AU5175800A - Induction of vascular endothelial growth factor (vegf) by the serine/threonine protein kinase akt - Google Patents

Induction of vascular endothelial growth factor (vegf) by the serine/threonine protein kinase akt Download PDF

Info

Publication number
AU5175800A
AU5175800A AU51758/00A AU5175800A AU5175800A AU 5175800 A AU5175800 A AU 5175800A AU 51758/00 A AU51758/00 A AU 51758/00A AU 5175800 A AU5175800 A AU 5175800A AU 5175800 A AU5175800 A AU 5175800A
Authority
AU
Australia
Prior art keywords
vegf
akt
nucleic acid
protein
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU51758/00A
Other versions
AU773450B2 (en
Inventor
Kenneth Clark
Kun Guo
Yuri Ivashchenko
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aventis Pharmaceuticals Inc
Original Assignee
Aventis Pharmaceuticals 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 Aventis Pharmaceuticals Inc filed Critical Aventis Pharmaceuticals Inc
Publication of AU5175800A publication Critical patent/AU5175800A/en
Application granted granted Critical
Publication of AU773450B2 publication Critical patent/AU773450B2/en
Assigned to AVENTIS PHARMACEUTICALS INC. reassignment AVENTIS PHARMACEUTICALS INC. Amend patent request/document other than specification (104) Assignors: AVENTIS PHARMACEUTICALS PRODUCTS INC.
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/01Phosphotransferases with an alcohol group as acceptor (2.7.1)
    • C12Y207/01037Protein kinase (2.7.1.37)
    • 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
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Enzymes And Modification Thereof (AREA)

Description

WO 00/77190 PCTUSOO/15098 -1 INDUCTION OF VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) BY THE SERINE/THREONINE PROTEIN KINASE AKT FIELD OF THE INVENTION The present invention relates to methods and compositions for therapeutic angiogenesis. More especially, this invention is directed to induction of expression of the angiogenic protein vascular endothelial growth factor (VEGF) by the serine/threonine protein kinase Akt. Preferably, the compositions and methods according to the invention comprise a nucleic acid encoding Akt, and administration thereof. BACKGROUND OF THE INVENTION Angiogenesis Angiogenesis is a biological process resulting in the development of new blood vessels. The process involves various cell-cell, cell-matrix and cell-cytokine interactions (Melillo et al., 1997, Cardiovascular Research 35:480-489; Lewis et al., 1997, Cardiovascular Research 35:490-497). Formation of a new vascular network is normally rare in adult organisms. However, new vasculature may occur under a variety of pathologic conditions, including ischemia, inflammation, wound healing, tumor growth, diabetic retinopathy, rheumatoid arthritis, psoriasis and chronic wounds. Therapeutic angiogenesis involves the deliberate stimulation of new blood vessel development using appropriate angiogenic growth factors. Therefore, therapeutic angiogenesis may be used to treat a variety of ischemic conditions or to stimulate wound healing. Ischemic conditions may affect the heart, lower limbs, skin flaps, peripheral nerves, bone, or grafts. Ischemic conditions include cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, peripheral arterial disease, intermittent claudication, ischemic cardiomyopathy and myocardial ischemia (W097/14307). A variety of factors have been demonstrated to have angiogenic activity. Among these factors are basic and acidic fibroblast growth factors (bFGF and aFGF), FGF-5 (US Pat. 5,661,133), endothelial cell growth factor (Pu et al., 1993, Circulation 88:208-2156), angiopoietin and VEGF (for reviews see Melillo et al., 1997 and Lewis et al., 1997). Angiogenesis has also been suggested as essential for the growth and persistence of solid tumors and their metastases (US Pat. 5,854,205). In order to stimulate angiogenesis, tumors may upregulate the production of a variety of angiogenic factors, including VEGF. VEGF Vascular endothelial cell growth factors are a group of angiogenic polypeptides that are WO 00/77190 PCTUSOO/15098 -2 members of the platelet-derived growth factor family of proteins. These protein are glycosylated cationic dimers having a molecular weight of approximately 46-48 kDa. Unlike other angiogenic factors, VEGF is preceded by a natural signal sequence enabling its secretion from intact cells. Alternative names for VEGF include vascular permeability factor (VPF) and c-fos induced growth factor (FIGF). Several forms of VEGF have been identified, including VEGF1 2 I (US Pat. 5,219,739), VEGFi 65 (US Pat. 5,332,672), VEGF 89 (US Pat. 5,240,848), VEGF 2 0 6 , VEGF-2 (W095/24473; W096/39515), VEGF-B (US Pats. 5,607,918 and 5,840,693), and VEGF-D (W097/12972). Various forms of VEGF have been shown to be mitogenic for vascular endothelial cells and to enhance collateral blood vessel formation and blood flow in ischemic tissue. Transition metal ions, such as CoCb, have been shown to enhance expression of the VEGF gene and to stimulate vascularization (US. Pat. 5,480,975). Akt The AKT proteins are serine/threonine protein kinases that play a role in apoptosis (programmed cell death). Recently, two intracellular signaling pathways involved in the regulation of cell survival/death have been studied. Activation of apoptotic stimulating kinasel (ASK1) leads to apoptosis in various cell types (Ichijo et al. 1997), while a pathway involving phosphoinositide 3 kinase (P13K) and Akt leads to cytoprotection. It has been demonstrated that the activity of ASKI is induced by tumor necrosis factor-alpha (TNFa) treatment or Fas ligation (Ichijo et al. 1997, Chang et al. 1998). Overexpression of ASKI dominant negative mutants inhibit apoptosis induced by TNFa or Fas ligation, indicating that ASKI plays important roles during TNFa or Fas ligation-induced apoptotic cell death. The molecular mechanism by which ASKI induces apoptosis is not clear. It has been shown that ectopic expression of ASK1 leads to activation of various stress-activated signaling pathways, such as the MKK4/JNK and MKK6/p3 8 pathways, which may mediate ASK 1-induced apoptosis (Ichijo et al. 1997). The PI3K/Akt pathway also appears important for regulating cell survival/cell death (Kulik et al. Franke et al 1997, Kauffmann-Zeh et al, Hemmings 1997. Dudek et al. 1997). Survival factors, such as platelet derived growth factor (PDGF), nerve growth factor (NGF) and insulin-like growth factor-I (IGF-1), promote cell survival under various conditions by inducing the activity of P13K (Kulik et al. 1997, Hemmings 1997). Activated P13K leads to the production of phosphatidylinositol (3,4,5)-triphosphate (Ptdlns(3,4,5)-P3), which in turn binds to and induces the activity of a AH/PH domain containing serine/threonine kinase, Akt (Franke et al 1995, Hemmings 1997b, Downward 1998, Alessi et al. 1996). Specific inhibitors of P13K or dominant negative Akt mutants abolish survival-promoting activity of these growth factors or cytokines. In addition, introduction of WO 00/77190 PCTIUSOO/15098 -3 constitutively active P13K or Akt mutants promotes cell survival under conditions in which cells normally undergo apoptotic cell death (Kulik et al. 1997, Dudek et al. 1997). These observations demonstrate that the PI3K/Akt pathway plays important roles for regulating cell survival or apoptosis. Two isoforms of human Akt protein kinases, Aktl and Akt2, have been identified in the literature (Staal. 1987). A third form of human Akt, designated Akt3, is described in US Provisional application number 60/125,108). Yet another isoform of Akt is described in Nakatani et al., 1999 (Biochem. Biophys. Res. Comm. 257, 906-910). A rat Akt sequence has also been identified (Konishi et al. 1995). Serine-473 in the C-terminus of human Aktl has been shown to be critical for its regulation (Stokeo et al. 1997; Stephens et al. 1998). Upon growth factor stimulation, P13K is activated. The product of P13K, Ptdlns(3.4.5)-P binds Aktl, and causes translocation of Aktl from the cytoplasm to the proximity of the inner cytoplasmic membrane, where it becomes phosphorylated at residues Thr308 and Ser473 (Downward, 1998). Phosphorylation of these residues is critical for the activation of Aktl. A recently identified protein kinase, PDKl, has been shown to be responsible for the phosphorylation of Thr308, while the kinase(s) which phosphorylates Ser473 has not yet been identified (Stokeo et al. 1997, Stephens et al. 1998). Gene Therapy Gene therapy involves correcting a deficiency or abnormality (mutation, aberrant expression, and the like) by introduction of genetic information into a patient, such as into an affected cell or organ of the patient. This genetic information may be introduced either in vitro into a cell, the modified cell then being reintroduced into the body, or directly in vivo into an appropriate site. In this regard, different techniques of cell transfection and of gene transfer have been described in the literature (see Roemer and Friedman, Eur. J. Biochem. 208 (1992) 211), including transfection of "naked DNA" and various techniques involving complexes of DNA and DEAE-dextran (Pagano et al., J.Virol. 1 (1967) 891), of DNA and nuclear proteins (Kaneda et al., Science 243 (1989) 375), of DNA and lipids (Felgner et al., PNAS 84 (1987) 7413), the use of liposomes (Fraley et al., J.Biol.Chem. 255 (1980) 10431) and the like. More recently, the use of viruses as vectors for the transfer of genes has emerged as a promising alternative to physical transfection techniques. In this regard, different viruses have been tested for their capacity to infect certain cell populations, including retroviruses, herpes viruses, adeno-associated viruses, and adenoviruses. Gene therapy for angiogenesis, specifically employing a sequence encoding a VEGF has been proposed (Melillo et al., 1997; Lewis et al., 1997). Intraarterial or intramuscular administration of a plasmid comprising the cDNA for VEGF 6 S increases collateral blood flow in an ischemic rabbit hindlimb model (Tsurumi et al., 1996, Circulation 94:3281-3290; Takeshita et al., 1996, Biochem.
WO 00/77190 PCT/USOO/15098 -4 Biophys. Res. Commun. 227:628-635). Plasmids comprising the cDNAs for human VEGF1I2,VEGF 65 , and VEGFi 89 have also been shown to have angiogenic activity in this model (Takeshita et al., 1996, Lab Invest. 75:487-501; W097/14307). Similarly, a replication defective adenovirus comprising a VEGF 65 coding sequence induces neovascularization in mice (Muhlhauser et al., 1995, Circ. Res. 77: 1077-1086). In human patients, a plasmid comprising a sequence for
VEGF
165 has been shown to induce angiogenesis in ischemic limbs (Isner et al., 1996, Lancet 348: 370-374) and in the heart (see Time magazine, January 11, 1999, pp. 68-73). The present invention relates to an alternative method for stimulating angiogenesis, rather than by direct administration of a VEGF coding sequence. More specifically, Applicants have unexpectedly discovered that VEGF production is induced by the protein Akt. Therefore, the present invention is directed to stimulating expression of a VEGF in a cell by introducing an Akt protein into the cell. Preferably, the cells are present in a patient suffering from an ischemic condition, and the result is beneficial collateral blood vessel formation in the patient. The citation of any reference herein should not be construed as an admission that such reference is available as "Prior Art" to the instant application. SUMMARY OF THE INVENTION The present invention relates to methods and compositions for stimulating expression of a VEGF in a cell. More specifically, Applicants have unexpectedly discovered that Akt proteins are able to stimulate VEGF expression. In its most preferred aspect, the cells are present in a patient suffering from an ischemic condition, and the result is beneficial collateral blood vessel formation in the patient. Therefore, a first subject of the invention relates to a method of inducing expression of VEGF in a cell by administering to the cell an Akt protein. The protein may be any Akt protein. Preferably, the Akt protein is a human Akt protein. More preferably, the Akt protein is human Aktl, Akt2 or Akt3. The VEGF produced upon administration of an Akt protein may be any form of VEGF capable of stimulating angiogenesis. Preferably, the VEGF is VEGF 1 2 i, VEGF 1 6 5, VEGF 189 , VEGF 2 0 6 , VEGF-2, VEGF-B, or VEGF-D. In one aspect, Akt protein is administered to cells. In a preferred embodiment, a nucleic acid encoding an Akt protein, and operatively associated with an expression control sequence, is administered to cells. The nucleic acid may be part of a plasmid or viral vector. Preferred viral vectors are retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus. The Akt protein, or nucleic acid encoding an Akt protein, may be administered alone or in WO 00/77190 PCTIUSOO/15098 -5 combination with a transition metal ion and/or a vasodilator. The Akt protein, or a nucleic acid encoding an Akt protein may also be administered with a nucleic acid encoding a second angiogenic factor operatively associated with an expression control sequence. Preferred angiogenic factors include a VEGF, acidic fibroblast growth factor, basic fibroblast growth factor, endothelial cell growth factor, or an angiopoietin. In another aspect, more than one form of Akt protein may be administered to the cell. This invention also relates to a method of inducing expression of VEGF in cells of a patient suffering from an ischemic condition by administering to the patient an Akt protein. The ischemic condition may be cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, or ischemic, idiopathic or hypertrophic cardiomyopathy. The protein may be any Akt protein. Preferably, the Akt protein is Akt I, Akt2 or Akt3. The VEGF produced upon administration of an Akt protein may be any form of VEGF capable of stimulating angiogenesis. Preferably, the VEGF is VEGF121, VEGFi 6 5 , VEGF 8 9 , VEGF 2 0 6 , VEGF-2, VEGF-B, or VEGF-D. In one aspect, Akt protein is administered to the patient. In a preferred embodiment, a nucleic acid encoding an Akt protein, and operatively associated with an expression control sequence, is administered to the patient. The nucleic acid may be part of a plasmid or viral vector. Preferred viral vectors are retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus. In a most preferred embodiment, a nucleic acid encoding the Akt protein is administered directly into cardiac tissue by transepicardial surgical administration or by percutaneous delivery using a catheter. The Akt protein, or nucleic acid encoding an Akt protein, may be administered to the patient alone or in combination with a transition metal ion and/or a vasodilator. The Akt protein, or a nucleic acid encoding an Akt protein may also be administered to a patient with a nucleic acid encoding a second angiogenic factor operatively associated with an expression control sequence. Preferred angiogenic factors include a VEGF, acidic fibroblast growth factor, basic fibroblast growth factor, endothelial cell growth factor, or an angiopoietin. In another aspect, more than one form of Akt protein may be administered to the patient. The present invention also relates to pharmaceutical compositions comprising a nucleic acid encoding an Akt protein, a transition metal and/or a vasodilator and a pharmaceutically acceptable vehicle. The nucleic acid may be part of a plasmid or viral vector. Preferred viral vectors are retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus. In another aspect, the invention relates to a method of inhibiting angiogenesis in a patient suffering from a tumor by inhibiting the level of Akt activity in the patient, thereby inhibiting production of VEGF. The level of Akt may be decreased by introducing an Akt antisense nucleic acid WO 00/77190 PCTIUS0O/15098 -6 into cells of the patient under conditions wherein the antisense nucleic acid hybridizes under intracellular conditions to an Akt mRNA. The level of Akt can also be decreased by introducing an intracellular binding protein, such as a single chain Fv antibody (scFv), that specifically binds Akt into a patient's cell at a level sufficient to bind to and inactivate Akt. In another embodiment, Akt activity can be decreased by administering a nucleic acid encoding a dominant negative form of an Akt. Preferably, the antisense nucleic acid, intracellular binding protein or nucleic acid encoding therefor, or dominant negative are administered directly to tumor cells. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1: Construction of activated Akt3 mutant. Figure 1 A: Schematic presentation of activated Akt3: Full length coding sequence of human Akt3 was fused in frame with the Myristylation signal from human Src gene (Myr) in the N terminal, and fused in frame with the HA-tag in the C-terminus (HA). (see the Examples) Figure IB: Ectopic expression of activated Akt3 in HEK293 cells. HEK293 cells were transfected with either CMV6-MyrAkt3HA or expression plasmid (CMV6) alone. 24 hours after transfections, cell lysates were prepared and subjected to immunoblotting with a-HA antibodies. Figure IC: Activated Akt3 possesses Akt activity. HEK293 cells were transfected with expression plasmid for activated Akt3 (MyrAkt3HA) or expression vector alone (CMV6). 24 hours after transfections, cell lysates were prepared and subjected to immunoprecipitation with anti-HA antibodies. Akt3 kinase activities of immunopellets were measured by using substrate peptide derived from GSK3. Bkgd: background level from non-transfected cells; CMV6: CMV6 transfected cells; Akt3cak: cells transfected with expression plasmid for constitutively activated Akt3 (CMV6-MyrAkt3HA). (see the Examples). Figure 2: Akt increases VEGF-165 secretion from HeLa cells HeLa cells were transfected with expression plasmid for activated mouse Aktl (Aktl), activated human Akt3 (Akt3) or CMV6 vector alone. One day after transfection, cells were switched to low mitogen media (DMEM-0.5%FBS). 16 hours later, culture media was collected for VEGF ELISA assay. Lane A: cells (6-well tissue culture dish) transfected with 0.4 pIg CMV6 vector DNA; Lane Aktl: cells (6-well tissue culture dish) transfected with 0.4 pg CMV6 mAktlcak expression plasmid; Lane Akt3: cells (in 6-well tissue culture dish) transfected with expression plasmid for activated human Akt3 (Akt3).
WO 00/77190 PCT/USOO/15098 -7 Figure 3: Akt increases VEGF-165 expression in human coronary smooth muscle cells and human skeletal muscle cells Figure 3A: Human skeletal muscle cells (HSKMCs) were infected with recombinant adenoviruses for green fluorescence protein (AV-GFP), constitutively active mouse Aktl (AV mAktlcak) or constitutively active human Akt3 (AV-hAkt3cak) at the concentration of 3x10 8 VP/ml overnight (VP: viral particles). One day after infection, media were collected and VEGF levels in the media were measured by ELISA assay. Figure 3B: Human coronary artery smooth muscle cells (HCASMCs) were infected AV-GFP, AV mAktlcak, AV-hAkt3cak at the concentration of 3xl0 8 VP/ml overnight. One day after infections, media were collected and VEGF levels in the media were measured by ELISA assay for human VEGF. Figure 3C: HCASMCs were infected with indicated viruses at concentration of 3x10VP/ml overnight. As a control, non-infected cells were switched to hypoxia condition. One day later, total RNA was isolated from these cells and VEGF expression were detected by Northern blot analysis. Figure 4: Akt increase VEGF expression in rat cardiomyocytes Neonatal cardiomyocytes were infected with recombinant adenoviruses for green fluorescence protein (AV-GFP), constitutively active mouse Aktl (AV-mAktlcak) or constitutively active human Akt3 (AV-hAkt3cak) at a concentration of 3x10'VP/ml overnight. As a control, non-infected cells were subjected hypoxia treatment for 24 hours. One day after infection, total RNA was isolated and VEGF expression was detected by Northern blot analysis. DETAILED DESCRIPTION OF THE INVENTION The present invention advantageously provides methods and compositions for stimulating expression of a VEGF in a cell. Therefore, the invention enables the treatment of ischemic disease in patients by providing the means and methodology for stimulating collateral blood vessel formation in ischemic tissue. More specifically, Akt proteins are shown herein to stimulate the expression of the angiogenic protein VEGF. Therefore, a first subject of the invention relates to stimulation of VEGF expression in cells by introducing an Akt protein to the cells. In a preferred aspect, a nucleic acid encoding the Akt protein is administered to cells. The invention also relates to the treatment of a patient suffering from an ischemic condition by administering to the patient an Akt protein. Preferably, a nucleic acid encoding the Akt protein is administered to the patient, and the result is beneficial collateral blood vessel formation in ischemic tissue in the patient.
WO 00/77190 PCT/USOO/15098 -8 The various aspects of the invention will be set forth in greater detail in the following sections. This organization into various sections is intended to facilitate understanding of the invention, and is in no way intended to be limiting thereof. Definitions The following defined terms are used throughout the present specification, and should be helpful in understanding the scope and practice of the present invention. In a specific embodiment, the term "about" or "approximately" means within 20%, preferably within 10%, and more preferably within 5% of a given value or range. A "nucleic acid" is a polymeric compound comprised of covalently linked subunits called nucleotides. Nucleic acid includes polyribonucleic acid (RNA) and polydeoxyribonucleic acid (DNA), both of which may be single-stranded or double-stranded. DNA includes cDNA, genomic DNA, synthetic DNA, and semi-synthetic DNA. A "gene" refers to an assembly of nucleotides that encode a polypeptide, and includes cDNA and genomic DNA nucleic acids. A "recombinant DNA molecule" is a DNA molecule that has undergone a molecular biological manipulation. A "vector" is any means for the transfer of a nucleic acid into a host cell. A vector may be a replicon to which another DNA segment may be attached so as to bring about the replication of the attached segment. A "replicon" is any genetic element (e.g., plasmid, phage, cosmid, chromosome, virus) that functions as an autonomous unit of DNA replication in vivo, i.e., capable of replication under its own control. The term "vector" includes both viral and nonviral means for introducing the nucleic acid into a cell in vitro, ex vivo or in vivo. Viral vectors include retrovirus, adeno-associated virus, pox, baculovirus, vaccinia, herpes simplex, Epstein-Barr and adenovirus vectors, as set forth in greater detail below. Non-viral vectors include plasmids, liposomes, electrically charged lipids (cytofectins), DNA-protein complexes, and biopolymers. In addition to a nucleic acid, a vector may also contain one or more regulatory regions, and/or selectable markers useful in selecting, measuring, and monitoring nucleic acid transfer results (transfer to which tissues, duration of expression, etc.). A "cloning vector" is a replicon, such as plasmid, phage or cosmid, to which another DNA segment may be attached so as to bring about the replication of the attached segment. Cloning vectors may be capable of replication in one cell type, and expression in another ("shuttle vector"). A "cassette" refers to a segment of DNA that can be inserted into a vector at specific restriction sites. The segment of DNA encodes a polypeptide of interest, and the cassette and restriction sites are designed to ensure insertion of the cassette in the proper reading frame for transcription and translation.
WO 00/77190 PCT/USOO/15098 -9 A cell has been "transfected" by exogenous or heterologous DNA when such DNA has been introduced inside the cell. A cell has been "transformed" by exogenous or heterologous DNA when the transfected DNA effects a phenotypic change. The transforming DNA can be integrated (covalently linked) into chromosomal DNA making up the genome of the cell. A "nucleic acid molecule" refers to the phosphate ester polymeric form of ribonucleosides (adenosine, guanosine, uridine or cytidine; "RNA molecules") or deoxyribonucleosides (deoxyadenosine, deoxyguanosine, deoxythymidine, or deoxycytidine; "DNA molecules"), or any phosphoester anologs thereof, such as phosphorothioates and thioesters, in either single stranded form, or a double-stranded helix. Double stranded DNA-DNA, DNA-RNA and RNA-RNA helices are possible. The term nucleic acid molecule, and in particular DNA or RNA molecule, refers only to the primary and secondary structure of the molecule, and does not limit it to any particular tertiary forms. Thus, this term includes double-stranded DNA found, inter alia, in linear or circular DNA molecules (e.g., restriction fragments), plasmids, and chromosomes. In discussing the structure of particular double-stranded DNA molecules, sequences may be described herein according to the normal convention of giving only the sequence in the 5' to 3' direction along the nontranscribed strand of DNA (i.e., the strand having a sequence homologous to the mRNA). A "recombinant DNA molecule" is a DNA molecule that has undergone a molecular biological manipulation. A nucleic acid molecule is "hybridizable" to another nucleic acid molecule, such as a cDNA, genomic DNA, or RNA, when a single stranded form of the nucleic acid molecule can anneal to the other nucleic acid molecule under the appropriate conditions of temperature and solution ionic strength (see Sambrook et al., supra). The conditions of temperature and ionic strength determine the "stringency" of the hybridization. For preliminary screening for homologous nucleic acids, low stringency hybridization conditions, corresponding to a Tm of 55', can be used, e.g., 5x SSC, 0.1% SDS, 0.25% milk, and no formamide; or 30% formamide, 5x SSC, 0.5% SDS). Moderate stringency hybridization conditions correspond to a higher Tm, e.g., 40% formamide, with 5x or 6x SCC. High stringency hybridization conditions correspond to the highest Tm, e.g., 50% formamide, 5x or 6x SCC. Hybridization requires that the two nucleic acids contain complementary sequences, although depending on the stringency of the hybridization, mismatches between bases are possible. The appropriate stringency for hybridizing nucleic acids depends on the length of the nucleic acids and the degree of complementation, variables well known in the art. The greater the degree of similarity or homology between two nucleotide sequences, the greater the value of Tm for hybrids of nucleic acids having those sequences. The relative stability (corresponding to higher Tm) of nucleic acid hybridizations decreases in the following order: RNA:RNA, DNA:RNA, DNA:DNA. For hybrids of greater than 100 nucleotides in length, equations for calculating Tm have been derived (see Sambrook WO 00/77190 PCT/USOO/15098 -10 et al., supra, 9.50-0.51). For hybridization with shorter nucleic acids, i.e., oligonucleotides, the position of mismatches becomes more important, and the length of the oligonucleotide determines its specificity (see Sambrook et al., supra, 11.7-11.8). Preferably a minimum length for a hybridizable nucleic acid is at least about 10 nucleotides; preferably at least about 15 nucleotides; and more preferably the length is at least about 20 nucleotides. In a specific embodiment, the term "standard hybridization conditions" refers to a Tm of 55 0 C, and utilizes conditions as set forth above. In a preferred embodiment, the Tm is 60'C; in a more preferred embodiment, the Tm is 65 0 C. As used herein, the term "oligonucleotide" refers to a nucleic acid, generally of at least 18 nucleotides, that is hybridizable to a genomic DNA molecule, a cDNA molecule, or an mRNA molecule encoding Akt. Oligonucleotides can be labeled, e.g., with 32 P-nucleotides or nucleotides to which a label, such as biotin, has been covalently conjugated. In one embodiment, a labeled oligonucleotide can be used as a probe to detect the presence of a nucleic acid encoding Akt. In another embodiment, oligonucleotides (one or both of which may be labeled) can be used as PCR primers, either for cloning full length or a fragment of Akt, or to detect the presence of nucleic acids encoding Akt. In a further embodiment, an oligonucleotide of the invention can form a triple helix with an Akt DNA molecule. Generally, oligonucleotides are prepared synthetically, preferably on a nucleic acid synthesizer. Accordingly, oligonucleotides can be prepared with non-naturally occurring phosphoester analog bonds, such as thioester bonds, etc. A DNA "coding sequence" is a double-stranded DNA sequence which is transcribed and translated into a polypeptide in a cell 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 translation stop codon at the 3' (carboxyl) terminus. A coding sequence can include, but is not limited to, prokaryotic sequences, cDNA from eukaryotic mRNA, genomic DNA sequences from eukaryotic (e.g., mammalian) DNA, and even synthetic DNA sequences. If the coding sequence is intended for expression in a eukaryotic cell, a polyadenylation signal and transcription termination sequence will usually be located 3' to the coding sequence. Transcriptional and translational control sequences are DNA regulatory sequences, such as promoters, enhancers, terminators, and the like, that provide for the expression of a coding sequence in a host cell. In eukaryotic cells, polyadenylation signals are control sequences. A "promoter sequence" is a DNA regulatory region capable of binding RNA polymerase in a cell and initiating transcription of a downstream (3' direction) coding sequence. For purposes of defining the present invention, the promoter sequence is bounded at its 3' terminus by the transcription initiation site and extends upstream (5' direction) to include the minimum number of WO 00/77190 PCT/USOO/15098 - 11 bases or elements necessary to initiate transcription at levels detectable above background. Within the promoter sequence will be found a transcription initiation site (conveniently defined for example, by mapping with nuclease Si), as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase. A coding sequence is "under the control" of transcriptional and translational control sequences in a cell when RNA polymerase transcribes the coding sequence into mRNA, which is then trans-RNA spliced (if the coding sequence contains introns) and translated into the protein encoded by the coding sequence. As used herein, the term "homologous" in all its grammatical forms and spelling variations refers to the relationship between proteins that possess a "common evolutionary origin," including proteins from superfamilies (e.g., the immunoglobulin superfamily) and homologous proteins from different species (e.g., myosin light chain, etc.) (Reeck et al., 1987, Cell 50:667). Such proteins (and their encoding genes) have sequence homology, as reflected by their high degree of sequence similarity. Accordingly, the term "sequence similarity" in all its grammatical forms refers to the degree of identity or correspondence between nucleic acid or amino acid sequences of proteins that may or may not share a common evolutionary origin (see Reeck et al., supra). However, in common usage and in the instant application, the term "homologous," when modified with an adverb such as "highly," may refer to sequence similarity and not a common evolutionary origin. In a specific embodiment, two DNA sequences are "substantially homologous" or "substantially similar" when at least about 50% (preferably at least about 75%, and most preferably at least about 90 or 95%) of the nucleotides match over the defined length of the DNA sequences. Sequences that are substantially homologous can be identified by comparing the sequences using standard software available in sequence data banks, or in a Southern hybridization experiment under, for example, stringent conditions as defined for that particular system. Defining appropriate hybridization conditions is within the skill of the art. See, e.g., Maniatis et al., supra; DNA Cloning, Vols. I & II, supra; Nucleic Acid Hybridization, supra. An "antisense nucleic acid" is a sequence of nucleotides that is complementary to the sense sequence. Antisense nucleic acids can be used to down regulate or block the expression of the polypeptide encoded by the sense strand. Transcriptional and translational control sequences are DNA regulatory sequences, such as promoters, enhancers, terminators, and the like, that provide for the expression of a coding sequence in a host cell. In eukaryotic cells, polyadenylation signals are additional types of control sequences. A "signal sequence" is included at the beginning of the coding sequence of a protein to be WO 00/77190 PCT/USOO/15098 -12 expressed on the surface of a cell. This sequence encodes a signal peptide, N-terminal to the mature polypeptide, that directs the host cell to translocate the polypeptide. The term "translocation signal sequence" is used herein to refer to this sort of signal sequence. Translocation signal sequences can be found associated with a variety of proteins native to eukaryotes and prokaryotes, and are often functional in both types of organisms. "Regulatory region" means a nucleic acid sequence which regulates the expression of a second nucleic acid sequence. A regulatory region may include sequences which are naturally responsible for expressing a particular nucleic acid (a homologous region) or may include sequences of a different origin which are responsible for expressing different proteins or even synthetic proteins (a heterologous region). In particular, the sequences can be sequences of eukaryotic or viral genes or derived sequences which stimulate or repress transcription of a gene in a specific or non-specific manner and in an inducible or non-inducible manner. Regulatory regions include origins of replication, RNA splice sites, promoters, enhancers, transcriptional termination sequences, signal sequences which direct the polypeptide into the secretory pathways of the target cell, and promoters. A regulatory region from a "heterologous source" is a regulatory region which is not naturally associated with the expressed nucleic acid. Included among the heterologous regulatory regions are regulatory regions from a different species, regulatory regions from a different gene, hybrid regulatory sequences, and regulatory sequences which do not occur in nature, but which are designed by one having ordinary skill in the art. "Heterologous" DNA refers to DNA not naturally located in the cell, or in a chromosomal site of the cell. Preferably, the heterologous DNA includes a gene foreign to the cell. "Homologous recombination" refers to the insertion of a foreign DNA sequence into another DNA molecule, e.g., insertion of a vector in a chromosome. Preferably, the vector targets a specific chromosomal site for homologous recombination. For specific homologous recombination, the vector will contain sufficiently long regions of homology to sequences of the chromosome to allow complementary binding and incorporation of the vector into the chromosome. Longer regions of homology, and greater degrees of sequence similarity, may increase the efficiency of homologous recombination. A "polypeptide" is a polymeric compound comprised of covalently linked amino acid residues. Amino acids have the following general structure: H R-C-COOH
NH
2 Amino acids are classified into seven groups on the basis of the side chain R: (1) aliphatic side WO 00/77190 PCT/USOO/15098 -13 chains, (2) side chains containing a hydroxylic (OH) group, (3) side chains containing sulfur atoms, (4) side chains containing an acidic or amide group, (5) side chains containing a basic group, (6) side chains containing an aromatic ring, and (7) proline, an imino acid in which the side chain is fused to the amino group. A polypeptide of the invention preferably comprises at least about 14 amino acids. A "protein" is a polypeptide which plays a structural or functional role in a living cell. A "variant" of a polypeptide or protein is any analogue, fragment, derivative, or mutant which is derived from a polypeptide or protein and which retains at least one biological property of the polypeptide or protein. Different variants of the polypeptide or protein may exist in nature. These variants may be allelic variations characterized by differences in the nucleotide sequences of the structural gene coding for the protein, or may involve differential splicing or post-translational modification. The skilled artisan can produce variants having single or multiple amino acid substitutions, deletions, additions, or replacements. These variants may include, inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids, (b) variants in which one or more amino acids are added to the polypeptide or protein, (c) variants in which one or more of the amino acids includes a substituent group, and (d) variants in which the polypeptide or protein is fused with another polypeptide such as serum albumin. The techniques for obtaining these variants, including genetic (suppressions, deletions, mutations, etc.), chemical, and enzymatic techniques, are known to persons having ordinary skill in the art. If such allelic variations, analogues, fragments, derivatives, mutants, and modifications, including alternative mRNA splicing forms and alternative post-translational modification forms result in derivatives of the polypeptide which retain any of the biological properties of the polypeptide, they are intended to be included within the scope of this invention. A "heterologous protein" refers to a protein not naturally produced in the cell. Two amino acid sequences are "substantially homologous" or "substantially similar" when greater than about 40% of the amino acids are identical, or greater than 60% are similar (functionally identical). Preferably, the similar or homologous sequences are identified by alignment using, for example, the GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wisconsin) pileup program. The term "corresponding to" is used herein to refer to similar or homologous sequences, whether the exact position is identical or different from the molecule to which the similarity or homology is measured. A nucleic acid or amino acid sequence alignment may include spaces. Thus, the term "corresponding to" refers to the sequence similarity, and not the numbering of the amino acid residues or nucleotide bases.
WO 00/77190 PCT/USOO/15098 -14 Genes Encoding Akt Proteins The present invention contemplates the use of an Akt protein or polypeptide, or a nucleic acid encoding an Akt protein or polypeptide to stimulate expression of VEGF in cells. Preferably, the Akt is a human Akt3 protein or polypeptide, including a full length, or naturally occurring form of Akt, or any fragment thereof capable of stimulating expression of VEGF. As used herein, "Akt" refers to Akt polypeptide, and "akt" refers to a gene encoding an Akt polypeptide. Various mouse and human Akt sequences are known in the art (see Coffer et al., 1991, Eur. J. Biochem. 201:475-481; Jones et al., 1991, Proc. Natl. Acad. Sci. 88:4171-4175; Bellacosa et al., 1993, Oncogene, 8:745-754; GenBank Accession Nos. M63167, X61037 and X65687; and US Provisional application number 60/125,108). Preferably, the Akt is human Aktl (SEQ ID NO: 11), Akt2 (SEQ ID NO:12) or Akt3 (SEQ ID NO:2). A preferred Akt according to the invention comprises an amino acid sequence as shown in SEQ ID NO: 2. A preferred nucleic acid according to the invention encodes an amino acid sequence as shown in SEQ ID NO: 2, SEQ ID NO: 11 or SEQ ID NO: 12. More preferably, the nucleic acid comprises a sequence as depicted in SEQ ID NO: 1. The Akt can also be derived from a non-human source. In accordance with the present invention there may be employed conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (herein "Sambrook et al., 1989"); DNA Cloning: A Practical Approach, Volumes I and II (D.N. Glover ed. 1985); Oligonucleotide Synthesis (M.J. Gait ed. 1984); Nucleic Acid Hybridization [B.D. Hames & S.J. Higgins eds. (1985)]; Transcription And Translation [B.D. Hames & S.J. Higgins, eds. (1984)]; Animal Cell Culture [R.I. Freshney, ed. (1986)]; Immobilized Cells And Enzymes [lRL Press, (1986)]; B. Perbal, A Practical Guide To Molecular Cloning (1984); F.M. Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (1994). A gene encoding an Akt, whether genomic DNA or cDNA, can be isolated from any source, particularly from a human cDNA or genomic library. General methods for obtaining an akt gene are well known in the art, as described above (see, e.g., Sambrook et al., 1989, supra). Accordingly, any animal cell potentially can serve as the nucleic acid source for the molecular cloning of a akt gene. The DNA may be obtained by standard procedures known in the art from cloned DNA (e.g., a DNA "library"), and preferably is obtained from a cDNA library prepared from tissues with high level expression of the protein (e.g., heart, pancreas and skeletal muscle cDNA), by chemical synthesis, by cDNA cloning, or by the cloning of genomic DNA, or fragments thereof, purified from the desired cell (See, for example, Sambrook et al., 1989, supra; Glover, D.M. (ed.), WO 00/77190 PCT/USOO/15098 -15 1985, DNA Cloning: A Practical Approach, MRL Press, Ltd., Oxford, U.K. Vol. I, II). Clones derived from genomic DNA may contain regulatory and intron DNA regions in addition to coding regions; clones derived from cDNA will not contain intron sequences. Whatever the source, the gene should be molecularly cloned into a suitable vector for propagation of the gene. Once the DNA fragments are generated, identification of the specific DNA fragment containing the desired akt gene may be accomplished in a number of ways. For example, DNA fragments may be screened by nucleic acid hybridization to a labeled probe (Benton and Davis, 1977, Science 196:180; Grunstein and Hogness, 1975, Proc. Natl. Acad. Sci. U.S.A. 72:3961). Those DNA fragments with substantial homology to the probe will hybridize. As noted above, the greater the degree of homology, the more stringent hybridization conditions can be used. In a specific embodiment, Northern hybridization conditions are used to identify mRNA splicing variants of an akt gene. Further selection can be carried out on the basis of the properties of the gene, e.g., if the gene encodes a protein product having the isoelectric, electrophoretic, amino acid composition, or partial amino acid sequence of Akt protein as disclosed herein. Thus, the presence of the gene may be detected by assays based on the physical, chemical, or immunological properties of its expressed product. For example, cDNA clones, or DNA clones which hybrid-select the proper mRNAs, can be selected which produce a protein that, e.g., has similar or identical electrophoretic migration, isoelectric focusing or non-equilibrium pH gel electrophoresis behavior, proteolytic digestion maps, or antigenic properties as known for Akt. In a specific embodiment, the expressed protein is recognized by a polyclonal antibody that is generated against an epitope specific for human Akt. The present invention also relates to the use of genes (e.g., cDNAs) encoding allelic variants, splicing variants, analogs, and derivatives of Akt, that have the ability to stimulate the expression of a VEGF. The production and use of Akt derivatives and analogs are within the scope of the present invention. Such variants, analogs, derivatives and homologs should retain the ability to stimulate expression of a VEGF. Akt derivatives can be made by altering encoding nucleic acid sequences by substitutions, additions or deletions that provide for functionally equivalent molecules. Preferably, derivatives are made that have enhanced or increased functional activity relative to native Akt. Due to the degeneracy of nucleotide coding sequences, other DNA sequences which encode substantially the same amino acid sequence as a akt gene, including an amino acid sequence that contains a single amino acid variant, may be used in the practice of the present invention. These include but are not limited to allelic genes, homologous genes from other species, and nucleotide sequences comprising all or portions of akt genes which are altered by the substitution of different WO 00/77190 PCT/USOO/15098 -16 codons that encode the same amino acid residue within the sequence, thus producing a silent change. Likewise, the Akt derivatives of the invention include, but are not limited to, those containing, as a primary amino acid sequence, all or part of the amino acid sequence of a Akt protein including altered sequences in which functionally equivalent amino acid residues are substituted for residues within the sequence resulting in a conservative amino acid substitution. For example, one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity, which acts as a functional equivalent, resulting in a silent alteration. Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs. For example, the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine. Amino acids containing aromatic ring structures are phenylalanine, tryptophan, and tyrosine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine. The positively charged (basic) amino acids include arginine, lysine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Such alterations will not be expected to affect apparent molecular weight as determined by polyacrylamide gel electrophoresis, or isoelectric point. Particularly preferred substitutions are: - Lys for Arg and vice versa such that a positive charge may be maintained; - Glu for Asp and vice versa such that a negative charge may be maintained; - Ser for Thr such that a free -OH can be maintained; and - Gln for Asn such that a free CONH, can be maintained. Amino acid substitutions may also be introduced to substitute an amino acid with a particularly preferable property. For example, a Cys may be introduced in order to add a potential site for disulfide bridges with another Cys. A His may be introduced as a particularly "catalytic" site (i.e., His can act as an acid or base and is the most common amino acid in biochemical catalysis). Pro may be introduced because of its particularly planar structure, which induces p-turns in the protein's structure. The genes encoding Akt derivatives and analogs of the invention can be produced by various methods known in the art. The manipulations which result in their production can occur at the gene or protein level. For example, the cloned Akt gene sequence can be modified by any of numerous strategies known in the art (Sambrook et al., 1989, supra). The sequence can be cleaved at appropriate sites with restriction endonuclease(s), followed by further enzymatic modification if desired, isolated, and ligated in vitro. In the production of the gene encoding a derivative or analog of Akt, care should be taken to ensure that the modified gene remains within the same translational reading frame as the Akt gene, uninterrupted by translational stop signals, in the gene region where the WO 00/77190 PCT/USOO/15098 -17 desired activity is encoded. Additionally, the Akt-encoding nucleic acid sequence can be mutated in vitro or in vivo, to create and/or destroy translation, initiation, and/or termination sequences, or to create variations in coding regions and/or form new restriction endonuclease sites or destroy preexisting ones, to facilitate further in vitro modification. Preferably, such mutations enhance the functional activity of the mutated Akt gene product. Any technique for mutagenesis known in the art can be used, including but not limited to, in vitro site-directed mutagenesis (Hutchinson, C., et al., 1978, J. Biol. Chem. 253:6551; Zoller and Smith, 1984, DNA 3:479-488; Oliphant et al., 1986, Gene 44:177; Hutchinson et al., 1986, Proc. Natl. Acad. Sci. U.S.A. 83:710), use of TAB® linkers (Pharmacia), etc. PCR techniques are preferred for site directed mutagenesis (see Higuchi, 1989, "Using PCR to Engineer DNA", in PCR Technology: Principles and Applications for DNA Amplification, H. Erlich, ed., Stockton Press, Chapter 6, pp. 61-70). The identified and isolated gene can then be inserted into an appropriate cloning vector. A large number of vector-host systems known in the art may be used. Possible vectors include, but are not limited to, plasmids or modified viruses, but the vector system must be compatible with the host cell used. Examples of vectors include, but are not limited to, E. coli, bacteriophages such as lambda derivatives, or plasmids such as pBR322 derivatives or pUC plasmid derivatives, e.g., pGEX vectors, pmal-c, pFLAG, etc. The insertion into a cloning vector can, for example, be accomplished by ligating the DNA fragment into a cloning vector which has complementary cohesive termini. However, if the complementary restriction sites used to fragment the DNA are not present in the cloning vector, the ends of the DNA molecules may be enzymatically modified. Alternatively, any site desired may be produced by ligating nucleotide sequences (linkers) onto the DNA termini; these ligated linkers may comprise specific chemically synthesized oligonucleotides encoding restriction endonuclease recognition sequences. Recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, etc., so that many copies of the gene sequence are generated. Preferably, the cloned gene is contained on a shuttle vector plasmid, which provides for expansion in a cloning cell, e.g., E. coli, and facile purification for subsequent insertion into an appropriate expression cell line, if such is desired. For example, a shuttle vector, which is a vector that can replicate in more than one type of organism, can be prepared for replication in both E. coli and Saccharomyces cerevisiae by linking sequences from an E. coli plasmid with sequences form the yeast 2p plasmid. Expression of Akt Polypeptides The nucleotide sequence coding for Akt, or antigenic fragment, derivative or analog thereof, WO 00/77190 PCTUSOO/15098 -18 or a functionally active derivative, including a chimeric protein, thereof, can be inserted into an appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted protein-coding sequence. Such elements are termed herein a "promoter." Thus, the nucleic acid of the invention is operationally associated with a promoter in an expression vector of the invention. Both cDNA and genomic sequences can be cloned and expressed under control of such regulatory sequences. An expression vector also preferably includes a replication origin. The necessary transcriptional and translational signals can be provided on a recombinant expression vector, or they may be supplied by the native gene encoding the Akt and/or its flanking regions. In one preferred embodiment, the expression of Akt is restricted to cardiomyocytes using a cardiac specific promoter and/or a vector with specific tropism for cardiac cells. Potential host-vector systems include but are not limited to mammalian cell systems infected with virus (e.g., vaccinia virus, adenovirus, etc.); insect cell systems infected with virus (e.g., baculovirus); microorganisms such as yeast containing yeast vectors; or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA. The expression elements of vectors vary in their strengths and specificities. Depending on the host-vector system utilized, any one of a number of suitable transcription and translation elements may be used. A recombinant Akt protein, or functional fragment, derivative, chimeric construct, or analog thereof, may be expressed chromosomally, after integration of the coding sequence by recombination. In this regard, any of a number of amplification systems may be used to achieve high levels of stable gene expression (See Sambrook et al., 1989, supra). A cell containing a recombinant vector comprising the nucleic acid encoding an Akt may be cultured in an appropriate cell culture medium under conditions that provide for expression of Akt by the cell. Any of the methods previously described for the insertion of DNA fragments into a cloning vector may be used to construct expression vectors containing a gene consisting of appropriate transcriptional/translational control signals and the protein coding sequences. These methods may include in vitro recombinant DNA and synthetic techniques and in vivo recombination (genetic recombination). A nucleic acid encoding an Akt polypeptide may be operably linked and controlled by any regulatory region, i.e., promoter/enhancer element known in the art, but these regulatory elements must be functional in the host target tumor selected for expression. The regulatory regions may comprise a promoter region for functional transcription in the host cell, as well as a region situated 3' of the gene of interest, and which specifies a signal for termination of transcription and a polyadenylation site. All these elements constitute an expression cassette.
WO 00/77190 PCTUSOO/15098 -19 Promoters that may be used in the present invention include both constitutive promoters and regulated (inducible) promoters. The promoter may be naturally responsible for the expression of the nucleic acid. It may also be from a heterologous source. In particular, it may be promoter sequences of eukaryotic or viral genes. For example, it may be promoter sequences derived from the genome of the cell which it is desired to infect. Likewise, it may be promoter sequences derived from the genome of a virus, such as adenovirus (E1A and MLP), cytomegalovirus, or Rous Sarcoma Virus. In addition, the promoter may be modified by addition of activating or regulatory sequences or sequences allowing a tissue-specific or predominant expression (enolase and GFAP promoters and the like). Moreover, when the nucleic acid does not contain promoter sequences, it may be inserted. Some promoters useful for practice of this invention are ubiquitous promoters (e.g., HPRT, vimentin, actin, tubulin), intermediate filament promoters (e.g., desmin, neurofilaments, keratin, GFAP), therapeutic gene promoters (e.g., MDR type, CFTR, factor VIII), tissue-specific promoters (e.g., actin promoter in smooth muscle cells), promoters which are preferentially activated in dividing cells, promoters which respond to a stimulus (e.g., steroid hormone receptor, retinoic acid receptor), tetracycline-regulated transcriptional modulators, cytomegalovirus (CMV) immediate-early, retroviral LTR, metallothionein, SV-40, adenovirus Ela, and adenovirus major late (MLP) promoters. Tetracycline-regulated transcriptional modulators and CMV promoters are described in WO 96/01313, US 5,168,062 and 5,385,839, the contents of which are incorporated herein by reference. More specifically, expression of Akt protein may be controlled by any promoter/enhancer element known in the art, but these regulatory elements must be functional in the host selected for expression. Promoters which may be used to control gene expression include, but are not limited to, the SV40 early promoter region (Benoist and Chambon, 1981, Nature 290:304-3 10), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Cell 22:787 797), the herpes thymidine kinase promoter (Wagner et al., 1981, Proc. NatI. Acad. Sci. U.S.A. 78:1441-1445), the regulatory sequences of the metallothionein gene (Brinster et al., 1982, Nature 296:39-42); prokaryotic expression vectors such as the p-lactamase promoter (Villa-Kamaroff, et al., 1978, Proc. Natl. Acad. Sci. U.S.A. 75:3727-3731), or the tac promoter (DeBoer, et al., 1983, Proc. Natl. Acad. Sci. U.S.A. 80:21-25); see also "Useful proteins from recombinant bacteria" in Scientific American, 1980, 242:74-94; promoter elements from yeast or other fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, alkaline phosphatase promoter; and the animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639-646; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol. 50:399-409; MacDonald, 1987, Hepatology 7:425-515); insulin gene control WO 00/77190 PCT/USOO/15098 -20 region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115-122), immunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al., 1984, Cell 38:647-658; Adames et al., 1985, Nature 318:533-538; Alexander et al., 1987, Mol. Cell. Biol. 7:1436-1444), mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Cell 45:485-495), albumin gene control region which is active in liver (Pinkert et al., 1987, Genes and Devel. 1:268-276), alpha-fetoprotein gene control region which is active in liver (Krumlauf et al., 1985, Mol. Cell. Biol. 5:1639-1648; Hammer et al., 1987, Science 235:53-58), alpha 1-antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel. 1:161-171), beta-globin gene control region which is active in myeloid cells (Mogram et al., 1985, Nature 315:338-340; Kollias et al., 1986, Cell 46:89-94), myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712), myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286), and gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al., 1986, Science 234:1372-1378). Preferably, expression of Akt is restricted to cardiomyocytes using either a cardiac specific promoter, or a vector with a specific tropism for cardiac cells. Expression vectors containing a nucleic acid encoding a Akt protein can be identified by five general approaches: (a) PCR amplification of the desired plasmid DNA or specific mRNA, (b) nucleic acid hybridization, (c) presence or absence of selection marker gene functions, (d) analyses with appropriate restriction endonucleases, and (e) expression of inserted sequences. In the first approach, the nucleic acids can be amplified by PCR to provide for detection of the amplified product. In the second approach, the presence of a foreign gene inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted marker gene. In the third approach, the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "selection marker" gene functions (e.g., P-galactosidase activity, thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, etc.) caused by the insertion of foreign genes in the vector. In another example, if the nucleic acid encoding Akt is inserted within the "selection marker" gene sequence of the vector, recombinants containing the Akt insert can be identified by the absence of the gene function. In the fourth approach, recombinant expression vectors are identified by digestion with appropriate restriction enzymes. In the fifth approach, recombinant expression vectors can be identified by assaying for the activity, biochemical, or immunological characteristics of the gene product expressed by the recombinant, provided that the expressed protein assumes a functionally active conformation.
WO 00/77190 PCT/USOO/15098 -21 A wide variety of host/expression vector combinations may be employed in expressing Akt DNA sequences. Useful expression vectors, for example, may consist of segments of chromosomal, non-chromosomal and synthetic DNA sequences. Suitable vectors include derivatives of SV40 and known bacterial plasmids, e.g., E. coli plasmids col El, pCR1, pBR322, pMal-C2, pET, pGEX (Smith et al., 1988, Gene 67:31-40), pMB9 and their derivatives, plasmids such as RP4; phage DNAS, e.g., the numerous derivatives of phage 1, e.g., NM989, and other phage DNA, e.g., M13 and filamentous single stranded phage DNA; yeast plasmids such as the 2m plasmid or derivatives thereof; vectors useful in eukaryotic cells, such as vectors useful in insect or mammalian cells; vectors derived from combinations of plasmids and phage DNAs, such as plasmids that have been modified to employ phage DNA or other expression control sequences; and the like. For example, in a baculovirus expression systems, both non-fusion transfer vectors, such as but not limited to pVL941 (BamHl cloning site; Summers), pVL1393 (BamH1, SmaI, XbaI, EcoR1, NotI, XmaIII, BglII, and PstI cloning site; Invitrogen), pVL 1392 (BglII, PstI, NotI, XmaI, EcoRI, XbaI, SmaI, and BamH1 cloning site; Summers and Invitrogen), and pBlueBacIII (BamH1, BglII, PstI, NcoI, and HindIII cloning site, with blue/white recombinant screening possible; Invitrogen), and fusion transfer vectors, such as but not limited to pAc700 (BamH 1 and KpnI cloning site, in which the BamH1 recognition site begins with the initiation codon; Summers), pAc701 and pAc702 (same as pAc700, with different reading frames), pAc360 (BamHI cloning site 36 base pairs downstream of a polyhedrin initiation codon; Invitrogen(195)), and pBlueBacHisA, B, C (three different reading frames, with BamH1, BglII, PstI, NcoI, and HindII cloning site, an N-terminal peptide for ProBond purification, and blue/white recombinant screening of plaques; Invitrogen (220)) can be used. Mammalian expression vectors contemplated for use in the invention include vectors with inducible promoters, such as the dihydrofolate reductase (DHFR) promoter, e.g., any expression vector with a DHFR expression vector, or a DHFR/methotrexate co-amplification vector, such as pED (PstI, SalI, SbaI, SmaI, and EcoRI cloning site, with the vector expressing both the cloned gene and DHFR; see Kaufman, Current Protocols in Molecular Biology, 16.12 (1991). Alternatively, a glutamine synthetase/methionine sulfoximine co-amplification vector, such as pEE14 (HindI, XbaI, SmaI, SbaI, EcoRI, and BclI cloning site, in which the vector expresses glutamine synthase and the cloned gene; Celltech). In another embodiment, a vector that directs episomal expression under control of Epstein Barr Virus (EBV) can be used, such as pREP4 (BamH 1, Sf1I, XhoI, NotI, NheI, HindI, NheI, Pvull, and KpnI cloning site, constitutive Rous Sarcoma Virus Long Terminal Repeat (RSV-LTR) promoter, hygromycin selectable marker; Invitrogen), pCEP4 (BamH 1, SfiI, AhoI, NotI, NheI, HindIII, NheI, PvuIl, and KpnI cloning site, constitutive human cytomegalovirus (hCMV) immediate early gene, hygromycin selectable marker; Invitrogen), pMEP4 (KpnI, PvuI, NheI, HindII, WO 00/77190 PCT/USOO/15098 -22 NotI, AhoI, SfiI, BamHl cloning site, inducible methallothionein Ha gene promoter, hygromycin selectable marker: Invitrogen), pREP8 (BamH 1, AhoI, NotI, Hindfl, NheI, and KpnI cloning site, RSV-LTR promoter, histidinol selectable marker; Invitrogen), pREP9 (KpnI, NheI, HindIII, NotI, XhoI, SfiI, and BamHI cloning site, RSV-LTR promoter, G418 selectable marker; Invitrogen), and pEBVHis (RSV-LTR promoter, hygromycin selectable marker, N-terminal peptide purifiable via ProBond resin and cleaved by enterokinase; Invitrogen). Selectable mammalian expression vectors for use in the invention include pRc/CMV (HindIll, BstXI, NotI, SbaI, and ApaI cloning site, G418 selection; Invitrogen), pRc/RSV (HindIII, Spel, BstXI, NotI, XbaI cloning site, G418 selection; Invitrogen), and others. Vaccinia virus mammalian expression vectors (see, Kaufman, 1991, supra) for use according to the invention include but are not limited to pSC 1 (SmaI cloning site, TK- and p gal selection), pMJ601 (SalI, SmaI, AflI, NarI, BspMII, BamHI, ApaI, NheI, Sacl, KpnI, and HindIll cloning site; TK- and p-gal selection), and pTKgptFlS (EcoRI, PstI, SalI, AccI, HindII, SbaI, BamHI, and Hpa cloning site, TK or XPRT selection). Yeast expression systems can also be used according to the invention to express Akt protein. For example, the non-fusion pYES2 vector (XbaI, SphI, ShoI, NotI, GstXI, EcoRI, BstXI, BamH 1, SacI, Kpn1, and HindIII cloning sit; Invitrogen) or the fusion pYESHisA, B, C (XbaI, SphI, ShoI, NotI, BstXI, EcoRI, BamH 1, SacI, KpnI, and HindIII cloning site, N-terminal peptide purified with ProBond resin and cleaved with enterokinase; Invitrogen), to mention just two, can be employed according to the invention. Once a particular recombinant DNA molecule is identified and isolated, several methods known in the art may be used to propagate it. Once a suitable host system and growth conditions are established, recombinant expression vectors can be propagated and prepared in quantity. As previously explained, the expression vectors which can be used include, but are not limited to, the following vectors or their derivatives: human or animal viruses such as vaccinia virus or adenovirus; insect viruses such as baculovirus; yeast vectors; bacteriophage vectors (e.g., lambda), and plasmid and cosmid DNA vectors, to name but a few. In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Different host cells have characteristic and specific mechanisms for the translational and post-translational processing and modification of proteins. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. Expression in yeast can produce a biologically active product. Expression in eukaryotic cells can increase the likelihood of "native" folding. Moreover, expression in mammalian cells can provide a tool for reconstituting, or constituting, Akt activity. Furthermore, different vector/host expression systems may affect WO 00/77190 PCT/USOO/15098 -23 processing reactions, such as proteolytic cleavages, to a different extent. Vectors are introduced into the desired host cells by methods known in the art, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, lipofection (lysosome fusion), use of a gene gun, or a DNA vector transporter (see, e.g., Wu et al., 1992, J. Biol. Chem. 267:963-967; Wu and Wu, 1988, J. Biol. Chem. 263:14621 14624; Hartmut et al., Canadian Patent Application No. 2,012,311, filed March 15, 1990). Soluble forms of the protein can be obtained by collecting culture fluid, or solubilizing inclusion bodies, e.g., by treatment with detergent, and if desired sonication or other mechanical processes, as described above. The solubilized or soluble protein can be isolated using various techniques, such as polyacrylamide gel electrophoresis (PAGE), isoelectric focusing, 2-dimensional gel electrophoresis, chromatography (e.g., ion exchange, affinity, immunoaffinity, and sizing column chromatography), centrifugation, differential solubility, immunoprecipitation, or by any other standard technique for the purification of proteins. Gene Therapy and Transgenic Vectors As discussed above, the present invention relates to the ability of Akt proteins to stimulate expression of VEGF, a protein that induces angiogenesis. Therefore, the present invention includes gene therapy by the administration of a nucleic acid encoding an Akt protein to a patient suffering from an ischemic condition. Ischemic conditions may include cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, peripheral arterial disease, intermittent claudication, myocardial ischemia, or ischemic, idiopathic or hypertrophic cardiomyopathy. A nucleic acid encoding Akt, where appropriate incorporated in vectors, and the pharmaceutical compositions containing them, may be used for the treatment of ischemic tissue. They may be used for the transfer and expression of genes in vivo in any type of ischemic tissue, especially the heart. The treatment can, moreover, be targeted in accordance with the pathology to be treated (transfer to a particular tissue can, in particular, be determined by the choice of a vector, and expression by the choice of a particular promoter). The nucleic acids or vectors of the invention are advantageously used for the production in humans or animals, in vivo and intracellularly, of Akt proteins capable of stimulating the expression of VEGF proteins. The present invention thus makes it possible to treat specifically, locally and effectively ischenia. A nucleic acid encoding an Akt protein can be administered alone or in combination with a nucleic acid encoding an angiogenic factor. Known angiogenic factors include basic and acidic fibroblast growth factors (bFGF and aFGF), FGF-5 (US Pat. 5,661,133), endothelial cell growth factor (Pu et al., 1993, Circulation 88:208-2156), angiopoietin and VEGF (for reviews see Melillo et al., WO 00/77190 PCT/USOO/15098 -24 1997 and Lewis et al., 1997). Several forms of VEGF have been identified, including VEGFi 2 I (US Pat. 5,219,739), VEGF 65 (US Pat. 5,332,672), VEGFI 8 9 (US Pat. 5,240,848), VEGF 2 0 6 , VEGF-2 (W095/24473; W096/39515), VEGF-B (US Pats. 5,607,918 and 5,840,693), and VEGF-D (W097/12972). A nucleic acid encoding an Akt protein can also be administered in combination with a transition metal ion, such as CoCl2, which has been shown to enhance expression of the VEGF gene and to stimulate vascularization (US. Pat. 5,480,975). As discussed above, a "vector" is any means for the transfer of a nucleic acid according to the invention into a host cell. Preferred vectors are viral vectors, such as retroviruses, herpes viruses, adenoviruses, and adeno-associated viruses. Thus, a gene encoding an Akt protein or polypeptide is introduced in vivo, ex vivo, or in vitro using a viral vector or through direct introduction of DNA. Expression in targeted tissues can be effected by targeting the transgenic vector to specific cells, such as with a viral vector or a receptor ligand, or by using a tissue-specific promoter, or both. Viral vectors commonly used for in vivo targeting and therapy procedures are DNA-based vectors and retroviral vectors. Methods for constructing and using viral vectors are known in the art [see, e.g., Miller and Rosman, BioTechniques 7:980-990 (1992)]. Preferably, the viral vectors are replication defective, that is, they are unable to replicate autonomously in the target cell. In general, the genome of the replication defective viral vectors which are used within the scope of the present invention lack at least one region which is necessary for the replication of the virus in the infected cell. These regions can either be eliminated (in whole or in part), be rendered non-functional by any technique known to a person skilled in the art. These techniques include the total removal, substitution (by other sequences, in particular by the inserted nucleic acid), partial deletion or addition of one or more bases to an essential (for replication) region. Such techniques may be performed in vitro (on the isolated DNA) or in situ, using the techniques of genetic manipulation or by treatment with mutagenic agents. Preferably, the replication defective virus retains the sequences of its genome which are necessary for encapsulating the viral particles. DNA viral vectors include an attenuated or defective DNA virus, such as but not limited to herpes simplex virus (HSV), papillomavirus, Epstein Barr virus (EBV), adenovirus, adeno-associated virus (AAV), vaccinia virus, and the like. Defective viruses, which entirely or almost entirely lack viral genes, are preferred. Defective virus is not replication competent after introduction into a cell, and thus does not lead to a productive viral infection. Use of defective viral vectors allows for administration to cells in a specific, localized area, without concern that the vector can infect other cells. Thus, a specific tissue can be specifically targeted. Examples of particular vectors include, but are not limited to, a defective herpes virus 1 (HSV 1) vector [Kaplitt et al., Molec. Cell. Neurosci. 2:320-330 (1991)], defective herpes virus vector lacking a glyco-protein L gene [Patent Publication WO 00/77190 PCT/USOO/15098 -25 RD 371005 A], or other defective herpes virus vectors [International Patent Publication No. WO 94/21807, published September 29, 1994; International Patent Publication No. WO 92/05263, published April 2, 1994]: an attenuated adenovirus vector, such as the vector described by Stratford Perricaudet et al. [J. Clin. Invest. 90:626-630 (1992); see also La Salle et al., Science 259:988-990 (1993)]; and a defective adeno-associated virus vector [Samulski et al., J. Virol. 61:3096-3101 (1987); Samulski et al., J. Virol. 63:3822-3828 (1989); Lebkowski et al., Mol. Cell. Biol. 8:3988-3996 (1988)]. Preferably, for in vivo administration, an appropriate immunosuppressive treatment is employed in conjunction with the viral vector, e.g., adenovirus vector, to avoid immuno-deactivation of the viral vector and transfected cells. For example, immunosuppressive cytokines, such as interleukin-12 (IL-12), interferon-y (IFN-y), or anti-CD4 antibody, can be administered to block humoral or cellular immune responses to the viral vectors [see, e.g., Wilson, Nature Medicine (1995)]. In addition, it is advantageous to employ a viral vector that is engineered to express a minimal number of antigens. Naturally, the invention contemplates delivery of a vector that will express a therapeutically effective amount of Akt for gene therapy applications. The phrase "therapeutically effective amount" is used herein to mean an amount sufficient to cause an improvement in a clinically significant ischemic condition in a host. Adenovirus vectors In a preferred embodiment, the vector is an adenovirus vector. Adenoviruses are eukaryotic DNA viruses that can be modified to efficiently deliver a nucleic acid of the invention to a variety of cell types. Various serotypes of adenovirus exist. Of these serotypes, preference is given, within the scope of the present invention, to using type 2 or type 5 human adenoviruses (Ad 2 or Ad 5) or adenoviruses of animal origin (see W094/26914). Those adenoviruses of animal origin which can be used within the scope of the present invention include adenoviruses of canine, bovine, murine (example: Mavl, Beard et al., Virology 75 (1990) 81), ovine, porcine, avian, and simian (example: SAV) origin. Preferably, the adenovirus of animal origin is a canine adenovirus, more preferably a CAV2 adenovirus (e.g. Manhattan or A26/61 strain (ATCC VR-800), for example). Preferably, the replication defective adenoviral vectors of the invention comprise the ITRs, an encapsidation sequence and the nucleic acid of interest. Still more preferably, at least the El region of the adenoviral vector is non-functional. The deletion in the El region preferably extends from nucleotides 455 to 3329 in the sequence of the Ad5 adenovirus (PvuII-BglII fragment) or 382 to 3446 (HinflI-Sau3A fragment). Other regions may also be modified, in particular the E3 region (W095/02697), the E2 region (W094/28938), the E4 region (W094/28152, W094/12649 and WO 00/77190 PCT/USOO/15098 -26 W095/02697), or in any of the late genes L1-L5. In a preferred embodiment, the adenoviral vector has a deletion in the El region (Ad 1.0). Examples of El-deleted adenoviruses are disclosed in EP 185,573, the contents of which are incorporated herein by reference. In another preferred embodiment, the adenoviral vector has a deletion in the El and E4 regions (Ad 3.0). Examples of El/E4-deleted adenoviruses are disclosed in W095/02697 and W096/22378, the contents of which are incorporated herein by reference. In still another preferred embodiment, the adenoviral vector has a deletion in the El region into which the E4 region and the nucleic acid sequence are inserted (see FR94 13355, the contents of which are incorporated herein by reference). The replication defective recombinant adenoviruses according to the invention can be prepared by any technique known to the person skilled in the art (Levrero et al., Gene 101 (1991) 195, EP 185 573; Graham, EMBO J. 3 (1984) 2917). In particular, they can be prepared by homologous recombination between an adenovirus and a plasmid which carries, inter alia, the DNA sequence of interest. The homologous recombination is effected following cotransfection of the adenovirus and plasmid into an appropriate cell line. The cell line which is employed should preferably (i) be transformable by the said elements, and (ii) contain the sequences which are able to complement the part of the genome of the replication defective adenovirus, preferably in integrated form in order to avoid the risks of recombination. Examples of cell lines which may be used are the human embryonic kidney cell line 293 (Graham et al., J. Gen. Virol. 36 (1977) 59) which contains the left-hand portion of the genome of an Ad5 adenovirus (12%) integrated into its genome, and cell lines which are able to complement the El and E4 functions, as described in applications W094/26914 and W095/02697. Recombinant adenoviruses are recovered and purified using standard molecular biological techniques, which are well known to one of ordinary skill in the art. Adeno-associated virus vectors The adeno-associated viruses (AAV) are DNA viruses of relatively small size which can integrate, in a stable and site-specific manner, into the genome of the cells which they infect. They are able to infect a wide spectrum of cells without inducing any effects on cellular growth, morphology or differentiation, and they do not appear to be involved in human pathologies. The AAV genome has been cloned, sequenced and characterised. It encompasses approximately 4700 bases and contains an inverted terminal repeat (ITR) region of approximately 145 bases at each end, which serves as an origin of replication for the virus. The remainder of the genome is divided into two essential regions which carry the encapsulation functions: the left-hand part of the genome, which contains the rep gene involved in viral replication and expression of the viral genes; and the right-hand part of the genome, which contains the cap gene encoding the capsid proteins of the virus.
WO 00/77190 PCT/USOO/15098 -27 The use of vectors derived from the AAVs for transferring genes in vitro and in vivo has been described (see WO 91/18088; WO 93/09239; US 4,797,368, US 5,139,941, EP 488 528). These publications describe various AAV-derived constructs in which the rep and/or cap genes are deleted and replaced by a gene of interest, and the use of these constructs for transferring the said gene of interest in vitro (into cultured cells) or in vivo, (directly into an organism). The replication defective recombinant AAVs according to the invention can be prepared by cotransfecting a plasmid containing the nucleic acid sequence of interest flanked by two AAV inverted terminal repeat (ITR) regions, and a plasmid carrying the AAV encapsulation genes (rep and cap genes), into a cell line which is infected with a human helper virus (for example an adenovirus). The AAV recombinants which are produced are then purified by standard techniques. The invention also relates, therefore, to an AAV-derived recombinant virus whose genome encompasses a sequence encoding a nucleic acid encoding an Akt3 flanked by the AAV ITRs. The invention also relates to a plasmid encompassing a sequence encoding a nucleic acid encoding an Akt3 flanked by two ITRs from an AAV. Such a plasmid can be used as it is for transferring the nucleic acid sequence, with the plasmid, where appropriate, being incorporated into a liposomal vector (pseudo-virus). Retrovirus vectors In another embodiment the gene can be introduced in a retroviral vector, e.g., as described in Anderson et al., U.S. Patent No. 5,399,346; Mann et al., 1983, Cell 33:153; Temin et al., U.S. Patent No. 4,650,764; Temin et al., U.S. Patent No. 4,980,289; Markowitz et al., 1988, J. Virol. 62:1120; Temin et al., U.S. Patent No. 5,124,263; EP 453242, EP178220; Bernstein et al. Genet. Eng. 7 (1985) 235; McCormick, BioTechnology 3 (1985) 689; International Patent Publication No. WO 95/07358, published March 16, 1995, by Dougherty et al.; and Kuo et al., 1993, Blood 82:845. The retroviruses are integrating viruses which infect dividing cells. The retrovirus genome includes two LTRs, an encapsulation sequence and three coding regions (gag, pol and env). In recombinant retroviral vectors, the gag, pol and env genes are generally deleted, in whole or in part, and replaced with a heterologous nucleic acid sequence of interest. These vectors can be constructed from different types of retrovirus, such as, HIV, MoMuLV ("murine Moloney leukaemia virus" MSV ("murine Moloney sarcoma virus"), HaSV ("Harvey sarcoma virus"); SNV ("spleen necrosis virus"); RSV ("Rous sarcoma virus") and Friend virus. Defective retroviral vectors are disclosed in W095/02697. In general, in order to construct recombinant retroviruses containing a nucleic acid sequence, a plasmid is constructed which contains the LTRs, the encapsulation sequence and the coding sequence. This construct is used to transfect a packaging cell line, which cell line is able to supply in trans the retroviral functions which are deficient in the plasmid. In general, the packaging cell lines WO 00/77190 PCT/USOO/15098 -28 are thus able to express the gag, pol and env genes. Such packaging cell lines have been described in the prior art, in particular the cell line PA317 (US4,861,719); the PsiCRIP cell line (W090/02806) and the GPenvAm-12 cell line (WO89/07150). In addition, the recombinant retroviral vectors can contain modifications within the LTRs for suppressing transcriptional activity as well as extensive encapsulation sequences which may include a part of the gag gene (Bender et al., J. Virol. 61 (1987) 1639). Recombinant retroviral vectors are purified by standard techniques known to those having ordinary skill in the art. Retroviral vectors can be constructed to function as infections particles or to undergo a single round of transfection. In the former case, the virus is modified to retain all of its genes except for those responsible for oncogenic transformation properties, and to express the heterologous gene. Non infectious viral vectors are prepared to destroy the viral packaging signal, but retain the structural genes required to package the co-introduced virus engineered to contain the heterologous gene and the packaging signals. Thus, the viral particles that are produced are not capable of producing additional virus. Targeted gene delivery is described in International Patent Publication WO 95/28494, published October 1995. Non-viral vectors Alternatively, the vector can be introduced in vivo by lipofection. For the past decade, there has been increasing use of liposomes for encapsulation and transfection of nucleic acids in vitro. Synthetic cationic lipids designed to limit the difficulties and dangers encountered with liposome mediated transfection can be used to prepare liposomes for in vivo transfection of a gene encoding a marker [Felgner, et. al., Proc. Nati. Acad. Sci. U.S.A. 84:7413-7417 (1987); see Mackey, et al., Proc. Natl. Acad. Sci. U.S.A. 85:8027-8031 (1988); Ulmer et al., Science 259:1745-1748 (1993)]. The use of cationic lipids may promote encapsulation of negatively charged nucleic acids, and also promote fusion with negatively charged cell membranes [Felgner and Ringold, Science 337:387-388 (1989)]. Particularly useful lipid compounds and compositions for transfer of nucleic acids are described in International Patent Publications W095/18863 and W096/17823, and in U.S. Patent No. 5,459,127. The use of lipofection to introduce exogenous genes into the specific organs in vivo has certain practical advantages. Molecular targeting of liposomes to specific cells represents one area of benefit. It is clear that directing transfection to particular cell types would be particularly advantageous in a tissue with cellular heterogeneity, such as pancreas, liver, kidney, and the brain. Lipids may be chemically coupled to other molecules for the purpose of targeting [see Mackey, et. al., supra]. Targeted peptides, e.g., hormones or neurotransmitters, and proteins such as antibodies, or non peptide molecules could be coupled to liposomes chemically.
WO 00/77190 PCT/USOO/15098 -29 Other molecules are also useful for facilitating transfection of a nucleic acid in vivo, such as a cationic oligopeptide (e.g., International Patent Publication W095/21931), peptides derived from DNA binding proteins (e.g., International Patent Publication W096/25508), or a cationic polymer (e.g., International Patent Publication W095/21931). It is also possible to introduce the vector in vivo as a naked DNA plasmid (see U.S. Patents 5,693,622, 5,589,466 and 5,580,859). Naked DNA vectors for gene therapy can be introduced into the desired host cells by methods known in the art, e.g., transfection, electroporation, microinjection, transduction, cell fusion, DEAE dextran, calcium phosphate precipitation, use of a gene gun, or use of a DNA vector transporter [see, e.g., Wu et al., J. Biol. Chem. 267:963-967 (1992); Wu and Wu, J. Biol. Chem. 263:14621-14624 (1988); Hartmut et al., Canadian Patent Application No. 2,012,311, filed March 15, 1990; Williams et al., Proc. Nati. Acad. Sci. USA 88:2726-2730 (1991)]. Receptor mediated DNA delivery approaches can also be used [Curiel et al., Hum. Gene Ther. 3:147-154 (1992); Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)]. Preferred naked DNA vectors include pCOR plasmids having a conditional origin of replication (see W097/10343), and minicircle plasmids lacking an origin of replication and a marker gene (see W096/26270). Pharmaceutical Compositions and Delivery The present invention also relates to a pharmaceutical compositions. Such compositions may comprise an Akt protein or polypeptide or a nucleic acid encoding an Akt protein or polypeptide, as defined above, and a pharmaceutically acceptable carrier or vehicle. The compositions of the invention are particularly suitable for formulation of biological material for gene therapy. Thus, in a preferred embodiment, the composition comprises a nucleic acid encoding a human Akt protein or polypeptide. The composition may comprise an Akt protein, or nucleic acid encoding an Akt protein. The composition may, in addition, comprise a nucleic acid encoding an angiogenic factor. Known angiogenic factors include basic and acidic fibroblast growth factors (bFGF and aFGF), FGF-5 (US Pat. 5,661,133), endothelial cell growth factor (Pu et al., 1993, Circulation 88:208-2156), angiopoietin and VEGF (for reviews see Melillo et al., 1997 and Lewis et al., 1997). Nucleic acids encoding several forms of VEGF have been identified, including VEGF 12 (US Pat. 5,219,739), VEGF 6 S (US Pat. 5,332,672), VEGF 189 (US Pat. 5,240,848), VEGF 2 0 6 , VEGF-2 (W095/24473; W096/39515), VEGF-B (US Pats. 5,607,918 and 5,840,693), and VEGF-D (W097/12972). The composition may also comprise a transition metal ion, such as CoCl 2 , which has been shown to enhance expression of the VEGF gene and to stimulate vascularization (US. Pat. 5,480,975). The Akt protein, or nucleic WO 00/77190 PCT/USOO/15098 -30 acid encoding an Akt protein may also be administered in conjunction with a vasodilator. Examples of vasodilators include nitrovasodilators (e.g. nitroprusside, nitroglycerin), non-specific vasodilators (e.g. hyrdralizine, papaverine), adenosine receptor agonists, calcium channel blocking agents, alpha blockers (e.g. prazosin), endogenous vasodilator peptides or related peptide analogs (e.g. substance P, CGRP), K channel activators, ACE inhibitors or angiotensin receptor blockers, endothelin receptor blockers or ECE inhibitors, and vasodilator prostaglandins. Any vector, viral or non-viral, of the invention will preferably be introduced in vivo in a pharmaceutically acceptable vehicle or carrier. The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a human. Preferably, as used herein, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous solution saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. The pharmaceutical compositions of the invention may be formulated for the purpose of topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous, intraocular, and the like, administration. Preferably, the pharmaceutical compositions contain pharmaceutically acceptable vehicles for an injectable formulation. These can be, in particular, sterile, isotonic saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride, and the like, or mixtures of such salts), or dry, in particular lyophilized, compositions which, on addition, as appropriate, of sterilized water or of physiological saline, enable injectable solutions to be formed. The compositions may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, allow the constitution of injectable solutions. The preferred sterile injectable preparations can be a solution or suspension in a nontoxic parenterally acceptable solvent or diluent. Examples of pharmaceutically acceptable carriers or vehicles are saline, buffered saline, isotonic saline (e.g., monosodium or disodium phosphate, sodium, WO 00/77190 PCTIUSOO/15098 -31 potassium, calcium or magnesium chloride, or mixtures of such salts), Ringer's solution, dextrose, water, sterile water, glycerol, ethanol, and combinations thereof. 1,3-butanediol and sterile fixed oils are conveniently employed as solvents or suspending media. Any bland fixed oil can be employed including synthetic mono- or di-glycerides. Fatty acids such as oleic acid also find use in the preparation of injectables. The doses of nucleic acids of the invention, either alone or incorporated in a vector, used for administration can be adjusted in accordance with different parameters, and in particular in accordance with the mode of administration used, the pathology in question, the gene to be expressed or the desired treatment period. Generally speaking, in the case of the recombinant viruses according to the invention, these are formulated and administered in the form of doses of between 104 and 1014 pfu, and preferably 106 to 1010 pfu. The term pfu (plaque forming unit) corresponds to the infectious power of a solution of virus, and is determined by infection of a suitable cell culture and measurement, generally after 48 hours, of the number of infected cell plaques. The techniques of determination of the pfu titre of a viral solution are well documented in the literature. The composition of the invention may be introduced parenterally or transmucosally, e.g., orally, nasally, or rectally, or transdermally. Preferably, administration is parenteral, e.g., via intravenous injection, and also including, but is not limited to, intra-arteriole, intramuscular, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial administration. The administration of the composition may introduced by injection directly into the site to be treated, in particular, into the heart. The preferred route of administration to the heart is by direct injection into the heart (US Pats. 5,693,622 or 5,661,133). The heart can be imaged using any of the techniques available in the art, such as magnetic resonance imaging or computer-assisted tomography, and the therapeutic composition administered by stereotactic injection, for example, into ischemic regions of the myocardium. Preferably, expression of Akt is restricted to cardiomyocytes using either a cardiac specific promoter, or a vector with a specific tropism for cardiac cells. Administration to the heart can also occur through the use of a catheter. Various porous balloon, double balloon and hydrogel catheters are described in US patents 5,851,521, 5,652,225, 5,328,470, 5,698,531, 5,707,969, 5,830,879, and 5,674,192. In yet another embodiment, a composition comprising an Akt polypeptide, or nucleic acid encoding the polypeptide, can be delivered in a controlled release system. For example, the nucleic acid or polypeptide may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump may be WO 00/77190 PCT/US0O/15098 -32 used [see Langer, supra; Sefton, CRC Crit. Ref Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J Med. 321:574 (1989)]. In another embodiment, polymeric materials can be used [see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Press: Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley: New York (1984); Ranger and Peppas, J Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., JNeurosurg. 71:105 (1989)]. In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, i.e., the heart, thus requiring only a fraction of the systemic dose [see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)]. Other controlled release systems are discussed in the review by Langer [Science 249:1527-1533 (1990)]. Thus, the compositions of the invention can be delivered by intravenous, intraarterial, intraperitoneal, intramuscular, or subcutaneous routes of administration. Alternatively, the compositions, properly formulated, can be administered by nasal or oral administration. A constant supply of the biological material can be ensured by providing a therapeutically effective dose (i.e., a dose effective to induce metabolic changes in a subject) at the necessary intervals, e.g., daily, every 12 hours, etc. These parameters will depend on the severity of the disease condition being treated, other actions, such as diet modification, that are implemented, the weight, age, and sex of the subject, and other criteria, which can be readily determined according to standard good medical practice by those of skill in the art. The present invention also relates to a method of inhibiting angiogenesis in a patient suffering from a tumor by inhibiting the level of Akt activity in the patient, thereby inhibiting production of VEGF. The level of Akt may be decreased by introducing an Akt antisense nucleic acid into cells of the patient under conditions wherein the antisense nucleic acid hybridizes under intracellular conditions to an Akt mRNA. The level of Akt can also be decreased by introducing an intracellular binding protein, such as a single chain Fv antibody (scFv), that specifically binds Akt into a patient's cell at a level sufficient to bind to and inactivate Akt. In another embodiment, Akt activity can be decreased by administering a nucleic acid encoding a dominant negative form of an Akt. Preferably, the antisense nucleic acid, intracellular binding protein or nucleic acid encoding therefor, or dominant negative are administered directly to tumor cells. An antisense sequence according to the invention is complementary to the sequence encoding an Akt protein and down-regulates or blocks expression of the Akt protein. A preferred embodiment comprises an antisense polynucleotide molecule. Preparation and use of antisense polynucleotides, DNA encoding antisense RNA molecules and use of oligo and genetic antisense is disclosed in WO WO 00/77190 PCTIUSOO/15098 -33 92/15680, the entire contents of which are incorporated herein by reference. Antisense nucleic acids of the invention are preferably RNA capable of specifically hybridizing with all or part of DNA sequence encoding an Akt protein, or the corresponding messenger RNA. The antisense sequence of the present invention may be derived from DNA sequences whose expression in the cell produces RNA complementary to all or part of a human Akt mRNA. These antisense sequences can be prepared by expression of all or part of a sequence encoding an Akt protein in the opposite orientation (EP 140 308). Any length of the antisense sequence is suitable for practice of the invention so long as it is capable of down-regulating or blocking expression of the Akt. Preferably, the antisense sequence is at least 20 nucleotides in length. In another aspect of this preferred embodiment the nucleic acid encodes antisense RNA molecules. In this embodiment, the nucleic acid is operably linked to signals enabling expression of the nucleic acid sequence and is introduced into a cell utilizing, preferably, recombinant vector constructs, which will express the antisense nucleic acid once the vector is introduced into the cell. Examples of suitable vectors includes plasmids, adenoviruses, adeno-associated viruses, retroviruses, and herpes viruses. A second embodiment of the present invention's method of specifically inhibiting angiogenesis through the inhibition of Akt activity comprises expression of a nucleic acid sequence encoding an intracellular binding protein capable of selectively interacting with the Akt within a transfected cell. WO 94/29446 and WO 94/02610, the entire contents of which are incorporated herein by reference, disclose cellular transfection with genes encoding an intracellular binding protein. An intracellular binding protein includes any protein capable of selectively interacting, or binding, with Akt in the cell in which it is expressed and of neutralizing the function of bound Akt protein. Preferably, the intracellular binding protein is an antibody or a fragment of an antibody. WO 94/02610 discloses preparation of antibodies and identification of the nucleic acid encoding a particular antibody. Using an Akt protein, or a fragment thereof, a monoclonal antibody specific for the protein is prepared according to techniques known to those skilled in the art. A vector comprising the nucleic acid encoding an intracellular binding protein, or a portion thereof, and capable of expression in a host cell is subsequently prepared for use in the method of this invention. Suitable vectors and methods of delivering nucleic acids encoding intracellular binding proteins to cells containing Akt include those discussed above. The nucleic acid sequence encoding an Akt intracellular binding protein may additionally comprise a sequence encoding a localization signal for targeting the intracellular binding protein to the cellular location of Akt and/or a sequence enabling insertion of the intracellular binding protein into the plasma membrane. The localization signal or insertion sequence can be located anywhere on the intracellular binding protein, so long as it does not WO 00/77190 PCT/US0O/15098 -34 interfere with binding to the Akt protein. Examples of localization signals are disclosed in WO 94/02610. In another embodiment, Akt activity can be decreased by administering a nucleic acid encoding a dominant negative form of an Akt. Examples of dominant negative forms of Akt are described in Fujio et al., 1999 (J. Biol. Chem. 274(23):16349-16354), Wang et al., 1999 (Mol. Cell Biol. 19(6): 4008-4018), Jiang et al., 1999 (Proc. Natl. Acad. Sci. 96(5): 2077-2081), and Gerber et al., 1998 (J. Biol. Chem. 273(46): 30336-30343). An organism in whom administration of a biological material within the scope of the invention is administered is preferably a human, but can be any animal. Thus, as can be readily appreciated by one of ordinary skill in the art, the methods and pharmaceutical compositions of the present invention are particularly suited to administration to any animal, particularly a mammal, and including, but by no means limited to, domestic animals, such as feline or canine subjects, farm animals, such as but not limited to bovine, equine, caprine, ovine, and porcine subjects, wild animals (whether in the wild or in a zoological garden), research animals, such as mice, rats, rabbits, goats, sheep, pigs, dogs, cats, etc., avian species, such as chickens, turkeys, songbirds, etc., i.e., for veterinary medical use. The present invention may be better understood by reference to the following non-limiting Examples, which are provided as exemplary of the invention. EXAMPLES General molecular biology techniques The methods traditionally used in molecular biology, such as preparative extractions of plasmid DNA, centrifugation of plasmid DNA in a caesium chloride gradient, agarose or acrylamide gel electrophoresis, purification of DNA fragments by electroelution, protein extraction with phenol or phenol/chloroform, ethanol or isopropanol precipitation of DNA in a saline medium, transformation in Escherichia coli, and the like, are well known to a person skilled in the art and are amply described in the literature [Maniatis T. et al., "Molecular Cloning, a Laboratory Manual", Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1982; ( 2 "d Ed. 1989); Ausubel F.M. et al. (eds), "Current Protocols in Molecular Biology", John Wiley & Sons, New York, 1987]. Conventional cloning vehicles include pBR322 and pUC type plasmids and phages of the M13 series. These may be obtained commercially (Bethesda Research Laboratories). For ligation, DNA fragments may be separated according to their size by agarose or acrylamide gel electrophoresis, extracted with phenol or with a phenol/chloroform mixture, WO 00/77190 PCTUSOO/15098 -35 precipitated with ethanol and then incubated in the presence of phage T4 DNA ligase (Biolabs) according to the supplier's recommendations. The filling in of 5' protruding ends may be performed with the Klenow fragment of E. coli DNA polymerase I (Biolabs) according to the supplier's specifications. The destruction of 3' protruding ends is performed in the presence of phage T4 DNA polymerase (Biolabs) used according to the manufacturer's recommendations. The destruction of 5' protruding ends is performed by a controlled treatment with SI nuclease. Mutagenesis directed in vitro by synthetic oligodeoxynucleotides may be performed according to the method developed by Taylor et al. [Nucleic Acids Res. 13 (1985) 8749-8764] using the kit distributed by Amersham. The enzymatic amplification of DNA fragments by PCR [Polymerase-catalyzed Chain Reaction, Saiki R.K. et al., Science 230 (1985) 1350-1354; Mullis K.B. and Faloona F.A., Meth. Enzym. 155 (1987) 335-350] technique may be performed using a "DNA thermal cycler" (Perkin Elmer Cetus) according to the manufacturer's specifications. Verification of nucleotide sequences may be performed by the method developed by Sanger et al. [Proc. Natl. Acad. Sci. USA, 74 (1977) 5463-5467] using the kit distributed by Amersham. Plasmid DNAs may be purified by the Qiagen Plasmid Purification System according to the manufacture's instruction. Example 1: Cloning of Human Akt3 This example describes the cloning of a nucleic acid encoding Akt3 protein. Example 1.1: cDNA library screeningfor Akt3 A data base search revealed that one human cDNA clone contains a stretch of human cDNA sequence that is homologous to, but different from human Aktl and Akt2. To isolate the full length coding sequence of this previously unknown human Akt isoform (herein named human Akt3), a human heart cDNA library was screened with a cDNA probe corresponding to the 5'-UTR and coding region for the N-terminus of human Akt3. A human cDNA clone (ID# 479072) was purchased (Genome System Inc.). One fragment of this DNA, which covers part of the 5'-UTR (untranslated region) and part of the 5'-coding sequence of human Akt3, was amplified by polymerase chain reaction (PCR) using the following primers: AKT3-5'UTR-F3 (5' TCC AAA CCC TAA AGC TGA TAT CAC 3'; SEQ ID NO:3) and AKT3-C R1 (5' CCT GGA TAG CTT CTG TCC ATT C 3'; SEQ ID NO:4). A cDNA probe was labeled with [a-p32]dCTP using a Random Primer DNA labeling kit (Boerhinger Mannheim) according to the manufacture's instructions. The probe was purified using a Bio-Rad chromatography spin column according to the manufacture's instruction.
WO 00/77190 PCT/USOO/15098 -36 Over one million phage clones were initially used for cDNA phage library screening (Clonetech, Cat# HL5027t). Host cells XL 1-B were inoculated at 37'C overnight in LB media (supplemented with 20 mg/ml tetracycline, 0.2% maltose and 10mM MgCl2). Phage infection and membrane lifting were carried out as described in Maniatis, 1989. Membranes were denatured, renatured and baked, then pre-hybridized with hybridization solution for 4 hours at 65'C. A denatured form of the p32-labeled probe (heat denatured for 10 minutes) was added to the membranes for overnight hybridization. After hybridization, membranes were serially washed with 2XSSC/0. 1 %SDS, 1XSSC/0.1%SDS, and 0.5XSSC/0.1%SDS at 65'C. Membranes were air-dried and exposed to Kodak X-ray films. After this primary screening, positive clones were selected for secondary and tertiary screening. Resulting positive phages were purified, and phage DNA converted into plasmid DNA using BM25.8-25 host cells according to the manufacture's (Boerhinger Mannheim) instructions. Two positive clones were chosen for complete sequencing and further characterization. One of these clones (clone #9) comprises part of the 5'-UTR and the N-terminal coding sequence (aa I to 127) of human Akt3. A second clone (clone #1) comprises most of the human Akt3 sequence (aa 15 to the C-terminus) and 3'-UTR. A full length cDNA sequence was formed by the fusion of these two partial sequences. A complete sequence encoding a human Akt3 is shown in SEQ ID NO: 1. The corresponding amino acid sequence is shown in SEQ ID NO:2. Akt3 is shorter that Aktl and Akt2, and there is no significant homology between Akt3 and Aktl or Akt2 at the C-terminus of the molecules. In particular, the last 14 amino acids in the C-terminal portion of human Akt-3 are different from those present in human Aktl and Akt2. Example 2: Construction of Akt3 expression plasmids This example describes the construction of an expression plasmid for activated Akt3. First two partial cDNA clones (clone #1 and clone #9, described above) were fused to obtain a full length AKT3 coding sequence. A DNA comprising the human Src myristylation sequence was fused to the N-terminus of the full length Akt3 sequence. An HA-tag sequence was fused to the C-terminus of the full length Akt3 sequence (for detection of expression). The sequence for this chimeric MyrAkt3HA was placed under the control of a CMV promoter. The complete construct is called CMV6 MyrAkt3HA (Figure IA). Example 2.1: CMV6-MyrAktHA This example describes the construction of plasmids capable of expressing Akt3 and a constitutively active form of human Akt3. A full length Akt3 coding sequence was obtained by PCR amplification of clone #1 using the following primers: hAKT3cl9-PCR5(F): (5'- ATG AGC GAT GTT ACC ATT GTG AAA GAA GGT TGG GTT CAG AAG AGG GGA GAA TAT ATA AAA AAC TGG AGG CCA AG - 3'; SEQ ID NO:5), which contains the coding sequence of the first 24 WO 00/77190 PCTIUSOO/15098 -37 amino acids of Akt3, and hAKT3 cll-PCR3 @: (5' - TTA TTT TTT CCA GGT ACC CAG CAT GCC - 3'; SEQ ID NO:6). To make the constitutively active Akt3 form, the coding sequence of full length Akt3 was PCR amplified by using the following primers: MyrAKT3Ha-F1(5' - GCG CGC GAA TTC CCA CCA TGG GTA GCA ACA AGA GCA AGC CCA AGG ATG CCA GCC AGC GGC GCC GCA GCA GCG ATG TTA CCA TTG TGA AAG - 3'; SEQ ID NO:7), which contains the Kozak sequence (CCA CC), the myristylation sequence from human src (underlined) and the first 8 amino acids of human Akt3 (in bold), and MyrAKT3Ha-R (5' - GCG CGC GGG CCC TTA GGC GTA GTC GGG GAC GTC GTA CGG GTA TTT TTT CCA GTT ACC CAG CAT GCC - 3'; SEQ ID NO:8), which contains the coding sequence of an HA tag (in bold). The PCR product was digested with EcoR 1/Apa 1 and subcloned into the EcoR 1/Apa 1 sites of pCDNA3.1 producing pCDNA3 Myr-Akt-HA. The coding sequence of MyrAktHA was also PCR amplified and subcloned into the Kpn 1/ EcoR 1 sites of the vector CMV6. The primers used for PCR reaction were: CMV6-AKT3cat F (5' - CGG GGT ACC ACC ATG GGT AGC AAC AAG AGC AAG CCC AAG GAT GCC AGC CAG - 3'; SEQ ID NO:9), and CMV6-AKT3cat-R (5' - CCG GAA TTC TTA GGC GTA GTC GGG GAC GTC - 3'; SEQ ID NO:10). The plasmid was verified by sequencing. Example 2.2: Expression of Human AKT3 This example describes the expression of human AKT3 in tissue culture. HEK293 cells and COS-7 cells were maintained in DME media supplemented with 10% fetal bovine serum (FBS). Cells were grown in 37 0 C, 5%CO 2 incubator. The plasmid CMV6-[MyrAkt3HA] was transiently transfected into HEK293 cells. As a control, HEK293 cells were transfected with the CMV6 vector. One day prior to either transfection, cells were split to a density of 0.2x10 6 /Cm2. Transfections were carried out using LipofectAmine (Gibco BRL) according to the manufacture's instruction. Briefly, DNA was mixed in DME media (without serum or antibiotics). LipofectAmine was added (DNA:LipofectAmine =1mg : 4ml). After brief mixing, the DNA/LipofectAmine mixture was kept at room temperature for 30 minutes. Cells were washed with 1xPBS, and exposed to the DNA/LipofectAmine mixture for 3 hours. After transfection, cells were washed two times with 1xPBS and switched to DMEM-10%FBS media. Twenty-four hours after transfection, cells were lysed. Lysates were immunoprecipitated with anti-HA antibodies, and the kinase activity of the immunopellets was determined using peptides derived from GSK-3, a downstream target for Aktl (Cross et al. 1995). In vitro kinase assays for Akt were carried out according to Cross et al (Cross et al, 1995) 24 hours post-transfection. Cells were washed twice in lxPBS solution, and lysed in lysis buffer (50mM Tris/HCl, pH 7.4, 1mM EDTA, 1mM EGTA, 0.5mM Na 3
VO
4 , 0.1% p-mercaptoethanol, 1% Triton X-100, 50mM NaF, 5mM Sodium WO 00/77190 PCT/USOO/15098 -38 pyrophosphate, 10mM sodium glycerophosphate, 0.5mM PMSF, 2ug/ml aprotinin, 2mg/ml leupeptin, and 1mM microcystin). Insoluble materials were cleared by centrifugation at 4'C for 15 minutes. Cell lysates were incubated with polyclonal anti-HA antibodies (BABCO) for 1 hour at 4'C while on a rotating platform. Protein A-Agarose beads were added to lysates for 1 hour. After immunoprecipitation, pellets were washed three times with washing solution A (lysis buffer supplemented with 0.5M NaCl), three times with washing solution B (50mM Tris/HCI, pH7.4, 0.03% Brij35, 0.1mM EGTA and 0.1% p-mercaptoethanol), and three times with kinase buffer (20mM MOPS, pH7.2, 25mM sodium P-glycerophosphate pH7.0, 1mM Na 3
VO
4 , 1mM DTT). After washing, pellets were resuspended in 40d kinase reaction mixture [100mM ATP, 0.lmg/ml Crosstide substrate peptide (UBI), 20mM MgCl2, 10mM protein kinase A inhibitor/PKI (UBI), and 1OmCi (g-32P)-ATP]. Reactions were carried out at 30'C for 30 minutes. After completion of the reactions, mixtures were briefly centrifuged, and 30u1 of the supernatant was loaded onto a p81 nitrocellulose paper circle (Gibco BRL). Nitrocellulose papers were washed three times with 180mM phosphoric acid (10 minutes for each washing), and two times with acetone (2 minutes for each washing). The radioactivity of the paper was monitored by Scintillation Counting Machine. Kinase activity present in CMV6[MyrAkt3HA] transfected samples was 20 times higher than that present in cells transfected with the control vector CMV6, which is similar to the background level observed for this assay (Figure 1B). To test the expression of MyrAkt3HA in transfected cells, lysates prepared from transfected cells were subjected to immunobloting with anti-HA antibodies. Cell lysates were prepared as described above, and electrophoresed on SDS polyacrylamide gels. Proteins were transferred to nitrocellulose membranes, which was then treated with blocking solution (1xPBS, 0.2% Tween 20, 5% non-fat dry milk) overnight at 4 0 C. Membranes were incubated with mouse monoclonal anti-NA antibodies (1:500 dilution in blocking solution) for 3 hours at room temperature. After washing three times with blocking solution (15 minutes each), membranes were incubated with HRP-conjugated rabbit anti-mouse IgG antibodies (1:1000 dilution in blocking solution) for 1 hour at room temperature. After washing three times in blocking solution (10 minutes each) and three times in 1xPBS supplemented with 0.2% Tween 20, membranes were developed in ECL (PIERCE) according to the manufacture's instruction, and exposed to Kodak X-ray film. As shown in Figure IC, a strong ~60KD band (similar to the size of MyrAkt1HA, data not shown) is present in CMV6-[MyrAkt3HA] transfected samples, but not in CMV6 transfected samples (negative control). Taken together, these data demonstrate that transfection with CMV6-[MyrAkt3HA] results in functional Akt activity. Example 3: Stimulation of VEGF Expression Example 3.1: Cell culture WO 00/77190 PCTUSOO/15098 -39 HeLa cells (ATCC) were maintained in DME media supplemented with 10% fetal bovine serum (FBS). Cells were grown in 37 0 C, 5%CO2 incubator. Human skeletal muscle cells (HSKMCs) and human coronary smooth muscle cells (HCSMCs) were purchased from Clonetics Corporation. Neonatal rat cardiomyocytes were isolated using a Myocyte Isolation System (Worthington Biochemical Co.). Briefly, hearts collected from 1- to 3-day old rats were minced, digested with trypsin (final concentration 50 pg/ml) overnight at 4'C, followed by digestion with collagenase at 37'C for 45 minutes. After trituration, mixtures were filtered through cell strainer. After brief centrifugation, cells were resuspended in plating media (DMEM:M199 = 4:1, 10% heat-inactivated horse serum, 5% fetal bovine serum, 1x insulin-transferrin-selenium supplement (Gibco BRL), and lxGentamicin, 100 jig/ml BrdU) at density of 0.3x10 6 cells/ml). Cells were switched to low mitogen medium (DMEM:M199 = 4:1, lxGentamicin) 24 hours later. Example 3.2: Transfection One day prior to transfection, cells were split at density of 0.2x 106 cells/cm 2 . Transfections were carried out using lipofectAmine (Gibco BRL) according to the manufacture's instruction. Briefly, indicated DNAs were mixed in DME media (without serum or antibiotics), and lipofectAmine was added (DNA:LipofectAmine =1 ig : 4 pl). After being briefly vortexed, the DNA/lipofectAmine mixture was kept at room temperature for 30 minutes. Cells were then washed with lxPBS, and exposed to the DNA/lipofectAmine mixture for 3 hours. After transfection, cells were washed two times with 1xPBS and switched to DMEM-10%FBS media. 3.3 Recombinant adenovirus construction A recombinant adenovirus containing constitutively active human Akt3 (hAkt3cak) was constructed as described in Crouzet et al. (1997) (Proc. Natl. Acad. Sci. USA. Vol. 94, 1414-1419). A cDNA for constitutively active human Akt3 (comprising a myristylation sequence from c-src ar the N-terminus) was subeloned into pXL2996 (this plasmid is called pXL2996-hAkt3cak). The expression cassette for hAkt3cak from pXL2996-hAkt3cak was subcloned into the shuttle vector pXL3474. This shuttle plasmid for hAkt3cak and the plasmid DNA for adenoviral-bgal (pXL3215) were introduced into bacteria JM83 cells by electroporation. After double homologous recombination, plasmid DNA for adenoviral-hAkt3cak was purified by CsCl. This DNA was linearized by digestion with the restriction enzyme Pac and transfected into 293 cells using lipofectAmine. Three weeks after transfection, recombinant adenovirus containing hAkt3cak (AV-hAKT3cak) was collected, and amplified in 293 cells. Viral titer was determined using a cytoplasmic toxicity assay (CPA). A recombinant adenovirus containing constitutively active mouse akt 1 (AV-mAkt 1 cak) was prepared using standard methodology (discussed above) and provided by Dr. Kenneth Walsh (Boston,
MA).
WO 00/77190 PCTIUSOO/15098 -40 Prior to viral infection, viruses were diluted in tissue culture media at the concentration of 3x10 7 /ml). Iml of virus-containing media was added to each well of a 6-well tissue culture plate, and 8ml of virus-containing medium was added to each 100-mm culture dish. After overnight infection, excess viruses in the media were washed off with lxPBS, and cells were switched to normal media. Example 3.4: ELISA assay Human VEGF Elisa assays were performed using a VEGF-165 ELISA detection kit (purchased form R&D Systems, Inc., cat. DVEOO). Culture media was collected and cleared by brief centrifugation. Samples were added to each appropriate well after addition of assay diluent RD 1W. The plate was then allowed to incubate for two hours at room temperature. Each well was then washed with wash buffer three times. Following this, each well was treated with the provided conjugated anti-VEGF at room temperature for two hours. At this point the same aforementioned wash step was repeated. The substrate was added and incubated for twenty minutes at room temperature. Optical density was determined via a microplate reader set to 450nm, and wavelength correction was set at 540nm. Example 3.5: RNA isolation and Northern blotting: Total RNA was isolated using Ultraspec RNA Isolation reagents (Biotecx).Briefly, 1ml of Ultraspec solution was added to cells in 100mm tissue culture plates. Cells were scraped from the plate and transferred into RNase-free Eppendorf tubes. After addition of 2 0 0 pl of chloroform, mixtures were vortexed and centrifuged at 4"C. The aqueous solution (upper layer) was collected, and RNA was precipitated with equal volume of iso-propanol. After washing with 70% ethanol and drying, RNA was dissolved in DEPC-treated water. 20pug of total RNA was separated on 1% agarose gel. After electrophoresis and transfer, the blot was UV-cross linked. Hybridized was performed with a p32-labeled DNA probe, generated using a Random Primer DNA labeling kit (Boehringer Mannheim). After hybridization at 65'C, the blot was washed sequentially with 2xSSC/0.1%SDS and 0.lxSSC/0.1%SDS, and exposed to Kodak X-ray film overnight. Example 3.6: Akt increases the expression of VEGF from transfected cells HeLa cells were transfected with an expression plasmid for activated mouse Aktl (CMV6 mAktlcak), activated human Akt3 (CMV6-hAkt3) or CMV6 vector (as a control). After transfection cells were switched to low mitogen media (DMEM supplemented with 0.5% fetal bovine serum). 16 hours later, media from transfected cells was collected and subjected to ELISA for human VEGF-165. As shown in Figure 2, the VEGF level in the media of Aktl- or Akt3-transfected cells is significantly higher than that present in the media of vector CMV6 transfected cells (as control). These data demonstrated that constitutively active Aktl or Akt3 induces VEGF-165 expression in HELA cells.
WO 00/77190 PCTUSOO/15098 -41 Example 3.7: A V-mAktlcak and A V-hAkt3cak induce VEGF expression in human skeletal muscle and human smooth muscle cells Human skeletal muscle cells (HSKMCs) and human coronary smooth muscle cells (HCASMCs) were infected with recombinant adneoviruses expressing either active mouse Aktl (AV mAktlcak) or constitutively active human Akt3 (AV-hAKT3cak). As a control, cells were infected with AV-GFP, which drives the expression of green fluorescence protein. One day after infection, culture media was collected, and the VEGF level in the media measured by ELISA. As shown in Figure 3A, both AV-mAKTlcak and AV-hAKT3cak significantly increase VEGF-165 expression in HSKMCs, while AV-GFP infection had little or no effect. Further, as shown in Figure 3B, AV mAktl and AV-hAkt3cak induce VEGF-165 from human coronary artery smooth muscle cells (HCASMCs). In order to evaluate the effect of Akt on VEGF messenger RNA, HCASMCs were infected with adenovirus expressing mAktlcak, hAkt3cak, or GFP. As a positive control, cells were switched to hypoxic conditions for 24 hours. Total RNA was isolated and subjected to Northern blot analysis for VEGF. As shown in Figure 3C, hypoxia treatment dramatically induces the expression VEGF. In addition, AV-mAktlcak and AV-hAkt3cak, but not AV-GFP, significantly increase the mRNA level of VEGF. These data indicate that Akt increases VEGF expression by increasing the level of mRNA. Example 3.8: A V-mAKT1 cak and A V-hAKt3cak induces VEGF expression in cardiomyocytes Neonatal rat cardiomyocytes were infected with adenovirus encoding mAkt 1 cak (AV mAktlcak), mouse wild type Aktl (AV-mAktlwt), hAkt3cak (AV-hAkt3cak) or AV-GFP (as a control). As a positive control, non-infected cells were incubated under hypoxic conditions for 24 hours. One day after infection, RNA from these cells were isolated, and VEGF expression was detected by Northern blot analysis. As shown in Figure 3, AV-mAKtlcak or AV-hAkt3cak significantly increased the expression of VEGF in cardiomyocytes, while AV-GFP or AV-mAktlwt has little or no effect on VEGF- 165 expression. The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and the accompanying figures. Such modifications are intended to fall within the scope of the appended claims. It is further to be understood that all base sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids or polypeptides are approximate, and are provided for description. Various publications are cited herein, the disclosures of which are incorporated by reference in their entireties.
WO 00/77190 PCT/USOO/15098 -42 Aams JM. et al. 1998. The Bcl-2 portein family: arbiters of cell survival. Science. Vol281(5381): 1322-1326. Alessi DR. et al. 1996. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. Vol15(23): 6541-6551. Barinaga M. 1998a. Is apoptosis key in Alzheimer's disease? Science Vol281: 1303-1304. Barinaga M. 1998b. Stroke-damaged neurons may commit cellular sucide. Science Vo1281: 1302 1303. Chang HY et al. 1998. Activation of apoptosis signal- regulating kinase (ASKI) by the adapter protein DAXX. Science. Vol281 (5384): 1860-1863. Cross DA. et al.1995. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature Vol378(6559):785-789. Downward J. 1998. Mechanisms and consequences of activation of protein kinase B/Akt. Curr. Opin. Cell Biol. Vol10(2): 262-267 Dudek H. et al. 1997. Regulation of neuronal survival by the serine-threonine protein kinase Akt. Science Vol275(5300): 661-665. Franke TF. et al. 1995. The protein kinase encoded by the Akt proto-oncogene is a target of PDGF activated phosphatidylinositol 3-kinase. Cell Vol8l(5): 727-736. Franke TF. et al. 1997. P13K: downstream AKTion blocks apoptosis. Cell Vol88(4): 435-437. Hemmings BA. 1997a. Akt signalling: linking membrane events to life and death decisions. Science Vol275(5300): 628-630. Hemmings BA. 1997b. Ptdlns(3.4.5)P3 gets its message across. Science Vol277: 534. Ichijo H. et al. 1997. Induction of apoptosis by ASKI, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science. Vol 275: 90-94. Kauffmann-Zeh A. et al. 1997. Suppression of c-Myc-induced apoptosis by Ras signalling through PI(3)K and PKB. Nature Vol385(6616): 544-548. Klippel A. et al.1997. A specific product of phosphatidylinositol 3-kinase directly activates the protein kinase Akt through its pleckstrin homology domain. Mol. Cell Biol. Vol1 7(1): 338 344. Konishi H. et al. 1995. Molecular cloning and characterization of a new member of the RAC protein kinase family: association of the pleckstrin homology domain of three types of RAC protein kinase with protein kinase C subspecies and beta gamma subunits of G proteins. Biochem. Biophys. Res. Comm. Vol. 216(2): 526-534. Kulik G. et al. 1997. Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol. Cell Biol. Vol17(3): 1595-1606.
WO 00/77190 PCT/USOO/15098 -43 MacLellan WR. et al. 1998. Death by design: programmed cell death in cardiovascular biology and diseases. Circ. Res. Vol81(2): 137-144. Okubo Y. et al. 1998. Insulin-like growth factor-i inhibits the stress-activated protein kinase/c-Jun N terminal kinase. JBiol. Chem. Vol273: 25961-25966. Pullen N. et al. 1998. Phosphorylation and activation of p70S6K by PDK1. Science Vol279: 707-710. Sambrook J., Fritsch EF. & Maniatis T. 1989. Moleucular Cloning ( 2 "d edition). Staal SP. 1987. Molecular cloning of the Akt oncogene and its human homologues AKT1 and AKT2: amplification of AKT1 in a primary human gastric adenocarcinoma. Proc. Natl. Acad. Sci. USA. Vol84(14): 5034-5037. Stephens L. et al. 1998. Protein kinase B kinases that mediated phosphatidylinositol 3,4,5 triphosphate-dependent activation of protein kinase B. Science Vol279: 710-714. Stokoe D. et al. 1997. Dual role of phosphatidylinositol-3,4,5-triphosphate in the activation of protein kinase B. Science Vol277: 567-570. Thornberry NA. et al. 1998. Caspases: enemies within. Science Vol281: 1312-1316.

Claims (42)

1. A method of inducing expression of VEGF in a cell, the method comprising administering to the cell an Akt protein.
2. The method according to claim 1, wherein the Akt protein is selected from the group consisting of Aktl, Akt2 and Akt3.
3. The method according to claim 2, wherein the Akt protein is Akt3.
4. The method according to claim 1, wherein the VEGF is selected from the group consisting of VEGF 2 1, VEGFI 6 s, VEGFi 89 , VEGF 2 0 6 , VEGF-2, VEGF-B, and VEGF-D.
5. The method according to claim 1, wherein the administering comprises introducing into the cell a nucleic acid encoding the Akt protein operatively associated with an expression control sequence.
6. The method according to claim 5, wherein the nucleic acid is part of a plasmid or viral vector.
7. The method according to claim 6, wherein the nucleic acid is part of a plasmid.
8. The method according to claim 6, wherein the viral vector is selected from the group consisting of retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus.
9. The method according to claim 5, wherein the Akt is constitutively expressed within the cell.
10. The method according to claim 1, further comprising administration of a transition metal ion and/or a vasodilator.
11. The method according to claim 5, further comprising administering a nucleic acid encoding a second angiogenic factor operatively associated with an expression control sequence.
12. The method according to claim 11, wherein the second angiogenic factor is selected from the group consisting of a VEGF, acidic fibroblast growth factor, basic fibroblast growth factor, endothelial cell growth factor, and an angiopoietin.
13. The method according to claim 12, wherein the VEGF is selected from the group consisting of VEGF 2 1, VEGFi 65 , VEGF 18 9 , VEGF 2 06 , VEGF-2, VEGF-B, and VEGF-D.
14. The method according to claim 12, wherein the second angiogenic factor is endothelial cell growth factor.
15. The method according to claim 2, wherein at least two forms of Akt protein are administered to the cell.
16. The method according to claim 1, wherein the cell is in a patient suffering from an ischemic condition.
17. The method according to claim 16, wherein the ischemic condition is cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, or ischemic, idiopathic or hypertrophic cardiomyopathy. WO 00/77190 PCTIUSOO/15098 -45
18. A method of inducing expression of VEGF in cells of a patient suffering from an ischemic condition, the method comprising administering to the patient an Akt protein.
19. The method according to claim 18, wherein the Akt protein is selected from the group consisting of Aktl, Akt2 and Akt3.
20. The method according to claim 19, wherein the Akt protein is Akt3.
21. The method according to claim 18, wherein the VEGF is selected from the group consisting of VEGF1 2 i, VEGFi 65 , VEGFs 9 , VEGF 2 0 6 , VEGF-2, VEGF-B, and VEGF-D.
22. The method according to claim 18, wherein a nucleic acid encoding the Akt protein operatively associated with an expression control sequence is administered to the patient.
23. The method according to claim 22, wherein the nucleic acid is part of a plasmid or viral vector.
24. The method according to claim 23, wherein the nucleic acid is part of a plasmid.
25. The method according to claim 23, wherein the viral vector is selected from the group consisting of retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus.
26. The method according to claim 22, wherein the Akt is constitutively expressed within the cell.
27. The method according to claim 18, further comprising administration of a transition metal ion and/or a vasodilator to the patient.
28. The method according to claim 18, wherein the ischemic condition is cerebrovascular ischemia, renal ischemia, pulmonary ischemia, limb ischemia, myocardial ischemia, or ischemic, idiopathic or hypertrophic cardiomyopathy.
29. The method according to claim 22, further comprising administering a nucleic acid encoding a second angiogenic factor operatively associated with an expression control sequence.
30. The method according to claim 29, wherein the second angiogenic factor is selected from the group consisting of a VEGF, acidic fibroblast growth factor, basic fibroblast growth factor, endothelial cell growth factor, and an angiopoietin.
31. The method according to claim 30, wherein the VEGF is selected from the group consisting of VEGF1 2 i, VEGFi 65 , VEGFi 8 9 , VEGF 2 0 6 , VEGF-2, VEGF-B, and VEGF-D.
32. The method according to claim 30, wherein the second angiogenic factor is endothelial cell growth factor.
33. The method according to claim 16, wherein at least two forms of Akt protein are administered to the patient.
34. A pharmaceutical composition comprising a nucleic acid encoding an Akt protein, a transition metal and/or a vasodilator, and a pharmaceutically acceptable vehicle.
35. The composition according to claim 34, wherein the nucleic acid is part of a plasmid or viral WO 00/77190 PCT/USOO/15098 -46 vector.
36. The composition according to claim 35, wherein the nucleic acid is part of a plasmid.
37. The composition according to claim 55, wherein the viral vector is selected from the group consisting of retrovirus, adenovirus, adeno-associated virus, herpes virus, and vaccinia virus.
38. A method of inhibiting angiogenesis in a patient suffering from a tumor, the method comprising inhibiting the level of Akt activity in the patient, thereby inhibiting production of VEGF.
39. The method according to claim 38, comprising introducing an Akt antisense nucleic acid into cells of the patient under conditions wherein the antisense nucleic acid hybridizes under intracellular conditions to an Akt mRNA.
40. The method according to claim 38, comprising introducing an intracellular binding protein that specifically binds Akt into a patient's cells at a level sufficient to bind to and inactivate Akt.
41. The method according to claim 40, wherein the intracellular binding protein is a single chain Fv antibody (scFv).
42. The method according to claim 38, comprising introducing a nucleic acid encoding a dominant negative form of an Akt.
AU51758/00A 1999-06-11 2000-06-01 Induction of vascular endothelial growth factor (VEGF) by the serine/threonine protein kinase AKT Ceased AU773450B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US13872499P 1999-06-11 1999-06-11
US60/138724 1999-06-11
GB9926058 1999-11-03
GB9926058 1999-11-03
PCT/US2000/015098 WO2000077190A2 (en) 1999-06-11 2000-06-01 Induction of vascular endothelial growth factor (vegf) by the serine/threonine protein kinase akt

Publications (2)

Publication Number Publication Date
AU5175800A true AU5175800A (en) 2001-01-02
AU773450B2 AU773450B2 (en) 2004-05-27

Family

ID=26316052

Family Applications (1)

Application Number Title Priority Date Filing Date
AU51758/00A Ceased AU773450B2 (en) 1999-06-11 2000-06-01 Induction of vascular endothelial growth factor (VEGF) by the serine/threonine protein kinase AKT

Country Status (17)

Country Link
EP (1) EP1187911A2 (en)
JP (1) JP2003530818A (en)
KR (1) KR20020012270A (en)
CN (1) CN1360629A (en)
AU (1) AU773450B2 (en)
BR (1) BR0011503A (en)
CA (1) CA2376630A1 (en)
CZ (1) CZ20014444A3 (en)
HK (1) HK1041500A1 (en)
HU (1) HUP0201663A3 (en)
IL (1) IL146730A0 (en)
MX (1) MXPA01012748A (en)
NO (1) NO20016025L (en)
NZ (1) NZ516054A (en)
PL (1) PL352859A1 (en)
SI (1) SI20978A (en)
WO (1) WO2000077190A2 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1144600T1 (en) * 1999-03-19 2008-12-31 Aventis Pharma Inc Akt-3 nucleic acids, polypeptides, and uses thereof
US6881555B2 (en) 1999-03-19 2005-04-19 Aventis Pharmaceuticals Inc. AKT nucleic acids, polypeptides, and uses thereof
HUP0200799A3 (en) * 1999-04-16 2004-11-29 Univ Yale New Haven Enos mutations useful for gene therapy and therapeutic screening
US6187586B1 (en) * 1999-12-29 2001-02-13 Isis Pharmaceuticals, Inc. Antisense modulation of AKT-3 expression
US20020102260A1 (en) 2000-03-02 2002-08-01 Marc Achen Methods for treating neoplastic disease characterized by vascular endothelial growth factor D expression, for screening for neoplastic disease or metastatic risk, and for maintaining vascularization of tissue
WO2001064235A1 (en) * 2000-03-02 2001-09-07 Ludwig Institute For Cancer Research Methods for treating, screening for, and detecting cancers expressing vascular endothelial growth factor d
US7531318B2 (en) 2004-08-20 2009-05-12 Board Of Regents, The University Of Texas System Screening of agents for activity against ischemic myocardial insults

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1144600T1 (en) * 1999-03-19 2008-12-31 Aventis Pharma Inc Akt-3 nucleic acids, polypeptides, and uses thereof

Also Published As

Publication number Publication date
KR20020012270A (en) 2002-02-15
PL352859A1 (en) 2003-09-08
MXPA01012748A (en) 2002-07-02
CN1360629A (en) 2002-07-24
CZ20014444A3 (en) 2002-05-15
JP2003530818A (en) 2003-10-21
AU773450B2 (en) 2004-05-27
EP1187911A2 (en) 2002-03-20
BR0011503A (en) 2002-03-05
IL146730A0 (en) 2002-07-25
CA2376630A1 (en) 2000-12-21
HUP0201663A3 (en) 2005-01-28
WO2000077190A2 (en) 2000-12-21
HUP0201663A2 (en) 2002-08-28
HK1041500A1 (en) 2002-07-12
NO20016025D0 (en) 2001-12-10
WO2000077190A3 (en) 2001-04-12
NO20016025L (en) 2002-02-08
NZ516054A (en) 2004-04-30
SI20978A (en) 2003-02-28

Similar Documents

Publication Publication Date Title
US7662628B2 (en) Human Akt3 nucleic acid and uses thereof
IL190270A (en) Use of akt3 nucleic acids for the manufacture of a medicament for inhibiting cell death, treating myocardial infarction or ischemia reperfusion injury
AU773450B2 (en) Induction of vascular endothelial growth factor (VEGF) by the serine/threonine protein kinase AKT
AU2005203749A1 (en) Variants of Traf2 which act as an inhibitor of TNF-ALPHA (TNF alpha) signaling pathway
US20030044895A1 (en) Nucleic acids of the human ABCA5, ABCA6, ABCA9, AND ABCA10 Genes, vectors containing such nucleic acids, and uses thereof
US7332593B2 (en) Variants of TRAF2 which act as an inhibitor of TNF-alpha (TNFα) signaling pathway
MXPA01009431A (en) Akt nucleic acids, polypeptides, and uses thereof

Legal Events

Date Code Title Description
TC Change of applicant's name (sec. 104)

Owner name: AVENTIS PHARMACEUTICALS INC.

Free format text: FORMER NAME: AVENTIS PHARMACEUTICALS PRODUCTS INC.

FGA Letters patent sealed or granted (standard patent)