a human, diagnosed with, at risk for developing, or presenting clinical symptoms of one or more bleeding disorders, such as Factor VII deficiency or hemophilia. The invention provides methods for treating or reducing the severity or extent of Factor Vπ polypeptide deficiency in a human manifesting one or more of the disorders linked to a deficiency in Factor VII polypeptide in cells and tissues of a human in need thereof. In a general sense, the method involves administration of at least a first composition that comprises a rAAV-based genetic construct that encodes one or more Factor VII peptides, polypeptides, or proteins in a pharmaceutically-acceptable vehicle to the animal in an amount and for a period of time sufficient to prevent, treat, or ameliorate one or more symptoms of the Factor VII deficiency, defect, disorder, disease, or dysfunction in the animal suspected of suffering from one or more disorders linked to a deficiency in Factor VII. The invention provides for superior advantages over the currently-existing products as it results in long-term expression of the Factor VII protein in the patient from a single, or few doses. Current products are extremely unstable and often in short supply as it is extremely difficult and costly to produce. Delivery of a gene therapy therapeuticum such as recombinant Factor VII via recombinant adeno-associated virus (rAAV)-mediated gene transfer overcomes the need for frequent and costly administration of exogenous Factor VII polypeptides, or administration of plasma, plasma concentrates, or plasma extracts containing the Factor VII polypeptide. Because activated Factor VII acts through the extrinsic pathway of the clotting cascade and circumvents the necessity for functional Factor V_H, the present therapy method provides a vital alternate form of treatment for hemophiliacs which are refractory to current replacement therapies. Additional benefits of the present therapy include the elimination of a need for repeated injections or exogenous Factor VH polypeptide. In fact, recipients of the current therapy would receive multiple benefits including, for example, the increased efficacy of treating the disease than with the currently available modalities, and an overall reduced healthcare cost, as repeated injections of the purified protein are obviated by the gene therapy methods provided herein. Many patients with hemophilia A are not able to control their disease with current replacement therapies and are left with few or no options. This invention provides a cost- effective option, with improved patient compliance, more convenient dosage and administration regimens, and less repeated injections. The recombinant AAV- Factor VII compositions disclosed herein also lower expenses due to far fewer dosings and increased stability and longevity of treatment. A single dose of recombinant AAV has been shown to produce therapeutic proteins over the course of weeks, months, and even years.
2.1 RAAV-FACTOR VII VECTOR COMPOSITIONS In a first embodiment, the invention provides an rAAV vector comprising a polypeptide that comprises at least a first nucleic acid segment that encodes a mammalian Factor VII peptide or polypeptide, and in particular, a biologically-active Factor VII (FVII) polypeptide, or biologically-active fragment thereof, operably linked to at least a first promoter capable of expressing the nucleic acid segment in a suitable host cell transformed with such a vector. In preferred embodiments, the nucleic acid segment encodes a mammalian, and in particular, a human Factor VII polypeptide, such as for example, one or more of the polypeptides as disclosed in SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, and SEQ ID NO: 17.
In addition to therapeutic polynucleotides and polypeptides of human origin, the invention also encompasses treatment modalities involving the use of one or more other
mammalian Factor VII genes or proteins, as may be desirable in the treatment of humans, or
other mammals (such as for example, in veterinary medicine therapies), and as such, the
rAAV vectors may comprise sequences of murine, porcine, feline, canine, bovine, ovine,
equine, epine, caprine, or lupine origin. In an example presented herein as an illustrative
embodiment of the practice of the invention, the rAAV-Factor VII constructs comprise at least a first genetic sequence that encodes a human Factor VII peptide, polypeptide, or
protein, to provide therapeutic levels of the selected protein, e.g., Factor VII, to the transfected cells.
Alternatively, the therapeutic constructs of the invention may encompass nucleic acid segments that encode modified hemophilia-inhibitory polypeptides obtained from any
mammalian origin, and engineered by the hand of man to produce more desirable properties
or characteristsics. For example, nucleic acids, peptides, and polypeptides of murine, primate, ovine, porcine, bovine, equine, epine, caprine, canine, feline, and/or lupine origin,
may be used in their native or unmodified form, but also may be modified or site-specifically
mutagenized, and/or otherwise genetically modified to be expressed in human cells such that
their Factor VII biological activity is retained, increased, or prolonged.
Preferred rAAV vector backbones for the practice of the present invention include,
but are not limited to, rAAV serotype 1 (rAAVl), rAAV serotype 2 (rAAV2), rAAV
serotype 3 (rAAV3), rAAV serotype 4 (rAAV4) and rAAV serotype 5 (rAAV5) and rAAV
serotype 6 (rAAVό). The polynucleotides comprised in the vectors and viral particles of the present invention preferably comprise at least a first constitutive or inducible promoter operably linked to the nucleic acid segments disclosed herein. Such promoters may be homologous or heterologous promoters, and may be operatively positioned upstream of the nucleic acid segment encoding the therapeutic polypeptide of interest, such that the expression of the segment is under the control of the promoter. The construct may comprise a single promoter, or alternatively, two or more promoters may be used to facilitate expression of the therapeutic gene sequence. Exemplary promoters useful in the practice of the invention include, but are in no way limited to, those promoter sequences that are operable in mammalian, and in particular, human host cells, tissues, and organs, such as for example, a
CMV promoter, a β-actin promoter, a hybrid CMV promoter, a hybrid β-actin promoter, an
EF1 promoter, a Ula promoter, a Ulb promoter, a Tet-inducible promoter and a VP16-LexA promoter being particularly useful in the practice of the invention. In illustrative embodiments, a polynucleotide encoding a therapeutic polypeptide was placed under the
control of the chicken β-actin (CBA) promoter and used to produce therapeutically effective
levels of the encoded polypeptide when suitable host cells were transformed with the genetic construct. The polynucleotides comprised in the vectors and viral particles of the present invention may also further optionally comprise one or more native, synthetic, homologous, heterologous, or hybrid enhancer or 5' regulatory elements, for example, a CMV enhancer, a synthetic enhancer, or a liver- or tissue-specific enhancer operably linked to the therapeutic polypeptide-encoding nucleic acid segments disclosed herein. The polynucleotides and nucleic acid segments comprised within the vectors and viral particles of the present invention may also further optionally comprise one or more intron sequences. The polynucleotides comprised in the vectors and viral particles of the present invention may also further optionally comprise one or more native, synthetic, homologous, heterologous, or hybrid post-transcriptional or 3' regulatory elements operably positioned relative to the therapeutic polypeptide-encoding nucleic acid segments disclosed herein to provide greater expression, stability, or translation of the encoded polypeptides. One such example is the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE), operably positioned downstream of the therapeutic gene of interest. In illustrative embodiments, the invention concerns administration of one or more biologically-active neovascularization-inhibitory peptides or polypeptides that comprise an at least 20, at least 40, at least 60, at least 80, at least 100, at least 120, at least 140, at least 160, at least 180, at least 200, at least 220, at least 240, at least 260, at least 280, at least 300, or more contiguous amino acid sequence from one or more of the polypeptide sequences disclosed in Section 5.2 hereinbelow and particularly those polypeptides as recited in any one of SEQ ID NO:2, SEQ ID NO:4. SEQ ID NO:6. SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, or SEQ ID NO:17. Likewise, in additional illustrative embodiments, the invention concerns administration of one or more biologically-active Factor VII polypeptides that are encoded by a nucleic acid segment that comprises at least 30, at least 60, at least 90, at least 120, at least 150, at least 180, at least 210, at least 240, at least 270, at least 300, at least 330, at least 360, at least 390, at least 420, at least 450, at least 480, at least 510, at least 540, at least 570, or at least 600, 700, 800, or 900, or more contiguous nucleic acid residues, up to and including substantially full-length, and full-length sequences from the DNA sequences disclosed in Section 5.2 hereinbelow and particularly those DNA sequences as recited in any one of SEQ ID NO:l, SEQ ID NO:3. SEQ ID NO:5. SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:ll, or SEQ ID NO:13. Exemplary adeno-associated viral vector constructs and polynucleotides of the present invention include those that comprise, consist essentially of, or consist of at least a first nucleic acid segment that encodes a peptide or polypeptide that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the sequence of SEQ ID NO:2, SEQ ID NO:4. SEQ ID NO:6. SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO:15, SEQ ID NO:16, or SEQ ID NO: 17, wherein the peptide or polypeptide has Factor VII activity when administered to, and expressed in, a suitable mammalian cell. Exemplary polynucleotides of the present invention also include those sequences that comprise, consist essentially of, or consist of at least a first nucleic acid segment that encodes a polypeptide that is at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about
92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the amino acid sequence of any one of SEQ ID NO:2, SEQ ID NO:4. SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, or SEQ ID NO: 17, wherein the peptide or polypeptide encoded by the nucleic acid segment has Factor VII activity when administered to a mammalian eye. Particularly preferred adeno-associated viral vector constructs and polynucleotides of the present invention include those that comprise, consist essentially of, or consist of at least a first nucleic acid segment that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the sequence of any one of SEQ ID NO:l, SEQ ID NO:3. SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13, wherein the segment encodes a peptide or polypeptide that has Factor VII activity when administered to, and expressed in, a suitable mammalian cell. Exemplary polynucleotides of the present invention also include those sequences that comprise, consist essentially of, or consist of at least a first nucleic acid segment that is at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about
93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to the sequence of any one of SEQ ID NO:l, SEQ ID NO:3. SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13, wherein the peptide or polypeptide encoded by the nucleic acid segment has Factor VII activity when administered to a mammalian eye.
2.2 RAAV VIRAL PARTICLES AND VIRIONS, AND HOST CELLS COMPRISING THEM Other aspects of the invention concern rAAV particles and virions that comprise the vectors of the present invention, pluralities of such particles and virions, as well as pharmaceutical compositions and host cells that comprise one or more of the rAAV vectors disclosed herein, such as for example pharmaceutical formulations of the rAAV vectors or virions intended for administration to a mammal through suitable means, such as, by intramuscular, intravenous, or direct injection to selected cells, tissues, or organs of the mammal, for example, to the muscle tissue, the circulatory system, or directly to one or more organs of the selected mammal, such as for example, by direct administration to the liver, or to liver cells. Typically, such compositions will be formulated with pharmaceutically- acceptable excipients, buffers, diluents, adjuvants, or carriers, as described hereinbelow, and may further comprise one or more liposomes, lipids, lipid complexes, microspheres, microparticles, nanospheres, or nanoparticle formulations to facilitate administration to the selected organs, tissues, and cells for which therapy is desired.
Further aspects of the invention include mammalian host cells, and pluralities thereof that comprise one or more of the adeno-associated viral vectors, virions, or viral particles as disclosed herein. Particularly preferred cells are human host cells, and in particular, human bone, blood, liver, pancreatic, kidney, muscle, heart, lung, epithelial, endothelial, or vascular cells. 0 2.3 THERAPEUTIC KITS AND PHARMACEUTICAL COMPOSITIONS The compositions of the invention also will optionally further comprising at least a first pharmaceutical vehicle, and particularly those formulations that are acceptable for administration to a human through one or more conventional routes of administration, such 5 as for example, oral, nasal, inhalation, trasndermal, intravenous, subcutaneous, or intramuscular administration. The compositions of the invention may also further comprise one or more liposomes, lipids, proteins, peptides, polypeptides, nucleic acids, polysaccharides, antibodies, antigens, antigen binding fragments, enzymes, lipid complexes, or at least a first detectable label, marker, or tag. The rAAV particles may be conjugated or 0 otherwise associated with one or more surfaces of a micro- or nanoparticle, such as for example, the nanospheres and microspheres described herein.
Therapeutic kits for treating or ameliorating the symptoms of hemophilia, bleeding or clotting disorders, or other condition resulting from defect, deficiency, or dysfunction of the native Factor FII polypeptide in a mammal are also part of the present invention. Such kits
!5 typically comprise one or more of the disclosed AAV vector constructs, virion or virus particles, host cells, or therapeutic AAV compositions described herein, and instructions for using the kit. Another important aspect of the present invention concerns methods of use of the
disclosed vectors, virions, compositions, and host cells described herein in the preparation of
medicaments for treating or ameliorating the symptoms of a bleeding disorder, clotting
disorder, hemophilia, or other conditions resulting from a defect, deficiency, or dysfunction
of Factor VII polypeptide in a mammal. Such methods generally involve administration to a
mammal, or human in need thereof, one or more of the disclosed vectors, virions, host cells, or compositions, in an amount and for a time sufficient to treat or ameliorate the symptoms
of such a defect, dysfunction, or deficiency in the affected mammal. The methods may also
encompass prophylactic treatment of animals suspected of having such conditions, or administration of such compositions to those animals at risk for developing such conditions
either following diagnosis, or prior to the onset of symptoms. Such symptoms may include,
but are not limited to, increased bleeding time, increased clotting time, or hemophilia in affected animals.
Another aspect of the invention concerns compositions that comprise one or more of
the disclosed adeno-associated viral vectors, virions, viral particles, and host cells as
described herein. Pharmaceutical compositions comprising such are particularly
contemplated to be useful in therapy, and particularly in the preparation of medicaments for
treating Factor VII deficiency, dysfunction, or defect in affected mammals, and humans in
particular.
The present invention also provides for a host cell that comprises the rAAV-Factor
VII compositions disclosed herein. Preferably, such host cells are mammalian cells, with
human host cells being particular preferred. For example, the host cell may be a human pancreas, kidney, muscle, epithelial, endothelial, perivascular, liver, heart, lung, brain, blood, bone, or nerve cell.
2.4 THERAPEUTIC METHODS
5 The invention also provides methods for delivering therapeutically-effective amounts of a biologically-active Factor VII polypeptide to a mammal in need thereof. Such methods generally comprise at least the step of providing or administering to such a mammal, one or more of the AAV-Factor VII compositions disclosed herein. For example, the method may involve providing to such a mammal, one or more of the rAAV vectors,0 virions, viral particles, host cells, or pharmaceutical compositions as described herein.
Preferably such providing or such administration will be in an amount and for a time effective to provide a therapeutically-effective amount of one or more of the Factor VII peptides or polypeptides disclosed herein to selected cells, tissues, or organs of the mammal, and in particular, therapeutically-effective levels to the cells, tissues, or organs of 5 the mammal. Such methods may include systemic injection(s) of the therapeuticum, or may even involve direct or indirect administration, injection, or introduction of the therapeutic compositions to particular cells, tissues, or organs of the mammal, such as for example, by direct injection into muscle or liver tissues.
The invention also provides methods of treating, ameliorating the symptoms, and
!0 reducing the severity of Factor VII deficiency in an animal. These methods generally involve at least the step of providing to an animal in need thereof, one or more of the rAAV vector compositions disclosed herein in an amount and for a time effective to treat the Factor VII deficiency or other related dysfunction in the animal. As described above, such methods may involve systemic injection(s) of the therapeuticum, or may even involve direct or indirect administration, injection, or introduction of the therapeutic compositions to particular cells, tissues, or organs of the animal. In one embodiment, the invention provides a method for treating, preventing, or ameliorating the symptoms of a Factor VII protein, peptide, or polypeptide deficiency or dysfunction in a mammal. The method generally involves administering to a mammal in need thereof, one or more of the disclosed rAAV-Factor VII vector compositions disclosed herein, in an amount and for a time sufficient to treat, prevent, or ameliorate the symptoms of the Factor VII deficiency or the bleeding disorder or dysfunction, such as e.g., hemophilia, in the mammal. In preferred embodiments, the mammal is a human that is has, is at risk for developing, or has been diagnosed with one or more diseases, disorders, or dysfunctions that result from the deficiency or lack of one or more Factor VII peptides, polypeptides, or proteins normally present in a healthy subject. In such cases, the compositions of the invention may be administered to the patient in an amount and for a time sufficient to treat or prevent the symptoms of the Factor VII deficiency or dysfunction through a single dose, or by administration of a plurality of doses given over a relatively short, or even relatively long period of therapy. The patient may require only one or two administrations of the disclosed rAAV constructs to achieve relatively short-term, relatively medium-term, or even relatively long-term treatment. For example, one or two administrations of the disclosed compositions may provide sufficient therapeutic levels of the Factor VII composition for a period of several days, several weeks, or several months. Alternatively, three or four administrations of the disclosed compositions either over a relatively short, or relatively long administration period, may provide sufficient therapeutic levels of the Factor VII composition for a period of several weeks, several months, several years, or even tens of years, up to and including the natural lifetime of the treated mammal. When relatively short-term therapy is warranted, the therapeutic effectiveness of a single administration or of multiple administrations of the disclosed compositions may
persist for a period of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days or more, and even up to an
including a period of about 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days or more. When
relatively medium-term therapy is warranted, the therapeutic effectiveness of a single
administration or of multiple administrations of the disclosed compositions may persist for a
period of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks or more, and even up to an including a
period of about 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 weeks or more, such as for example,
about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, or about 30 weeks or more, and even up to an including a period of about 31, 32, 33, 34, 35, 36,
37, 38, 39, or 40 weeks or more. Likewise, when relatively long-term therapy is warranted, the therapeutic effectiveness of a single administration or of multiple administrations of the
disclosed compositions may persist for a period of about 41, 42, 43, 44, 45, 46, 47, 48, 49, or
50 weeks or more, and even up to an including a period of about 51, 52, 53, 54, 55, 56, 57, 58, 59, or 60, 70, 80, 90, 100, or even 200 or 300 weeks or more. As such, the inventors
contemplate that particular therapeutic regimens involving one or more of the compositions
disclosed herein will provide a biologically- effective amount of the Factor VII peptide,
polypeptide, or protein, to the individual to which such compositions have been
administered, for periods of at least about 1 month, at least about 2 months, at least about 3
months, at least about 4 months, at least about 5 months, at least about 6 months, at least
about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at
least about 11 months, at least about 12 months 4 and up to and including periods of therapy
that persist in the treated individual for periods of at least about 1 year, at least about 2 years,
at least about 3 years, at least about 4 years, year, at least about 6 years, at least about 7 years, at least about 8 years, at least about 9 years, or even at least about 10 or more years, up to and including the natural lifetime of the treated individual. The rAAV-Factor VII compositions disclosed herein may be administered by any of the conventional drug delivery methods, such as for example, orally, intranasally,
5 transdermally, intramuscularly, intravenously, subcutaneously, intrathecally, intraperitoneally, or by absorption, inhalation or direct injection into at least a first organ or at least a first tissue of the patient as may be required. Exemplary organs and tissues which may find particular benefit through administration of one or more of the compositions disclosed herein include, but are not limited to, the vascular or circulatory system, the0 pancreas, liver, heart, lung, brain, kidney, joint, bone, neural, and muscles. In yet another embodiment, the invention provides kits for treating, preventing, or ameliorating the symptoms of a Factor VII protein, peptide, or polypeptide-related deficiency or disorder in a mammal, comprising (i) one or more rAAV-Factor VII composition disclosed herein; and (ii) instructions for using the kit in diagnostic, therapeutic, or5 prophylactic treatment regimens.
3. BRIEF DESCRIPTION OF THE DRAWING The following drawing forms part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by !0 reference to the following description taken in conjunction with the accompanying drawing, in which like reference numerals identify like elements, and in which:
FIG. 1 shows that functional Factor VII can be produced in vitro. The chromogenic Coaset assay was used to determine the levels of functional Factor VII secreted. Factor VIII KO plasma was used as a positive control as these mice have been shown to produce normal levels
5 of Factor VH. 4. DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS Illustrative embodiments of the invention are described below. In the interest of clarity, not all features of an actual implementation are described in this specification. It will of course be appreciated that in the development of any such actual embodiment, numerous implementation-specific decisions must be made to achieve the developers' specific goals, such as compliance with system-related and business-related constraints, which will vary from one implementation to another. Moreover, it will be appreciated that such a development effort might be complex and time-consuming, but would nevertheless be a routine undertaking for those of ordinary skill in the art having the benefit of this disclosure. The practical feature of this invention is the ability to treat hemophilia patients that are resistant to current therapy. AAV-mediated gene delivery has demonstrated long-term correction of the diseased state with a minimal pathology. Optimally, a single delivery of AAV- Factor VH should result in disease correction for the lifetime of the individual.
4.1 ADENO-ASSOCIATED VIRUS Adeno-associated virus (AAV) is a single-stranded DNA parvo virus with a 4.7 kb genome and a particle diameter of approximately 20 nm. The AAV genome is flanked by two identical inverted terminal repeat (ITR) sequences (Lusby et al, 1980). These ITRs provide all the cw-acting sequence required for replication, packaging and integration (Samulski et al, 1989). There are two large open reading frames (Srivastava et al, 1983). The open reading frame in the right half of the genome (cap) encodes 3 overlapping coat proteins (VP1, VP2 and VP3). The open reading frame in the left half (rep gene) encodes 4 regulatory proteins with overlapping sequences which are known as Rep proteins (Rep78, Rep68, Rep52 and j?ej_>40), because frame shift mutations at most locations within the open reading frame inhibit viral DNA replication (Hermonat et al, 1984). The Rep proteins are multi-functional DNA binding proteins. The functions of the Rep proteins in viral DNA replication include helicase activity and a site-specific, strand-specific endonuclease (nicking) activity (Ni et al, 1994). AAV infects a broad spectrum of vertebrates from birds to humans, although in nature specific types are species specific (Berns, 1996). In humans AAV can infect a large variety of cells derived from different tissues. The infection of AAV is ubiquitous within the population with about 90% of adults being seropositive (Cukor et al, 1983). In spite of its omnipresence, AAV has never been associated with any human disease. In this sense, rAAV is the safest of the currently used gene therapy vectors. Because of its propensity to establish latency and because it has not been implicated as a pathogen, AAV has been of considerable interest as a potential vector for human gene therapy (Flotte and Ferkol, 1997; Flotte and Carter, 1995). In general, rAAV vectors are produced by deleting the viral coding sequences and substituting the fransgene of interest under control of a non-AAV promoter between the two AAV inverted terminal repeats (ITRs). When the rep and cap proteins are expressed in trans in Ad-infected cells, rAAV genomes can be efficiently packaged. Considerations in the development of AAV as a vector have included difficulties in attaining high vector titers and the limited insertional capacity (>5 kb). Although these issues can still be improved, recently developed packaging techniques for high titer and Ad- contamination free vectors, and strategies to overcome the packaging limitation, have dramatically impacted the applications of rAAV (Zolotukhin et al, 1999; Duan et al, 2000;
Yan et al, 2000). Unlike adenovirus vectors, rAAV vectors are remarkably nonimmunogenic with little host response (Jooss et al, 1998; Song et al, 1998). In addition to the above unique features, rAAV have mediated long-term fransgene expression in a wide variety of tissues, including muscle (Song et al, 1998; Kessler et al, 1996; Xiao et al, 1996; Clark et al, 1997; Snyder et al, 1997a), lung (Flotte et al, 1993), liver (Snyder et al, 1997b; Xiao et al, 1998; Song et al, 2001a; Xu et al, 2001), brain (Kaplitt et al, 1994) and eye (Flannery et al, 1997). Thus rAAV vectors appear to have significant advantages over other commonly used viral vectors. Six serotypes of AAV have been cloned and sequenced. Of the six AAV serotypes, serotype 2 (AAV2) is the best-characterized and has been predominantly used in gene transfer studies. Membrane-associated heparan sulfate proteoglycan is the primary receptor for AAV
type 2 (Summerford and Samulski, 1998). Human fibroblast growth factor receptor 1 and αyβs
integrin are co-receptors for AAV2 (Qing et al, 1999; Summerford et al, 1999). Serotypes 1 and 6 share >99% amino acid homology in their capsid proteins. Sequence analysis supports a recombination event between seroType I and 2. Comparison of the serotype capsid amino acid sequences suggests that serotypes, 1, 2, and 3 share homology across the three capsids in accord with heparan sulfate binding (Summerford and Samulski, 1998). In contrast, AAV type 4 and 5 are the most divergent of the six AAV serotypes, exhibiting only 60% homology to AAV2 or to each other. AAV4 and AAV5 require different sialic acid-containing glycoproteins for binding and transduction of target cells. The different tropisms of AAV serotypes provide opportunities to optimize the transduction efficiency in different target cells. Data showed that of the serotypes, AAV1 mediated the highest fransgene expression in skeletal muscle and murine islets (Chao et α/., 2000). The AAV life cycle is quite unusual (Berns and Linden, 1995). AAV binds to cells via a heparan sulfate proteoglycan receptor (Summerford and Samulski, 1998). Once attached, AAV
entry is dependent upon the presence of a co-receptor, which may consist of either the fibroblast
growth factor receptor (FGF-R) (Qing et al, 1999) or the v5 integrin molecule (Summerford
et al, 1999). Cells infected with AAV and a helper virus (or another adjunctive agent, such as
UV irradiation or treatment with genotoxic agents) will undergo productive replication of AAV
prior to cell lysis, which is induced by the helper rather than by AAV. Human cells infected with AAV alone, however, will become persistently infected (Berns et al, 1975). This latency
pathway often results in colinear integration of AAV sequences within the host cell genome
(Cheung et al, 1980), often within a specific site on human chromosome 19, the AAVS1 site
(Kotin etα/., 1990; Kotin et α/., 1991; Kotin etα/., 1992; Samulski etal, 1991; Samulski, 1993.
While this site is not strictly homologous to AAV, it contains consensus elements required for
binding and nicking by the AAV Rep protein, a non-structural protein that is also involved in
productive replication and in transcriptional regulation of the virus (Weitzman et al, 1994;
Giraud et al, 1994; Giraud et al, 1995; Linden et al, 1996). Once AAV is integrated, it will
remain stable within infected cells for prolonged periods of time, up to 100 passages (Hoggan et al, 1972). Episomal forms of the virus may also be present for extended periods in some
circumstances (Afione et al, 1996; Kearns et al, 1996; Flotte et al., 1994). If latently infected cells are subsequently infected with a helper virus, the genome will be excised and productive
AAV replication and packaging will ensue (Senapathy et al, 1984; Afione et al, 1996). The AAV genome consists of two 145-nucleotide inverted terminal repeat (ITR)
sequences, each an identical palindrome at either terminus of the virus, flanking the two AAV
genes, rep and cap (Tratschin et al , 1984). The rep gene is transcribed from two promoters, the
p5 promoter (at map position 5) and the pl9 promoter (map position 19), which is embedded
within tlie coding sequence of the longer Rep proteins. In each case, both the spliced and
unspliced transcripts are processed and translated. This allows for the production of 4 Rep
proteins, Rep78, Rep68, Rep52, and Rep40. Rep78 and Rep68 are multifunctional DNA
binding proteins which possess helicase activity and site-specific, strand-specific nickase
activity, both of which are required for terminal resolution of replicating AAV genomes (Im and
Muzyczka, 1990). The long Rep proteins are also capable of binding to the chromosomal target
sequence for AAV integration, the AAVS1 site, and these proteins are required for normal
integration into this site. Finally, Rep78/68 are potent bi-functional transcription regulators that generally activate transcription from AAV promoters during productive infection and repress their transcription during latent infection (Pereira and Muzyczka, 1997; Pereira et al, 1997). The shorter Rep proteins, Rep52 and Rep40 act as modifier proteins for long Rep transcriptional activities, and are required for sequestration of single-stranded AAV genomes into capsids during productive infection. The AAV cap gene is transcribed from the p40 promoter. The 5' end of the mRNA transcript from p40 contains an intron which can utilize either of two downstream splice acceptor sites. The longer of the two processed messages contains an ATG codon which is used in the translation of VP1, the longest of the three AAV capsid proteins. The shorter mRNA can initiate translation using either a non-canonical ACG start codon, which represents the start of
VP2, or an ATG codon further downstream, which comprises the N-terminal Met of VP3 (Trempe and Carter, 1988). VP3 is the shortest and most abundant of the AAV capsid proteins, but all three are required for the production of infectious virions.
4.2 RECOMBINANT AAV VECTORS Recombinant AAV (rAAV) vectors have been developed by replacement of the viral coding sequences with fransgene of interest (Tratschin et al, 1984; Hermonat and Muzyczka, 1984). The ITR sequences must be retained in rAAV since these serve as origins for viral DNA replication and contain the packaging signals. The maximum packaging capacity of rAAV is approximately 5 kb, including the ITRs, the fransgene, its promoter, and polyadenylation signal
(Flotte et al, 1992; Dong et al, 1996). The full exploitation of rAAV for gene transfer has been limited in the past primarily by the packaging and purification process. In particular, contamination of rAAV vector preparations with wild-type AAV has been found to alter the biological behavior of the vector, and limitations on the titers and infectivity of the vectors have limited their widespread use on the past. Recent advances in the packaging and purification technology have resulted in a dramatic improvement in the expression levels that have been
achievable in vivo. In particular, the use of adenoviral plasmids and of complementing rep gene
expression constructs that express less of the longer Rep proteins (Rep68/78) has resulted in a
substantial improvement in the efficiency of vector production on a per cell basis (Xiao et al,
1998; Li et al, 1997). The availability of packaging cell lines has also resulted in a substantial
improvement in the scale-ability of the packaging process (Clark et al, 1996; Flotte et al, 1995;
Gao et al, 1998). Finally, the availability of several column chromatography methods,
including heparin sulfate affinity column chromatography, has allowed for the elimination of CsCl banding, which in turn appears to have enhanced the infectivity of output particles
(Zolotukhin et al , 1999). rAAV vectors are uniquely suitable for in vivo gene therapy for genetic and metabolic disorders, since they are non-toxic (Flotte et al, 1993; Conrad et al, 1996; Flotte and Carter,
1998), highly efficient when used at high titers, relatively non-immunogenic (Jooss et al, 1998;
Hernandez et al, 1999; Beck et al, 1999), and very stable in their pattern of expression. The utility of rAAV vectors for in vitro and in vivo gene transfer has now been well established.
There appear to be important tissue specific differences in rAAV effects, however. rAAV
vectors have been found to be particularly efficient for gene transfer into terminally
differentiated cells such as neurons (Kaplitt et al, 1994; McCown et al, 1996; Peel et al, 1997;
Mandel et al, 1997), myofibers (Xiao et al, 1996; Kessler et al, 1996; Clark et al, 1997;
Fisher et al, 1997; Song et al, 1998), and photoreceptor cells (Flannery et al, 1997; Lewin et
al, 1998; Zolotukhin et al, 1996; Rolling et al, 1999). Gene transfer to the bronchial
epithelium has also been demonstrated (Flotte et al, 1993; Conrad et al, 1996; Afione et al,
1996; Flotte et al, 1998; Halbert et al, 1998), although the efficiency of transduction remains
relatively low. Likewise, rAAV transduction of hepatocytes has also been studied, and has been
found to be efficient enough to provide a potential therapeutic strategy for hemophilia B, by providing persistent and therapeutic concentrations of human Factor IX in mice (Snyder et al, 1997). However, in that study, in situ hybridization results indicated that only 5% of hepatocytes had been transduced (Miao et al, 1998). In the case of each of these two cell types, recent evidence has shown that the efficiency can be enhanced by altering the capsid to incorporate ligands for a receptor that is abundantly expressed on the cell surface and by optimizing the promoter usage (Wu et al, 2000; Virella- Lowell et al, 1999). Similar manipulations are also advantageous in pancreatic islet cells. Recent reports of severe dose-related clinical adverse events due to adenovirus, although not directly reflective of rAAV, underscore the necessity of minimizing the dose of vector whenever possible.
4.3 RAAV THERAPY FOR HUMAN DISEASES During recent years, viral vector-based human gene therapy approaches have been developed as potentially effective, alternative treatment modalities for a variety of diseases. In particular, adeno-associated virus 2 (AAV), a defective parvo virus of human origin, has been demonstrated to be one such promising vector. AAV is an ideal vector for viral-based human gene therapy because it has not been associated with any known pathology and post-infection, the viral genome integrates into the human chromosome (Muzyczka, 1992). Recombinant adeno-associated virus (rAAV) vectors have important utility as vehicles for the in vivo delivery of polynucleotides to target host cells (Kessler etal, 1996; Koeberl etal, 1997; Kotin, 1994;
Xiao et al, 1996). rAAV vectors are useful vector for efficient and long-term gene transfer in a variety of mammalian tissues, e.g., lung (Flotte et ah, 1993), muscle (Kessler et al, 1996; Xiao etal, 1996; Clark etal, 1997; Fisher etal, 1997), brain (Kaplitt etal, 1994; Klein etal, 1998) retina (Flannery etal, 1997; Lewin etal, 1998), and liver (Snyder, 1997). It has also been shown that rAAV can evade the immune response of the host by failing to transduce dendritic cells (Jooss et al, 1998). Clinical trials have been initiated for several important mammalian diseases including hemophilia B, muscular dystrophy and cystic fibrosis (Flotte etal, 1996; Wagner etal, 1998; Flotte and Carter, 1995; Kay et al, 2000). As with most gene therapy vectors, obstacles in the efficient use of rAAV vectors for a variety of disease models include sub-therapeutic levels of transduction and the ability to target the site(s) of gene transfer.
4.4 PROMOTERSANDENHANCERS Recombinant AAV vectors form important aspects of the present invention. The term
"expression vector or construct" means any type of genetic construct containing a nucleic acid in which part or all of the nucleic acid encoding sequence is capable of being transcribed. In preferred embodiments, expression only includes transcription of the nucleic acid, for example, to generate a biologically-active Factor VII polypeptide product from a transcribed gene. Particularly useful vectors are contemplated to be those vectors in which the nucleic acid segment to be transcribed is positioned under the transcriptional control of a promoter. A "promoter" refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. The phrases "operatively positioned," "under control" or "under transcriptional control" means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene. In preferred embodiments, it is contemplated that certain advantages will be gained by positioning the coding DNA segment under the control of a recombinant, or heterologous, promoter. As used herein, a recombinant or heterologous promoter is intended to refer to a promoter that is not normally associated with a biologically-active Factor Vπ gene in its natural environment. Such promoters may include promoters normally associated with other genes, and/or promoters isolated from any bacterial, viral, eukaryotic, or mammalian cell. Naturally, it will be important to employ a promoter that effectively directs the expression of the biologically-active Factor VH-encoding DNA segment in the cell type, organism, or even animal, chosen for expression. The use of promoter and cell type combinations for protein expression is generally known to those of skill in the art of molecular biology, for example, see Sambrook etal. (1989), incorporated herein by reference. The promoters employed may be constitutive, or inducible, and can be used under the appropriate conditions to direct high-level expression of the introduced DNA segment. At least one module in a promoter functions to position the start site for RNA synthesis. The best-known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl fransferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation. Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upsfream of the start site, although a number of promoters have been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
In the tk promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either co-operatively or independently to activate transcription. The particular promoter that is employed to control the expression of a nucleic acid is not believed to be critical, so long as it is capable of expressing the biologically-active Factor VII polypeptide-encoding nucleic acid segment in the targeted cell. Thus, where a human cell is targeted, it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell. Generally speaking, such a promoter might include either a human or viral promoter, such as a beta- actin, CMV, an HSV promoter, or even a human tissue-specific or otherwise inducible promoter. In certain aspects of the invention, the chicken beta-actin promoter has been demonstrated to be particularly desirable in some embodiments disclosed herein. In various other embodiments, the human cytomegalovirus (CMV) immediate early gene promoter, the SV40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression of transgenes. The use of other viral or mammalian cellular or bacterial phage promoters that are well known in the art to achieve expression of a fransgene is contemplated as well, provided that the levels of expression are sufficient for a given purpose. Tables 1 and 2 below list several elements/promoters that may be employed, in the context of the present invention, to regulate the expression of the present biologically- active therapeutic polypeptide-encoding nucleic acid segments comprised within the rAAV-
Factor VII vectors and compositions of the present invention. This list is not intended to be exhaustive of all the possible elements involved in the promotion of fransgene expression, but merely to be exemplary thereof. Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of DNA with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins. The basic distinction between enhancers and promoters is operational. An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. On the other hand, a promoter must have one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization. Additionally any promoter/enhancer combination (as per the Eukaryotic Promoter Data Base EPDB) could also be used to drive expression. Use of a T3, T7 or SP6 cytoplasmic expression system is another possible embodiment. Eukaryotic cells can support cytoplasmic transcription from certain bacterial promoters if the appropriate bacterial polymerase is provided, either as part of the delivery complex or as an additional genetic expression construct.
TABLE 1 PROMOTER AND ENHANCER ELEMENTS
PROMOTER/ENHANCER REFERENCES
Immunoglobulin Heavy Chain Banerji et al, 1983; Gilles et al, 1983; Grosschedl and Baltimore, 1985; Atchison and Perry, 1986, 1987; Imler et α/., 1987; Weinberger et al, 1984; Kiledjian et al, 1988; Porton et α/.; 1990
Immunoglobulin Light Chain Queen and Baltimore, 1983; Picard and Schaffner, 1984
T-Cell Receptor Luria et al, 1987; Winoto and Baltimore, 1989; Redondo et α/.; 1990 PROMOTER ENHANCER REFERENCES
HLA DQ a and DQ β Sullivan and Peterlin, 1987
β-Interferon Goodboum et al, 1986; Fujita et al, 1987; Goodboum and Maniatis, 1988
Interleukin-2 Greene et al, 1989
Interleukin-2 Receptor Greene et al, 1989; Lin et al, 1990
MHC Class II 5 Koch et al, 1989
MHC Class II HLA-Dra Sherman et al, 1989
β-Actin Kawamoto etal, 1988; Ng etal; 1989
Muscle Creatine Kinase Jaynes etal, 1988; Horlick and Benfield, 1989; Johnson et al. , 1989
Prealbumin (Transthyretin) Costa et al, 1988
Elastase I Ornitz et /., 1987
Metallothionein Karin et al, 1987; Culotta and Hamer, 1989
Collagenase Pinkert et /., 1987; Angel et al, 1987a
Albumin Gene Pinkert etal, 1987; Tronche etal, 1989, 1990
α-Fetoprotein Godbout etal, 1988; Campere and Tilghman, 1989
t-Globin Bodine and Ley, 1987; Perez-Stable and Constantini, 1990
β-Globin Trudel and Constantini, 1987
e-fos Cohen et al, 1987 PROMOTER/ENHANCER REFERENCES
c-HA-ras Triesman, 1986; Deschamps etal, 1985
Insulin Edlund et α/,, 1985
Neural Cell Adhesion Molecule Hirsch et al. , 1990 (NCAM)
0"1-Antitrypain Latimer et α/., 1990
H2B (TH2B) Histone Hwang et al, 1990
Mouse or Type I Collagen Ripe et al, 1989
Glucose-Regulated Proteins Chang et al, 1989 (GRP94 and GRP78)
Rat Growth Hormone Larsen et α/., 1986
Human Serum Amyloid A (SAA) Edbrooke et al. , 1989
Troponin I (TN I) Yutzey et α/., 1989
Platelet-Derived Growth Factor Pech et al. , 1989
Duchenne Muscular Dystrophy Klamut et al. , 1990
SV40 Banerji et al, 1981; Moreau et al, 1981; Sleigh and Lockett, 1985; Firak and Subramanian, 1986; Herr and Clarke, 1986; Imbra and Karin, 1986; Kadesch and Berg, 1986; Wang and Calame, 1986; Ondek et al, 1987; Kuhl et al, 1987; Schaffner et al, 1988 PROMOTER ENHANCER REFERENCES
Polyoma Swartzendruber and Lehman, 1975; Vasseur et al. , 1980; Katinka et /., 1980, 1981; Tyndell et al, 1981; Dandolo et al, 1983; de Villiers et al, 1984; Hen et al, 1986; Satake et al, 1988; Campbell and Villarreal, 1988
Retrovimses Kriegler and Botchan, 1982, 1983; Levinson et al, 1982; Kriegler etal, 1983, 1984a, b, 1988; Bosze et al, 1986; Miksicek et al, 1986; Celander and Haseltine, 1987; Thiesen et al, 1988; Celander et al, 1988; Choi etal, 1988; Reisman and Rotter, 1989
Papilloma Vims Campo etal, 1983; Lusky etal, 1983; Spandidos and Wilkie, 1983; Spalholz et al, 1985; Lusky and Botchan, 1986; Cripe et al, 1987; Gloss et al, 1987; Hirochika et al, 1987; Stephens and Hentschel, 1987
Hepatitis B Vims Bulla and Siddiqui, 1986; Jameel and Siddiqui, 1986; Shaul and Ben-Levy, 1987; Spandau and Lee, 1988; Vannice and Levinson, 1988
Human Immunodeficiency Vims Muesing et al. , 1987; Hauber and Cullan, 1988; Jakobovits et al, 1988; Feng and Holland, 1988; Takebe et al, 1988; Rosen et al, 1988; Berkhout et al, 1989; Laspia et al, 1989; Sharp and Marciniak, 1989; Braddock et /., 1989 PROMOTER/ENHANCER REFERENCES
Cytomegalovirus Weber et al, 1984; Boshart et al, 1985; Foecking and Hofstetter, 1986
Gibbon Ape Leukemia Vims Holbrook et al, 1987; Quinn et al, 1989
TABLE 2 INDUCIBLE ELEMENTS
ELEMENT INDUCER REFERENCES
MT II Phorbol Ester (TFA) Palmiter et al, 1982; Haslinger
Heavy metals and Karin, 1985; Searle et al, 1985; Stuart et al, 1985; Imagawa et al, 1987, Karin et al, 1987; Angel et al, 1987b; McNeall et α/. , 1989
MMTV (mouse mammary Glucocorticoids Huang et al. , 1981; Lee et al. , tumor vims) 1981 ; Majors and Varmus, 1983; Chandler et al, 1983; Lee et al, 1984; Ponta et al, 1985; Sakai et /., 1988
β-Interferon poly(rI)x Tavernier et al, 1983 poly(rc)
Adenovirus 5 E2 Ela Imperiale and Nevins, 1984
Collagenase Phorbol I Angel et al, 1987a ELEMENT INDUCER REFERENCES
Stromelysin Phorbol Ester (TPA) Angel et al, 1987b
SV40 Phorbol Ester (TPA) Angel et al, 1987b
Murine MX Gene Interferon, Newcastle Disease Vims
GRP78 Gene A23187 Resendez et al, 1988 -2-Macroglobulin IL-6 Kunz et α/., 1989
Vimentin Serum Rittling et α/., 1989
MHC Class I Gene H-2κb Interferon Blanar et α/., 1989
HSP70 Ela, SV40 Large T Antigen Taylor et al. , 1989; Taylor and Kingston, 1990a, b
Proliferin Phorbol Ester-TPA Mordacq and Linzer, 1989
Tumor Necrosis Factor FMA Hensel et α/., 1989
Thyroid Stimulating Thyroid Hormone Chatterjee et α/., 1989 Hormone a Gene
As used herein, the terms "engineered" and "recombinant" cells are intended to refer to a cell into which an exogenous DNA segment, such as DNA segment that leads to the transcription of a biologically-active Factor VII polypeptide or a ribozyme specific for such a biologically-active Factor VII polypeptide product, has been introduced. Therefore, engineered cells are distinguishable from naturally occurring cells, which do not contain a recombinantly introduced exogenous DNA segment. Engineered cells are thus cells having DNA segment introduced through the hand of man. To express a biologically-active Factor VII encoding gene in accordance with the
present invention one would prepare an rAAV expression vector that comprises a biologically- active Factor VH polypeptide-encoding nucleic acid segment under the confrol of one or more
promoters. To bring a sequence "under the control of a promoter, one positions the 5' end of the transcription initiation site of the transcriptional reading frame generally between about
1 and about 50 nucleotides "downstream" of (i.e., 3' of) the chosen promoter. The "upstream"
promoter stimulates transcription of the DNA and promotes expression of the encoded
polypeptide. This is the meaning of "recombinant expression" in this context. Particularly
preferred recombinant vector constructs are those that comprise an rAAV vector. Such vectors are described in detail herein.
4.5 PHARMACEUTICAL COMPOSITIONS In certain embodiments, the present invention concerns formulation of one or more of
the rAAV compositions disclosed herein in pharmaceutically acceptable solutions for
administration to a cell or an animal, either alone or in combination with one or more other modalities of therapy, and in particular, for therapy of the mammalian pancreas and tissues
thereof, such as for example, islet cells.
It will also be understood that, if desired, nucleic acid segments, RNA, DNA or PNA
compositions that express one or more of the biologically-active Factor VII therapeutic gene
products as disclosed herein may be administered in combination with other agents as well, such
as, e.g., proteins or polypeptides or various pharmaceutically-active agents, including one or
more systemic or direct administrations of therpeutic proteins, polypeptides, peptides, antisense
compounds, ribozymes, or biologically active fragments, or variants thereof. In fact, there is
virtually no limit to other components that may also be included, given that the additional agents
do not cause a significant adverse effect upon contact with the target cells or host tissues. The disclosed rAAV-Factor VH compositions may thus be delivered along with various other agents as required in the particular instance. Such compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein. Likewise, such compositions may further comprise substituted or derivatized RNA, DNA, or PNA compositions. Formulation of pharmaceutically-acceptable excipients and carrier solutions is well- known to those of skill in the art, as is the development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular administration and formulation. Typically, these formulations may contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 70% or 80% or more of the weight or volume of the total formulation. Naturally, the amount of active compound(s) in each therapeutically- useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable. In certain circumstances it will be desirable to deliver the AAV vector-based therapeutic constructs in suitably formulated pharmaceutical compositions disclosed herein either subcutaneously, intraopancreatically, parenterally, intravenously, intramuscularly, intrathecally, or even orally, infraperitoneally, or by nasal inhalation, including those modalities as described in U. S. Patent 5,543,158; U. S. Patent 5,641,515 and U. S. Patent 5,399,363 (each specifically incorporated herein by reference in its entirety). Solutions of the active compounds as freebase or pharmacologically acceptable salts may be prepared in sterile water and may also suitably mixed with one or more surfactants, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U. S. Patent 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. For administration of an injectable aqueous solution, for example, the solution may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035- 1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human admimsfration, preparations should meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologies standards. Sterile injectable solutions are prepared by incorporating the active AAV vector- delivered biologically-active Factor VII polypeptide-encoding polynucleotides in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The AAV vector compositions disclosed herein may also be formulated in a neutral or salt form. Pharmaceutically-acceptable salts, include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the
dosage formulation and in such amount as is therapeutically effective. The formulations are
easily administered in a variety of dosage forms such as injectable solutions, drag-release
capsules, and the like. As used herein, "carrier" includes any and all solvents, dispersion media, vehicles,
coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents,
buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents
for pharmaceutical active substances is well known in the art. Except insofar as any
conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into
the compositions. The phrase "pharmaceutically-acceptable" refers to molecular entities and compositions
that do not produce an allergic or similar untoward reaction when administered to a human, and in particular, when administered to human cells that express LDLR polypeptides. The
preparation of an aqueous composition that contains a protein as an active ingredient is well
understood in the art. Typically, such compositions are prepared as mjectables, either as liquid
solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to
injection can also be prepared. The preparation can also be emulsified.
4.6 LIPOSOME-, NANOCAPSULE-, AND MICROPARTICLE-MEDIATED DELIVERY
In certain embodiments, the inventors contemplate the use of liposomes, nanocapsules,
nanospheres, nanoparticles, nanofibers, microparticles, microspheres, microfibers, lipid
particles, vesicles, and the like, for the introduction of the rAAV-Factor VII vectors of the
present invention into suitable hosts, and in particular, humans. A microparticle is well
understood in the art to be "a small unit of material with a phase boundary separating it from its surroundings, such as a solid fragment or liquid droplet suspended in blood or water. The size of microparticles are typically on the order of 5 mm or less in diameter, and they may assume any shape, such as spherical, polygonal, fiber-like, or simply a fractured piece of a larger structure. Nanoparticles are understood to encompass even smaller particles, typically those having diameters in the 5 micron or less range. These particles can be made by a wide variety of known methods such as suspension, emulsion or dispersion polymerization. Ball-milling, or grinding (for instance of a larger piece cooled to low temperature if not brittle at room temperature) can be used to create smaller fragments. Aerosol spraying and solidification by cooling or photo cross-linking can also be employed to prepare microparticles or nanoparticles of preferred sizes and compositions. Ultrasonication, for example, may be used to disperse one liquid in another. Many microparticles can be obtained directly from commercial sources (for example, Bangs Laboratories) or simply made by precipitating iron oxides and using those particles without polymers, or co-precipitation them with other solids. In additions, combinations of materials may also be used to prepare suitable microspheres. For example, polymers of degradable solids (polylactic acid, polyglycolic acid, and their copolymers) or hydrogels such as dextran or starch or alginate, or non-degradable hydrogels such as polyhydroxyethyl methacrylate (PHEMA), or even non-degradable solids like polystyrene or hydroxy apatite or plaster of paris may also be used, either alone, or in combination with one or more magnetic field-responsive compounds (e.g., magnetite) to form mixed microparticles. Of course, even finely milled particles of magnetic metals (e.g., iron metal) may also be used as particulate substrates, owing to their highly desirable magnetic properties. Such nanoparticle- and microparticle-associated formulations may be preferred for the introduction of pharmaceutically acceptable formulations of the rAAV-Factor VII vectors disclosed herein. Likewise, the inventors also contemplate the use of other delivery regimens to improve introduction of the rAAV vectors into suitable animals. Such delivery regimens may include, for example, the use of nanocapsules, liposomes, lipid particles, and the like. The formation and use of liposomes for pharmaceutical administration is generally known to those of skill in the art (see for example, Couvreur et al, 1977; Couvreur, 1988; Lasic, 1998; which describes the use of liposomes and nanocapsules in the targeted antibiotic therapy for intracellular bacterial infections and diseases). Recently, liposomes were developed with improved serum stability and circulation half-times (Gabizon and Papahadjopoulos, 1988; Allen and Choun, 1987; U. S. Patent 5,741,516, specifically incorporated herein by reference in its entirety). Further, various methods of liposome and liposome like preparations as potential dmg carriers have been reviewed (Takakura, 1998; Chandran et al, 1997; Margalit, 1995; U. S.
Patent 5,567,434; U. S. Patent 5,552,157; U. S. Patent 5,565,213; U. S. Patent 5,738,868 and U. S. Patent 5,795,587, each specifically incorporated herein by reference in its entirety). Exemplary microparticles and nanoparticles, and methods for their synthesis have been described in a variety of patents including, for example, U.S. Patents 6,383,470, 6,346,274, 6,238,294, 6,284,280, 6,331,310, 6,254,890, 6,177,088, 5,972,707, 6,2587,588, and 6,361,994, each specifically incorporated herein by reference in its entirety. Magnetically responsive microparticles are useful in biological techniques requiring the separation of bound from free fractions. Magnetically responsive particles useful in immunoassays, for the separation of cells, as magnetic resonance imaging agents, etc. have been described in U.S. Pat. Nos. 3,215,572, 4,452,773, 4,795,698, 4,770,183, 4,695,392,
4,329,241, 4,230,685, 4,177,253, 5,069,216, 5,091,206, and 5,705,628, 5,597,530, all incorporated herein by reference. The targeting of pharmaceutical substrates to particular portions of the body using magnetic field-responsive microparticles has also gained clinical importance in recent years. In particular, the use of magnetically responsive microparticles may be used to selectively target one or more regions of the body by administration of the pharmaceutically-active ingredients bound to the particles, and then placing the body in a magnetic field, and using this field to enrich the population of magnetic microbeads in the focus of the magnetic field. U. S. Patent
6,178,871 (specifically incorporated herein by reference in its entirety) illustrates the use of
external magnetic fields to target magnetic materials within an animal.
Such biologically-active magnetic particles may find use in a variety of preparative and
diagnostic techniques, including those described herein. Among these is high gradient magnetic
separation (HGMS) which uses a magnetic field to separate magnetic particles from suspension.
In instances where these particles are attached to biological materials of interest (e.g., cells, drugs), the material of interest may thereby be separated from other materials not bound to the
magnetic particles. Because of their magnetic properties, these materials also function as
contrast agents for magnetic resonance imaging. As used herein, the term "resuspendable coated particle" refers to a finely divided solid,
which forms a colloidal suspension and may be separated from the suspension and subsequently resuspended. "Magnetic" encompasses material which may or may not be permanently
magnetic, which also may be paramagnetic or superparamagnetic but which in all cases exhibits
a response in a magnetic field, i.e., is magnetically responsive. "Disrupted" particles are those
which are too small to contain a complete magnetic domain or, alternatively, whose Brownian
energy exceeds their magnetic moment. Generally, such particles are less than 0.03 mu. in size. Liposomes have been used effectively to introduce genes, drags (Heath and Martin,
1986; Heath et al, 1986; Balazsovits et al, 1989; Fresta and Puglisi, 1996), radiotherapeutic
agents (Pikul et al, 1987), enzymes (Imaizumi et al, 1990a; Imaizumi et al, 1990b), viruses
(Faller and Baltimore, 1984), transcription factors and allosteric effectors (Nicolau and
Gersonde, 1979) into a variety of cultured cell lines and animals. In addition, several successful
clinical frails examining the effectiveness of liposome-mediated drag delivery have been completed (Lopez-Berestein et al, 1985a; 1985b; Coune, 1988; Sculier et al, 1988).
Furthermore, several studies suggest that the use of liposomes is not associated with autoimmune responses, toxicity or gonadal localization after systemic delivery (Mori and
Fukatsu, 1992). Liposomes are formed from phospholipids that are dispersed in an aqueous medium and
spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles
(MLVs). MLVs generally have diameters of from 25 nm to 4 μm. Sonication of MLVs results
in the formation of small unilamellar vesicles (SUVs) with diameters in the range of 200 to 500
A, containing an aqueous solution in the core. Liposomes bear resemblance to cellular membranes and are contemplated for use in connection with the present invention as carriers for the peptide compositions. They are widely
suitable as both water- and lipid-soluble substances can be entrapped, i.e. in the aqueous spaces and within the bilayer itself, respectively. It is possible that the drug-bearing liposomes may
even be employed for site-specific delivery of active agents by selectively modifying the
liposomal formulation. In addition to the teachings of Couvreur et al. (1977; 1988), the following information
may be utilized in generating liposomal formulations. Phospholipids can form a variety of
structures other than liposomes when dispersed in water, depending on the molar ratio of lipid to
water. At low ratios the liposome is the preferred stracture. The physical characteristics of
liposomes depend on pH, ionic strength and the presence of divalent cations. Liposomes can
show low permeability to ionic and polar substances, but at elevated temperatures undergo a
phase transition which markedly alters their permeability. The phase fransition involves a
change from a closely packed, ordered structure, known as the gel state, to a loosely packed,
less-ordered structure, known as the fluid state. This occurs at a characteristic phase-transition
temperature and results in an increase in permeability to ions, sugars and drugs. In addition to temperature, exposure to proteins can alter the permeability of liposomes.
Certain soluble proteins, such as cytochrome c, bind, deform and penetrate the bilayer, thereby
causing changes in permeability. Cholesterol inhibits this penetration of proteins, apparently by packing the phospholipids more tightly. It is contemplated that the most useful liposome
formations for antibiotic and inhibitor delivery will contain cholesterol.
The ability to trap solutes varies between different types of liposomes. For example,
MLVs are moderately efficient at trapping solutes, but SUVs are extremely inefficient. SUVs
offer the advantage of homogeneity and reproducibility in size distribution, however, and a compromise between size and trapping efficiency is offered by large unilamellar vesicles
(LUVs). These are prepared by ether evaporation and are three to four times more efficient at solute entrapment than MLVs.
In addition to liposome characteristics, an important determinant in entrapping
compounds is the physicochemical properties of the compound itself. Polar compounds are trapped in the aqueous spaces and nonpolar compounds bind to the lipid bilayer of the vesicle.
Polar compounds are released through permeation or when the bilayer is broken, but nonpolar compounds remain affiliated with the bilayer unless it is dismpted by temperature or exposure to
lipoproteins. Both types show maximum efflux rates at the phase transition temperature.
Liposomes interact with cells via four different mechanisms: Endocytosis by phagocytic
cells of the reticuloendothelial system such as macrophages and neutrophils; adsorption to the
cell surface, either by nonspecific weak hydrophobic or electrostatic forces, or by specific
interactions with cell-surface components; fusion with the plasma cell membrane by insertion of
the lipid bilayer of the liposome into the plasma membrane, with simultaneous release of
liposomal contents into the cytoplasm; and by transfer of liposomal lipids to cellular or
subcellular membranes, or vice versa, without any association of the liposome contents. It often is difficult to determine which mechanism is operative and more than one may operate at the
same time. The fate and disposition of intravenously injected liposomes depend on their physical
properties, such as size, fluidity, and surface charge. They may persist in tissues for h or days, depending on their composition, and half lives in the blood range from min to several h. Larger
liposomes, such as MLVs and LUVs, are taken up rapidly by phagocytic cells of the
reticuloendothelial system, but physiology of the circulatory system restrains the exit of such
large species at most sites. They can exit only in places where large openings or pores exist in the capillary endothelium, such as the sinusoids of the liver or spleen. Thus, these organs are
the predominate site of uptake. On the other hand, SUVs show a broader tissue distribution but still are sequestered highly in the liver and spleen. In general, this in vivo behavior limits the
potential targeting of liposomes to only those organs and tissues accessible to their large size. These include the blood, liver, spleen, bone marrow, and lymphoid organs.
Targeting is generally not a limitation in terms of the present invention. However, should specific targeting be desired, methods are available for this to be accomplished.
Antibodies may be used to bind to the liposome surface and to direct the antibody and its drag
contents to specific antigenic receptors located on a particular cell-type surface. Carbohydrate
determinants (glycoprotein or glycolipid cell-surface components that play a role in cell-cell
recognition, interaction and adhesion) may also be used as recognition sites as they have
potential in directing liposomes to particular cell types. Mostly, it is contemplated that
intravenous injection of liposomal preparations would be used, but other routes of
administration are also conceivable.
Alternatively, the invention provides for pharmaceutically acceptable nanocapsule
formulations of the AAV vector-based polynucleotide compositions of the present invention.
Nanocapsules can generally entrap compounds in a stable and reproducible way (Henry- Michelland etal, 1987; Quintanar-Guerrero et al, 1998; Douglas et al, 1987). To avoid side effects due to intracelrular polymeric overloading, such ulfrafine particles (sized around 0.1 μm) should be designed using polymers able to be degraded in vivo. Biodegradable polyalkyl- cyanoacrylate nanoparticles that meet these requirements are contemplated for use in the present 5 invention. Such particles may be are easily made, as described (Couvreur etal, 1980; Couvreur, 1988; zur Muhlen et al, 1998; Zambaux et al. 1998; Pinto-Alphandry et al, 1995 and U. S. Patent 5,145,684, specifically incorporated herein by reference in its entirety).
4.7 ADDITIONAL MODES OF DELIVERY0 In addition to the methods of delivery described above, the following techniques are also contemplated as alternative methods of delivering the disclosed rAAV vector based polynucleotide compositions to a target cell or animal. Sonophoresis (i.e., ultrasound) has been used and described in U. S. Patent 5,656,016 (specifically incorporated herein by reference in its entirety) as a device for enhancing the rate and efficacy of drag permeation into and through the5 circulatory system. Other drag delivery alternatives contemplated are intraosseous injection (U. S. Patent 5,779,708), microchip devices (U. S. Patent 5,797,898), ophthalmic formulations (Bourlais et al, 1998), transdermal matrices (U. S. Patent 5,770,219 and U. S. Patent 5,783,208) and feedback-controlled delivery (U. S. Patent 5,697,899), each specifically incorporated herein by reference in its entirety.0 4.8 THERAPEUTIC AND DIAGNOSTIC KITS The invention also encompasses one or more compositions together with one or more pharmaceutically-acceptable excipients, carriers, diluents, adjuvants, and/or other components, as may be employed in the formulation of particular rAAV-polynucleotide delivery
!5 formulations, and in the preparation of therapeutic agents for administration to a mammal, and in particularly, to a human, for one or more of the deficiencies, dysfunctions, or abnormalities described herein. In particular, such kits may comprise one or more of the disclosed microsphere-conjugated rAAV compositions in combination with instructions for using the viral vector in the treatment of such disorders in a mammal, and may typically further include containers prepared for convenient commercial packaging. As such, preferred animals for administration of the pharmaceutical compositions disclosed herein include mammals, and particularly humans. Other preferred animals include murines, bovines, equines, ovines, epines, porcines, canines, felines, and other animals of veterinary significance which may benefit from the therapeutic and/or prophylactic methods disclosed herein. The composition may include partially or significantly purified rAAV compositions, either alone, or in combination with one or more additional active ingredients, which may be obtained from natural or recombinant sources, or which may be obtainable naturally or either chemically synthesized, or alternatively produced in vitro from recombinant host cells expressing DNA segments encoding such additional active ingredients. Therapeutic kits may also be prepared that comprise at least one of the compositions disclosed herein and instructions for using the composition as a therapeutic agent. The container means for such kits may typically comprise at least one vial, test tube, flask, bottle, syringe or other container means, into which the disclosed rAAV composition(s) may be placed, and preferably suitably aliquoted. Where a second biologically-active therapeutic polypeptide composition is also provided, the kit may also contain a second distinct container means into which this second composition may be placed. Alternatively, the plurality of biologically-active therapeutic polypeptide compositions may be prepared in a single pharmaceutical composition, and may be packaged in a single container means, such as a vial, flask, syringe, bottle, or other suitable single container means. The kits of the present invention will also typically include a means for containing the vial(s) in close confinement for commercial sale, such as, e.g., injection or blow-molded plastic containers into which the desired vial(s) are retained.
4.9 METHODS OF NUCLEIC ACID DELIVERY AND DNA TRANSFECTION In some embodiments, it may be desirable to use other methods for the transfer of expression constructs into target mammalian cells. Some of these techniques may be successfully adapted for in vivo or ex vivo use, as discussed below. Likewise, in some applications, it may be desirable to transfer a naked DNA expression construct into cells using methods such as particle bombardment. This method depends on the ability to accelerate DNA coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them. Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical current, which in turn provides the motive force. The microprojectiles used have consisted of biologically inert substances such as tungsten or gold beads. In certain embodiments, it is contemplated that one or more polynucleotide compositions disclosed herein will be used to transfect an appropriate host cell. Technology for introduction of nucleic acids into cells is well known to those of skill in the art. These include calcium phosphate precipitation (Graham and Van Der Eb, 1973; Chen and Okayama, 1987; Rippe et al, 1990) DEAE-dexfran (Gopal, 1985), elecfroporation (Wong and Neumann, 1982; Fromm et al , 1985; Tur-Kaspa et al. , 1986; Potter et al. , 1984; Suzuki et al, 1998; Vanbever et al, 1998), direct microinj ection (Capecchi, 1980; Harland and Weinfraub, 1985), DNA-loaded liposomes (Nicolau and Sene, 1982; Fraley et al, 1979; Takakura, 1998) and lipofectamine- DNA complexes, cell sonication (Fechheimer et al, 1987), gene bombardment using high velocity microprojectiles (Yang et al, 1990; Klein et al, 1992), and receptor-mediated fransfection (Curiel et al, 1991; Wagner et al, 1992; Wu and Wu, 1987; Wu and Wu, 1988). Some of these techniques may be successfully adapted for in vivo or ex vivo use.
4.10 COMPLEMENTARITY AND HOMOLOGY OF POLYNUCLEOTIDE SEQUENCES The term "substantially complementary," when used to define either amino acid or
nucleic acid sequences, means that a particular subject sequence, for example, an nucleotide
sequence, is substantially complementary to all or a portion of the selected target sequence, and
thus will specifically bind to a portion of the selected target sequence, such as for example, in a
hybridization methodology. As such, typically the sequences will be highly complementary to the selected target sequence, and will have no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 base
mismatches throughout the complementary portion of the sequence (e.g., sequences that are
about 90% homologous, about 91% homologous, about 92% homologous, about 93% homologous, about 94% homologous, about 95% homologous, about 96% homologous, about
97% homologous, about 98% homologous, or about 99% homologous, etc.). In many instances, it may be desirable for the sequences to be exact matches, i.e. be completely complementary
(e.g., 100% homologous or 100% complementary) to the selected sequence to which the
nucleotide sequence specifically binds, and therefore have zero mismatches along the
complementary stretch. For example, in the case of oligonucleotides that bind to a specific
cellular mRNA (e.g., antisense molecules), highly complementary sequences will typically bind
quite specifically to the target sequence region of the candidate mRNA sequence and will
therefore be highly efficient in reducing, and/or even inhibiting the translation of the target
mRNA sequence into polypeptide product. Such methodologies are particularly preferred for
therapies in which it is desirable to reduce, alter, or eliminate expression of one or more
polypeptides in a selected cell through the introduction of one or more rAAV- vectored antisense
oligonucleotides. In such cases, substantially complementary oligonucleotide sequences will typically be greater than about 80 percent complementary (or '% exact-match') to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and will, more preferably be greater than about 85 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds. In certain aspects, as described above, it will be desirable to have even more substantially complementary oligonucleotide sequences for use in the practice of the invention, and in such instances, the oligonucleotide sequences will be greater than about 90 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and may in certain embodiments be greater than about 95 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and even up to and including 96%, 97%, 98%, 99%, and even 100% exact match complementary to all or a portion of the target mRNA to which the designed oligonucleotide specifically binds. Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG). The GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences. The preferred default parameters for the GAP program include:
(1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps. 4.11 PEPTIDE NUCLEIC ACID COMPOSITIONS In certain embodiments, the inventors contemplate the use of peptide nucleic acids (PNAs) in the practice of the methods of the invention. PNA is a DNA mimic in which the nucleobases are attached to a pseudopeptide backbone (Good and Nielsen, 1997). PNAs may be utilized in a number of methods that traditionally have used RNA or DNA. Often PNA sequences perform better in techniques than the corresponding RNA or DNA sequences and have utilities that are not inherent to RNA or DNA. An excellent review of PNA including methods of making, characteristics of, and methods of using, is provided by Corey (1997) and is incorporated herein by reference. As such, in certain embodiments, one may prepare PNA sequences that are complementary to one or more portions of the particular selected mRNA sequence, and such PNA compositions may be used to regulate, alter, decrease, or reduce the translation of the specific mRNA, and thereby alter the level of polypeptide encoded by the targeted mRNA in a host cell to which such PNA compositions have been administered. PNAs have 2-aminoethyl-glycine linkages replacing the normal phosphodiester backbone of DNA (Nielsen et al , 1993 ; Hanvey et al , 1992; Hyrup and Nielsen, 1996; Nielsen,
1995). This chemistry has three important consequences: firstly, in contrast to DNA or phosphorothioate oligonucleotides, PNAs are neutral molecules; secondly, PNAs are achiral, which avoids the need to develop a stereoselective synthesis; and thirdly, PNA synthesis uses standard Boc (Dueholm et al, 1992) or Fmoc (Bonham et al, 1995) protocols for solid-phase peptide synthesis, although other methods, including a modified Merrifield method, have been
used (Christensen et al, 1995).
PNA monomers or ready-made oligomers are commercially available from PerSeptive
Biosystems (Framingham, MA, USA). PNA syntheses by either Boc or Fmoc protocols are
straightforward using manual or automated protocols (Norton et al, 1995). 4.12 NUCLEIC ACID AMPLIFICATION AND SITE-SPECIFIC MUTAGENESIS
In certain embodiments, it may be desirable to prepared modified nucleotide compositions, such as, for example, in the generation of the nucleic acid segments that encode either parts of the AAV vector itself, or the promoter, or even the therapeutic gene delivered by
5 such rAAV vectors. Various means exist in the art, and are routinely employed by the artisan to generate modified nucleotide compositions.
Site-specific mutagenesis is a technique useful in the preparation of individual peptides, or biologically functional equivalent polypeptides, through specific mutagenesis of the underlying polynucleotides that encode them. The technique, well-known to those of skill in the 0 art, further provides a ready ability to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the DNA. Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a 5 primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Mutations may be employed in a selected polynucleotide sequence to improve, alter, decrease, modify, or change the properties of the polynucleotide itself, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide. 0 In certain embodiments of the present invention, the inventors contemplate the mutagenesis of the disclosed polynucleotide sequences to alter the activity or effectiveness of such viral vector constructs in a transformed host cell. Likewise in certain embodiments, the inventors contemplate the mutagenesis of the viral genome itself to facilitate improved infectivity, replication, stability, activity, or viral titers, as well as efficiency of transfection both
!5 in vitro and/or in vivo. h general, site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector or melting apart of two strands of a double-stranded vector which includes within its sequence a DNA sequence which encodes the desired polypeptide(s). An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to
DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected which include recombinant vectors bearing the mutated sequence arrangement. The preparation of sequence variants of the selected polynucleotide segments using site- directed mutagenesis is provided as a means of producing potentially useful species and is not meant to be limiting, as there are other ways in which sequence variants of polypeptides and the DNA sequences encoding them may be obtained. For example, recombinant vectors encoding a desired polypeptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants. Specific details regarding these methods and protocols are found in the teachings of Maloy etal, 1994; Segal, 1976; Prokop and Bajpai, 1991; Kuby, 1994; and Maniatis et al, 1982, each incorporated herein by reference, for that purpose. As used herein, the term "oligonucleotide directed mutagenesis procedure" refers to template-dependent processes and vector-mediated propagation that result in an increase in the concentration of a specific nucleic acid molecule relative to its initial concentration, or in an increase in tlie concentration of a detectable signal, such as amplification. As used herein, the term "oligonucleotide directed mutagenesis procedure" is intended to refer to a process that involves the template-dependent extension of a primer molecule. The term template dependent process refers to nucleic acid synthesis of an RNA or a DNA molecule wherein the sequence of
the newly synthesized strand of nucleic acid is dictated by the well-known rules of complementary base pairing (see, for example, Watson, 1987). Typically, vector mediated
methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector,
the clonal amplification of the vector, and the recovery of the amplified nucleic acid fragment.
Examples of such methodologies are provided by U. S. Patent 4,237,224, specifically
incorporated herein by reference in its entirety. Nucleic acids, used as a template for
amplification methods, may be isolated from cells according to standard methodologies (Sambrook et al, 1989). The nucleic acid may be genomic DNA or fractionated or whole cell
RNA. Where RNA is used, it may be desired to convert the RNA to a complementary DNA. In
one embodiment, the RNA is whole cell RNA and is used directly as the template for amplification.
Pairs of primers that selectively hybridize to nucleic acids corresponding to the ribozymes or conserved flanking regions are contacted with the isolated nucleic acid under
conditions that permit selective hybridization. The term "primer," as defined herein, is meant to
encompass any nucleic acid that is capable of priming the synthesis of a nascent nucleic acid in a template-dependent process. Typically, primers are oligonucleotides from ten to twenty base
pairs in length, but longer sequences can be employed. Primers may be provided in double-
stranded or single-stranded form, although the single-stranded form is preferred. Once hybridized, the nucleic acid:primer complex is contacted with one or more
enzymes that facilitate template-dependent nucleic acid synthesis. Multiple rounds of
amplification, also referred to as "cycles," are conducted until a sufficient amount of
amplification product is produced.
Next, the amplification product is detected. In certain applications, the detection may be
performed by visual means. Alternatively, the detection may involve indirect identification of the product via chemiluminescence, radioactive scintigraphy of incorporated radiolabel or fluorescent label or even via a system using electrical or thermal impulse signals (Affymax technology). A number of template dependent processes are available to amplify the marker sequences present in a given template sample. One of the best-known amplification methods is the polymerase chain reaction (referred to as PCR™) which is described in detail in U. S.
Patent Nos. 4,683,195, 4,683,202 and 4,800,159, and each incorporated herein by reference in entirety. Briefly, in PCR™, two primer sequences are prepared that are complementary to regions on opposite complementary strands of the marker sequence. An excess of deoxynucleoside triphosphates is added to a reaction mixture along with a DNA polymerase, e.g., Tag polymerase. If the marker sequence is present in a sample, the primers will bind to the marker and the polymerase will cause the primers to be extended along the marker sequence by adding on nucleotides. By raising and lowering the temperature of the reaction mixture, the extended primers will dissociate from the marker to form reaction products, excess primers will bind to the marker and to the reaction products and the process is repeated. A reverse transcriptase PCR amplification procedure may be performed in order to quantify the amount of mRNA amplified. Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et al, 1989. Alternative methods for reverse transcription utilize thermostable, RNA-dependent DNA polymerases. These methods are described in WO 90/07641, filed December 21, 1990, incorporated herein by reference in its entirety. Polymerase chain reaction methodologies are well known in the art. Another method for amplification is the ligase chain reaction ("LCR"), disclosed in Eur. Pat. Appl. No. 320308, incorporated herein by reference in its entirety. In LCR, two complementary probe pairs are prepared, and in the presence of the target sequence, each pair will bind to opposite complementary strands of the target such that they abut. In the presence of a ligase, the two probe pairs will link to form a single unit. By temperature cycling, as in PCR, bound ligated units dissociate from the target and then serve as "target sequences" for ligation of excess probe pairs. U. S. Patent 4,883,750 describes a method similar to LCR for binding probe pairs to a target sequence.
Qbeta Replicase (QβR), described in Intl. Pat. Appl. Publ. No. PCT/US87/00880,
incorporated herein by reference, may also be used as still another amplification method in the present invention. In this method, a replicative sequence of RNA that has a region complementary to that of a target is added to a sample in the presence of an RNA polymerase. The polymerase will copy the replicative sequence that can then be detected. An isothermal amplification method, in which restriction endonucleases and ligases are used to achieve the amplification of target molecules that contain nucleotide 5'-[alpha-thio]- triphosphates in one strand of a restriction site may also be useful in the amplification of nucleic acids in the present invention. Strand Displacement Amplification (SD A), described in U. S. Patent Nos. 5,455,166,
5,648,211, 5,712,124 and 5,744,311, each incorporated herein by reference, is another method of carrying out isothermal amplification of nucleic acids which involves multiple rounds of strand displacement and synthesis, i.e., nick translation. A similar method, called Repair Chain Reaction (RCR), involves annealing several probes throughout a region targeted for amplification, followed by a repair reaction in which only two of the four bases are present. The other two bases can be added as biotinylated derivatives for easy detection. A similar approach is used in SDA. Target specific sequences can also be detected using a cyclic probe reaction (CPR). In CPR, a probe having 3' and 5' sequences of non-specific DNA and a middle sequence of specific RNA is hybridized to DNA that is present in a sample. Upon hybridization, the reaction is treated with RNase H, and the products of the probe identified as distinctive products that are released after digestion. The original template is annealed to another cycling probe and the reaction is repeated. Still another amplification methods described in Great Britain Patent 2202328, and in Intl. Pat. Appl. Publ. No. PCT/US89/01025, each of which is incorporated herein by reference in its entirety, may be used in accordance with the present invention. In the former application, "modified" primers are used in a PCR-like, template- and enzyme-dependent synthesis. The primers may be modified by labeling with a capture moiety (e.g. , biotin) and/or a detector moiety (e.g., enzyme). In the latter application, an excess of labeled probes is added to a sample. In the presence of the target sequence, the probe binds and is cleaved catalytically. After cleavage, the target sequence is released intact, available to be bound by excess probe.
Cleavage of the labeled probe signals the presence of the target sequence. Other nucleic acid amplification procedures include transcription-based amplification systems (TAS), including nucleic acid sequence based amplification (NASBA) and 3SR Gingeras et al, PCT Application WO 88/10315, incorporated herein by reference. In NASBA, the nucleic acids can be prepared for amplification by standard phenol/chloroform extraction, heat denaturation of a clinical sample, treatment with lysis buffer and minispin columns for isolation of DNA and RNA or guanidinium chloride extraction of RNA. These amplification techniques involve annealing a primer that has target specific sequences. Following polymerization, DNA/RNA hybrids are digested with RNase H while double stranded DNA molecules are heat denatured again. In either case the single stranded DNA is made fully double stranded by addition of second target specific primer, followed by polymerization. The double-stranded DNA molecules are then multiply transcribed by an RNA polymerase such as T7 or SP6. In an isothermal cyclic reaction, the RNA's are reverse transcribed into single stranded DNA, which is then converted to double-stranded DNA, and then transcribed once again with an RNA polymerase such as T7 or SP6. The resulting products, whether truncated or complete, indicate target specific sequences. Davey et al, Eur. Pat. Appl. No. 329822 (incorporated herein by reference in its entirety) disclose a nucleic acid amplification process involving cyclically synthesizing single- stranded RNA ("ssRNA"), ssDNA, and double-stranded DNA (dsDNA), which may be used in accordance with the present invention. The ssRNA is a template for a first primer oligonucleotide, which is elongated by reverse transcriptase (RNA-dependent DNA polymerase). The RNA is then removed from the resulting DNA:RNA duplex by the action of ribonuclease H (RNase H, an RNase specific for RNA in duplex with either DNA or RNA). The resultant ssDNA is a template for a second primer, which also includes the sequences of an
RNA polymerase promoter (exemplified by T7 RNA polymerase) 5' to its homology to the template. This primer is then extended by DNA polymerase (exemplified by the large "Klenow" fragment of E. coli DNA polymerase I), resulting in a double-stranded DNA ("dsDNA") molecule, having a sequence identical to that of the original RNA between the primers and having additionally, at one end, a promoter sequence. This promoter sequence can be used by the appropriate RNA polymerase to make many RNA copies of the DNA. These copies can then re-enter the cycle leading to very swift amplification. With proper choice of enzymes, this amplification can be done isothermally without addition of enzymes at each cycle. Because of the cyclical nature of this process, the starting sequence can be chosen to be in the form of either DNA or RNA. Miller et al, PCT Application WO 89/06700 (incorporated herein by reference in its entirety) disclose a nucleic acid sequence amplification scheme based on the hybridization of a promoter/primer sequence to a target single-stranded DNA ("ssDNA") followed by transcription of many RNA copies of the sequence. This scheme is not cyclic, i.e., new templates are not produced from the resultant RNA transcripts. Other amplification methods include "RACE" and "one-sided PCR" (Frohman, 1990 incorporated by reference). Methods based on ligation of two (or more) oligonucleotides in the presence of nucleic acid having the sequence of the resulting "di-oligonucleotide", thereby amplifying the di- oligonucleotide, may also be used in the amplification step of the present invention. Following any amplification, it may be desirable to separate the amplification product from the template and the excess primer for the purpose of determining whether specific amplification has occurred. In one embodiment, amplification products are separated by agarose, agarose-acrylamide or polyacrylamide gel electrophoresis using standard methods (Sambrook et al. , 1989). Alternatively, chromatographic techniques may be employed to effect separation. There are many kinds of chromatography which may be used in the present invention: adsorption, partition, ion exchange and molecular sieve, and many specialized techniques for using them including column, paper, thin-layer and gas chromatography. Amplification products must be visualized in order to confirm amplification of the marker sequences. One typical visualization method involves staining of a gel with ethidium bromide and visualization under UV light. Alternatively, if the amplification products are integrally labeled with radio- or fluorometrically-labeled nucleotides, the amplification products can then be exposed to x-ray film or visualized under the appropriate stimulating spectra, following separation. In one embodiment, visualization is achieved indirectly. Following separation of amplification products, a labeled, nucleic acid probe is brought into contact with the amplified marker sequence. The probe preferably is conjugated to a chromophore but may be radiolabeled. In another embodiment, the probe is conjugated to a binding partner, such as an antibody or biotin, and the other member of the binding pair carries a detectable moiety. In one embodiment, detection is by Southern blotting and hybridization with a labeled probe. The techniques involved in Southern blotting are well known to those of skill in the art and can be found in many standard books on molecular protocols (Sambrook etal, 1989). Briefly, amplification products are separated by gel electrophoresis. The gel is then contacted with a membrane, such as nitrocellulose, permitting transfer of the nucleic acid and non- covalent binding. Subsequently, the membrane is incubated with a chromophore-conjugated probe that is capable of hybridizing with a target amplification product. Detection is by exposure of the membrane to x-ray film or ion-emitting detection devices. One example of the foregoing is described in U. S. Patent 5,279,721, incorporated by reference herein, which discloses an apparatus and method for the automated electrophoresis and transfer of nucleic acids. The apparatus permits electrophoresis and blotting without external manipulation of the gel and is ideally suited to carrying out methods according to the present invention.
4.13 BIOLOGICAL FUNCTIONAL EQUIVALENTS Modification and changes may be made in the stracture of the polynucleotides and polypeptides of the present invention and still obtain a functional molecule that still possesses desirable characteristics. As mentioned above, it is often desirable to introduce one or more mutations into a specific polynucleotide and/or encoded polypeptide sequence. In certain circumstances, the resulting encoded polypeptide sequence is altered by this mutation, or in other cases, the sequence of the polypeptide is unchanged by one or more mutations in the encoding polynucleotide. When it is desirable to alter the amino acid sequence of a polypeptide to create an equivalent, or even an improved, second-generation molecule, the amino acid changes may be achieved by changing one or more of the codons of the encoding DNA sequence, according to Table 3. For example, certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the polynucleotide sequences disclosed herein, without appreciable loss of their biological utility or activity.
TABLE 3
Amino Acids Codons
Alanine Ala A GCA GCC GCG GCU Cysteine Cys C UGC UGU Aspartic acid Asp D GAC GAU Glutamic acid Glu E GAA GAG Phenylalanine Phe UUC UUU Glycine Gly G GGA GGC GGG GGU Histidine His H CAC CAU Isoleucine He I AUA AUC AUU Lysine Lys K AAA AAG Leucine Leu L UUA UUG CUA CUC CUG CUU Methionine Met M AUG Asparagine Asn N AAC AAU Proline Pro CCA CCC CCG CCU Glutamine Gin Q CAA CAG Arginine Arg R AGA AGG CGA CGC CGG CGU Serine Ser S AGC AGU UCA UCC UCG UCU Threonine Thr T ACA ACC ACG ACU Valine Val V GUA GUC GUG GUU Tryptophan Trp W UGG Tyrosine Tyr Y UAC UAU
In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporate herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary stracture of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like. Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics (Kyte and Doolittle, 1982), these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5). It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e. still obtain a biological functionally equivalent protein. In making such changes,
the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those that
are within +1 are particularly preferred, and those within +0.5 are even more particularly
preferred. It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U. S. Patent 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein. As detailed in U. S. Patent 4,554,101, the following hydrophilicity values have been
assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ± 1); glutamate
(+3.0 + 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4);
proline (-0.5 ± 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine
(-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4). It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein. In such changes, the substitution of amino acids whose hydrophilicity values are within
+2 is preferred, those that are within +1 are particularly preferred, and those within ±0.5 are
even more particularly preferred. As outlined above, amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions which take several of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
4.14 EXEMPLARY DEFINITIONS In accordance with the present invention, polynucleotides, nucleic acid segments, nucleic acid sequences, and the like, include, but are not limited to, DNAs (including and not limited to genomic or extragenomic DNAs), genes, peptide nucleic acids (PNAs) RNAs
(including, but not limited to, rRNAs, mRNAs and tRNAs), nucleosides, and suitable nucleic acid segments either obtained from native sources, chemically synthesized, modified, or otherwise prepared in whole or in part by the hand of man. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and compositions similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and compositions are described herein. For purposes of the present invention, the following terms are defined below: A, an: In accordance with long standing patent law convention, the words "a" and "an" when used in this application, including the claims, denotes "one or more". Expression: The combination of intracellular processes, including transcription and translation undergone by a polynucleotide such as a stmctural gene to synthesize the encoded peptide or polypeptide. Promoter: a term used to generally describe the region or regions of a nucleic acid sequence that regulates transcription. Regulatory Element: a term used to generally describe the region or regions of a nucleic acid sequence that regulates transcription. Structural gene: A gene or sequence region that is expressed to produce an encoded peptide or polypeptide. Transformation: A process of introducing an exogenous polynucleotide sequence (e.g. , a vector, a recombinant DNA or RNA molecule) into a host cell or protoplast in which that exogenous nucleic acid segment is incorporated into at least a first chromosome or is capable of autonomous replication within the transformed host cell. Transfection, elecfroporation, and naked nucleic acid uptake all represent examples of techniques used to transform a host cell with one or more polynucleotides. Transformed cell: A host cell whose nucleic acid complement has been altered by the introduction of one or more exogenous polynucleotides into that cell. Transgenic cell: Any cell derived or regenerated from a transformed cell or derived from a transgenic cell, or from the progeny or offspring of any generation of such a transformed host cell. Vector: A nucleic acid molecule (typically comprised of DNA) capable of replication in a host cell and/or to which another nucleic acid segment can be operatively linked so as to bring about replication of the attached segment. A plasmid, cosmid, or a virus is an exemplary vector. The terms "substantially corresponds to", "substantially homologous", or "substantial identity" as used herein denotes a characteristic of a nucleic acid or an amino acid sequence, wherein a selected nucleic acid or amino acid sequence has at least about 70 or about 75 percent sequence identity as compared to a selected reference nucleic acid or amino acid sequence. More typically, the selected sequence and the reference sequence will have at least about 76, 77, 78, 79, 80, 81, 82, 83, 84 or even 85 percent sequence identity, and more preferably at least about 86, 87, 88, 89, 90, 91, 92, 93, 94, or 95 percent sequence identity. More preferably still, highly homologous sequences often share greater than at least about 96,
97, 98, or 99 percent sequence identity between the selected sequence and the reference sequence to which it was compared. The percentage of sequence identity may be calculated over the entire length of the sequences to be compared, or may be calculated by excluding small deletions or additions which total less than about 25 percent or so of the chosen reference sequence. The reference sequence may be a subset of a larger sequence, such as a portion of a gene or flanking sequence, or a repetitive portion of a chromosome. However, in the case of sequence homology of two or more polynucleotide sequences, the reference sequence will typically comprise at least about 18-25 nucleotides, more typically at least about 26 to 35 nucleotides, and even more typically at least about 40, 50, 60, 70, 80, 90, or even 100 or so nucleotides. Desirably, which highly homologous fragments are desired, the extent of percent identity between the two sequences will be at least about 80%, preferably at least about 85%, and more preferably about 90% or 95% or higher, as readily determined by one or more of the sequence comparison algorithms well-known to those of skill in the art, such as e.g., the FASTA program analysis described by Pearson and Lipman (1988). The term "naturally occurring" as used herein as applied to an object refers to the fact that an object can be found in nature. For example, a polypeptide or polynucleotide sequence that is present in an organism (including vimses) that can be isolated from a source in nature and which has not been intentionally modified by the hand of man in a laboratory is naturally-occurring. As used herein, laboratory strains of rodents that may have been selectively bred according to classical genetics are considered naturally occurring animals. As used herein, a "heterologous" is defined in relation to a predetermined referenced gene sequence. For example, with respect to a stmctural gene sequence, a heterologous promoter is defined as a promoter which does not naturally occur adjacent to the referenced stmctural gene, but which is positioned by laboratory manipulation. Likewise, a heterologous gene or nucleic acid segment is defined as a gene or segment that does not naturally occur adjacent to the referenced promoter and/or enhancer elements. "Transcriptional regulatory element" refers to a polynucleotide sequence that activates transcription alone or in combination with one or more other nucleic acid sequences. A transcriptional regulatory element can, for example, comprise one or more promoters, one or more response elements, one or more negative regulatory elements, and/or one or more enhancers. As used herein, a "transcription factor recognition site" and a "transcription factor binding site" refer to a polynucleotide sequence(s) or sequence motif(s) which are identified as being sites for the sequence-specific interaction of one or more transcription factors, frequently taking the form of direct protein-DNA binding. Typically, transcription factor binding sites can be identified by DNA footprinting, gel mobility shift assays, and the like, and/or can be predicted on the basis of known consensus sequence motifs, or by other methods known to those of skill in the art. As used herein, the term "operably linked" refers to a linkage of two or more polynucleotides or two or more nucleic acid sequences in a functional relationship. A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the coding sequence. Operably linked means that the DNA sequences being linked are typically contiguous and, where necessary to join two protein coding regions, contiguous and in reading frame. However, since enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not contiguous. "Transcriptional unit" refers to a polynucleotide sequence that comprises at least a first stmctural gene operably linked to at least a first cw-acting promoter sequence and optionally linked operably to one or more other -acting nucleic acid sequences necessary for efficient transcription of the stmctural gene sequences, and at least a first distal regulatory element as may be required for the appropriate tissue-specific and developmental franscription of the stmctural gene sequence operably positioned under the confrol of the promoter and/or enhancer elements, as well as any additional cis sequences that are necessary for efficient transcription and translation (e.g., polyadenylation site(s), mRNA stability controlling sequence(s), etc. The term "substantially complementary," when used to define either amino acid or nucleic acid sequences, means that a particular subject sequence, for example, an oligonucleotide sequence, is substantially complementary to all or a portion of the selected sequence, and thus will specifically bind to a portion of an mRNA encoding the selected sequence. As such, typically the sequences will be highly complementary to the mRNA "target" sequence, and will have no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 base mismatches throughout the complementary portion of the sequence. In many instances, it may be desirable for the sequences to be exact matches, i.e. be completely complementary to the sequence to which the oligonucleotide specifically binds, and therefore have zero mismatches along the complementary stretch. As such, highly complementary sequences will typically bind quite specifically to the target sequence region of the mRNA and will therefore be highly efficient in reducing, and/or even inhibiting the translation of the target mRNA sequence into polypeptide product. Substantially complementary oligonucleotide sequences will be greater than about 80 percent complementary (or '% exact-match') to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and will, more preferably be greater than about 85 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds. In certain aspects, as described above, it will be desirable to have even more substantially complementary oligonucleotide sequences for use in the practice of the invention, and in such instances, the oligonucleotide sequences will be greater than about 90 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and may in certain embodiments be greater than about 95 percent complementary to the corresponding mRNA target sequence to which the oligonucleotide specifically binds, and even up to and including 96%, 97%, 98%, 99%, and even 100% exact match complementary to all or a portion of the target mRNA to which the designed oligonucleotide specifically binds. Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG). The GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences. The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps. As used herein, the terms "gene transfer" and "gene delivery" refer to methods or systems for reliably inserting a particular polynucleotide sequence (e.g., a nucleic acid segment DNA) into targeted cells. In particularly preferred embodiments, the nucleotide sequence comprises at least a portion of biologically active Factor VII. As used herein, the terms "vector," and "gene transfer vector" refer to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, vims, virion, etc., which is capable of replication when associated with the proper control sequences and/or which can transfer nucleic acid sequences between cells. Thus, the term includes cloning and expression vectors, as well as viral vectors. As used herein, the terms "host" and "expression host" refer to organisms and/or cells which harbor an exogenous DNA sequence (e.g., via transfection), an expression vector or vehicle, as well as organisms and/or cells that are suitable for use in expressing a recombinant gene or protein. It is not intended that the present invention be limited to any particular type of cell or organism. Indeed, it is contemplated that any suitable organism and/or cell will find use in the present invention as a host. As used herein, the terms "viral replicons" and "viral origins of replication" refer to viral DNA sequences that allow for the extrachromosomal replication of a vector in a host cell expressing the appropriate replication factors. In some embodiments, vectors which contain either the SV40 or polyoma virus origin of replication replicate to high copy number, while vectors which contain the replicons from bovine papillomaviras or Epstein-Barr virus replicate extrachromosomally at low copy number may be utilized in other embodiments. As used herein, the term "wild type" ("wt") refers to a gene or gene product which has the characteristics of that gene or gene product when isolated from a naturally occurring source. A wild-type gene is that which is most frequently observed in a population and is thus arbitrarily designed the "normal" or "wild-type" form of the gene. In contrast, the term "modified" or "mutant" refers to a gene or gene product which displays modifications in sequence and or functional properties (i.e., altered characteristics) when compared to the wild-type gene or gene product. It is noted that naturally-occurring mutants can be isolated; these are identified by the fact that they have altered characteristics when compared to the wild-type gene or gene product. The term "heterologous" as it relates to nucleic acid sequences such as coding sequences and control sequences, denotes sequences that are not normally joined together, and/or are not normally associated with a particular cell. Thus, a "heterologous" region of a nucleic acid construct or a vector is a segment of nucleic acid within or attached to another nucleic acid molecule that is not found in association with the other molecule in nature. For example, a heterologous region of a nucleic acid constmct could include a coding sequence flanked by sequences not found in association with the coding sequence in nature. Another example of a heterologous coding sequence is a construct where the coding sequence itself is not found in nature (e.g., synthetic sequences having codons different from the native gene).
Similarly, a cell transfected with a construct which is not normally present in the cell would
be considered heterologous for purposes of this invention. Allelic variation or naturally
occurring mutational events do not give rise to heterologous DNA, as used herein.
As used herein, "coding sequence" or a sequence which "encodes" a particular
antigen, is a nucleic acid sequence which is transcribed (in the case of DNA) and translated (in the case of mRNA) into a polypeptide in vitro or in vivo, when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a franslation stop codon at the 3' (carboxy) terminus. A coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and even synthetic DNA sequences. A transcription termination sequence will usually be located 3' to the coding sequence. The term DNA "control sequences" refers collectively to regulatory elements such as promoter sequences, polyadenylation signals, transcription termination sequences, upstream regulatory domains, origins of replication, internal ribosome entry sites ("IRES"), enhancers, and the like, which collectively provide for the replication, transcription and translation of a coding sequence in a recipient cell. Not all of these control sequences need always be present so long as the selected coding sequence is capable of being replicated, transcribed and translated in an appropriate recipient cell. "Operably linked" or "operatively linked" refers to an arrangement of elements wherein the components so described are configured so as to perform their usual function. Thus, control sequences operably linked to a coding sequence are capable of effecting the expression of the coding sequence. The control sequences need not be contiguous with the coding sequence, so long as they function to direct the expression thereof. Thus, for example, intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered "operably linked" or "operatively linked" to the coding sequence. As used herein, the term "isolated" when used in relation to a nucleic acid, as in "an isolated oligonucleotide" or "isolated polynucleotide" refers to a nucleic acid sequence that is identified and separated from at least one contaminant nucleic acid with which it is ordinarily associated in its natural source. Isolated nucleic acid is such present in a form or setting that is different from that in which it is found in nature. In contrast, non-isolated nucleic acids are nucleic acids such as DNA and RNA found in the state they exist in nature. For example, a given DNA sequence (e.g., a gene) is found on the host cell chromosome in proximity to neighboring genes; RNA sequences, such as a specific mRNA sequence encoding a specific protein, are found in the cell as a mixture with numerous other mRNAs which encode a multitude of proteins. The isolated nucleic acid, oligonucleotide, or polynucleotide may be present in single-stranded or double-stranded form. When an isolated nucleic acid, oligonucleotide or polynucleotide is to be utilized to express a protein, the oligonucleotide or polynucleotide will contain at a minimum the sense or coding strand (i.e., the oligonucleotide or polynucleotide may single-stranded), but may contain both the sense and anti-sense strands (i.e., the oligonucleotide or polynucleotide may be double-stranded). As used herein, the term "purified" or "to purify" refers to the removal of contaminants from a sample. For example, antibodies may be purified by removal of contaminating non-immunoglobulin proteins; they may also purified by the removal of immunoglobulin that does not bind the antigen of interest (e.g., at least a portion of Factor VII). The removal of non-immunoglobulin proteins and/or the removal of immunoglobulins that do not bind the antigen of interest (e.g., at least a portion of Factor VII) results in an increase in the percent of desired antigen-reactive immunoglobulins in the sample. In another example, recombinant polypeptides of Factor VII are expressed in bacterial host cells and the polypeptides are purified by the removal of host cell proteins; the percent of recombinant polypeptides is thereby increased in the sample. A "composition comprising a given polynucleotide sequence" as used herein refers broadly to any composition containing the given polynucleotide sequence. The composition may comprise an aqueous solution. As used herein, the term "at risk" is used in references to individuals who are at risk for experiencing hemorrhagic episodes. In particularly preferred embodiments, the individuals are hemophiliacs with mild, moderate, or severe hemophilia. As used herein, the term "subject" refers to any animal (i.e., vertebrates and invertebrates), while the term "vertebrate subject" refers to any member of the subphylum
Chordata. It is intended that the term encompass any member of this subphylum, including, but not limited to humans and other primates, rodents (e.g., mice, rats, and guinea pigs), lagomorphs (e.g., pikas, hares, rabbits), bovines (e.g., cattle), ovines (e.g., sheep), caprines
(e.g., goats), porcines (e.g., swine), equines (e.g., horses), canines (e.g., dogs, wolves), felines (e.g., lions, tigers, cheetahs, domestic cats), domestic fowl (e.g., chickens, turkeys, ducks, geese, other gallinaceous birds, etc.), as well as feral or wild animals, including, but not limited to, such animals as ungulates, including members of the Artiodactyla (e.g., antelope, deer, etc.) and the Perissodactyla (e.g., rhinoceros, tapir, horse, zebra, etc.), etc. It is not intended that the term be limited to a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are encompassed by the term. As defined herein, a "therapeutically effective amount" or "therapeutic effective dose" is an amount or dose of AAV vectors or AAV virions capable of producing sufficient amounts of Factor VII to decrease the time it takes for a subject's blood to clot.
5. EXAMPLES The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
5.1 EXAMPLE 1 - RAAV COMPOSITIONS EXPRESSING FACTOR VII POLYPEPTIDES Replacement therapy using plasma-derived concentrates or recombinant factor VIII administered after bleeding episodes is the current mode of therapy for hemophilia A.
Although prophylaxis with protein has been shown to significantly reduce spontaneous bleeds, freatment efficacy is limited by the short half-life of FVIII in vivo, high production costs, repeated intravenous administrations, and development of host antibodies to the therapeutic protein. Gene therapy approaches may circumvent these problems, and as little as 2% of normal FVIII activity can result in therapeutic effects (Connelly et al, 1996; Kay and High, 1999). Adeno-associated virus 2 (AAV)-based vectors have emerged at the forefront of gene therapy. Previously, AAV has been used effectively for the treatment of factor IX (FIX) deficiency in both murine and canine models (Koeberl et al, 1997; Snyder et al, 1997; Chao et al, 1999; Herzog et al, 1999; Hagsfrom et al, 2000), leading to current phase I clinical trials. The present example describes the use of a recombinant AAV-based vector system for the treatment of a clinically relevant murine model of hemophilia A. This study demonstrates expression of Factor VII polypeptide from rAAV vectors expressing the FVII gene, administered either intravenously or intramuscularly.
5.1.2 METHODS FOR PREPARING RAAV VECTORS rAAV is most often produced by co-transfection of rAAV vector plasmid and wt AAV helper plasmid into Ad-infected 293 cells (Hermonat and Muzyczka, 1984). Recent improvements in AAV helper design (Li et al, 1997) as well as construction of non-infectious mini-Ad plasmid helper (Grimm et al, 1998; Xiao et al, 1998; Salvetti, 1998) have eliminated the need for Ad infection, and made it possible to increase the yield of rAAV up to 105 particles per fransfected cell in a cmde lysate. Scalable methods of rAAV production that do not rely on DNA transfection have also been developed (Cbiorini et al, 1995; frioue and Russell, 1998;
Clark et al, 1995). These methods, which generally involve the construction of producer cell lines and helper virus infection, are suitable for high- volume production. The conventional protocol for downstream purification of rAAV involves the stepwise precipitation of rAAV using ammonium sulfate, followed by two or preferably, three rounds of CsCl density gradient centrifugation. Each round of CsCl centrifugation involves fractionation of the gradient and probing fractions for rAAV by dot-blot hybridization or by PCR™ analysis. The AAV vectors and rAAV virions of the present invention can be produced using standard methodology known to those of skill in the art. Such methods typically involve one or more steps such as: (a) introducing an AAV vector into an appropriate mammalian host cell; (b) introducing an AAV helper construct into the host cell, where the helper construct includes AAV coding regions capable of being expressed in the host cell to complement AAV helper functions missing from the AAV vector; (c) introducing one or more helper viruses and/or accessory function vectors into the host cell, wherein the helper virus and/or accessory function vectors provide accessory functions capable of supporting efficient recombinant AAV ("rAAV") virion production in the host cell; and (d) culturing the host cell to produce rAAV virions. The AAV vector, AAV helper construct and the helper vims or accessory function vector(s) can be introduced into the host cell either simultaneously or serially, using standard transfection techniques. 5.1.3 INCORPORATION OF RAAV VECTORS INTO CELLS In various embodiments of the invention, DNA may be delivered to a suitable mammalian host cell as an expression vector construct. Preferred gene therapy vectors of the present invention are generally viral vectors. Adeno-associated virus (AAV) is particularly attractive for gene transfer because it does not induce any pathogenic response and can integrate into the host cellular chromosome (Kotin et al, 1990). The AAV terminal repeats (TRs) are the only essential cw-components for the chromosomal integration (Muzyczka and McLaughin, 1988). These TRs are reported to have promoter activity (Flotte et al, 1993). They may promote efficient gene transfer from the cytoplasm to the nucleus or increase the stability of plasmid DNA and enable longer-lasting gene expression. Studies using recombinant plasmid
DNAs containing AAV TRs have attracted considerable interest. AAV-based plasmids have been shown to drive higher and longer fransgene expression than the identical plasmids lacking the TRs of AAV in most cell types (Shafron et al, 1998). AAV (Ridgeway, 1988; Hermonat and Muzyczka, 1984) is a paroviras, discovered as a contamination of adenoviral stocks. It is a ubiquitous virus (antibodies are present in 85% of the
U. S. human population) that has not been linked to any disease. It is also classified as a dependovims, because its replication is dependent on the presence of a helper vims, such as adenovirus. There are several factors that prompted researchers to study the possibility of using rAAV as an expression vector. One is that the requirements for delivering a gene to integrate into the host chromosome are surprisingly few. It is necessary to have the 145-bp ITRs, which are only 6% of the AAV genome. This leaves room in the vector to assemble a 4.5-kb DNA insertion. AAV is also a good choice of delivery vehicles due to its safety. There is a relatively complicated rescue mechanism: not only wild type adenovirus but also AAV genes are required to mobilize rAAV. Likewise, AAV is not pathogenic and not associated with any disease. The removal of viral coding sequences minimizes immune reactions to viral gene expression, and therefore, rAAV does not evoke an inflammatory response. AAV therefore, represents an ideal candidate for delivery of the present anti-hemophilia constructs. Refrovimses have promise as gene delivery vectors due to their ability to integrate their genes into the host genome, transferring a large amount of foreign genetic material, infecting a broad spectrum of species and cell types and of being packaged in special cell-lines. Of course, in using viral delivery systems, one may desire to purify the virion sufficiently to render it essentially free of undesirable contaminants, such as defective interfering viral particles or endotoxins and other pyrogens such that it will not cause any untoward reactions in the cell, animal or individual receiving the vector construct. One preferred means of purifying the vector involves the use of buoyant density gradients, such as cesium chloride gradient centrifugation, heparin affinity chromatography (Clark et al, 1999), or non-ionic iodixinol gradients followed by heparin affinity chromatography (Zolotukhin et al, 1999). The titer of AAV in a given sample may be determined using any one of the methods routinely accepted in the AAV arts. For example, the inventors routinely use the methods of
QC-PCR™ or infectious center assay, as described in detail in the Examples and by Zolotul in
et al. (1999), to determine the titer of a viral stock. Likewise, the infectivity of a given AAV sample may be determined using any one of the methods routinely accepted in the AAV arts. For example, the inventors routinely use the methods of Hermonat and Muzyczka (1984) or Clark et al. (1999) to determine the infectivity of a given AAV stock. 5.1.4 MATERIALS AND METHODS
5.1.4.1 CELLS, PLASMIDS, AND VIRUSES Adenoviras-transformed human embryonic kidney cells (293 cells), the human hepatocellular carcinoma cell line Hep G2, the human cervical carcinoma cell line HeLa, and the mouse myoblast cell line C2C12 may be obtained from the American Type Culture
Collection (ATCC, Manassas, VA). Monolayer cells may be maintained at 37°C, 5% CO2 in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and antibiotics. The albumin promoter may be obtained from K. Zaret (Foxchase, Philadelphia, PA). A truncated hybrid cytomegalovirus immediate-early enhancer chicken b- actin (CBA) promoter may be generated by using _3sj_> 1201 to delete 700 bp from the 39 end of the promoter, according to standard cloning methods. rAAV plasmids pCMVp-/αcZ, UF5 (pCMVp-GFP), and UF11 (pCBAp-GFP) have been described previously (Kessler et al., 1996; Klein et al, 1998). rAAV plasmids containing the appropriate DNA sequence encoding the desired mammalian Factor VII polypeptide may be generated by standard cloning methods. The splice donor/acceptor (SD and SA, respectively) elements in pCBA-
HC-SD and pSALC may be cloned from the intron region of the pCI cloning vector (Stratagene, La Jolla, CA). rAAV vectors may be generated, purified, and titered at the University of Florida (Gainesville, FL) Vector Core Laboratory as previously described (Zolotukhin et al, 1999).
5.1.4.2 IN VITRO TRANSFECTION AND TRANSDUCTION In vitro fransfections may be performed by the calcium phosphate-DNA coprecipitation method as described by Sambrook et al. (1989). For in vitro infections, cells may be infected with the Factor VII vectors at the indicated multiplicities of infection (MOIs) and coinfected with wild-type adenovirus type 5 at an MOI of 1. Forty-eight hours posttransfection or infection, conditioned medium may be assayed for the presence of functional FVII by the Coatest kit assay (Chromogenix, Milan, Italy).
For the cotransduction studies, HeLa or 293 cells may be infected with the indicated vectors at the indicated MOIs. In addition, HeLa cells, may also be coinfected with wild-type adenovirus type 5 at an MOI of 1. HeLa cells (48 hr postinfection) and 293 cells (72 hr postinfection) may be subjected to fluorescence-activated cell sorting (FACS) analysis at a suitable facility, such as the University of Florida Interdisciplinary Center for Biotechnology Research (ICBR) Flow Cytometry Core (FACScan; BD Immunocytometry Systems, San Jose, CA). /αcZ-positive cells may be identified with an ImaGene Red C12RG lacZ gene expression kit, as per the kit protocol (Molecular Probes,
Eugene, OR).
5.1.4.3 ANIMALS A suitable model of hemophilia A Factor VII deficiency, such as for example a mouse FVII knockout [KO] mouse may be utilized in the practice of the present invention.
One-day-old FVII KO mice may be administered a suitable concentration of infectious particles (IP) (for example about 2.4 x 108) via the superficial temporal vein as described by Sands and Barker (1999). Seven-month-old FVII KO mice may also be administered a suitable concentration of IP via intramuscular injection into the gastrocnemius of the hind leg. Beginning, for example, about 4 weeks postinj ection, plasma may then be collected from tail vein bleeds and functional mFVII activity determined by a suitable Factor VII assay, such as the Coatest assay. Plasma samples are obtained by tail bleeding of anesthetized animals followed by the immediate addition of sodium citrate to a final concentration of about 0.38% (wt./vol.). Samples are centrifuged at 2000 3 g for 10 min at 25°C. The plasma fraction is collected and frozen immediately. Samples are thawed quickly at 37°C immediately before testing. Serial dilutions of pooled normal C57BL/6 mouse plasma diluted in pooled FVII KO mouse plasma is then used to derive the standard curve. Inhibitor formation is detected by Bethesda assay as described previously, with the following modifications (Kasper et ah, 1975). Treated mouse plasma is mixed with an equivalent volume of pooled normal C57BL/6 mouse plasma and incubated at 37°C for 2 hr. The residual mFVII activity of each sample is plotted on the established Bethesda Inhibitor Assay standard curve to determine the anti-mFVII inhibitor titer.
5.1.4.4 IMMUNOHISTOCHEMISTRY Tissue samples are fixed in 10% buffered formalin, embedded, and sectioned (4-mm thickness). Sections are immunostained with a suitable anti-mFVII antibody, the mouse-on- mouse (M.O.M.) immunostaining kit (Vector Laboratories, Burlingame, CA), and 3,39- diaminobenzidine (DAB), and then counterstained with Gill's hematoxylin (Vector Laboratories). Photographs are taken with a light microscope (Zeiss, Thomwood, NY), camera (Olympus America, Melville, NY) and MagnaFire digital recording system (Pro
Image Digital, Teddington, Middlesex, UK). Relative transduction efficiency is determined as described by Nakai et al. (2002). Briefly, the number of positively stained cells and total number of cells are counted. Ten fields of approximately 500 nuclei per field are counted for each section.
5.1.4.5 WESTERN BLOT AND SEMIQUANTITATIVE WESTERN BLOT Plasma samples are subjected to cryoprecipitation as described (Bi et al, 1996).
Cryoprecipitate samples were analyzed by the Laemmli gel method, using precast 8-16%
Tris-glycine sodium dodecyl sulfate (SDS)-polyacrylamide minigels (Invitrogen, Carlsbad, CA). Western transfer is performed with a Novex Western blot module (Invitrogen) and Hybond ECL nitrocellulose (Amersham Pharmacia Biotech, Piscataway, NJ). Blots are hybridized with a suitable anti-FVII antibody (Bi et al, 1996; Sarkar et al, 2000) (diluted 1 :50) and horseradish peroxidase (HRP)-conjugated secondary antibody (diluted 1:1000) (Amersham Pharmacia Biotech). Hybridization is detected with the ECL Plus Western blotting detection system (Amersham Pharmacia Biotech). For semiquantitative Western blot, equivalent amounts of treated animal plasma or standard samples containing 10, 25, 50, 75, and 100% normal C57BL/6 plasma is subjected to cryoprecipitation and analyzed by the Laemmli gel method, using precast 8% Tris-glycine SDS-polyacrylamide minigels (Invitrogen). Western transfer and hybridization visualization is performed as described above. Autoradiographs are scanned with an Astra 6450 scanner (UMAX Technologies,
Dallas, TX) and optical density is determined with Scion Image Release Beta 4.0.2 software (Scion, Frederick, MD). The standard curve is determined by plotting optical density versus percent normal FVII.
5.1.2 RESULTS FIG. 1 shows that functional Factor VII can be produced in vitro. The chromogenic Coaset assay was used to determine the levels of functional Factor VII secreted. Factor VIII KO plasma was used as a positive control as these mice have been shown to produce normal levels of Factor VII.
5.2 EXAMPLE 2 - ILLUSTRATIVE THERAPEUTIC POLYPEPTIDE SEQUENCES USEFUL IN THE PRACTICE OF THE PRESENT INVENTION AND DNA SEQUENCES ENCODING SUCH AMINO Aero SEQUENCES 5.2.1 DNA SEQUENCE ENCODING HUMAN FACTOR VII FROM GENBANK
5 AF466933 (SEQ ID NO:l) TCAACAGGCAGGGGCAGCACTGCAGAGATTTCATCATGGTCTCCCAGGCCCTCAGGCTC CTCTGCCTTCTGCTTGGGCTTCAGGGCTGCCTGGCTGCAGGCGGGGTCGCTAAGGCCTC AGGAGGAGAAACACGGGACATGCCGTGGAAGCCGGGGCCTCACAGAGTCTTCGTAACCC AGGAGGAAGCCCACGGCGTCCTGCACCGGCGCCGGCGCGCCAACGCGTTCCTGGAGGAG0 CTGCGGCCGGGCTCCCTGGAGAGGGAGTGCAAGGAGGAGCAGTGCTCCTTCGAGGAGGC CCGGGAGATCTTCAAGGACGCGGAGAGGACGAAGCTGTTCTGGATTTCTTACAGTGATG GGGACCAGTGTGCCTCAAGTCCATGCCAGAATGGGGGCTCCTGCAAGGACCAGCTCCAG TCCTATATCTGCTTCTGCCTCCCTGCCTTCGAGGGCCGGAACTGTGAGACGCACAAGGA TGACCAGCTGATCTGTGTGAACGAGAACGGCGGCTGTGAGCAGTACTGCAGTGACCACA5 CGGGCACCAAGCGCTCCTGTCGGTGCCACGAGGGGTACTCTCTGCTGGCAGACGGGGTG TCCTGCACACCCACAGTTGAATATCCATGTGGAAAAATACCTATTCTAGAAAAAAGAAA TGCCAGCAAACCCCAAGGCCGAATTGTGGGGGGCAAGGTGTGCCCCAAAGGGGAGTGTC CATGGCAGGTCCTGTTGTTGGTGAATGGAGCTCAGTTGTGTGGGGGGACCCTGATCAAC ACCATCTGGGTGGTCTCCGCGGCCCACTGTTTCGACAAAATCAAGAACTGGAGGAACCT
!0 GATCGCGGTGCTGGGCGAGCACGACCTCAGCGAGCACGACGGGGATGAGCAGAGCCGGC GGGTGGCGCAGGTCATCATCCCCAGCACGTACGTCCCGGGCACCACCAACCACGACATC GCGCTGCTCCGCCTGCACCAGCCCGTGGTCCTCACTGACCATGTGGTGCCCCTCTGCCT GCCCGAACGGACGTTCTCTGAGAGGACGCTGGCCTTCGTGCGCTTCTCATTGGTCAGCG GCTGGGGCCAGCTGCTGGACCGTGGCGCCACGGCCCTGGAGCTCATGGTGCTCAACGTG
5 CCCCGGCTGATGACCCAGGACTGCCTGCAGCAGTCACGGAAGGTGGGAGACTCCCCAAA TATCACGGAGTACATGTTCTGTGCCGGCTACTCGGATGGCAGCAAGGACTCCTGCAAGG GGGACAGTGGAGGCCCACATGCCACCCACTACCGGGGCACGTGGTACCTGACGGGCATC GTCAGCTGGGGCCAGGGCTGCGCAACCGTGGGCCACTTTGGGGTGTACACCAGGGTCTC CCAGTACATCGAGTGGCTGCAAAAGCTCATGCGCTCAGAGCCACGCCCAGGAGTCCTCC TGCGAGCCCCATTTCCCTAGCCCA
5.2.2 AMINOACIDSEQUENCEOFHUMANFACTORVIIFROMGENBANK AF466933 (SEQIDNO:2)
MVSQALRLLCLLLGLQGCLAAGGVAKASGGETRDMP KPGPHRVFVTQEEAHGVLHRRR RANAFLEELRPGSLERECKEEQCSFEEAREIFKDAERTKLF ISYSDGDQCASSPCQNG GSCKDQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSCRCHEG YSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGRIVGGKVCPKGECPWQVLLLVNGAQ LCGGTLINTIWVVSAAHCFDKIKNWRNLIAVLGEHDLSEHDGDEQSRRVAQVIIPSTYV PGTTNHDIALLRLHQPVVLTDHVVPLCLPERTFSERTLAFVRFSLVSGWGQLLDRGATA LELMVLNVPRLMTQDCLQQSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYR GTWYLTGIVSWGQGCATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP (From Rieder et al . , unpublished)
5.2.3 DNASEQUENCEENCODINGHUMANFACTORVIIVARIANT1 FROMGENBANKNM_000131 (SEQIDNO:3) ATGGTCTCCCAGGCCCTCAGGCTCCTCTGCCTTCTGCTTGGGCTTCAGGGCTGCCTGGCTGC AGGCGGGGTCGCTAAGGCCTCAGGAGGAGAAACACGGGACATGCCGTGGAAGCCGGGGCCTC I
ACAGAGTCTTCGTAACCCAGGAGGAAGCCCACGGCGTCCTGCACCGGCGCCGGCGCGCCAAC
GCGTTCCTGGAGGAGCTGCGGCCGGGCTCCCTGGAGAGGGAGTGCAAGGAGGAGCAGTGCTC
CTTCGAGGAGGCCCGGGAGATCTTCAAGGACGCGGAGAGGACGAAGCTGTTCTGGATTTCTT ACAGTGATGGGGACCAGTGTGCCTCAAGTCCATGCCAGAATGGGGGCTCCTGCAAGGACCAG
CTCCAGTCCTATATCTGCTTCTGCCTCCCTGCCTTCGAGGGCCGGAACTGTGAGACGCACAA GGATGACCAGCTGATCTGTGTGAACGAGAACGGCGGCTGTGAGCAGTACTGCAGTGACCACA CGGGCACCAAGCGCTCCTGTCGGTGCCACGAGGGGTACTCTCTGCTGGCAGACGGGGTGTCC TGCACACCCACAGTTGAATATCCATGTGGAAAAATACCTATTCTAGAAAAAAGAAATGCCAG CAAACCCCAAGGCCGAATTGTGGGGGGCAAGGTGTGCCCCAAAGGGGAGTGTCCATGGCAGG TCCTGTTGTTGGTGAATGGAGCTCAGTTGTGTGGGGGGACCCTGATCAACACCATCTGGGTG GTCTCCGCGGCCCACTGTTTCGACAAAATCAAGAACTGGAGGAACCTGATCGCGGTGCTGGG CGAGCACGACCTCAGCGAGCACGACGGGGATGAGCAGAGCCGGCGGGTGGCGCAGGTCATCA TCCCCAGCACGTACGTCCCGGGCACCACCAACCACGACATCGCGCTGCTCCGCCTGCACCAG CCCGTGGTCCTCACTGACCATGTGGTGCCCCTCTGCCTGCCCGAACGGACGTTCTCTGAGAG GACGCTGGCCTTCGTGCGCTTCTCATTGGTCAGCGGCTGGGGCCAGCTGCTGGACCGTGGCG CCACGGCCCTGGAGCTCATGGTGCTCAACGTGCCCCGGCTGATGACCCAGGACTGCCTGCAG CAGTCACGGAAGGTGGGAGACTCCCCAAATATCACGGAGTACATGTTCTGTGCCGGCTACTC GGATGGCAGCAAGGACTCCTGCAAGGGGGACAGTGGAGGCCCACATGCCACCCACTACCGGG GCACGTGGTACCTGACGGGCATCGTCAGCTGGGGCCAGGGCTGCGCAACCGTGGGCCACTTT GGGGTGTACACCAGGGTCTCCCAGTACATCGAGTGGCTGCAAAAGCTCATGCGCTCAGAGCC ACGCCCAGGAGTCCTCCTGCGAGCCCCATTTCCCTAG
5.2.4 AMINO ACID SEQUENCE OF HUMAN FACTOR VII VARIANT 1 FROM GENBANK NM_000131 (SEQ ID NO:4) MVSQALRLLCLLLGLQGCLAAGGVAKASGGETRDMPWKPGPHRVFVTQEEAHGVLHRRR RANAFLEELRPGSLERECKEEQCSFEEAREIFKDAERTKLF ISYSDGDQCASSPCQNG GSCKDQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSCRCHEG YSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGRIVGGKVCPKGECP QVLLLVNGAQ LCGGTLINTIWVVSAAHCFDKIKNWRNLIAVLGEHDLSEHDGDEQSRRVAQVIIPSTYV PGTTNHDIALLRLHQPVVLTDHVVPLCLPERTFSERTLAFVRFSLVSGWGQLLDRGATA LELMVLNVPRLMTQDCLQQSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYR GTWYLTGIVSWGQGCATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP
(from Toso et al . , Biochem . J. , 369 (Pt 3):563-571, 2003). 5 5.2.5 DNA SEQUENCEENCODINGHUMANFACTORVIIVARIANT2 FROMGENBANKNM_019616 (SEQIDNO:5) ATGGTCTCCCAGGCCCTCAGGCTCCTCTGCCTTCTGCTTGGGCTTCAGGGCTGCCTGGC TGCAGTCTTCGTAACCCAGGAGGAAGCCCACGGCGTCCTGCACCGGCGCCGGCGCGCCA
10 ACGCGTTCCTGGAGGAGCTGCGGCCGGGCTCCCTGGAGAGGGAGTGCAAGGAGGAGCAG TGCTCCTTCGAGGAGGCCCGGGAGATCTTCAAGGACGCGGAGAGGACGAAGCTGTTCTG GATTTCTTACAGTGATGGGGACCAGTGTGCCTCAAGTCCATGCCAGAATGGGGGCTCCT GCAAGGACCAGCTCCAGTCCTATATCTGCTTCTGCCTCCCTGCCTTCGAGGGCCGGAAC TGTGAGACGCACAAGGATGACCAGCTGATCTGTGTGAACGAGAACGGCGGCTGTGAGCA
15 GTACTGCAGTGACCACACGGGCACCAAGCGCTCCTGTCGGTGCCACGAGGGGTACTCTC TGCTGGCAGACGGGGTGTCCTGCACACCCACAGTTGAATATCCATGTGGAAAAATACCT ATTCTAGAAAAAAGAAATGCCAGCAAACCCCAAGGCCGAATTGTGGGGGGCAAGGTGTG CCCCAAAGGGGAGTGTCCATGGCAGGTCCTGTTGTTGGTGAATGGAGCTCAGTTGTGTG GGGGGACCCTGATCAACACCATCTGGGTGGTCTCCGCGGCCCACTGTTTCGACAAAATC
»0 AAGAACTGGAGGAACCTGATCGCGGTGCTGGGCGAGCACGACCTCAGCGAGCACGACGG GGATGAGCAGAGCCGGCGGGTGGCGCAGGTCATCATCCCCAGCACGTACGTCCCGGGCA CCACCAACCACGACATCGCGCTGCTCCGCCTGCACCAGCCCGTGGTCCTCACTGACCAT GTGGTGCCCCTCTGCCTGCCCGAACGGACGTTCTCTGAGAGGACGCTGGCCTTCGTGCG CTTCTCATTGGTCAGCGGCTGGGGCCAGCTGCTGGACCGTGGCGCCACGGCCCTGGAGC
5 TCATGGTGCTCAACGTGCCCCGGCTGATGACCCAGGACTGCCTGCAGCAGTCACGGAAG GTGGGAGACTCCCCAAATATCACGGAGTACATGTTCTGTGCCGGCTACTCGGATGGCAG CAAGGACTCCTGCAAGGGGGACAGTGGAGGCCCACATGCCACCCACTACCGGGGCACGT GGTACCTGACGGGCATCGTCAGCTGGGGCCAGGGCTGCGCAACCGTGGGCCACTTTGGG GTGTACACCAGGGTCTCCCAGTACATCGAGTGGCTGCAAAAGCTCATGCGCTCAGAGCC ACGCCCAGGAGTCCTCCTGCGAGCCCCATTTCCCTAG 5 5.2.6 AMINO ACID SEQUENCE OF HUMAN FACTOR VII VARIANT 2 FROM GENBANK NM_019616 (SEQ ID NO:6) MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRRRRANAFLEELRPGSLERECKEEQ CSFEEAREIFKDAERTKLF ISYSDGDQCASSPCQNGGSCKDQLQSYICFCLPAFEGRN0 CETHKDDQLICVNENGGCEQYCSDHTGTKRSCRCHEGYSLLADGVSCTPTVEYPCGKIP ILEKRNASKPQGRIVGGKVCPKGECPWQVLLLVNGAQLCGGTLINTI VVSAAHCFDKI KN RNLIAVLGEHDLSEHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPVVLTDH VVPLCLPERTFSERTLAFVRFSLVSGWGQLLDRGATALELMVLNVPRLMTQDCLQQSRK VGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYRGT YLTGIVSWGQGCATVGHFG5 VYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP (from Toso et al . , Biochem . J. , 369 (Pt 3):563-571, 2003).
5.2.7 DNASEQUENCEENCODINGRATFACTORVIIFROMGENBANK NM_152846 (SEQIDNO:7)
»0 ATGGTTCCACAGACTCACGGACTGCTGCTTCTCTACTTTCTGCTCCAGCTCCAGGGACCCCT AGGGGCTGTGGTTTTCATAACCCAGGAGGAAGCACACGGTGTCCTACACAGGCAAAGGCGTG CCAACTCACTCCTAGAGGAGCTTTGGTCTAGCTCCTTGGAGAGGGAGTGCAATGAAGAGCGG TGCTCCTTTGAGGAGGCCCGAGAGATCTTCAAGAGCCCTGAGAGAACCAAGCAGTTCTGGAC TATTTACAGCGATGGCGACCAGTGTGCCTCGAATCCATGTCAGAACGGGGGTACCTGCCAGG
5 ATCACCTCAAGTCTTATGTCTGCTTCTGCCCCCTAGACTTTGAGGGCCGGAACTGTGAGAAA AACAAGAATGAGCAGCTGATCTGTGCAAATGAAAATGGTGACTGTGACCAGTACTGCAGGGA CCACGTAGGGACCAAGCGTACCTGTAGCTGTCACGAGGACTACGTGCTGCAGCCAGATGAGG TGTCCTGCAAACCAAAAGTTGAGTACCCATGCGGGAGAATACCTGTTGTAGAAAAAAGAAAC TTCAGCAGACCCCAAGGCCGGATTGTGGGAGGCTATGTGTGCCCCAAAGGGGAGTGCCCATG GCAGGCTGTGCTGAAATTCAATGAGGCATTGCTGTGTGGGGCCGTCCTGCTGGACACCAGAT 5 GGATAGTAACTGCAGCCCACTGCTTCGATAAATTCGGGAAATTGGTAAACATCACAGTGGTG TTGGGTGAACACGACTTCAGTGAGAAGGAGGGGACTGAGCAAGTACGGCTGGTGGAACAGGT CATCATGCCCAACAAGTACACCCGCGGCAGGACTGACCATGACATCGCCCTGGTCCGCCTTC ACCGGCCTGTAACCTTCACTGACTACGTGGTACCTCTGTGTCTGCCTGAACGGGCCTTCTCC GAGAACACCCTAGCCAGCATCCGCTTCTCGAGGGTCAGCGGCTGGGGCCAGCTACTGGACCG0 TGGTGCCACAGCTCTGGAGCTCATGGTCATCGAGGTGCCCCGGCTGATGACCCAGGACTGCC TGGAGCATGCCAAACACAGTGCTAACACCCCCAGAATCACGGAGAACATGTTCTGCGCCGGC TACATGGACGGCACCAAGGACGCCTGCAAGGGTGACAGTGGAGGCCCACACGCCACCCACTA CCATGGCACTTGGTATCTGACAGGTGTGGTCAGCTGGGGGGAGGGCTGTGCAGCTATCGGCC ACATCGGGGTGTACACCAGGGTCTCCCAGTACATAGACTGGCTGGTCAAATACATGGACTCC5 AAGCTCCGGGTTGGGATTTCTCGAGTCTCCCTACTGTAG
5.2.8 AMINO ACID SEQUENCE OF RAT FACTOR VII FROM GENBANK NM_152846 (SEQ ID NO:8) MVPQTHGLLLLYFLLQLQGPLGAVVFITQEEAHGVLHRQRRANSLLEELWSSSLERECNEER
!0 CSFEEAREIFKSPERTKQFWTIYSDGDQCASNPCQNGGTCQDHLKSYVCFCPLDFEGRNCEK NKNEQLICANENGDCDQYCRDHVGTKRTCSCHEDYVLQPDEVSCKPKVEYPCGRIPVVEKRN FSRPQGRIVGGYVCPKGECP QAVLKFNEALLCGAVLLDTRWIVTAAHCFDKFGKLVNITVV LGEHDFSEKEGTEQVRLVEQVIMPNKYTRGRTDHDIALVRLHRPVTFTDYVVPLCLPERAFS ENTLASIRFSRVSGWGQLLDRGATALELMVIEVPRLMTQDCLEHAKHSANTPRITENMFCAG
5 YMDGTKDACKGDSGGPHATHYHGT YLTGVVSWGEGCAAIGHIGVYTRVSQYID LVKYMDS KLRVGISRVSLL 5.2.9 DNA SEQUENCE ENCODING DANIO FACTOR VII FROM GENBANK NMJ31819 (SEQ ID NO:9)
ATGAGTCTGCTGCTTGTGTTTTCTCTGCTCTGGAGTCTCCATTACTGCCATTCAGCAGCAGT GTTCGTGCACAGAGATGAAGCTCACGAGGTGTTGATCAGGAGCAAAAGAGCCAACTCAGGCT GGTTTGAGGAGCTGAAGACGGGGAATCTGGAGCGCGAGTGTCTGGAGGAGAAATGCTCGTAT GAGGAGGCGCGCGAGGTGTTCGAGCACACAGAGGCCACGAATGAGTTCTGGAAGATCTACGA TGTTAAAGATCACTGCGCATCCAGTCCATGTGAGCATGACGGGCTCTGCACCACACAGAACG CGGACTCCTACATGTGTTTGTGTGCGCCGGGCTTCAGCGGACGCCACTGTGAGCAATCGATT GGAGACGTTCTCGACTCCTGTCTGCATGATAACGGCGGCTGCGAACACTTCTGCACGGAGCA GGACGGACGGAGAAACTGCTCCTGCGCAGACGGGTATTACCTAGATAACAGCGGGCAGAAGT GCCGGAGTCACGAGGTGTTTCCATGTGGGAAGGTTCCTCTCCTGCAGGCTGGAAAAGCTGCG GATCATCAGGTGGATCTCAGATCTCGTATCGTTGGAGGATCTGAATGTCCTAAAGGTCACTG TCCGTGGCAGGTGCTGCTGAAGTACGGTGAGAAGGGTTTCTGTGGAGGTGTGATCTACAAGC CCACCTGGATCCTCACAGCTGCTCACTGCTTGGAAAAGCTCAAGGTCAAGTTCCTCAGGATA
GTGGCAGGTGAGCATGATCTGGAGGTGGACGAGGGCACGGAGCAGCTCATCCAGGTGGATCA
GATGTTCACACACCCTGCGTACGTGTCTGAGACAGCGGACAGTGACATCGCCCTGCTGCGTC
TGCGCACCCCCATCGTCTACAGTGTGTATGCGGTGCCGGTGTGTTTGCCGCTGCGGGAGATG
GCGGAGCGCGAGCTGTGGGCGGTCAGCAAACACACGGTGAGCGGCTGGGGCAAACGCAGCGA GGACGGGCCGACCTCTCGCCTGCTGCGCCGGCTGCTGGTGCCGCGCATCCGCACGCAGGAGT
GTGTGCAGGTCAGCAACCTCACGCTCACCAGCAACATGTTCTGCGCCGGATACATCGAGGGC
CGGCAGGACTCCTGTAAGGGTGACAGCGGCGGCCCGCTGGTGACCCGGTACCGAGACACCGC
CTTCCTACTGGGCATCGTGAGCTGGGGGAAAGGCTGCGCTCGCCCGGGCTCCTACGGCATCT
ACACACGCGTGTCCAACTACCTGCAGTGGATCCGACAAACAACCAACACCACGATACACTGA TGAAGACATGACCCGGGTGCATTGCTCATCAAGATTGCTACTCTTAGGTGAACAATTAACAA
ATATTAACTATTATAGTTAATGTTTGTAAAAAATAGCAAAATTATATTGAAAATAAAAAATA TTTATATTAATTATGAAGTGACGGCGATTACTTTAATTATCCAAGACGGTGTTATAGCCCAA AATACCCAATAGTTGAGCATCAGCTGCTTTCCTGACATCCTGTACATATTAGACTCGGATCT GATATTTTGCACAGGTTATATTGCATTTTTAGCAGGTATTTAATGATTTTGCTCTGATTAAT CAGGAGATGTGCAGCTCATTATCTCCATATTATTAATGCTCAACTGTAGTAAACACTCG 5 5.2.10 AMINOACID SEQUENCE OFDANIOFACTORVII FROM GENBANKNMJ31819 (SEQID NO:10) MSLLLVFSLLWSLHYCHSAAVFVHRDEAHEVLIRSKRANSGWFEELKTGNLERECLEEKCSY EEAREVFEHTEATNEF KIYDVKDHCASSPCEHDGLCTTQNADSYMCLCAPGFSGRHCEQSI0 GDVLDSCLHDNGGCEHFCTEQDGRRNCSCADGYYLDNSGQKCRSHEVFPCGKVPLLQAGKAA DHQVDLRSRIVGGSECPKGHCPWQVLLKYGEKGFCGGVIYKPTWILTAAHCLEKLKVKFLRI VAGEHDLEVDEGTEQLIQVDQMFTHPAYVSETADSDIALLRLRTPIVYSVYAVPVCLPLREM AERELWAVSKHTVSGWGKRSEDGPTSRLLRRLLVPRIRTQECVQVSNLTLTSNMFCAGYIEG RQDSCKGDSGGPLVTRYRDTAFLLGIVSWGKGCARPGSYGIYTRVSNYLQWIRQTTNTTIH5 (From Sheehan et al . , Proc . Na tl . Acad. Sci . USA, 98(15) :8768- 8773, 2001.)
5.2.11 DNASEQUENCEENCODINGMOUSEFACTORVIIFROMGENBANK NM_010172 (SEQIDNO:ll)
!0 ATGGTTCCACAGGCGCATGGGCTGCTGCTTCTCTGCTTTCTGCTCCAGCTCCAGGGACCTCT AGGGACTGCAGTTTTCATAACCCAGGAGGAAGCACATGGTGTCCTACACAGGCAAAGGCGTG CCAACTCACTCCTGGAGGAGCTTTGGCCCGGCTCTCTGGAGAGAGAGTGCAATGAGGAACAG TGCTCCTTTGAGGAGGCCCGGGAGATCTTCAAGAGCCCTGAGAGGACCAAGCAGTTCTGGAT TGTTTACAGTGATGGGGACCAGTGTGCCTCGAATCCATGTCAGAACGTAGGTACCTGCCAGG
5 ATCATCTCAAGTCTTACGTCTGCTTCTGCCTCCTAGACTTTGAGGGTCGGAACTGTGAGAAA AGCAAGAATGAGCAGCTGATCTGTGCAAATGAAAATGGTGACTGTGACCAGTACTGCAGGGA CCATGTAGGGACCAAGCGTACCTGTAGCTGTCATGAGGACTACACGCTACAGCCAGATGAGG TGTCCTGCAAACCAAAAGTTGAGTACCCGTGTGGGAGAATACCTGTTGTAGAAAAAAGAAAC TCCAGCAGCCGCCAAGGCCGCATTGTGGGAGGCAACGTGTGCCCCAAAGGGGAGTGTCCATG GCAGGCTGTGCTGAAAATCAATGGGTTATTGCTGTGTGGGGCCGTCCTGCTGGACGCCAGAT GGATAGTGACCGCAGCCCACTGCTTCGATAATATCCGCTACTGGGGAAACATCACAGTGGTG ATGGGTGAACATGACTTCAGTGAGAAGGATGGGGATGAGCAAGTACGACGGGTGACACAGGT CATCATGCCCGACAAGTACATCCGCGGCAAGATCAACCACGACATTGCCCTGCTCCGCCTTC ACCGGCCTGTGACCTTCACTGACTACGTGGTGCCCCTGTGTCTGCCTGAAAAGTCCTTCTCC GAGAACACCCTAGCCAGAATCCGCTTCTCAAGGGTCAGTGGCTGGGGCCAGCTACTGGACCG TGGTGCCACAGCCCTGGAACTCATGTCCATCGAGGTGCCCCGGCTGATGACCCAGGACTGTC TGGAGCACGCCAAGCACAGCTCTAACACCCCCAAAATCACAGAGAACATGTTCTGCGCTGGC TACATGGATGGTACCAAGGACGCCTGCAAGGGTGACAGCGGTGGCCCACATGCCACGCACTA CCATGGCACATGGTATCTGACAGGTGTGGTCAGCTGGGGGGAGGGCTGTGCAGCTATTGGTC ACATTGGGGTGTACACCAGGGTCTCCCAGTACATAGACTGGCTGGTCAGACACATGGACTCC AAGCTCCAGGTTGGGGTTTTCCGACTCCCACTACTGTAG
5.2.12 AMINO ACID SEQUENCE OF MOUSE FACTOR VII FROM GENBANK NM_010172 (SEQ ID NO: 12)
MVPQAHGLLLLCFLLQLQGPLGTAVFITQEEAHGVLHRQRRANSLLEELWPGSLERECNEEQ CSFEEAREIFKSPERTKQF IVYSDGDQCASNPCQNVGTCQDHLKSYVCFCLLDFEGRNCEK
SKNEQLICANENGDCDQYCRDHVGTKRTCSCHEDYTLQPDEVSCKPKVEYPCGRIPVVEKRN
SSSRQGRIVGGNVCPKGECPWQAVLKINGLLLCGAVLLDAR IVTAAHCFDNIRYWGNITVV
MGEHDFSEKDGDEQVRRVTQVIMPDKYIRGKINHDIALLRLHRPVTFTDYVVPLCLPEKSFS
ENT ARIRFSRVSGWGQLLDRGATALELMSIEVPRLMTQDCLEHAKHSSNTPKITENMFCAG YMDGTKDACKGDSGGPHATHYHGTWYLTGVVSWGEGCAAIGHIGVYTRVSQYIDWLVRHMDS
KLQVGVFRLPLL ( From Aasru and Prydz , Biochemistry Mosc . , 67 ( l ) : 25-32 , 2002 . )
5.2.13 DNA SEQUENCE ENCODING CHICKEN FACTOR VII FROM GENBANK AF465268 (SEQ ID NO:13)
ATGGTTTCCAGGCAGTGCGTGGCTTTGCTGCTCTGCTTCCCGCTGCTGGTTCCTCCTTCTCT GGAAGCAGTCTTTTTAAAGCAGGAAGAGGCAAACAGCATTTTTCAAAGGCACAGAAGAGCCA ATAGCTTCTTTGAAGAGATAAAGCTGGGGCCACTAGAGCGAGAATGCATAGAAGAAAAGTGT TCATTTGAGGAAGCAAGAGAGATCTACCGTGATGATGAGAGGACAAAAGAGTTCTGGCACAT CTATTCTGACCCCAACCAGTGTGACTCCAGCCCCTGTCAGAACGGAGGGAGCTGCGATGACC AGTTTCAGGATTATGTCTGCCGCTGTCCTCCGGAGTACGAGGGCAAAAGCTGTGAAACAGCT GTGGCCGAGAACCTGAAGTGCATTTACGACAACGGCGGCTGTGAGCAGTACTGTGCTGACGA GCAGTCTGAAAAACGAGTGTGCTTCTGTGCAGAGGGCTACGCTTTAGCGAGTGATGGAGTGT CCTGCATTCCCCAAGTGAAATACCCTTGTGGAACGATACCAGTGCTGGCAAGAAAGAATACA ACTGCTCAGGGGAGAATAGTAGGTGGTGTCACCTGTCCTCCGGGTGAATGTCCATGGCAAGC CCTTATAATACAGGATCAGAAAGGGAAATGTGGGGGTAGTCTGCTCTCACCAGAGTGGGTGG TGACTGCAGCTCATTGCCTGGACTACGCTCATTCCAAACAGCTCCGGGTGAGGCTGGGTGAA TACTCAGTAAAAGTTGCTGAGAAAACTGAGCAAGAAAGTGGAGTTAGCAAGATCATCAGGCA CGAAGAATACACCATTGGACAAGTCAATCATGACATTGCCCTCCTGAAGCTGGAAACACCCG TGAATCTCACCGATTTCGTTGTGCCAATATGTTTGCCTGAAAAACGGTTTGCAGTGTACGAG CTGTCCTCCATTAAGTTCTCAATGGTGAGCGGATGGGGACGGCTACTAGATGGAGGGGCTAC TTCTACTTTTCTGATGCGAGTTCATTTGCCCCGTGTAAAGACACAAGAATGTGAAAAGCAGG CTAATTTGAACATCACCGAGAATATGTTCTGTGCAGGAGACCTGACCGGTAAAAAAGACTCC TGCAAGGGAGACAGTGGTGGACCTCACGCTACAAAGTACAAGAACACCTGGTTTCTGACTGG GATTGTCAGCTGGGGAAAGGGTTGTGCTGTTGAAGGCAGCTACGGGGTGTACACAAGGGTAT
CCAGATACATCAACTGGTTG 5.2.14 AMINOACID SEQUENCE OF CHICKENFACTORVII FROMGENBANKAF465268 (SEQ ID NO:14)
MVSRQCVALLLCFPLLVPPSLEAVFLKQEEANSIFQRHRRANSFFEEIKLGPLERECIEEKC SFEEAREIYRDDERTKEF HIYSDPNQCDSSPCQNGGSCDDQFQDYVCRCPPEYEGKSCETA VAENLKCIYDNGGCEQYCADEQSEKRVCFCAEGYALASDGVSCIPQVKYPCGTIPVLARKNT TAQGRIVGGVTCPPGECPWQALIIQDQKGKCGGSLLSPE VVTAAHCLDYAHSKQLRVRLGE YSVKVAEKTEQESGVSKIIRHEEYTIGQVNHDIALLKLETPVNLTDFVVPICLPEKRFAVYE LSSIKFSMVSG GRLLDGGATSTFLMRVHLPRVKTQECEKQANLNITENMFCAGDLTGKKDS CKGDSGGPHATKYKNTWFLTGIVSWGKGCAVEGSYGVYTRVSRYINWLKRHME Davidson et al . , Unpublished)
5.2.15 AMINOACIDSEQUENCE OFRABBITFACTORVII FROMSWISSPROT143962 (SEQ IDNO:15) APQARGLGLCSLLALQASLAAVFITQEEAHSVLRRQRRANSFLEELRPGSLERECKEELCS FEEAREVFQSTERTKQF ITYNDGDQCASNPCQNGGSCEDQIQSYICFCLADFEGRNCEKNK NDQLICMYENGGCEQYCSDHVGSQRSCRCHEGYTLLPNGVSCTPTVDYPCGKVPALEKRGAS NPQGRIVGGKVCPKGECPWQAALMNGSTLLCGGSLLDTH VVSAAHCFDKLSSLRNLTIVLG EHDLSEHEGDEQVRHVAQLI PDKYVPGKTDHDIALLRLLQPAALTNNVVPLCLPERNFSES TLATIRFSRVSG GQLLYRGALARELMAIDVPRLMTQDCVEQSEHNPGSPEVTGNMFCAGYL DGSKDACKGDSGGPHATSYHGTYLTGVVSWGEGCARVGHVGVYTRVSRDTEWLSRLMRSKLH HGIQRHPFP 5.2.16 AMINO ACID SEQUENCE OF MOUSE MUTATED FACTOR VII FROM SWISSPROT AAG00449 (SEQ ID NO:16)
MVPQAHGLLLLCFLLQLQGPLGTAVFITQEEAHGVLHRQRRANSLLEEL PGSLERECNEEQ CSFEEAREIFKSPERTKQF IVYSDGDQCASNPCQNVGTCQDHLKSYVCFCLLDFEGRNCEK SKNEQLICANENGDCDQYCRDHVGTKRTCSCHEDYTLQPDEVSCKPKVEYPCGRIPVVEKRN SSSRQGRIVGGNVCPKGECPWQAVLKINGLLLCGAVLLDARWIVTAAHCFDNIRYWGNITVV MGEHDFSEKDGDEQVRRVTQVIMPDKYIRGKINHDIALLRLHRPVTFTDYVVPLCLPEKSFS ENTLARIRFSRVSG GQLLDRGATALELMSIEVPRL TQDCLEHAKHSSNTPKITENMFCAG YMDGTKDACAGDSGGPHATHYHGTWYLTGVVS GEGCAAIGHIGVYTRVSQYID LVRHMDS KLQVGVFRLPLLGSAEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCV VVDVSHEDPEVKFN YVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVS NKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ PENNYKTTPPVLDSDGSFFLYSKLTVDKSR QQGNVFSCSVMHEALHNHYTQKSLSLSPGK
5.2.17 AMINO ACID SEQUENCE OF BOVINE FACTOR VII FROM SWISSPROT P22457 (SEQ ID NO:17)
ANGFLEELLPGSLERECREELCSFEEAHEIFRNEERTRQFWVSYNDGDQCASSPCQNGGSCE DQLRSYICFCPDGFEGRNCETDKQSQLICANDNGGCEQYCGADPGAGRFC CHEGYALQADG VSCAPTVEYPCGKIPVLEKRNGSKPQGRIVGGHVCPKGECP QAMLKLNGALLCGGTLVGPA WVVSAAHCFERLRSRGNLTAVLGEHDLSRVEGPEQERRVAQIIVPKQYVPGQTDHDVALLQL AQPVALGDHVAPLCLPDPDFADQTLAFVRFSAVSG GQLLERGVTARKLMVVLVPRLLTQDC LQQSRQRPGGPVVTDNMFCAGYSDGSKDACKGDSGGPHATRFRGTWFLTGVySWGEGCAAAG HFGIYTRVSRYTA LRQLMGHPPSRQGFFQVPLLP 6. REFERENCES The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference in whole or in part: 5 U. S. Patent 3,215,572 U. S. Patent 4,177,253 U. S. Patent 4,230,685 U. S. Patent 4,237,224
10 U. S. Patent 4,329,241 U. S. Patent 4,452,773 U. S. Patent 4,554,101 U. S. Patent 4,683,195 U. S. Patent 4,683,202 5 U. S. Patent 4,695,392 U. S. Patent 4,770,183 U. S. Patent 4,795,698 U. S. Patent 4,800,159 U. S. Patent 4,883,750
!0 U. S. Patent 5,069,216
U. S. Patent 5,091,206 U. S. Patent 5,145,684 U. S. Patent 5,279,721 U. S. Patent 5,399,363
5 U. S. Patent 5,455,166 U. S. Patent 5,466,468 U. S. Patent 5,543,158 U. S. Patent 5,552,157 U. S. Patent 5,565,213 U. S. Patent 5,567,434 U. S. Patent 5,597,530 U. S. Patent 5,641,515 U. S. Patent 5,648,211 U. S. Patent 5,656,016 U. S. Patent 5,697,899 U. S. Patent 5,705,628 U. S. Patent 5,712,124 U. S. Patent 5,738,868 U. S. Patent 5,741,516 U. S. Patent 5,744,311 U. S. Patent 5,770,219
U. S. Patent 5,783,208
U. S. Patent 5,797,898
U. S. Patent 5,795,587 U. S. Patent 5,972,707
U. S. Patent 6,177,088 U. S. Patent 6,178,871 U. S. Patent 6,238,294 U. S. Patent 6,254,890 U. S. Patent 6,2587,588 U. S. Patent 6,284,280
U. S. Patent 6,331,310
U. S. Patent 6,346,274
U. S. Patent 6,361,994
U. S. Patent 6,383,470
European Patent Application 320308
European Patent Application 329822
Great Britain Patent 2202328
International Patent Application WO 88/10315
International Patent Application WO 89/06700
International Patent Application WO 90/07641 International Patent Application PCT/US87/00880
International Patent Application PCT/US 89/01025
Acton, Scherer, Lodish and Krieger, "Expression cloning of SR-BI, a CD36-related class B scavenger receptor," J Biol Chem., 269:21003-09, 1994.
Afione, Conrad, Kearns, Chunduru, Adams, Reynolds, Guggino, Cutting, Carter and Flotte,
"In vivo model of adeno-associated virus vector persistence and rescue," J Virol,
70:3235-41, 1996.
Afione, Wang, Walsh, Guggino and Flotte, "Delayed expression of adeno-associated virus vector DNA," Intervirology, 42:213-20, 1999.
Agbandje, McKenna, Rossman, Strassheim and Parrish, "Structure determination of feline panleuopenia virus empty particles," Proteins, 16:155-71, 1993.
Agbandje, Kajigaya, McKenna, Young and Rossman, "The structure of human parvovirus
B19 at 8 A resolution," Virol, 203:106-15, 1994. Agbandje-McKenna, Llamas-Saiz, Wang, Tattersall and Rossman, "Functional implications of the structure of the murine parvovirus, minute virus of mice," Structure, 6:1369- 81, 1998.
Aird, Edelberg, Weiler-Guettler, Simmons, Smith and Rosenberg, "Vascular bed-specific expression of an endothelial cell gene is programmed by the tissue microenvironment," J Cell Biol, 138:1117-24, 1997.
Akagi, Isaka, Akagi, Ikawa, Takenaka, Moriyama, Yamauchi, Horio, Ueda, Okabe and Imai, "Transcriptional activation of a hybrid promoter composed of cytomegalovirus enhancer and β-actin/β-globin gene in glomerular epithelial cells in vivo," Kidney Int., 51:1265-69, 1997.
Alexander, Russell, Spence and Miller, "Effects of gamma irradiation on the transduction of dividing and nondividing cells in brain and muscle of rats by adeno-associated virus vectors," Hum. Gene Ther., 7:841-50, 1996.
Allen and Choun, "Large unilamellar liposomes with low uptake into the reticuloendothelial system," FEBS Lett, 223 :42-46, 1987.
Angel, Bauman, Stein, Dellus, Rahmsdorf and Ηerrlich, "12-0-tetradecanoyl-phorbol-13- acetate induction of the human collagenase gene is mediated by an inducible enhancer element located in the 5' flanking region," Mol Cell. Biol, 7:2256, 1987a.
Angel, Imagawa, Chiu, Stein, Imbra, Rahmsdorf, Jonat, Ηerrlich and Karin, "Phorbol ester- inducible genes contain a common cis element recognized by a TPA-modulated transacting factor," Cell, 49:729, 1987b.
Antonarakis, Kazazian and Tuddenham, "Molecular etiology of factor VIII deficiency in hemophilia A," Hum. Mutat., 5:1-22, 1995.
Ardekani, Greenberger and Jahroudi, "Two repressor elements inhibit expression of the von Willebrand factor gene promoter in vitro," Thromb. Haemost., 80:488-94, 1998. Atchison and Perry, "Tandem kappa immunoglobulin promoters are equally active in the presence of the kappa enhancer: Implications for model of enhancer function," Cell, 46:253, 1986. Atchison and Perry, "The role of the kappa enhancer and its binding factor NF-kappa B in the developmental regulation of kappa gene franscription," Cell, 48:121, 1987.
Atchison, Casto and Hammon, "Electron microscopy of adenovirus-associated virus (AAV) in cell cultures," Virology, 29:353-57, 1966. Atkinson and Eisenbarth, "Type 1 diabetes: new perspectives on disease pathogenesis and treatment," Lancet, 358:221-29, 2001. Atkinson and Maclaren, "The pathogenesis of insulin-dependent diabetes mellitus," N. Engl J. Med., 331:1421-28, 1995. Auricchio, O'Connor, Hildinger and Wilson, "A single-step affinity column for purification of serotype-5 based adeno-associated viral vectors," Mol. Ther., 4:372-74, 2001. Balague, Zhou, Dai, Alemany, Josephs, Andreason, Hariharan, Sethi, Prokopenko, Jan, Lou, Hubert-Leslie, Ruiz and Zhang, "Sustained high-level expression of full-length human factor VIII and restoration of clotting activity in hemophilic mice using a minimal adenovirus vector," Blood, 95:820-28, 2000. Balazsovits, Mayer, Bally, Cullis, McDonell, Ginsberg and Falk, "Analysis of the effect of liposome encapsulation on the vesicant properties, acute and cardiac toxicities, and antitumor efficacy of doxorubicin," Cancer Chemother. Pharmacol, 23:81-86, 1989.
Banerji, Rusconi and Schaffher, "Expression of a β-globin gene is enhanced by remote SV40
DΝA sequences," Cell, 27:299, 1981. Banerji, Olson and Schaffher, "A lymphocyte-specific cellular enhancer is located downstream of the joining region in immunoglobulin heavy-chain genes," Cell, 35:729, 1983. Baranov, Glazkov, Kiselev, Ostapenko, Mikhailov, Ivaschenko, Sabetsky and Baranov, "Local and distant transfection of mdx muscle fibers with dystrophin and LacZ genes delivered in vivo by synthetic microspheres," Gene Ther., 6:1406-14, 1999. Barbis, Chang and Parrish, "Mutations adjacent to the dimple of the canine parvovirus capsid structure affect sialic acid binding," Virol, 191:301-08, 1992.
Bartlett, Kleinschmidt, Boucher and Samulski, "Targeted adeno-associated virus vector transduction of nonpermissive cells mediated by a bispecific F(ab'γ)2 antibody," Nat.
Biotechnol, 17:181-86, 1999. Bartlett, Wilcher and Samulski, "Infectious entry pathway of adeno-associated virus and adeno-associated virus vectors," J Virol, 74:2777-85, 2000.
Baskar, Smith, Ciment, Hoffmann, Tucker, Tenney, Colberg-Poley, Nelson and Ghazal, "Developmental analysis of the cytomegalovirus enhancer in transgenic animals," J Virol, 70:3215-26, 1996a. Baskar, Smith, Nilaver, Jupp, Hoffmann, Peffer, Tenney, Colberg-Poley, Ghazal and Nelson, "The enhancer domain of the human cytomegalovirus major immediate-early promoter determines cell type-specific expression in transgenic mice," J Virol, 70:3207-14, 1996b. Baudo, Redaelli, Caimi, Mostarda, Somaini and de Cataldo, "The continuous infusion of recombinant activated Factor Vila (rFVIIa) in patients with Factor VIII inhibitors activates the coagulation and fibrinolytic systems without clinical complications," Thromb. Res., 99:21-24, 2000. Beck, Jones, Chesnut, Walsh, Reynolds, Carter, Askin, Flotte and Guggino, "Repeated delivery of adeno-associated virus vectors to the rabbit airway," J. Virol, 73:9446-55, Benhamou, Mullen, Shaked, Bahmiller and Csete, "Decreased alloreactivity to human islets secreting recombinant viral interleukin 10," Transplantation, 62:1306-12, 1996. Berkhout, Silverman and Jeang, "tat frans-activates the human immunodeficiency virus through a nascent RNA target," Cell, 59:273, 1989. Berns and Bohenzky, "Adeno-associated viruses: an update," Adv. Virus Res., 32:243-306, 1987. Berns and Giraud, "Adenovirus and adeno-associated virus as vectors for gene therapy," Ann. N.Y. Acad. Scl, 772:95-104, 1995. Berns and Giraud, "Biology of adeno-associated virus," Curr. Top. Microbiol. Immunol, 218:1-23, 1996.
Berns and Linden, "The cryptic life style of adeno-associated virus," Bioessays, 17:237-45, 1995. Berns, Pinkerton, Thomas and Hoggan, "Detection of adeno-associated virus (AAV)-specific nucleotide sequences in DNA isolated from latently infected Detroit 6 cells," Virology, 68:556-60, 1975.
Bi, Lawler, Antonarakis, High, Gearhart and Kazazian, Jr., "Targeted disruption of the mouse factor VIII gene produces a model of haemophilia A," Nat. Genet., 10:119-21, 1995. Bi, Sarkar, Naas, Lawler, Pain, Shumaker, Bedian and Kazazian, Jr., "Further characterization of factor Vlll-deficient mice created by gene targeting: RNA and protein studies," Blood, 88:3446-50, 1996.
Blacklow, "Adeno-associated viruses of humans, p. 165-174," in PAR vo VIRUSES AND
HUMAN DISEASE, Pattison (ed.), CRC Press, Boca Raton, FL, 1988. Blacklow, Hoggan and Rowe, "Isolation of adenovirus-associated viruses from man," Proc. Natl Acad. Sci. USA, 58:1410-15, 1967. Blacklow, Hoggan and Rowe, "Serologic evidence for human infection with adenovirus- associated viruses," J Natl. Cancer Inst., 40:319-27, 1968a. Blacklow, Hoggan, Kapikian, Austin and Rowe, "Epidemiology of adenovirus-associated virus infection in a nursery population," Am. J. Epidemiol, 88:368-78, 1968b. Blacklow, Hoggan, Sereno, Brandt, Kim, Parrott and Chanock, "A seroepidemiologic study of adenovirus-associated virus infection in infants and children," Am. J. Epidemiol , 94:359-66, 1971a. Blacklow, Dolin and Hoggan, "Studies of the enhancement of an adenovirus-associated virus by herpes simplex virus," J Gen. Virol, 10:29-36, 1971b. Blanar, Baldwin, Flavell and Sharp, "A gamma-interferon-induced factor that binds the interferon response sequence of the MHC Class I gene, H-2Kb," EMBO J, 8:1139, 1989. Bodine and Ley, "An enhancer element lies 3' to the Human A gamma globin gene," EMBO J, 6:2997, 1987. Bonham, Brown, Boyd, Brown, Bruckenstein, Hanvey, Thomson, Pipe, Hassman, Bisi, et al, "An assessment of the antisense properties of RNase H-competen and steric-blocking oligomers," Nucleic Acids Res. , 23 : 1197-203, 1995. Boshart, Weber, Jahn, Dorsch-Hasler, Fleckenstein and Schaffner, "A very strong enhancer is located upstream of an immediate early gene of human cytomegalovirus," Cell, 41:521, 1985.
Bosze, Thiesen and Charnay, "A transcriptional enhancer with specificity for erythroid cells is located in the long terminal repeat of the friend murine leukemia virus," EMBO J., 5:1615, 1986. Bourlais, Acar, Zia, Sado, Needham and Leverge, "Ophthalmic drug delivery systems - recent advances," Prog. Retin Eye Res., 17:33-58, 1998. Braddock, Chambers, Wilson, Esnouf, Adams, Kingsman and Kingsman, "HIV-I tat activates presynthesized RNA in the nucleus," Cell, 58:269, 1989.
Bristol, Gallo-Penn, Andrews, Idamakanti, Kaleko and Connelly, "Adenovirus-mediated factor VIII gene expression results in attenuated anti-factor Vlll-specific immunity in hemophilia A mice compared with factor VIII protein infusion," Hum. Gene Ther., 12:1651-61, 2001.
Brown, Twells, Hey, Cox, Levy et al, "Isolation and characterization of LRP6, a novel member of the low density lipoprotein receptor gene family," Biochem. Biophys. Res. Commun., 248:879-88, 1998. Brown, Reading, Jones, Fitchett, Howl, Martin, Longland, Michelangeli, Dubrova and Brown, "Critical evaluation of EC V304 as a human endothelial cell model defined by genetic analysis and functional responses: a comparison with the human bladder cancer derived epithelial cell line T24/83," Lab. Invest., 80:37-45, 2000.
Bulla and Siddiqui, "The Hepatitis B virus enhancer modulates transcription of the Hepatitis B virus surface-antigen gene from an internal location," J. Virol, 62:1437, 1986.
Buller, Janik, Sebring and Rose, "Herpes simplex virus types 1 and 2 completely help adenovirus-associated virus replication," J Virol, 40:241-47, 1981.
Burcin, Schiedner, Kochanek, Tsai and O'Malley, "Adenovirus-mediated regulable target gene expression in vivo," Proc. Natl. Acad. Sci. USA, 96:355-60, 1999. Burton, Nakai, Colosi, Cunningham, Mitchell and Couto, "Coexpression of factor VIII heavy and light chain adeno-associated viral vectors produces biologically active protein," Proc. Natl. Acad. Sci. U.S.A., 96:12725-30, 1999.
Campbell and Villarreal, "Functional analysis of the individual enhancer core sequences of polyoma virus: cell-specific uncoupling of DNA replication from transcription," Mol. Cell. Biol, 8:1993, 1988. Campere and Tilghman, "Postnatal repression of the α-fetoprotein gene is enhancer independent," Genes andDev., 3:537, 1989.
Campo, Spandidos, Lang and Wilkie, "Transcriptional control signals in the genome of bovine papilloma virus Type 1," Nature, 303:77, 1983.
Cao, Zhao, Stangl, Hasegawa, Richardson, Parker and Hobbs, "Developmental and hormonal regulation of murine scavenger receptor, class B, type 1," Mol. Endocrinol, 13: 1460- 73, 1999.
Capecchi, "High efficiency transformation by direct microinj ection of DNA into cultured mammalian cells," Cell, 22:479-88, 1980.
Carew, Pollak, High and Bauer, "Severe factor VII deficiency due to a mutation disrupting an Spl binding site in the F.VII promoter," Blood, 92:1639-45, 1998.
Carroll, Rilo, Alejandro, Zeng, Khan, Fontes, Tzakis, Carr and Ricordi, "Long-term (> 3- year) insulin independence in a patient with pancreatic islet cell transplantation following upper abdominal exenteration and liver replacement for fibrolamellar hepatocellular carcinoma," Transplantation, 59:875-79, 1995.
Carter, "Adeno-associated virus vectors," Curr. Opin. Biotechnol, 3:533-39, 1992.
Carter and Flotte, "Development of adeno-associated virus vectors for gene therapy of cystic fibrosis," Curr. Top. Microbiol. Immunol, 218:119-44, 1996.
Carter, Khoury and Denhardt, "Physical map and strand polarity of specific fragments of adenovirus- associated virus DNA produced by endonuclease R-EcoRI," J Virol. ,
16:559-68, 1975.
Casto, Armstrong, Atchison and Hammon, "Studies on the relationship between adeno- associated virus type 1 (AAV-1) and adenoviruses. II. Inhibition of adenovirus plaques by AAV; its nature and specificity," Virol, 33:452-58, 1967. Celander and Haseltine, "Glucocorticoid regulation of murine leukemia virus transcription elements is specified by determinants within the viral enhancer region," J Virology, 61:269, 1987. Celander, Hsu and Haseltine, "Regulatory elements within the murine leukemia virus enhancer regions mediate glucocorticoid responsiveness," J Virology, 62: 1314, 1988.
Chandler, Maler and Yamamoto, "DNA sequences bound specifically by glucocorticoid receptor in vitro render a heterlogous promoter hormone responsive in vivo," Cell, 33:489, 1983. Chandran, Roy and Mishra, "Recent trends in drug delivery systems: liposomal drug delivery system - preparation and characterisation," Indian J. Exp. Biol, 35:801-09, 1997.
Chang, Erwin and Lee, "Glucose-regulated protein (GRP94 and GRP78) genes share common regulatory domains and are coordinately regulated by common trans-acting factors," Mol. Cell Biol, 9:2153, 1989. Chanson, Boerlin, Ajzenberg, Bachelot, Benito, Bringer, Caron, Charbonnel, Cortet, Delemer, Escobar- Jimenez, Foubert, Gaztambide, Jockenhoevel, Kuhn, Leclere, Lorcy, Perlemuter, Prestele, Roger, Rohmer, Santen, Sassolas, Scherbaum, Schopohl, Torres, Varela, Villamil and Webb, "Comparison of octreotide acetate LAR and lanreotide SR in patients with acromegaly," Clin. Endocrinol, 53:577-86, 2000. Chao and Walsh, "Induction of tolerance to human factor VIII in mice," Blood, 97:3311-12, 2001.
Chao, Samulski, Bellinger, Monahan, Nichols and Walsh, "Persistent expression of canine factor IX in hemophilia B canines," Gene Ther., 6:1695-1704, 1999. Chao, Liu, Rabinowitz, Li, Samulski and Walsh, "Several log increase in therapeutic fransgene delivery by distinct adeno-associated viral serotype vectors," Mol. Ther., 2:619-23, 2000. Chao, Mao, Bruce and Walsh, "Sustained expression of human factor VIII in mice using a parvovirus-based vector," Blood, 95:1594-99, 2000. Chao, Sun, Bruce, Xiao and Walsh, "Expression of human factor VIII by splicing between dimerized AAV vectors," Mol Ther., 5:716-22, 2002. Chapman and Rossman, "Structure, sequence, and function correlations among parvoviruses," Virol, 194:491-508, 1993. Chatterjee, Lee, Rentoumis and Jameson, "Negative regulation of the thyroid-stimulating hormone α gene by thyroid hormone: Receptor interaction adjacent to the TATA box," Proc. Natl. Acad. Sci. U.S.A., 86:9114, 1989. Chen and Okayama, "High-efficiency transformation of mammalian cells by plasmid DNA," Mol Cell. Biol, 7:2745-52, 1987. Cheung, Hoggan, Hauswirth and Berns, "Integration of the adeno-associated virus genome into cellular DNA in latently infected human Detroit 6 cells," J. Virol, 33:739-48, 1980. Chiorini, Yang, Liu, Safer and Kotin, "Cloning of adeno-associated virus type 4 (AAV4) and generation of recombinant AAV4 particles," J. Virol, 1 '1:6823-33, 1997. Chiorini, Kim, Yang and Kotin, "Cloning and characterization of adeno-associated virus type 5," J Virol, 73:1309-19, 1999. Choi, Chen, Kriegler and Roninson, "An altered pattern of cross-resistance in multi-drug- resistant human cells results from spontaneous mutations in the MDR-1 (P- glycoprotein) gene," Cell, 53:519, 1988. Christensen, Fitzpatrick, Gildea, Petersen, Hansen, Koch, Egholm, Buchardt, Nielsen, Coull, et al, "Solid-phase synthesis of peptide nucleic acids," J Pept. Sci., 1:175-83, 1995. Clark, Voulgaropoulou and Johnson, "A stable cell line carrying adenovirus-inducible rep and cap genes allows for infectivity titration of adeno-associated virus vectors," Gene
Ther., 3:1124-32, 1996. Clark, Sferra and Johnson. "Recombinant adeno-associated viral vectors mediate long-term fransgene expression in muscle," Hum. Gene Ther., 8:659-669, 1997.
Cohen, Walter and Levinson, "A repetitive sequence element 3' of the human c-Ha-rasl gene has enhancer activity," J Cell. Physiol, 5:75, 1987.
Connelly and Kaleko, "Haemophilia A gene therapy," Haemophilia, 4:380-88, 1998.
Connelly, Mount, Mauser, Gardner, Kaleko, McClelland and Lothrop, Jr., "Complete short- term correction of canine hemophilia A by in vivo gene therapy," Blood, 88:3846-53.
Conrad, Allen, Afione, Reynolds, Beck, Fee-Maki, Barrazza-Ortiz, Adams, Askin, Carter,
Guggino and Flotte, "Safety of single-dose administration of an adeno-associated virus (AAV)-CFTR vector in the primate lung," Gene Ther., 3:658-68, 1996.
Corey, "Peptide nucleic acids: expanding the scope of nucleic acid recognition," Trends Biotechnol, 15:224-29, 1997.
Costa, Lai, Grayson and Darnell, "The cell-specific enhancer of the mouse transthyretin
(prealbumin) gene binds a common factor at one site and a liver-specific factor(s) at two other sites," Mol. Cell. Biol, 8:81, 1988.
Coune, "Liposomes as drug delivery system in the treatment of infectious diseases: potential applications and clinical experience," Infection, 16:141-47, 1988.
Couvreur, Tulkens, Roland, Trouet and Speiser, "Nanocapsules, a new lysosomotropic carrier," FEBSLett., 84:323-26, 1977.
Couvreur, Kante, Lenaerts, Scailteur, Roland and Speiser, "Tissue distribution of antitumor drugs associated with polyalkylcyanoacrylate nanoparticles," J. Pharm. Sci., 69:199- 202, 1980. Couvreur, "Polyalkyleyanoacrylates as colloidal drug carriers," Crit. Rev. Ther. Drug Carrier Syst., 5:1-20, 1988. Cripe, Haugen, Turk, Tabatabai, Schmid, Durst, Gissmann, Roman and Turek, "Transcriptional regulation of the human papilloma virus- 16 E6-E7 promoter by a keratinocyte-dependent enhancer, and by viral E2 trans-activator and repressor gene products: Implications for cervical carcinogenesis," EMBOJ., 6:3745, 1987. Culotta and Hamer, "Fine mapping of a mouse metallothionein gene metal-response element," Mol. Cell. Biol, 9:1376, 1989. Cunningham and Wells, "High resolution epitope mapping of hGH-receptor interactions by alanine-scanning mutagenesis," Science, 244:1081-85, 1989.
Curiel, Agarwal, Wagner and Cotten, "Adenovirus enhancement of transferrin-polylysine- mediated gene delivery," Proc. Natl Acad. Sci. USA, 88:8850-54, 1991. D'Ovidio, Daddi, Suda, Grapperhause and Patterson, "Efficient naked plasmid cotransfection of lung grafts by extended lung/plasmid exposure time," Ann. Thorac. Surg., 71:1817-23, 2001.
Daly, Vogler, Levy, Haskins and Sands, "Neonatal gene transfer leads to widespread correction of pathology in a murine model of lysosomal storage disease," Proc. Natl Acad. Sci. USA., 96:2296-300, 1999. Daly, Ohlemiller, Roberts, Vogler and Sands, "Prevention of systemic clinical disease in MPS VII mice following AAV-mediated neonatal gene transfer," Gene Ther., 8:1291-98, 2001. Dandolo, Blangy and Kamen, "Regulation of polyma virus transcription in murine embryonal carcinoma cells," J Virology, 47:55, 1983. Datta, Chaddaha, Garber, Chung, Tytler, Dashti, Bradley, Gianturco and Anantharamaiah, "The receptor binding domain of apolipoprotein E, linked to a model class A amphipathic helix, enhances internalization and degradation of LDL by fibroblasts," Biochem., 39:213-220, 2000. Davidson, Stein, Heth, Martins, Kotin, Derksen, Zabner, Chodsi and Chiorini, "Recombinant adeno-associated virus type 2, 4, and 5 vectors: transduction of variant cell types and regions in the mammalian central nervous system," Proc. Natl Acad. Sci. USA, 97:3428-43, 2000. de Villiers, Schaffher, Tyndall, Lupton and Kamen, "Polyoma virus DNA replication requires an enhancer," Nature, 312:242, 1984. Deschamps, Meijlink and Verma, "Identification of a transcriptional enhancer element upstream from the proto-oncogene Fos," Science, 230: 1174, 1985.
Ding, Qin, Kotenko, Pestka and Bromberg, "A single amino acid determines the immunostimulatory activity of interleukin 10," J Exp. Med, 191:213-24, 2000. Dong, Fan and Frizzell, "Quantitative analysis of the packaging capacity of recombinant adeno- associated virus," Hum. Gene Ther., 7:2101-12, 1996. Duan, Sharma, Yang, Yue, Dudus, Zhang, Fisher and Engelhardt, "Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue," J Virol, 72:8568-77, 1998 [published erratum appears inJ Virol, 73:861, 1999]. Duan, Li, Kao, Yue, Pessin and Engelhardt, "Dynamin is required for recombinant adeno- associated virus type 2 infection," J Virol. , 73 : 10371 -76, 1999.
Duan, Yan, Yue and Engelhardt, "Structural analysis of adeno-associated virus transduction circular intermediates," Virology, 261:8-14, 1999. Ducholm, Motawia, Pedersen, Nielson and Lundt, "Synthesis of 3'-alkylthio-2',3'-dideoxy nucleosides with potential anti-HIV activity from 2-deoxy-D-ribose, using a phosphorous pentoxide reagent," Arch. Phαrm., 325:597-601, 1992. Dwarki, Belloni, Nijjar, Smith, Couto, Rabier, Clift, Berns and Cohen, "Gene therapy for hemophilia A: Production of therapeutic levels of human factor VIII in vivo in mice," Proc. Natl. Acad. Sci. U.S.A., 92:1023-27, 1995. Edbrooke, Burt, Cheshire and Woo, "Identification of s-acting sequences responsible for phorbol ester induction of human serum amyloid a gene expression via a nuclear- factor-κB-like transcription factor," Mol. Cell. Biol, 9:1908, 1989.
Edlund, Walker, Barr and Rutter, "Cell-specific expression of the rat insulin gene: Evidence for role of two distinct 5' flanking elements," Science, 230:912, 1985. Faller and Baltimore, "Liposome encapsulation of refrovirus allows efficient super infection of resistant cell lines," J Virol, 49:269-72, 1984.
Fechheimer, Boylan, Parker, Sisken, Patel and Zirnmer, "Transfection of mammalian cells with plasmid DNA by scrape loading and sonication loading," Proc. Natl. Acad. Sci. USA, 84:8463-67, 1987. Feng and Holland, "HIV-I tat trans-activation requires the loop sequence within tar," Nature, 334:6178, 1988.
Ferrari, Samulski, Shenk and Samulski, "Second-strand synthesis is a rate-limiting step for efficient transduction by recombinant adeno-associated virus vectors," J Virol, 70:3227-34, 1996. Ferreira, Assouline, Schwachtgen, Bahnak, Meyer and Kerbiriou-Nabias, "The role of the 5'- flanking region in the cell-specific transcription of the human von Willebrand factor gene," Biochem. J, 293:641-48, 1993. Firak and Subramanian, "Minimal transcription enhancer of simian virus 40 is a 74-base-pair sequence that has interacting domains," Mol. Cell. Biol, 6:3667, 1986. Fisher, Gao, Weitzman, DeMatteo, Burda and Wilson, "Transduction with recombinant adeno-associated virus for gene therapy is limited by leading-strand synthesis," J Virol, 70:520-32, 1996.
Fisher, Jooss, Alston, Yang, Haecker, High, Pathak, Raper and Wilson, "Recombinant adeno-associated virus for muscle directed gene therapy," Nat. Med., 3:306-12, 1997.
Fisher-Adams, Wong, Podsakoff, Forman and Chatterjee, "Integration of adeno-associated virus vectors in CD34 human hematopoietic progenitor cells after transduction," Blood, 88:492-504, 1996.
Flannery, Zolotukhin, Vaquero, La Vail, Muzyczka and Hauswirth, "Efficient photoreceptor- targeted gene expression in vivo by recombinant adeno-associated virus," Proc. Natl Acad. Sci. USA, 94:6916-21, 1997.
Flotte, "Prospects for virus-based gene therapy for cystic fibrosis," J. Bioenerg. Biomembr., 25:37-42, 1993.
Flotte and Carter, "Adeno-associated virus vectors for gene therapy," Gene Ther., 2:357-62, 1995.
Flotte and Carter, "Adeno-associated virus vectors for gene therapy of cystic fibrosis," Methods Enzymol, 292:717-32, 1998.
Flotte, Solow, Owens, Afione, Zeitlin and Carter, "Gene expression from adeno-associated virus vectors in airway epithelial cells," Am. J. Respir. Cell Mol. Biol, 7:349-56, 1992.
Flotte, Afione, Conrad, McGrath, Solow, Oka, Zeitlin, Guggino and Carter, "Stable in vivo expression of the cystic fibrosis transmembrane conductance regulator with an adeno- associated virus vector," Proc. Natl. Acad. Sci. USA, 90:10613-17, 1993. Flotte, Afione and Zeitlin, "Adeno-associated virus vector gene expression occurs in nondividing cells in the absence of vector DNA integration," Am. J. Respir. Cell Mol. Biol, 11:517-21, 1994. Flotte, Barraza-Ortiz, Solow, Afione, Carter and Guggino, "An improved system for packaging recombinant adeno-associated virus vectors capable of in vivo transduction," Gene Ther., 2:29-37, 1995. Flotte, Carter, Conrad, Guggino, Reynolds, Rosenstein, Taylor, Walden and Wetzel, "A phase I study of an adeno-associated virus-CFTR gene vector in adult CF patients with mild lung disease," Hum. Gene Ther., 7:1145-59, 1996. Flotte, Beck, Chesnut, Potter, Poirier and Zolotukhin, "A fluorescence video-endoscopy technique for detection of gene transfer and expression," Gene Ther., 5:166-73, 1998. Flotte, Agarwal, Wang, Song, Fenjves, Inverardi, Chesnut, Afione, Loiler, Wasserfall, Kapturczak, Ellis, Nick and Atkinson, "Efficient ex vivo transduction of pancreatic islet cells with recombinant adeno-associated virus vectors," Diabetes, 50:515-20, 2001.
Foecking and Hofstetter, "Powerful and versatile enhancer-promoter unit for mammalian expression vectors," Gene, 45:101-05, 1986. Fraley, Fornari and Kaplan, "Entrapment of a bacterial plasmid in phospholipid vesicles: Potential for gene transfer," Proc. Natl. Acad. Sci. USA, 76:3348-52, 1979. Fresta and Puglisi, "Application of liposomes as potential cutaneous drug delivery systems. In vitro and in vivo investigation with radioactively labelled vesicles," J Drug Target, 4:95-101, 1996. Frohman, "PCR Protocols: A Guide to Methods and Applications," Innis et al. (eds.), Academic Press, San Diego, p. 28, 1990.
-I l l- Fromm, Taylor and Walbot, "Expression of genes transferred into monocot and dicot plant cells by elecfroporation," Proc. Natl Acad. Sci. USA, 82:5824-28, 1985. Fry and Wood, "Gene therapy: potential applications in clinical transplantation," available only at http://www-ermm.cbcu.com.ac.uk/99000691a.pdf, 1999.
Fujita, Shibuya, Hotta, Yamanishi and Taniguchi, "Interferon-β gene regulation: tandemly repeated sequences of a synthetic 6-bp oligomer function as a virus-inducible enhancer," Cell, 49:357, 1987. Fukuda, Ohyama, Lowitz, Matsuo, Pasqualini, Ruoslahti and Fukuda, "A peptide mimic of E-selectin ligand inhibits sialyl Lewis X-dependent lung colonization of tumor cells," Cancer Res., 60:450-56, 2000.
Gabizon and Papahadjopoulos, "Liposomes formulations with prolonged circulation time in blood and enhanced uptake by tumors," Proc. Natl Acad. Sci. USA, 85:6949-53, 1988. Gallichan, Kafri, Krahl, Verma and Sarvetnick, "Lentivirus-mediated transduction of islet grafts with interleukin 4 results in sustained gene expression and protection from insulitis," Hum. Gene Ther., 9:2717-26, 1998. Gao, Qu, Faust, Engdahl, Xiao, Hughes, Zoltick and Wilson, "High-titer adeno-associated viral vectors from a Rep/Cap cell line and hybrid shuttle virus," Hum. Gene Ther., 9:2353-62, 1998. Giannoukakis, Rundert, Robbins and Trucco, "Targeting autoimmune diabetes with gene therapy," Diabetes, 48:2107-21, 1999. Gilles, Morris, Oi and Tonegawa, "A tissue-specific transcription enhancer element is lcoated in the major intron of a rearranged immunoglobulin heavy-chain gene," Cell, 33:717, 1983. Giraud, Winocour and Berns, "Site-specific integration by adeno-associated virus is directed by a cellular DNA sequence," Proc. Natl. Acad. Sci. USA, 91 :10039-43, 1994.
Giraud, Winocour and Berns, "Recombinant junctions formed by site-specific integration of adeno- associated virus into an episome," J Virol, 69:6917-24, 1995.
Girod, Ried, Wobus, Lahm, Leike, Kleinschmidt, Deleage and Hallek, "Genetic capsid modifications allow efficient re-targeting of adeno-associated virus type 2," Nat.
Med, 5:1438, 1999.
Gloss, Bernard, Seedorf and Klock, "The upstream regulatory region of the human papilloma virus- 16 contains an E2 protein-independent enhancer which is specific for cervical carcinoma cells and regulated by glucocorticoid hormones," EMBO J., 6:3735, 1987.
Gnatenko, Arnold, Zolotukhin, Nuovo, Muzyczka and Bahou, "Characterization of recombinant adeno-associated virus-2 as a vehicle for gene delivery and expression into vascular cells," J Investig. Med, 45:87-98, 1997.
Gnatenko, Saenko, Jesty, Cao, Hearing and Bahou, "Human factor VIII can be packaged and functionally expressed in an adeno-associated virus background: Applicability to haemophilia A gene therapy," Br. J. Haematol, 104:27-36, 1999.
Go, Castle, Barrett, Kastelein, Dang, Mosmann, Moore and Howard, "Interleukin 10, a novel
B cell stimulatory factor: unresponsiveness of X chromosome-linked immunodeficiency B cells," J Exp. Med., 172:1625-31, 1990.
Godbout, Ingram and Tilghman, "Fine-structure mapping of the three mouse α-fetoprotein gene enhancers," Mol Cell. Biol, 8:1169, 1988.
Good and Nielsen, "Progress in developing PNA as a gene-targeted drug," Antisense Nucleic
Acid Drug Dev., 7:431-37, 1997.
Goodboum and Maniatis, "Overlapping positive and negative regulatory domains of the human β-interferon gene," Proc. Natl. Acad. Sci. USA, 85:1447, 1988. Goodboum, Burstein and Maniatis, "The human β-interferon gene enhancer is under negative control," Cell, 45:601, 1986. Goodwin, Bittner, Peterson and Wong, "Single-dose toxicity study of hepatic infra-arterial infusion of doxorubicin coupled to a novel magnetically targeted drug carrier," Toxicol Sci, 60:177-83, 2001.
Gopal, "Gene transfer method for transient gene expression, stable transfection, and cotransfection of suspension cell cultures," Mol. Cell Biol. , 5:1188-90, 1985. Gossen, Bonin, Freundlieb and Bujard, "Inducible gene expression systems for higher eukaryotic cells," Curr. Opin. Biotechnol, 5:516-20, 1994. Graham and van der Eb, "Transformation of rat cells by DNA of human adenovirus 5," Virol, 54:536-39, 1973. Graham, Smiley, Russell and Nairn, "Characteristics of a human cell line transformed by DNA from human adenovirus type 5," J Gen. Virol, 36:59-74, 1977. Greelish, Su, Lankford, Burkman, Chen, Konig, Mercier, Desjardins, Mitchell, Zheng, Leferovich, Gao, Balice-Gordon, Wilson and Stedman, "Stable restoration of the sarcoglycan complex in dystrophic muscle perfused with histamine and a recombinant adeno-associated viral vector," N t. Med., 5:439-43, 1999. Greene, Bohnlein and Ballard, "HIV-1, and normal T-cell growth: Transcriptional strategies and surprises," Immunol. Today, 10:272, 1989. Gribskov and Burgess, "Sigma factors from E. coli, B. subtilis, phage SP01, and phage T are homologous proteins," Nucleic Acids Res. , 14:6745-63, 1986. Grifman, Trepel, Speece, Gilbert, Arap, Pasqualini and Weitzman, "Incorporation of rumor- targeting peptides into recombinant adeno-associated virus capsids," Mol. Ther., 3:964-75, 2001. Grimm, Kem, Pawlita, Ferrari, Samulski and Kleinschmidt, "Tifration of AAV-2 particles via a novel capsid ELISA: packaging of genomes can limit production of recombinant AAV-2," Gene Ther., 6:1322-30, 1999. Grosschedl and Baltimore, "Cell-type specificity of immunoglobulin gene expression is regulated by at least three DNA sequence elements," Cell, 41 :885, 1985.
Gmpping, Cnop, Van Schravendijk, Hannaert, Van Berkel and Pipeleers, "Low density lipoprotein binding and uptake by human and rat islet β cells," Endocrinology,
138:4064-68, 1997. Guan, Guillot and Aird, "Characterization of the mouse von Willebrand factor promoter," Blood, 94:3405-12, 1999.
Gupta and Hung, "Magnetically controlled targeted micro-carrier systems," Life Sci,. 1989;44(3):175-86. Hafeli, Sweeney, Beresford, Sim and Macklis, "Magnetically directed poly(lactic acid) 90Y- microspheres: novel agents for targeted intracavitary radiotherapy," J Biomed. Mater. Res., 28:901-18, 1994.
Hagstrom, Walter, Amatniek, Bluebond-Langner, Manno and High, "Prevalence of the factor V leiden mutation in children and neonates with thromboembolic disease," J Peds., 133:777-81, 1998. Hagstrom, Couto, Scallan, Burton, McClelland, Fields, Arruda, Herzog and High, "Improved muscle-derived expression of human coagulation factor IX from a skeletal actin/CMV hybrid enhancer/promoter," Blood, 95:2536-42, 2000. Halbert, Standaert, Wilson and Miller, "Successful readministration of adeno-associated virus vectors to the mouse lung requires transient immunosuppression during the initial exposure," J Virol, 72:9795-805, 1998. Hansen, Qing, Kwon, Mah and Srivastava, "Impaired intracellular trafficking of adeno- associated vims type 2 vectors limits efficient transduction of murine fibroblasts," J Virol, 74:992-96, 2000.
Hansen, Qing and Srivastava, "Infection of purified nuclei by adeno-associated vims 2," Mol. Ther., 4:289-96, 2001.
Hanvey, Peffer, Bisi, Thomson, Cadilla, Josey, Ricca, Hassman, Bonham, Au, et al, "Antisense and antigene properties of peptide nucleic acids," Science, 258:1481-85, 1992.1 ϊ
Harland and Weintraub, "Translation of mRNA injected into Xenopus oocytes is specifically inhibited by antisense RNA," J Cell Biol, 101:1094-99, 1985.
Haslinger and Karin, "Upstream promoter element of the human metallothionein-II gene can act like an enhancer element," Proc. Natl. Acad. Sci. USA, 82:8572, 1985.
Hauber and Cullen, "Mutational analysis of the trans-activiation-responsive region of the human immunodeficiency vims Type I long terminal repeat," J Virol, 62:673, 1988. Heath and Martin, "The development and application of protein-liposome conjugation techniques," Chem. Phys. Lipids, 40:347-58, 1986.
Heath, Lopez, Piper, Montgomery, Stem and Papahadjopoulos, "Liposome-mediated delivery of pteridine antifolates to cells: in vitro potency of methotrexate and its and gamma substituents," Biochim. Biophys. Ada, 862:72-80, 1986. Hen, Borrelli, Fromental, Sassone-Corsi and Chambon, "A mutated polyoma vims enhancer which is active in undifferentiated embryonal carcinoma cells is not repressed by adenovirus-2 El A products," Nature, 321 :249, 1986.
Hensel, Meichle, Pfizenmaier and Kronke, "PMA-responsive 5' flanking sequences of the human TNF gene," Lymphokine Res., 8:347, 1989. Hermonat and Muzyczka, "Use of adeno-associated vims as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue culture cells," Proc. Natl Acad. Sci. USA, 81:6466-70, 1984.
Hermonat, Labow, Wright, Berns and Muzyczka, "Genetics of adeno-associated vims: isolation and preliminary characterization of adeno-associated vims type 2 mutants," J Virol, 51:329-39, 1984.
Hernandez, Wang, Kearns, Loiler, Poirier and Flotte, "Latent adeno-associated vims infection elicits humoral but not cell-mediated immune responses in a nonhuman primate model," J Virol, 73:8549-58, 1999. Herr and Clarke, "The SV40 enhancer is composed of multiple functional elements that can compensate for one another," Cell, 45:461, 1986.
Herzog and High, "Problems and prospects in gene therapy for hemophilia," Curr. Opin. Hematol, 5:321-26, 1998.
Herzog, Yang, Couto, Hagstrom, Elwell, Bellinger, Fields, Read, Brinkhous, Nichols, Kurtzman and High, "Long-term correction of canine hemophilia B by AAV- mediated gene transfer of blood coagulation factor IX," Nature Med., 5:56-63, 1999.
Hey, Twells, Philips, Nakagawa, Brown et al, "Cloning of a novel member of the low- density lipoprotein receptor family," Gene, 216:103-11, 1998.
Hileman, Fromm, Weiler and Linhardt, "Glycosaminoglycan-protein interactions: definition of consensus sites in glycosaminoglycan binding proteins," Bioessays, 2:156-67, 1998.
Hirano, Yamashita, Nakagawa, Ohya, Matsuura, Tsukamoto, Okamoto, Matsuyama, Matsumoto, Miyagawa and Matsuzawa, "Expression of human scavenger receptor class B type I in cultured human monocyte-derived macrophages and atherosclerotic lesions," Circ. Res. , 85 : 108- 16, 1999. Hirochika, Browker and Chow, "Enhancers and trans-acting E2 transcriptional factors of papilloma vimses," J Virol, 61:2599, 1987.
Hirsch, Gaugler, Deagostini-Bauzin, Bally-Cuif and Gordis, "Identification of positive and negative regulatory elements governing cell-type-specific expression of the neural- cell-adhesion-molecule gene," Mol. Cell Biol. , 10: 1959, 1990.
Hoggan, Blacklow and Rowe, "Studies of small DNA vimses found in various adenovims preparations: physical, biological, and immunological characteristics," Proc. Natl.
Acad. Sci. USA, 55:1467-74, 1966. Hoggan, Shatkin, Blacklow, Koczot and Rose, "Helper-dependent infectious deoxyribonucleic acid from adenovims- associated vims," J Virol, 2:850-51, 1968.
Hoggan, Thomas, Thomas and Johnson, Presented at the 4th Lepetit Colloquium, Cocoyac,
Mexico, 1972.
Holbrook, Gulino and Ruscetti, "as-acting transcriptional regulatory sequences in the Gibbon ape leukemia vims (GALV) long terminal repeat," Virology, 157:211, 1987.
Holzknecht and Platt, "The fine cytokine line between graft acceptance and rejection," Nat. Med, 6:497-98, 2000.
Hoque, Ishizu, Matsumoto, Han, Arisaka, Takayama, Suzuki, Kato, Kanda, Watanabe and
Handa, "Nuclear transport of the major capsid protein is essential for adeno- associated vims capsid formation," J Virol, 73:7912-15, 1999.
Horlick and Benfield, "The upstream muscle-specific enhancer of the rat muscle creatine kinase gene is composed of multiple elements," Mol. Cell. Biol, 9:2396, 1989.
Hoyer, "Hemophilia A," N. Engl J. Med., 330:38-47, 1994.
Hsu, de Waal Malefyt, Fiorentino, Dang, Vieira, de Vries, Spits, Mosmann and Moore,
"Expression of interleukin- 10 activity by Epstein-Barr vims protein BCRF1," Science, 250:830-32, 1990. Huang, Osfrowski, Berard and Hagar, "Glucocorticoid regulation of the Ha-MuSV p21 gene conferred by sequences from mouse mammary tumor vims," Cell, 27:245, 1981.
Hussain, Strickland and Bakillah, "The Mammalian Low-Density Lipoprotein Receptor
Family," Annu. Rev. Nutr., 19:141-72, 1999.
Hwang, Lim and Chae, "Characterization of the s-phase-specific franscription regulatory elements in a DNA-replication-independent testis-specific H2B (TH2B) histone gene," Mol Cell. Biol, 10:585, 1990.
Hyrup and Nielsen, "Peptide nucleic acids (PNA): synthesis, properties and potential applications," Bioorg. Med. Chem., 4:5-23, 1996.
Idusogic, Rosen, Carmeliet, Collen and Castellino, "Nucleotide structure and characterization of the murine blood coagulation factor VII gene," Thromb. Haemost, 76:957-64, 1996.
Im and Muzyczka, "The AAV origin binding protein Rep68 is an ATP-dependent site- specific endonuclease with DNA helicase activity," Cell, 61:447-57, 1990.
Imagawa, Chiu and Karin, "Transcription factor AP-2 mediates induction by two different signal-transduction pathways: Protein kinase C and cAMP," Cell, 51:251, 1987.
Imaizumi, Woolworth, Fishman and Chan, "Liposome-enfrapped superoxide dismutase reduces cerebral infarction in cerebral ischemia in rats," Stroke, 21:1312-17, 1990a.
Imaizumi, Woolworth, Fishman and Chan, "Liposome-enfrapped superoxide dismutase ameliorates infarct volume in focal cerebral ischemia," Ada. Neurochirurgica Suppl,
51 :236-38, 1990b.
Imbra and Karin, "Phorbol ester induces the transcriptional stimulatory activity of the S V40 enhancer," Nature, 323:555, 1986.
Imler, Lemaire, Wasvlyk and Waslyk, "Negative regulation contributes to tissue specificity of the immunoglobulin heavy-chain enhancer," Mol. Cell. Biol, 7:2558, 1987. Imperiale and Nevins, "Adenovims 5 E2 transcription unit: An ElA-inducible promoter with an essential element that functions independently of position or orientation," Mol
Cell Biol, 4:875, 1984.
Ishii, Kim, Fujita, Endo, Saeki and Yamamoto, "cDNA cloning of a new low-density lipoprotein receptor-related protein and mapping of its gene (LRP3) to chromosome bands 19ql2-ql3.2," Genomics, 51:132-35, 1998.
Jacobsen, Madsen, Moestmp, Lund, Tommemp et al, "Molecular characterization of a novel human hybrid-type receptor that binds the alpha2-macroglobulin receptor-associated protein," J Biol Chem., 271 :31379-83, 1996.
Jaggar, Chan, Harris and Bicknell, "Endothelial cell-specific expression of tumor necrosis factor- from the KDR or E-selectin promoters following retroviral delivery," Hum.
Gene Ther., 8:2239-47, 1997.
Jakobovits, Smith, Jakobovits and Capon, "A discrete element 3' of human immunodeficiency vims 1 (HIV-1) and HIV-2 mRNA initiation sites mediates transcriptional activation by an HIV trans-activator," Mol. Cell. Biol, 8:2555, 1988.
Jameel and Siddiqui, "The human Hepatitis B vims enhancer requires transacting cellular factor(s) for activity," Mol. Cell. Biol, 6:710, 1986.
Jaynes, Johnson, Buskin, Gartside and Hauschka, "The muscle creatine kinase gene is regulated by multiple upstream elements, including a muscle-specific enhancer," Mol. Cell. Biol, 8:62, 1988.
Johannessen, Andreasen and Nordfang, "Decline of factor VIII and factor IX inhibitors during long-term treatment with NovoSeven," Blood Coagul Fibrinoloysis, 11 :239-
42, 2000.
Johnson, Wold and Hauschka, "Muscle creatine kinase sequence elements regulating skeletal and cardiac muscle expression in transgenic mice," Mol. Cell. Biol, 9:3393, 1989. Jones, Zou, Cowan and Kjeldgaard, "Improved methods for binding protein models in electron density maps and the location of errors in these models," Ada. Crystallograph. A, 47:110-19, 1991. Jooss, Yang, Fisher and Wilson, "Transduction of dendritic cells by DNA viral vectors directs the immune response to fransgene products in muscle fibers," J. Virol, 72:4212-23, 1998. Kadesch and Berg, "Effects of the position of the simian vims 40 enhancer on expression of multiple transcription units in a single plasmid," Mol. Cell Biol, 6:2593, 1986. Kaludov, Brown, Walters, Zabner and Chiorini, "Adeno-associated vims serotype 4 (AAV4) and AAV5 both require sialic acid binding for hemagglutination and efficient transduction but differ in sialic acid linkage specificity," J. Virol, 75:6884-93, 2001. Kalyanasundaram, Feinstein, Nicholson, Leong and Garver, "Coacervate microspheres as carriers of recombinant adenovimses," Cancer Gene Ther., 6:107-12, 2999. Kaplitt, Leone, Samulski, Xiao, Pfaff, O'Malley and During, "Long-term gene expression and phenotypic correction using adeno-associated vims vectors in the mammalian brain," N t. Genet, 8:148-54, 1994. Karin, Haslinger, Heguy, Dieflin and Cooke, "Metal-responsive elements act as positive modulators of human metallothionein-IIa enhancer activity," Mol. Cell. Biol, 7:606, 1987. Kasper, Aledort, Aronson, Counts, Edson, Van Eys, Fratantoni, Green, Hampton, Hilgartner, Levine, Lazerson, McMillan, Penner, Shapiro and Shulman, "Proceedings: A more uniform measurement of factor VIII inhibitors," Thromb. Diath. Haemorrk, 34:612, 1975. Katinka, Yaniv, Vasseur and Blangy, "Expression of polyoma early functions in mouse embryonal carcinoma cells depends on sequence rearrangements in the beginning of the late region," Cell, 20:393, 1980.
Katinka, Vasseur, Montreau, Yaniv and Blangy, "Polyoma DNA sequences involved in the control of viral gene expression in murine embryonal carcinoma cells," Nature,
290:720, 1981. Kaufman and Pipe, "Regulation of factor VIII expression and activity by von Willebrand factor," Thromb. Haemost, 82:201-08, 1999. Kawamoto, Makino, Niw, Sugiyama, Kimura, Anemura, Nakata and Kakunaga, "Identification of the human β-actin enhancer and its binding factor," Mol. Cell. Biol,
8:267, 1988. Kay and High, "Gene therapy for the hemophiliac," Proc. Natl. Acad. Sci. U.S.A., 96:9973- 75, 1999.
Kay, Manno, Ragni, Larson, Couto, McClelland, Glader, Chew, Tai, Herzog, Arruda, Johnson, Scallan, Skarsgard, Flake and High, "Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector," Nat.
Genet, 24:257-61, 2000.
Keams, Afione, Fulmer, Pang, Erikson, Egan, Landrum, Flotte and Cutting, "Recombinant adeno-associated vims (AAV-CFTR) vectors do not integrate in a site-specific fashion in an immortalized epithelial cell line," Gene Ther., 3:748-55, 1996.
Kenyon, Ranuncoli, Masetti, Chatzipetrou and Ricordi, "Islet transplantation: present and future perspectives," Diabetes Metab. Rev., 14:303-13, 1998.
Kessler, Podsakoff, Chen, McQuiston, Colosi, Matelis, Kurtzman and Byrne, "Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein," Proc. Natl Acad. Sci. USA, 93:14082-87, 1996. Kharkevich, Alyautdin and Filippov, "Employment of magnet-susceptible microparticles for the targeting of drugs," J Pharm. Pharmacol, 41:286-88, 1989. Kiledjian, Su and Kadesch, "Identification and characterization of two functional domains within the murine heavy-chain enhancer," Mol. Cell. Biol, 8:145, 1988. Klamut, Gangopadyhay, Worton and Ray, "Molecular and functional analysis of the muscle- specific promoter region of the Duchenne muscular dystrophy gene," Mol. Cell. Biol, 10:193, 1990. Klein, Wolf, Wu and Sanford, "High- velocity microprojectiles for delivering nucleic acids into living cells," Biotechnology, 24:384386, 1992. Klein, Meyer, Peel, Zolotukhin, Meyers, Muzyczka and King, "Neuron-specific transduction in the rat septohippocampal or nigrostriatal pathway by recombinant adeno-associated vims vectors," Exp. Neurol, 150:183-94, 1998. Koch, Benoist and Mathis, "Anatomy of a new B-cell-specific enhancer," Mol. Cell. Biol, 9:303, 1989. Koeberl, Alexander, Halbert, Russell and Miller, "Persistent expression of human clotting factor IX from mouse liver after intravenous injection of adeno-associated vims vectors," Proc. Nat 'I Acad. Sci. USA, 94:1426-31, 1997. Korhonen, Lahtinen, Halmekyto, Alhonen, Janne, Dumont and Alitalo, "Endothelial-specific gene expression directed by the tie gene promoter in vivo," Blood, 86:1828-35, 1995. Kotin, "Prospects for the use of adeno-associated vims as a vector for human gene therapy," Hum. Gene Ther., 5:793-801, 1994. Kotin, Siniscalco, Samulski, Zhu, Hunter, Laughlin, McLaughlin, Muzyczka, Rocchi and Berns, "Site-specific integration by adeno-associated virus," Proc. Natl. Acad. Sci. USA, 87:2211-15, 1990. Kotin, Menninger, Ward and Bems, "Mapping and direct visualization of a region-specific viral DNA integration site on chromosome 19ql3-qter," Genomics, 10:831-34, 1991.
Kotin, Linden and Bems, "Characterization of a preferred site on human chromosome 19q for integration of adeno-associated vims DNA by non-homologous recombination," EMBO J., 11 :5071-78, 1992.
Kovacs and Turowski, "Chemoembolization of oral and oropharyngeal cancer using a high- dose cisplatin crystal suspension and degradable starch microspheres," Oral Oncol, 38:87-95, 2002.
Kriegler and Botchan, "A retrovims LTR contains a new type of eukaryotic regulatory element," In: Eukaryotic Viral Vectors, Gluzman, Ed., Cold Spring Harbor, Cold
Spring Harbor Laboratory, NY, 1982.
Kriegler and Botchan, "Enhanced transformation by a simian vims 40 recombinant vims containing a Harvey murine sarcoma vims long terminal repeat," Mol. Cell. Biol. 3:325, 1983.
Kriegler, Perez and Botchan, "Promoter substitution and enhancer augmentation increases the penetrance of the SV40 a gene to levels comparable to that of the Harvey murine sarcoma virus Ras gene in morphologic transformation," In: Gene Expression, Hamer and Rosenberg, Eds., New York, Alan R. Liss, 1983.
Kriegler, Perez, Hardy and Botchan, "Transformation mediated by the SV40 T antigens: separation of the overlapping SV40 early genes with a retroviral vector," Cell,
38:483, 1984a.
Kriegler, Perez, Hardy and Botchan, "Viral integration and early gene expression both affect the efficiency of SV40 transformation of murine cells: Biochemical and biological characterization of an SV40 retrovims," In: Cancer Cells 2/Oncogenes and Viral Genes, Van de Woude, Levine, Topp and Watson, Eds., Cold Spring Harbor, Cold Spring Harbor Laboratory, 1984b. Kriegler, Perez, Defay, Albert and Liu, "A novel form of TNF/cachectin is a cell-surface cytotoxix transmembrane protein: Ramifications for the complex physiology of TNF," Cell, 53:45, 1988.
Kubo, Sugita, Shimose, Nitta, Ikuta and Murakami, "Targeted delivery of anticancer dmgs with intravenously administered magnetic liposomes in osteosarcoma-bearing hamsters," Int. J. Oncol, 17:309-15, 2000. Kuhl, De La Fuenta, Chaturvedi, Parinool, Ryals, Meyer and Weissman, "Reversible silencing of enhancers by sequences derived from the human IFN- promoter," Cell, 50:1057, 1987. Kunz, Zimmerman, Heisig and Heinrich, "Identification of the promoter sequences involved in the Interleukin-6-dependent expression of the rat α-2-macroglobulin gene," Nucl
Acids Res., 17:1121, 1989. Kurbatova, Fedorenko, Sokhov, Verkholomova and Shklovskaia, "Magnetically-guided anesthetics based on highly dispersed iron powders coated by polyacrylamide," Bioflzika, 47:331-37, 2002. Kyte and Doolittle, "A simple method for displaying the hydropathic character of a protein," J Mol. Biol, 157:105-32, 1982. Langer, "Biomaterials in dmg delivery and tissue engineering: one laboratory's experience," Ace. Chem. Res., 33:94-101, 2000. Larsen, Harney and Moore, "Repression medaites cell-type-specific expression of the rat growth hormone gene," Proc. Natl. Acad. Sci. U.S.A., 83:8283, 1986. Lasic, "Novel applications of liposomes," Trends Biotechnol, 16:307-21, 1998. Laspia, Rice and Mathews, "HIV-1 tat protein increases transcriptional initiation and stabilizes elongation," Cell, 59:283, 1989.
Latimer, Berger and Baumann, "Highly conserved upstream regions of the αrantitrypsin gene in two mouse species govern liver-specific expression by different mechanisms," Mol Cell. Biol, 10:760, 1990.
Lee, Hall, Ringold, Dobson, Luh and Jacob, "Functional analysis of the steroid hormone control region of mouse mammary tumor vims," Nucleic Acids Res., 12:4191-206, 1984.
Levinson, Khoury, VanDe Woude and Gmss, "Activation of SV40 genome by 72-base-pair tandem repeats of Moloney sarcoma vims," Nature, 295:79, 1982.
Lewin, Drenser, Hauswirth, Nishikawa, Yasumura, Flannery and LaVail, "Ribozyme rescue of photoreceptor cells in a transgenic rat model of autosomal dominant retinitis pigmentosa," Nat. Med, 4:967-71 (published erratum appears in Nat. Med., 4:1081)
1998.
Li, Samulski and Xiao, "Role for highly regulated rep gene expression in adeno-associated vims vector production," J Virol, 71:5236-43, 1997.
Lin, Cross, Halden, Dragos, Toledano and Leonard, "Delineation of an enhancer like positive regulatory element in the interleukin-2 receptor α-chain gene," Mol. Cell. Biol,
10:850, 1990.
Linden, Ward, Giraud, Winocour and Bems, "Site-specific integration by adeno-associated vims," Proc. Natl Acad. Sci. USA, 93:11288-94, 1996.
Liu, Nishikawa, Clemens and Huang, "Transfer of full-length Dmd to the diaphragm muscle of Dmd(mdx/mdx) mice through systemic administration of plasmid DNA," Mol.
Ther., 4:45-51, 2001. Loeb, Cordier, Harris, Weitzman and Hope, "Enhanced expression of transgenes from adeno- associated vims vectors with the woodchuck hepatitis vims posttranscriptional regulatory element: implications for gene therapy," Hum. Gene Ther., 10:2295-305, 1999. Lopez-Berestein, Fainstein, Hopfer, Mehta, Sullivan, Keating, Rosenblum, Mehta, Luna,
Hersh, et al, "Liposomal amphotericin B for the treatment of systemic fungal infections in patients with cancer: a preliminary study," J Infect. Dis., 2151 :704, 1985a.
Lopez-Berestein, McQueen and Mehta, "Protective effect of liposomal-amphotericin B against C. albicans infection in mice," Cancer Drug Delivery, 2:183, 1985b.
Luo, Paranya and Bischoff, "Noninflammatory expression of E-selectin is regulated by cell growth," Blood, 93:3785-91, 1999.
Luria, Gross, Horowitz and Givol, "Promoter ehancer elements in the rearranged α-chain gene of the human T-cell receptor," EMBO J., 6:3307, 1987.
Lusky and Botchan, "Transient replication of bovine papilloma vims Type 1 plasmids: cis and trans requirements," Proc. Natl. Acad. Sci. U.S.A., 83:3609, 1986.
Lusky, Berg, Weiher and Botchan, "Bovine papilloma vims contains an activator of gene expression at the distal end of the early transcription unit," Mol. Cell. Biol. 3:1108, 1983.
MacNeil, Suda, Moore, Mosmann and Zlotnik, "IL-10, a novel growth cofactor for mature and immature T cells," J Immunol, 145:4167-73, 1990.
Mah, Qing, Khuntirat, Ponnazhagan, Wang, Kube, Yoder and Srivastava, "Adeno-associated vims type 2-mediated gene transfer: role of epidermal growth factor receptor protein tyrosine kinase in fransgene expression," J Virol, 72:9835-43, 1998. Majors and Varmus, "A small region of the mouse mammary tumor vims long terminal repeat confers glucocorticoid hormone regulation on a linked heterologous gene," Proc. Natl Acad. Sci. U.S.A., 80:5866, 1983. Mandel, Spratt, Snyder and Leff, "Midbrain injection of recombinant adeno-associated vims encoding rat glial cell line-derived neurofrophic factor protects nigral neurons in a progressive 6-hydroxydopamine-induced degeneration model of Parkinson's disease in rats," Proc. Natl. Acad. Sci. USA, 94:14083-88, 1997. March, Madison and Trapnell, "Pharmacokinetics of adenoviral vector-mediated gene delivery to vascular smooth muscle cells: modulation by poloxamer 407 and implications for cardiovascular gene therapy," Hum. Gene Ther., 6:41-53, 1995.
Margalit, "Liposome-mediated drag targeting in topical and regional therapies," Crit. Rev. Ther. Drug Carrier Syst, 12:233-61, 1995. McCarty, Christensen and Muzyczka, "Sequences required for coordinate induction of adeno-associated vims pi 9 and p40 promoters by Rep protein," J Virol, 65:2936-45, 1991.
McCown, Xiao, Li, Breese and Samulski, "Differential and persistent expression patterns of CNS gene transfer by an adeno-associated vims (AAV) vector," Brain Res., 713:99- 107, 1996. McKenna, Olson, Chipman, Baker, Booth, Christensen, Aasted, Fox, Bloom, Wolfinburger and Agbandje-McKenna, "Three-dimensional stracture of Aleutian mink disease parvovirus: implications for disease pathogenicity," J Virol, 73:6882-91, 1999. McNeall, Sanchez, Gray, Chesterman and Sleigh, "Hyperinducible gene expression from a metallotionein promoter containing additional metal-responsive elements," Gene, 76:81, 1989. Miao, Snyder, Schowalter, Patijn, Donahue, Winther and Kay, "The kinetics of rAAV integration in the liver [letter]," Nat. Genet., 19:13-15, 1998. Miao, Νakai, Thompson, Storm, Chiu, Snyder and Kay, "Nonrandom transduction of recombinant adeno-associated vims vectors in mouse hepatocytes in vivo: cell cycling does not influence hepatocyte transduction," J Virol , 74:3793-803, 2000.
Mietus-Snyder, Glass and Pitas, "Transcriptional activation of scavenger receptor expression in human smooth muscle cells requires AP-1/c-Jun and C/EBPβ: both AP-1 binding and JNK activation are induced by phorbol esters and oxidative stress," Arterioscler. Thromb. Vase. Biol, 18:1440-49, 1998. Miksicek, Heber, Schmid, Danesch, Posseckert, Beato and Schutz, "Glucocorticoid responsiveness of the transcriptional enhancer of Moloney murine sarcoma vims," Cell, 46:203, 1986. Mittereder, March and Trapnell, "Evaluation of the concentration and bioactivity of adenovims vectors for gene therapy," J Virol, 70:7498-509, 1996. Montgomery and Gill, "Interactions between von Willebrand factor and factor VIII: Where did they first meet," J Pediatr. Hematol Oncol, 22:269-75, 2000. Moore, Vieira, Fiorentino, Trounstine, Khan and Mosmann, "Homology of cytokine synthesis inhibitory factor (IL-10) to the Epstein-Barr virus gene BCRFI," Science, 248:1230-34, 1990. Moore, Zborowski, Sun and Chalmers, "Lymphocyte fractionation using immunomagnetic colloid and a dipole magnet flow cell sorter," J Biochem. Biophys. Methods., 37:11- 33, 1998. Mordacq and Linzer, "Co-localization of elements required for phorbol ester stimulation and glucocorticoid repression of proliferin gene expression," Genes and Dev., 3:760, 1989. Moreau, Hen, Wasylyk, Everett, Gaub and Chambon, "The SV40 base-repair repeat has a striking effect on gene expression both in SV40 and other chimeric recombinants," Nucl Acids Res., 9:6047, 1981.
Mori and Fukatsu, "Anticonvulsant effect of DN- 1417 a derivative of thyrotropin-releasing hormone and liposome-enfrapped DN-1417 on amygdaloid-kindled rats," Epilepsia,
33:994-1000, 1992. Morwald, Yamazaki, Bujo, Kusunoki, Kanaki et al, "A novel mosaic protein containing
LDL receptor elements is highly conserved in humans and chickens," Arterioscler.
Thromb. Vase. Biol, 17:996-1002 (1997). Muesing, Smith and Capon, "Regulation of mRNA accumulation by a human immunodeficiency vims trans-activator protein," Cell, 48:691, 1987.
Muralidhar, Becerra and Rose, "Site-directed mutagenesis of adeno-associated virus type 2 structural protein initiation codons: effects on regulation of synthesis and biological activity," J Virol, 68:170-76, 1994. Muzyczka, "Use of adeno-associated virus as a general transduction vector for mammalian cells," Curr. Top. Microbiol Immunol, 158:97-129, 1992.
Muzyczka, Samulski, Hermonat, Srivastava and Bems, "The genetics of adeno-associated virus," cf . Exp. Med. Biol, 179:151-61, 1984.
Nakai, Herzog, Hagstrom, Walter, Kung, Yang, Tai, Iwaki, Kurtzman, Fisher, Colosi, Couto and High, "Adeno-associated viral vector-mediated gene transfer of human blood coagulation factor IX into mouse liver," Blood, 91 :4600-07, 1998.
Nakai, Yant, Storm, Fuess, Meuse and Kay, "Extrachromosomal recombinant adeno- associated vims vector genomes are primarily responsible for stable liver transduction in vivo," J. Virol, 75:6969-76, 2001. Nakai, Thomas, Storm, Fuess, Powell, Wright and Kay, "A limited number of transducible hepatocytes restricts a wide-range linear vector dose response in recombinant adeno- associated vims-mediated liver transduction," J Virol, 76:11343-49, 2002.
Needleman and Wunsch, "A general method applicable to the search for similarities in the amino acid sequence of two proteins," J Mol Biol, 48:443-53, 1970.
Nettelbeck, Jerome and Muller, "A strategy for enhancing the transcriptional activity of weak cell type-specific promoters," Gene Ther., 5:1656-64, 1998.
Nichols, Bellinger, Reddick, Smith, Koch, Davis, Sigman, Brinkhous, Griggs and Read, "The roles of von Willebrand factor and factor VIII in arterial thrombosis: Studies in canine von Willebrand disease and hemophilia A," Blood, 81 :2644-51 , 1993.
Nicklin, Buening, Dishart, de Alwis, Girod, Hacker, Thrasher, Ali, Hallek and Baker, "Efficient and selective aav2-mediated gene transfer directed to human vascular endothelial cells," Mol. Ther., 4:174-81, 2001.
Nicolau and Gersonde, "Incorporation of inositol hexaphosphate into intact red blood cells, I. fusion of effector-containing lipid vesicles with erythrocytes," Naturwissenschaften (Germany), 66:563-66, 1979.
Nicolau and Sene, "Liposome-mediated DNA transfer in eukaryotic cells," Biochem. Biophys. Ada, 721:185-90, 1982.
Nielsen, "DNA analogues with nonphosphodiester backbones," Annu. Rev. Biophys. Biomol Struct., 24:167-83, 1995.
Nielsen, Egholm, Berg and Buchardt, "Sequence specific inhibition of DNA restriction enzyme cleavage by PNA," Nucleic Acids Res. , 21 :197-200, 1993.
Ng, Gunning, Liu, Leavitt and Kedes, "Regulation of the human β-actin promoter by upstream and intron domains," Nuc. Acids Res., 17:601, 1989. Norton, Waggenspack, Vamum and Corey, "Targeting peptide nucleic acid-protein conjugates to stmctural features within duplex DNA," Bioorg. Med. Chem., 3:437-45, 1995. Ondek, Sheppard and Herr, "Discrete elements within the SV40 enhancer region display different cell-specific enhancer activities," EMBOJ., 6:1017, 1987.
Ornitz, Hammer, Davison, Brinster and Palmiter, "Promoter and enhancer elements from the rat elastase I gene function independently of each other and of heterologous enhancers," Mol. Cell. Biol, 7:3466, 1987. Palmiter, Chen and Brinster, "Differential regulation of metallothionein-thymidine kinase fusion genes in transgenic mice and their offspring," Cell, 29:701, 1982.
Patema, Moccetti, Mura, Feldon and Bueler, "Influence of promoter and WHV post- transcriptional regulatory element on AAV-mediated fransgene expression in the rat brain," Gene Ther., 7:1304-11, 2000. Papisov and Torchilin, "Prediction of the biological effect of magnetically regulated drugs," Antibiot. Khimioter., 33:751-57, 1988.
Papisov, Svel'ev, Segienko and Torchilin, "Biokinetics of magnetic carriers for directed transport of drags," Antibiot. Khimioter., 10:744-51, 1988. Patterson, Perrella, Hsieh, Yoshizumi, Lee and Haber, "Cloning and functional analysis of the promoter for KDR flk-1, a receptor for vascular endothelial growth factor," J Biol. Chem., 270:23111-18, 1995.
Pech, Rao, Robbins and Aaronson, "Functional identification of regulatory elements within the promoter region of platelet-derived growth factor 2," Mol. Cell. Biol, 9:396, 1989. Peel, Zolotukhin, Schrimsher, Muzyczka and Reier, "Efficient transduction of green fluorescent protein in spinal cord neurons using adeno-associated viras vectors containing cell type- specific promoters," Gene Ther., 4:16-24, 1997. Pereira and Muzyczka, "The cellular transcription factor SPl and an unknown cellular protein are required to mediate Rep protein activation of the adeno-associated viras pi 9 promoter," J Virol, 71:1747-56, 1997. Pereira, McCarty and Muzyczka, "The adeno-associated viras (AAV) Rep protein acts as both a repressor and an activator to regulate AAV transcription during a productive infection," J Virol, 71:1079-88, 1997.
Perez-Stable and Constantini, "Roles of fetal γ-globin promoter elements and the adult β- globin 3' enhancer in the stage-specific expression of globin genes," Mol. Cell. Biol, 10:1116, 1990. Perrey, Ishibashi, Kitamine, Osuga, Yagyu, Chen, Shionoiri, Izuka, Yahagi, Tamura, Ohashi,
Harada, Gotoda and Yamada, "The LDL receptor is the major pathway for β-VLDL uptake by mouse peritoneal macrophages," Atherosclerosis, 154 : 51 -60, 2001.
Picard and Schaffner, "A Lymphocyte-specific enhancer in the mouse immunoglobulin kappa gene," Nature, 307:83, 1984. Pikul, Parks and Schneider, "In vitro killing of melanoma by liposome-delivered intracellular irradiation," Arch. Surg, 122:1417-20, 1987. Pileggi, Molano, Berney, Cattan, Vizzardelli, Oliver, Fraker, Ricordi, Pastori, Bach and Inverardi, "Heme oxygenase-1 induction in islet cells results in protection from apoptosis and improved in vivo function after transplantation," Diabetes, 50:1983-91, 2001. Pinkert, Ornitz, Brinster and Palmiter, "An albumin enhancer located 10 kb upstream functions along with its promoter to direct efficient, liver-specific expression in transgenic mice," Genes andDev., 1:268, 1987. Pinto-Alphandary, Balland and Couvreur, "A new method to isolate polyalkylcyanoacrylate nanoparticle preparations," J. Drug Target, 3 : 167-69, 1995.
Pitas, "Expression of the acetyl low density lipoprotein receptor by rabbit fibroblasts and smooth muscle cells. Up-regulation by phorbol esters," J Biol. Chem., 265:12722-27, 1990.
Pitas, Boyles, Mahley and Bissell, "Uptake of chemically modified low density lipoproteins in vivo is mediated by specific endothelial cells," J Cell Biol. , 100: 103-17, 1985.
Pittman, Alderman, Tomkinson, Wang, Giles and Kaufman, "Biochemical, immunological, and in vivo functional characterization of B-domain-deleted factor VIII," Blood,
81 :2925-35, 1993.
Pober, "Immunobiology of human vascular endothelium," /mmwno/. Res., 19:225-32, 1999. Ponnazhagan, Mukherjee, Wang, Qing, Kube, Mah, Kurpad, Yoder, Srour and Srivastava,
"Adeno-associated viras type 2-mediated transduction in primary human bone marrow-derived CD34+ hematopoietic progenitor cells: donor variation and correlation of fransgene expression with cellular differentiation," J Virol, 71 :8262-
67, 1997.
Ponta, Kennedy, Skroch, Hynes and Groner, "Hormonal response region in the mouse mammary tumor vims long terminal repeat can be dissociated from the proviral pomoter and has enhancer properties," Proc. Natl. Acad. Sci. U.S.A., 82:1020, 1985.
Porton, Zaller, Lieberson and Eckhardt, "Immunoglobulin heavy-chain enliancer is required to maintain transfected γ2A gene expression in a pre-B-cell line," Mol. Cell. Biol , 10:1076, 1990. Potter, Weir and Leder "Enhancer-dependent expression of human K immunoglobulin genes introduced into mouse pre-B lymphocytes by elecfroporation," Proc. Natl. Acad. Sci. USA, 81 :7161-65, 1984.
Qian, Borovok, Bi, Kazazian, Jr. and Hoyer, "Inhibitor antibody development and T cell response to human factor VIII in murine hemophilia A," Thromb. Haemost, 81 :240-
44, 1999. Qing, Wang, Kube, Ponnazhagan, Bajpai and Srivastava, "Role of tyrosine phosphorylation of a cellular protein in adeno- associated viras 2-mediated fransgene expression,"
Proc. Nat 'I Acad. Sci. USA, 94:10879-84, 1997.
Qing, Mah, Hansen, Zhou, Dwarki and Srivastava, "Human fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated viras 2," Nat. Med., 5:71-77, 1999.
Queen and Baltimore, "Immunoglobulin gene transcription is activated by downstream sequence elements," Cell, 35:741, 1983. Quinn, Farina, Gardner, Krutzsch and Levens, "Multiple components are required for sequence recognition of the API site in the Gibbon ape leukemia vims enhancer,"
Mol. Cell. Biol, 9:4713, 1989.
Rabinovitch, Suzrez-Pinzon, Sorensen, Blaeackley, Power and Rajotte, "Combined therapy with interleukin-4 and interleukin- 10 inhibits autoimmune diabetes recurrence in syngeneic islet-transplanted non-obese diabetic mice: analysis of cytokine mRNA expression in the graft," Transplantation, 60:368-74, 1995.
Rabinowitz and Samulski, "Adeno-associated vims expression systems for gene transfer,"
Curr. Opin. Biotechnol, 9:470-75, 1998. Rabinowitz, Rolling, Conrath, Xiao and Samulski, "Cross-packaging of a single adeno- associated viras (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity," J Virol, 76:791-801, 2002.
Rabinowitz, Xiao and Samulski, "Insertional mutagenesis of AAV2 capsid and the production of recombinant viras," Virol, 265:274-85, 1999.
Ranuncoli, Cautero, Ricordi, Masetti, Molano, Inverardi, Alejandro and Kenyon, "Islet cell transplantation: in vivo and in vitro functional assessment of nonhuman primate pancreatic islets," Cell Transplant, 9:409-14, 2000.
Reddy, Sakhuja, Ganesh, Yang, Kayda, Brann, Pattison, Golightly, Idamakanti, Pinkstaff, Kaloss, Barjot, Chamberlain, Kaleko and Connelly, "Sustained human factor VIII expression in hemophilia A mice following systemic delivery of a gutless adenoviral vector," Mol. Ther., 5:63-73, 2002.
Redondo, Hata, Brocklehurst and Krangel, "A T-cell-specific transcriptional enhancer within the human T-cell receptor δ locus," Science, 247:1225, 1990.
Reisman and Rotter, "Induced expression from the Moloney murine leukemia virus long terminal repeat during differentiation of human myeloid cells is mediated through its transcriptional enhancer," Mol. Cell. Biol, 9:3571, 1989.
Remington's Pharmaceutical Sciences 15th Edition, pp. 1035-1038 and 1570-1580.
Resendez Jr., Wooden and Lee, "Identification of highly conserved regulatory domains and protein-binding sites in the promoters of the rat and human genes encoding the sfress- inducible 78-kilodalton glucose-regulated protein," Mol. Cell. Biol, 8:4579, 1988.
Rettenmaier, Stratton, Berman, Senyei, Widder, White and DiSaia, "Treatment of a syngeneic rat tumor with magnetically responsive albumin microspheres labeled with doxorabicin or protein A," Gynecol Oncol, 27:34-43, 1987. Ricordi, Lacy, Olack and Scharp, "Automated method for isolation of human pancreatic islets," Diabetes, 37:412-16, 1988. Ripe, Lorenzen, Brenner and Breindl, "Regulatory elements in the 5' flanking region and the first intron contribute to transcriptional confrol of the mouse α- l-type collagen gene," Mol. Cell. Biol, 9:2224, 1989.
Rippe, Brenner and Leffert, "DNA-mediated gene transfer into adult rat hepatocytes in primary culture," Mol. Cell Biol, 10:689-95, 1990. Rittling, Coutinho, Amarm and Kolbe, "AP-1/jun-binding sites mediate serum inducibility of the human vimentin promoter," Nuc. Acids Res., 17:1619, 1989. Robbins and Evans, "Prospects for treating autoimmune and inflammatory diseases by gene therapy," Gene Ther., 3:187-89, 1996. Rolling, Shen, Tabarias, Constable, Kanagasingam, Barry and Rakoczy, "Evaluation of adeno-associated virus-mediated gene transfer into the rat retina by clinical fluorescence photography," Hum. Gene Ther., 10:641-48, 1999. Rosen, Sodroski and Haseltine, "The location of cts-acting regulatory sequences in the human T-cell lymphotropic viras type III (HTLV-l l l/LAV) long terminal repeat," Cell, 41 :813, 1988. Rosenberg, Foster, Kaufman, Vokac, Moussalli, Kroner and Montgomery, "Infracellular trafficking of factor VIII to von Willebrand factor storage granules," J. Clin. Invest, 101:613-24, 1998.
Rossman, "The canyon hypothesis. Hiding the host cell receptor attachment site on a viral surface from immune surveillance," J Biol. Chem., 264:14587-90, 1989. Rudge, Peterson, Vessely, Koda, Stevens and Catterall, "Adsorption and desorption of chemotherapeutic drags from a magnetically targeted carrier (MTC)," J Control Release, 74:335-40, 2001. Ruffing, Zentgraf and Kleinschmidt, "Assembly of viraslike particles by recombinant structural proteins of adeno-associated vims type 2 in insect cells," J Virol, 66:6922- 30, 1992. Ruffing, Heid and Kleinschmidt, "Mutations in the carboxy terminus of adeno-associated vims 2 capsid proteins affect viral infectivity: lack of an RGD integrin-binding motif," J Gen. Virol, 75:3385-92, 1994. Russell, Miller and Alexander, "Adeno-associated vims vectors preferentially transduce cells in S phase," Proc. Nat'lAcad. Sci. USA, 91:8915-19, 1994. Russell, Alexander and Miller, "DNA synthesis and topoisomerase inhibitors increase transduction by adeno-associated viras vectors," Proc. Nat 'I Acad. Sci. USA, 92:5719-23, 1995. Sakai, Helms, Carlstedt-Duke, Gustafsson, Rottman and Yamamoto, "Hormone-mediated repression: A negative glucocorticoid-response element from the bovine prolactin gene," Genes andDev., 2:1144, 1988. Sambrook, Fritsch and Maniatis, "Molecular Cloning: A Laboratory Manual," Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1989. Samulski, "Adeno-associated viras: integration at a specific chromosomal locus," Curr. Opin. Genet. Dev., 3:74-80, 1993. Samulski, Zhu, Xiao, Brook, Housman, Epstein and Hunter, "Targeted integration of adeno- associated viras (AAV) into human chromosome 19," Embo. J, 10:3941-50, (published erratum appears in Embo. J, Mar;l 1:1228, 1992) 1991. Sands and Barker, "Percutaneous intravenous injection in neonatal mice," Lab. Anim. Sci., 49:328-30, 1999. Sanlioglu and Engelhardt, "Cellular redox state alters recombinant adeno-associated viras transduction through tyrosine phosphatase pathways," Gene Ther. , 6: 1427-37, 1999. Sarkar, Gao, Chirmule, Tazelaar and Kazazian, Jr., "Partial correction of murine hemophilia A with neo-antigenic murine factor VIII," Hum. Gene Ther., 11:881-94, 2000.
Satake, Furukawa and Ito, "Biological activities of oligonucleotides spanning the F9 point mutation within the enhancer region of polyoma viras DNA," J. Virology, 62:970, 1988.
Schaffher, Schirm, Muller-Baden, Wever and Schaffner, "Redundancy of information in enhancers as a principle of mammalian transcription control," J. Mol. Biol, 201 :81, 1988.
Schmidt- Wolf and Schmidt- Wolf, "Cytokines and gene therapy," Immunol Today, 16:173- 75, 1995.
Sculier, Coune, Meunier, Brassinne, Laduron, Hollaert, Collette, Heymans and Klastersky, "Pilot study of amphotericin B entrapped in sonicated liposomes in cancer patients with fungal infections," J Cancer Clin. Oncol, 24:52 -38, 1988.
Searle, Stuart and Palmiter, "Building a metal-responsive promoter with synthetic regulatory elements," Mol Cell Biol, 5:1480, 1985.
Senapathy, Tratschin and Carter, "Replication of adeno-associated viras DNA. Complementation of naturally occurring rep' mutants by a wild-type genome or an ori" mutant and correction of terminal palindrome deletions," J. Mol. Biol, 179:1-20, 1984. Senyei, Reich, Gonczy and Widder, "In vivo kinetics of magnetically targeted low-dose doxorabicin," J Pharm. Set, 70:389-91, 1981.
Shapiro, Lakey, Ryan, Korbutt, Toth, Warnock, Kneteman and Rajotte, "Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen," N. Engl. J. Med, 343:230-38, 2000. Sharp and Marciniak, "HIV Tar: An RΝA enhancer?" Cell, 59:229, 1989. Shaul and Ben-Levy, "Multiple nuclear proteins in liver cells are bound to Hepatitis B viras enhancer element and its upsfream sequences," EMBO J., 6:1913, 1987.
Sherman, Basta, Moore, Brown and Ting, "Class II box consensus sequences in the HLA- DRα gene: Transcriptional function and interaction with nuclear proteins," Mol. Cell. Biol, 9:50, 1989.
Shi, Arnold and Bartlett, "Insertional mutagenesis of the adeno-associated vims type 2 (aav2) capsid gene and generation of aav2 vectors targeted to alternative cell- surface receptors," Hum. Gene Ther., 12: 1697-711, 2001.
Sleigh and Lockett, "SV40 enhancer activation during retinoic-acid-induced differentiation of F9 embryonal carcinoma cells," J EMBO, 4:3831, 1985.
Snyder, Miao, Patijn, Spratt, Danos, Nagy, Gown, Winther, Meuse, Cohen, Thompson and Kay, "Persistent and therapeutic concentrations of human factor IX in mice after hepatic gene transfer of recombinant AAV vectors," Nat. Genet, 16:270-76, 1997.
Snyder, Miao, Meuse, Tubb, Donahue, Lin, Stafford, Patel, Thompson, Nichols, Read, Bellinger, Brinkhous and Kay, "Correction of hemophilia B in canine and murine models using recombinant adeno-associated viral vectors," Nat. Med. 5:64-70, 1999.
Song, Morgan, Ellis, Poirer, Chesnut, Wang, Brantly, Muzyczka, Byrne, Atkinson and Flotte, "Sustained secretion of human αl-antitrypsin from murine muscle transduced with adeno-associated vims vectors," Proc. Natl. Acad. Sci. USA, 95:14384-88, 1998.
Song, Embury, Laipis, Bems, Crawford and Flotte, "Stable therapeutic serum levels of human alpha- 1 antitrypsin (AAT) after portal vein injection of recombinant adeno- associated viras (rAAV) vectors," Gene Ther., 8:1299-306, 2001.
Spalholz, Yang and Howley, "Transactivation of a bovine papilloma viras transcriptional regulatory element by the E2 gene product," Cell, 42:183, 1985. Spandau and Lee, "Trans-activation of viral enhancers by the Hepatitis B viras X protein," J Virology, 62:427, 1988. Spandidos and Wilkie, "Host-specificities of papilloma viras, Moloney murine sarcoma virus and simian viras 40 enhancer sequences," EMBOJ., 2:1193, 1983. Srivastava, Lusby and Bems, "Nucleotide sequence and organization of the adeno-associated vims 2 genome," J Virol, 45:555-64, 1983. Stein and Stein, "Bovine aortic endothelial cells display macrophage-like properties towards acetylated 1251-labelled low density lipoprotein," Biochem. Biophys. Acta, 620:631- 35, 1980. Stephens and Hentschel, "The bovine papilloma viras genome and its uses as a eukaryotic vector," Biochem. J, 248:1, 1987. Stuart, Searle and Palmiter, "Identification of multiple metal regulatory elements in mouse metallothionein-I promoter by assaying synthetic sequences," Nature, 317:828, 1985. Sullivan and Peterlin, "Transcriptional enhancers in the HLA-DQ subregion," Mol. Cell. Biol, 7:3315, 1987.
Summerford and Samulski, "Membrane-associated heparan sulfate proteoglycan is a receptor for adeno-associated viras type 2 virions," J Virol, 72:1438-45, 1998.
Summerford, Bartlett and Samulski, "αvβ5 integrin: a co-receptor for adeno-associated viras type 2 virions," Nat. Med, 5:78-82, 1999. Sun, Li and Xiao, "Overcoming adeno-associated viras vector size limitation tlirough viral DΝA heterodimerization," Nat Med, 6:599-602, 2000. Suzuki, Shin, Fjuikura, Matsuzaki and Takata, "Direct gene transfer into rat liver cells by in vivo elecfroporation," FEBSLett, 425:436-40, 1998. Swartzendraber and Lehman, "Νeoplastic differentiation: Interaction of simian viras 40 and polyoma viras with murine teratocarcinoma cells," J Cell. Physiology, 85:179, 1975. Tagliavacca, Wang and Kaufman, "ATP-dependent dissociation of non-disulfide-linked aggregates of coagulation factor VIII is a rate-limiting step for secretion," Biochemistry, 39:1973-81, 2000.
Tahara, Mueller, Ricordi, Robbins and Lotze, "Islet cell transplantation facilitated by gene transfer," Transplant Proc. , 24:2975-76, 1992.
Takahashi, Sawasaki, Hata, Mukai and Goto, "Spontaneous fransformation and immortalization of human endothelial cells," In Vitro Cell Dev. Biol, 26:265-74, 1990.
Takakura, "Drag delivery systems in gene therapy," Nippon Rinsho, 56:691-95, 1998.
Takebe, Seiki, Fujisawa, Hoy, Yokota, Arai, Yoshida and Arai, "SRα promoter: An efficient and versatile mammalian cDNA expression system composed of the simian viras 40 early promoter and the R-U5 segment of human T-cell leukemia virus Type 1 long terminal repeat," Mol. Cell. Biol, 8:466, 1988.
Tavemier, Gheysen, Duerinck, Can der heyden and fiers, "Deletion mapping of the inducible promoter of human IFN-β gene," Nature, 301 :634, 1983.
Taylor and Kingston, "Factor substitution in a human HSP70 gene promoter: TATA- dependent and TAT A-independent interactions," Mol Cell. Biol, 10:165, 1990a.
Taylor and Kingston, "Ela trans-activation of human HSP70 gene promoter substitution mutants is independent of the composition of upsfream and TATA elements," Mol. Cell Biol, 10:176, 1990b.
Taylor, Solomon, Weiner, Paucha, Bradley and Kingston, "Stimulation of the human heat- shock protein 70 promoter in vitro by simian vims 40 large T antigen," J Biol. Chem., 264:15160, 1989.
Thiesen, Bosze, Henry and Charnay, "A DNA element responsible for the different tissue specificities of friend and Moloney retroviral enhancers," J Virology, 62:614, 1988. Tratschin, Miller and Carter, "Genetic analysis of adeno-associated vims: properties of delection mutants constructed in vitro and evidence for an adeno-associated viras replication function," J Virol, 51:611-19, 1984. Tratschin, West, Sandbank and Carter, "A human parvovirus, adeno-associated viras, as a eucaryotic vector: transient expression and encapsidation of the procaryotic gene for chloramphenicol acetyltransferase," Mol. Cell Biol, 4:2072-81, 1984. Trempe and Carter, "Alternate mRNA splicing is required for synthesis of adeno-associated viras VP1 capsid protein," J Virol, 62:3356-63, 1988. Tresnan, Southard, Weichert, Sgro and Parrish, "Analysis of the cell and erythrocyte binding activities of the dimple and canyon regions of the canine parvovirus capsid," Virol, 211:123-32, 1995. Triesman, "Identification of a protein-binding site that mediates transcriptional response to the c-fos gene to serum factors," Cell, 46:567-74, 1986. Tranche, Rollier, Bach, Weiss and Yaniv, "The rat albumin promoter: Cooperation with upstream elements is required when binding of APF/HNF 1 to the proximal element is partially impaired by mutation or bacterial methylation," Mol. Cell Biol, 9:4759, 1989. Tranche, Rollier, Herbomel, Bach, Cereghini, Weiss and Yaniv, "Anatomy of the rat albumin promoter," Mol. Biol. Med, 7:173, 1990. Trudel and Constantini, "A 3' enhancer contributes to the stage-specific expression of the human β-globin gene," Genes andDev., 6:954, 1987.
Tsao, Chapman, Agbandje, Keller, Smith, Wu, Luo, Smith, Rossman, Compans, et al, "The three-dimensional stracture of canine parvovims and its functional implications," Science, 251:1456-64, 1991. Tsao, Chapman, Wu, Agbandje, Keller and Rossman, "Stracture determination of monoclinic canine parvovirus," Ada. Crystallogr. B, 48:75-88, 1992. Tur-Kaspa, Teicher, Levine, Skoultchi and Shafritz, "Use of elecfroporation to infroduce biologically active foreign genes into primary rat hepatocytes," Mol. Cell Biol, 6:716-18, 1986.
Tyndell, La Mantia, Thacker, Favaloro and Kamen, "A region of the polyoma vims genome between the replication origin and late protein-coding sequences is required in cis for both early gene expression and viral DNA replication," Nuc. Acids. Res., 9:6231, 1981. Vanbever, Fouchard, Jadoul, De Morre, Preat and Marty, "In vivo noninvasive evaluation of hairless rat skin after high-voltage pulse exposure," Skin Parmacol Appl. Skin Physiol, 11:23-34, 1998. Vandendriessche, Vanslembrouck, Goovaerts, Zwinnen, Vanderhaeghen, Collen and Chuah, "Long-term expression of human coagulation factor VIII and correction of hemophilia A after in vivo retroviral gene transfer in factor Vlll-deficient mice," Proc. Natl Acad. Sci. U.S.A., 96:10379-84, 1999. Vannice and Levinson, "Properties of the human Hepatitis B vims enhancer: Position effects and cell-type nonspecificity," J. Virology, 62:1305, 1988. Varban, Rinninger, Wang, Fairchild-Huntress, Dunmore, Fang, Gosselin, Dixon, Deeds, Acton, Tall and Huszar, "Targeted mutation reveals a central role for SR-BI in hepatic selective uptake of high density lipoprotein cholesterol," Proc. Natl. Acad. Sci. USA, 95:4619-24, 1998. Vasseur, Kress, Montreau and Blangy, "Isolation and characterization of polyoma vims mutants able to develop in multipotential murine embryonal carcinoma cells," Proc. Natl Acad. Sci. U.S.A. , 77: 1068, 1980. Vestweber and Blanks, "Mechanisms that regulate the function of the selectins and their ligmds," Physiol Rev., 79:181-213, 1999.
Vincent-Lacaze, Snyder, Gluzman, Bohl, Lagarde and Danos, "Structure of adeno-associated viras vector DNA following transduction of the skeletal muscle," J Virol, 73:1949- 55, 1999.
Virella-Lowell, Song, Morgan and Flotte, "A CMV/β-actin hybrid promoter greatly improves recombinant adeno-associated viras (rAAV) vector expression in the murine lung,"
Ped. Pulmonol, SI 9:231, 1999. Virella-Lowell, Poirier, Chesnut, Brantly and Flotte, "Inhibition of recombinant adeno- associated vims (rAAV) transduction by bronchial secretions from cystic fibrosis patients," Gene Ther., 7: 1783-89, 2000. Voyta, Via, Butterfield and Zetter, "Identification and isolation of endothelial cells based on their increased uptake of acetylated-low density lipoprotein," J Cell Biol, 99:2034- 40, 1984.
Wagner, Zatloukal, Gotten, Kirlappos, Mechtler, Curiel and Bimstiel, "Coupling of adenovims to transferrin-polylysine/DNA complexes greatly enhances receptor- mediated gene delivery and expression of transfected genes," Proc. Natl. Acad. Sci.
USA, 89:6099-103, 1992.
Wagner, Reynolds, Moran, Moss, Wine, Flotte and Gardner, "Efficient and persistent gene transfer of AAV-CFTR in maxillary sinus," Lancet, 351 :1702-03, 1998.
Walters, Yi, Keshavjee, Brown, Welsh, Chiorini and Zabner, "Binding of adeno-associated vims type 5 to 2,3 -linked sialic acid is required for gene transfer," J Biol. Chem.,
276:20610-16, 2001. Wang and Calame, "SV40 enhancer-binding factors are required at the establishment but not the maintenance step of enhancer-dependent transcriptional activation," Cell, 47:241, 1986. Wang, Takabe, Bidlingmaier, 111 and Verma, "Sustained correction of bleeding disorder in hemophilia B mice by gene therapy," Proc. Natl Acad. Sci. U.S.A., 96:3906-10, 1999. Watson, "Post-radiolabeling for detecting DNA damage," Mutagenesis, 2:319-31, 1987. Weber, De Villiers and Schaffner, "An SV40 'enhancer trap' incorporates exogenous enhancers or generates enhancers from its own sequences," Cell, 36:983, 1984. Weger, Wendland, Kleinschmidt and Heilbronn, "The adeno-associated vims type 2 regulatory proteins Rep78 and Rep68 interact with the transcriptional coactivator PC4," J Virol, 73:260-69, 1999. Weinberger, Jat and Sharp, "Localization of a repressive sequence contributing to B-cell specificity in the immunoglobulin heavy-chain enhancer," Mol. Cell. Biol, 8:988, 1984.
Weir and Bonner-Weir, "Islet transplantation as a treatment for diabetes," J Am. Optom. Assoc, 69:727-32, 1998. Weiss, Bonneau and Liggitt, "Use of perfluorochemical liquid allows earlier detection of gene expression and use of less vector in normal lung and enhances gene expression in acutely injured lung," Mol Ther. , 3 :734-45, 2001.
Weitzman, Kyostio, Kotin and Owens, "Adeno-associated vims (AAV) Rep proteins mediate complex formation between AAV DNA and its integration site in human DNA," Proc. Natl. Acad. Sci. USA, 91:5808-12, 1994. Wiestuba, Kem, Weger, Grimm and Kleinschmidt, "Subcellular compartmentalization of adeno-associated vims type 2 assembly," J Virol. , 71:1341 -52, 1997. Winoto and Baltimore, "αβ-lineage-specific expression of the α T-cell receptor gene by nearby silencers," Cell, 59:649, 1989.
Wistuba, Weger, Kem and Kleinschmidt, "Intermediates of adeno-associated vims type 2 assembly identification of soluble complexes containing Rep and Cap proteins," J Virol, 69:5311-19, 1995.
Wong and Neumann, "Electric field mediated gene transfer," Biochim. Biophys. Res.
Commun., 107:584-87, 1982.
Wu and Wu, "Receptor-mediated in vitro gene transfections by a soluble DNA carrier system," J Biol. Chem., 262:4429-32, 1987. Wu and Wu, "Evidence for targeted gene delivery to HepG2 hepatoma cells in vitro,"
Biochemistry, 27:887-92, 1988. Wu, Xiao, Conlon, Hughes, Agbandje-McKenna, Ferkol, Flotte and Muzyczka, "Mutational analysis of the adeno-associated vims type 2 (AAV2) capsid gene and construction of
AAV2 vectors with altered tropism," J Virol, 74:8635-47, 2000.
Wyble, Hynes, Kucbibhotla, Marcus, Hallahan and Gewertz, "TNF-α and IL-1 upregulate membrane-bound and soluble E-selectin through a common pathway," J Surg. Res.,
73:107-12, 1997.
Xiao, Li and Samulski, "Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated vims vector," J Virol, 70:8098-108, 1996.
Xiao, Li and Samulski, "Production of high-titer recombinant adeno-associated viras vectors in the absence of helper adenovirus," J Virol, 72:2224-32, 1998.
Xu, Daly, Gao, Flotte, Song, Byrne, Sands and Parker Ponder, CMV-β-actin promoter directs higher expression from an adeno-associated viral vector in the liver than the cytomegalovirus or elongation factor lα promoter and results in therapeutic levels of human factor X in mice," Hum. Gene Ther., 12:563-73, 2001.
Yan, Zhang, Duan and Engelhardt, "Trans-splicing vectors expand the utility of adeno- associated virus for gene therapy," Proc. Natl. Acad. Sci. U.S.A., 97:6716-21, 2000. Yang and Kotin, "Glucose-responsive gene delivery in pancreatic Islet cells via recombinant adeno-associated viral vectors," Pharm. Res., 17:1056-61, 2000.
Yang, Burkholder, Roberts, Martinell and McCabe, "In vivo and in vitro gene transfer to mammalian somatic cells by particle bombardment," Proc. Natl. Acad. Sci. USA, 87:9568-72, 1990. Yang, Mamounas, Yu, Kennedy, Leaker, Merson, Wong-Staal, Yu and Barber, "Development of novel cell surface CD34-targeted recombinant adenoassociated viras vectors for gene therapy," Hum. Gene Ther., 9:1929-37, 1998.
Yang, Zhou, Zhang, Zidon, Ritchie and Engelhardt, "Concatamerization of adeno-associated viras circular genomes occurs through intermolecular recombination," J Virol, 73:9468-77, 1999.
Yutzey, Kline and Konieczny, "An internal regulatory element controls froponin I gene expression," Mol. Cell Biol, 9:1397, 1989.
Zabner, Seiler, Walters, Kotin, Fulgeras, Davidson and Chiorini, "Adeno-associated viras type 5 (AAV5) but not AAV2 binds to the apical surfaces of airway epithelia and facilitates gene transfer," J Virol , 74:3852-58, 2000.
Zambaux, Bonneaux, Gref, Maincent, Dellacherie, Alonso, Labrude and Vigneron, "Influence of experimental paparmeters on the characteristics of poly(lactic acid) nanoparticles prepared by a double emulsion method," J Controlled Release, 50:31- 40, 1998. Zatloukal, Gotten, Berger, Schmidt, Wagner and Bimstiel, "In vivo production of human factor VII in mice after intrasplenic implantation of primary fibroblasts fransfected by receptor-mediated, adenoviras-augmented gene delivery," Proc. Natl. Acad. Sci. U.S.A., 9\:5\48-52, 1994.
Zhou and Muzyczka, "In vitro packaging of adeno-associated viras DNA," J Virol,
72:3241-47, 1998.
Zhou, Cooper, Kang, Ruggieri, Heimfeld, Srivastava and Broxmeyer, "Adeno-associated viras 2-mediated high efficiency gene transfer into immature and mature subsets of hematopoietic progenitor cells in human umbilical cord blood," J Exp. Med., 179:1867-75, 1994.
Ziady, Perales, Ferkol, Gerken, Beegen, Perlmutter and Davis, "Gene transfer into hepatoma cell lines via the serpin enzyme complex receptor," Am. J. Physiol, 273(2 Pt l):G545-52, 1997.
Zolotukhin, Byrne, Mason, Zolotukhin, Potter, Chesnut, Summerford, Samulski and Muzyczka, "Recombinant adeno-associated viras purification using novel methods improves infectious titer and yield," Gene Ther., 6:973-85, 1999. Zolotukhin, Potter, Hauswirth, Guy and Muzyczka, "A 'humanized' green fluorescent protein cDNA adapted for high-level expression in mammalian cells," J Virol, 70:4646-54, 1996.
zur Muhlen, Schwarz and Mehnert, "Solid lipid nanoparticles (SLN) for controlled drag delivery—drag release and release mechanism," Eur. J. Pharm. Biopharm., 45:149-
55, 1998.
All of the compositions and methods disclosed and claimed herein can be made and
executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Claims

WHAT IS CLAIMED:
1. A recombinant adeno-associated viral vector comprising at least a first nucleic acid segment encoding a biologically-active Factor VII peptide, polypeptide or protein operably linked to at least a first promoter capable of expressing said segment in a mammalian host cell transformed with said vector.
2. The recombinant adeno-associated viral vector of claim 1, wherein said nucleic acid segment encodes a mammalian biologically-active Factor VII peptide, polypeptide or protein.
3. The recombinant adeno-associated viral vector of claim 2, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises a first contiguous sequence region of at least 60 amino acids from any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
4. The recombinant adeno-associated viral vector of claim 3, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises at least a first contiguous sequence region of at least 80 amino acids from any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
5. The recombinant adeno-associated viral vector of claim 4, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises at least a first contiguous sequence region of at least 100 amino acids from any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
6. The recombinant adeno-associated viral vector of claim 5, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises at least a first contiguous sequence region of at least 120 amino acids from any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
7. The recombinant adeno-associated viral vector of claim 6, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises the sequence of any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
The recombinant adeno-associated viral vector of claim 6, wherein said nucleic acid segment encodes a biologically-active Factor VII peptide, polypeptide or protein that comprises the sequence of any one of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO:15 or SEQ ID NO:17.
9. The recombinant adeno-associated viral vector of claim 1, wherein said nucleic acid segment encodes a biologically-active human Factor VII peptide, polypeptide or protein.
10. The recombinant adeno-associated viral vector of claim 9, wherein said nucleic acid segment encodes a biologically-active human Factor VII peptide, polypeptide or protein that comprises the sequence of SEQ ID NO:2.
11. The recombinant adeno-associated viral vector of claim 3, wherein said nucleic acid segment comprises at least a first contiguous sequence region of at least 180 nucleotides from any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l 1, or SEQ ID NO: 13.
12. The recombinant adeno-associated viral vector of claim 11, wherein said nucleic acid segment comprises at least a first contiguous sequence region of at least 210 nucleotides from any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l 1, or SEQ ID NO:13.
13. The recombinant adeno-associated viral vector of claim 12, wherein said nucleic acid segment comprises at least a first contiguous sequence region of at least 240 nucleotides from any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l 1, or SEQ ID NO:13.
14. The recombinant adeno-associated viral vector of claim 13, wherein said nucleic acid segment comprises at least a first contiguous sequence region of at least 270 nucleotides from any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13.
15. The recombinant adeno-associated viral vector of claim 14, wherein said nucleic acid segment comprises at least a first contiguous sequence region of at least 300 nucleotides from any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13.
16. The recombinant adeno-associated viral vector of claim 15, wherein said nucleic acid segment comprises the nucleotide sequence of any one of SEQ ID NO:l, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:l l, or SEQ ID NO:13.
17. The recombinant adeno-associated viral vector of claim 16, wherein said nucleic acid segment comprises the nucleotide sequence of SEQ ID NO: 1.
18. The recombinant adeno-associated viral vector of claim 1, wherein said promoter is a heterologous promoter.
19. The recombinant adeno-associated viral vector of claim 18, wherein said promoter is selected from the group consisting of a CMV promoter, a β-actin promoter, a hybrid CMV promoter, a hybrid β-actin promoter, an EF1 promoter, a Ula promoter, a Ulb promoter, a Tet-inducible promoter and a VP16-LexA promoter.
20. The recombinant adeno-associated viral vector of claim 19, wherein said promoter is a chicken β-actin promoter.
21. The recombinant adeno-associated viral vector of claim 1 , wherein said vector further comprises at least a first enhancer.
22. The recombinant adeno-associated viral vector of claim 21, wherein said vector further comprises a CMV enhancer, a synthetic enhancer, a muscle-specific enhancer, a liver-specific enhancer, or a tissue-specific enhancer.
23. The recombinant adeno-associated viral vector of claim 1, wherein said vector further comprises at least a first intron sequence.
24. The recombinant adeno-associated viral vector of claim 1, wherein said polynucleotide further comprises a 3' regulatory element operably linked to said nucleic acid segment.
25. The recombinant adeno-associated viral vector of claim 24, wherein said 3' regulatory element comprises a woodchuck hepatitis viras post-transcriptional regulatory element (WPRE).
26. The recombinant adeno-associated viral vector of claim 1, wherein said nucleic acid segment is obtained from a human, primate, murine, porcine, bovine, ovine, canine, feline, equine, epine, caprine, avian, or lupine source.
27. The recombinant adeno-associated viral vector of claim 1, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 85% identical to the amino acid sequence of any of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
28. The recombinant adeno-associated viral vector of claim 27, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 90% identical to the amino acid sequence of any of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 15 or SEQ ID NO: 17.
29. The recombinant adeno-associated viral vector of claim 28, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 95% identical to the amino acid sequence of any of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:15 or SEQ ID NO:17.
30. The recombinant adeno-associated viral vector of claim 29, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 98% identical to the amino acid sequence of any of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 15 or SEQ ID NO: 17.
31. The recombinant adeno-associated viral vector of claim 1, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 85% identical to the amino acid sequence of SEQ ID NO:2.
32. The recombinant adeno-associated viral vector of claim 31, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 88% identical to the amino acid sequence of SEQ ID NO:2.
33. The recombinant adeno-associated viral vector of claim 32, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 91% identical to the amino acid sequence of SEQ ID NO:2.
34. The recombinant adeno-associated viral vector of claim 33, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 94% identical to the amino acid sequence of SEQ ID NO:2.
35. The recombinant adeno-associated viral vector of claim 34, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 97% identical to the amino acid sequence of SEQ ID NO:2.
36. The recombinant adeno-associated viral vector of claim 1, comprised within an adeno-associated viral particle.
37. The recombinant adeno-associated viral vector of claim 36, wherein said viral particle is an adeno-associated viral serotype 1 (AAV1), serotype 2 (AAV2), serotype 3 (AAV3), serotype 4 (AAV4), serotype 5 (AAV5), or serotype 6 (AAV6) viral particle.
32. The recombinant adeno-associated viral vector of claim 31, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 88% identical to the amino acid sequence of SEQ ID NO:2.
33. The recombinant adeno-associated viral vector of claim 32, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 91% identical to the amino acid sequence of SEQ ID NO:2.
34. The recombinant adeno-associated viral vector of claim 33, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 94%o identical to the amino acid sequence of SEQ ID NO:2.
35. The recombinant adeno-associated viral vector of claim 34, wherein said nucleic acid segment encodes a biologically-active mammalian Factor VII polypeptide that is at least 97% identical to the amino acid sequence of SEQ ID NO:2.
36. The recombinant adeno-associated viral vector of claim 1, comprised within an adeno-associated viral particle.
37. The recombinant adeno-associated viral vector of claim 36, wherein said viral particle is an adeno-associated viral serotype 1 (AAV1), serotype 2 (AAV2), serotype 3 (AAV3), serotype 4 (AAV4), serotype 5 (AAV5), or serotype 6 (AAV6) viral particle.
-158-
38. The recombinant adeno-associated viral vector of claim 1, comprised within an isolated mammalian host cell.
39. A recombinant adeno-associated virion comprising the recombinant adeno-associated viral vector of claim 1.
40. A plurality of recombinant adeno-associated viral particles comprising the recombinant adeno-associated viral vector of claim 1.
41. A host cell comprising: (a) the recombinant adeno-associated viral vector of claim 1 ; (b) the virion of claim 39; or (c) the plurality of viral particles of claim 40.
42. The host cell of claim 41, wherein said host cell is a bone marrow, liver, kidney, spleen, endothelial, epithelial, heart, lung, pancreatic, cancer, tumor, bone, or blood cell.
43. The host cell of claim 42, wherein said host cell is a human cell.
44. The host cell of claim 43, wherein said host cell is a human liver cell.
-159-
45. A composition comprising: (a) the recombinant adeno-associated viral vector of claim 1; (b) the virion of claim 39; (c) the plurality of viral particles of claim 40, or the host cell of claim 41.
46. The composition of claim 45, further comprising a pharmaceutical excipient.
47. The composition of claim 45, further comprising a microparticle, nanoparticle, microsphere, nanosphere, liposome, lipid, or lipid complex.
48. The composition of claim 45, for use in therapy.
49. The composition of claim 48, for use in the therapy of hemophilia or Factor VII deficiency.
50. The composition of claim 49, for use in the therapy of human hemophilia or human Factor VII deficiency.
51. A kit for treating or ameliorating the symptoms of Factor VII deficiency in a mammal comprising (1) the recombinant adeno-associated viral vector of claim 1; the virion of -160- claim 39; the plurality of viral particles of claim 40; the host cell of claim 41; or the composition of claim 45; and (2) instructions for using said kit.
52. Use of the recombinant adeno-associated viral vector of claim 1, the virion of claim 39, the plurality of viral particles of claim 40, the host cell of claim 41, or the composition of claim 45, in the manufacture of a medicament for treating hemophilia, Factor VII deficiency, or a bleeding disorder in a mammal.
53. The use according to claim 52, wherein said vector, said virion, said particle, said host cell, or said composition is provided to said mammal by injection, infection, or direct administration to a cell, tissue, or organ of said mammal.
54. The use according to claim 53, wherein said mammal is human.
55. The use according to claim 54, wherein said mammal is a human that has, is suspected of having, or at risk for developing hemophilia A.
56. A method for providing an animal a biologically-active Factor VII peptide or polypeptide, said method comprising administering to said mammal: (a) the recombinant adeno-associated viral vector of claim 1; (b) the virion of claim 39; (c) the plurality of viral particles of claim 40; (c) the host cell of claim 41; or (d) the composition of claim 45, in an amount and for a time sufficient to provide said mammal with an effective amount of said biologically-active Factor VII peptide or polypeptide.
-161-
57. The method of claim 56, wherein said mammal has, is at risk for developing, or is diagnosed with hemophilia.
58. A method for treating or ameliorating the symptoms of a Factor VII polypeptide defect, deficiency or dysfunction in a mammal, said method comprising administering to said mammal: (a) the recombinant adeno-associated viral vector of claim 1; (b) the virion of claim 39; (c) the plurality of viral particles of claim 40; (c) the host cell of claim 41; or (d) the composition of claim 45, in an amount and for a time sufficient to treat or ameliorate the symptoms of said defect, deficiency or dysfunction in said mammal.
59. The method of claim 58, wherein said mammal has, is at risk for developing, or is diagnosed with hemophilia, a clotting deficiency, or a bleeding disorder.
60. A method for treating or ameliorating the symptoms of hemophilia in a mammal, said method comprising administering to said mammal: (a) the recombinant adeno- associated viral vector of claim 1; (b) the virion of claim 39; (c) the plurality of viral particles of claim 40; (c) the host cell of claim 41; or (d) the composition of claim 45, in an amount and for a time sufficient to treat or ameliorate the symptoms of hemophilia in said mammal.
-162-
61. The method of claim 60, wherein said mammal has, is at risk for developing, or is diagnosed with hemophilia A.
62. The method of claim 60, wherein said composition is administered to said human intramuscularly, intravenously, or by injection to at least one cell, tissue, or organ of said human.
63. The method of claim 60, wherein said vector, said virion, said particle, said cell, or said composition is provided to said mammal systemically, or by direct or indirect administration to a cell, tissue, or organ of said mammal.
64. The method of claim 63, wherein said vector, said virion, said particle, said cell, or said composition is provided to mammal by direct injection into the muscle tissue, or the liver of said mammal.
-163-
PCT/US2003/020746 2002-06-28 2003-06-30 Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a WO2005014775A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003304396A AU2003304396A1 (en) 2002-06-28 2003-06-30 Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US39272502P 2002-06-28 2002-06-28
US60/392,725 2002-06-28

Publications (2)

Publication Number Publication Date
WO2005014775A2 true WO2005014775A2 (en) 2005-02-17
WO2005014775A3 WO2005014775A3 (en) 2006-02-02

Family

ID=34134984

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/020746 WO2005014775A2 (en) 2002-06-28 2003-06-30 Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a

Country Status (2)

Country Link
AU (1) AU2003304396A1 (en)
WO (1) WO2005014775A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11965175B2 (en) 2017-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6132729A (en) * 1997-01-22 2000-10-17 Board Of Regents, The University Of Texas System Combined tissue factor and chemotherapeutic methods and compositions for coagulation and tumor treatment
US20030013189A1 (en) * 2000-04-28 2003-01-16 Wilson James M. Compositions and methods useful for non-invasive delivery of therapeutic molecules to the bloodstream
US20030228282A1 (en) * 2001-12-17 2003-12-11 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) serotype 8 sequences, vectors containing same, and uses therefor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6132729A (en) * 1997-01-22 2000-10-17 Board Of Regents, The University Of Texas System Combined tissue factor and chemotherapeutic methods and compositions for coagulation and tumor treatment
US20030013189A1 (en) * 2000-04-28 2003-01-16 Wilson James M. Compositions and methods useful for non-invasive delivery of therapeutic molecules to the bloodstream
US20030228282A1 (en) * 2001-12-17 2003-12-11 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) serotype 8 sequences, vectors containing same, and uses therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MARGARITIS V.R. ET AL: 'Long-term expression of activated FVII in vivo following AAV-mediated liver gene transfer: Implications for treatment with continuous infusion of recombinant activated FVII' BLOOD vol. 98, no. 11, November 2001, pages 696 - A, XP008056301 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11613727B2 (en) 2010-10-08 2023-03-28 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11773363B2 (en) 2010-10-08 2023-10-03 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11746319B2 (en) 2010-10-08 2023-09-05 Terumo Bct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US11708554B2 (en) 2013-11-16 2023-07-25 Terumo Bct, Inc. Expanding cells in a bioreactor
US11667876B2 (en) 2013-11-16 2023-06-06 Terumo Bct, Inc. Expanding cells in a bioreactor
US11795432B2 (en) 2014-03-25 2023-10-24 Terumo Bct, Inc. Passive replacement of media
US11667881B2 (en) 2014-09-26 2023-06-06 Terumo Bct, Inc. Scheduled feed
US11608486B2 (en) 2015-07-02 2023-03-21 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11634677B2 (en) 2016-06-07 2023-04-25 Terumo Bct, Inc. Coating a bioreactor in a cell expansion system
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor
US11629332B2 (en) 2017-03-31 2023-04-18 Terumo Bct, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11965175B2 (en) 2017-05-25 2024-04-23 Terumo Bct, Inc. Cell expansion

Also Published As

Publication number Publication date
AU2003304396A8 (en) 2005-02-25
AU2003304396A1 (en) 2005-02-25
WO2005014775A3 (en) 2006-02-02

Similar Documents

Publication Publication Date Title
AU2003295312B2 (en) Improved rAAV expression systems for genetic modification of specific capsid proteins
US20060078542A1 (en) Gel-based delivery of recombinant adeno-associated virus vectors
Wang et al. Immunity to adeno-associated virus-mediated gene transfer in a random-bred canine model of Duchenne muscular dystrophy
ES2629087T3 (en) AAV virions with reduced immunoreactivity and uses thereof
US20090111766A1 (en) Raav vector-based compositions and methods for the prevention and treatment of mammalian diseases
US20060093589A1 (en) Vp2-modified raav vector compositions and uses therefor
US20060292117A1 (en) Improved rAAv vectors
US20090298922A1 (en) Aav transduction of muscle tissue
US20100137211A1 (en) Methods and compositions for intra-articular coagulation proteins
JP2017531652A (en) Gene therapy based on AAV
US20060127358A1 (en) Raav expression systems and methods for enhancing transduction of mammalian neural cells
JP2002516345A (en) Adeno-associated virus vector-mediated expression of factor VIII activity
WO2005014775A2 (en) Raav compositions and methods for delivery of human factor vii polypeptides and treatment of hemophilia a
WO2024060463A1 (en) Improved human coagulation factor viii gene expression cassette and use thereof
US20210093735A1 (en) Methods and compositions for treatment of hemophilia
AU2004226961B2 (en) VP2-modified rAAV vector compositions and uses therefor
US20050271625A1 (en) RAAV-neprilysin compositions and methods of use
US20090068147A1 (en) Raav vector-based pro-opiomelanocortin compositions and methods of use
US20240076691A1 (en) Codon-optimized nucleic acid encoding the fix protein
CN117947040A (en) Expression cassette for target gene and application thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP