US20040126885A1 - Delivery vehicle for recombinant proteins - Google Patents

Delivery vehicle for recombinant proteins Download PDF

Info

Publication number
US20040126885A1
US20040126885A1 US10/701,331 US70133103A US2004126885A1 US 20040126885 A1 US20040126885 A1 US 20040126885A1 US 70133103 A US70133103 A US 70133103A US 2004126885 A1 US2004126885 A1 US 2004126885A1
Authority
US
United States
Prior art keywords
cell
cells
promoter
nucleic acid
platelet
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.)
Abandoned
Application number
US10/701,331
Inventor
Douglas Cines
Mortimer Poncz
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.)
Childrens Hospital of Philadelphia CHOP
University of Pennsylvania Penn
Original Assignee
Cines Douglas B.
Mortimer Poncz
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 Cines Douglas B., Mortimer Poncz filed Critical Cines Douglas B.
Priority to US10/701,331 priority Critical patent/US20040126885A1/en
Publication of US20040126885A1 publication Critical patent/US20040126885A1/en
Priority to US11/581,559 priority patent/US7939063B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: CHILDREN'S HOSPITAL OF PHILADELPHIA
Assigned to THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA reassignment THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CINES, DOUGLAS B
Assigned to THE CHILDREN'S HOSPITAL OF PHILADELPHIA reassignment THE CHILDREN'S HOSPITAL OF PHILADELPHIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PONCZ, MORTIMER
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: CHILDREN'S HOSPITAL OF PHILADELPHIA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0644Platelets; Megakaryocytes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/522Alpha-chemokines, e.g. NAP-2, ENA-78, GRO-alpha/MGSA/NAP-3, GRO-beta/MIP-2alpha, GRO-gamma/MIP-2beta, IP-10, GCP-2, MIG, PBSF, PF-4, KC
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/721Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • 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
    • 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
    • C12N2517/00Cells related to new breeds of animals
    • C12N2517/02Cells from transgenic animals
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/008Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian

Definitions

  • Hematopoietic stem cells are pluripotent cells present in the bone marrow, which divide to produce more specialized progenitor stem cells, i.e., lymphoid progenitors and myeloid progenitors.
  • Cells that are differentiated from the lymphoid progenitors in the bone marrow and that are found in the peripheral blood include B cells and T cells. From B cells are generated plasma cells; from T cells are generated activated T cells.
  • the common myeloid progenitor stem cells produce the granulocytes/macrophage progenitor cells and the megakaryocyte/erthyrocyte progenitor cells in the bone marrow.
  • Cells differentiated from the granulocyte/macrophage progenitors that are present in the blood include neutrophils, eosinophils, basophils, monocytes and immature dendritic cells. Monocytes further give rise to mast cells, macrophages and dendritic cells that are present in tissue and lymph nodes.
  • Cells differentiated from the megakaryocyte/erthrocyte progenitors include megakaryocytes and erythroblasts, which further differentiate into platelets and erythrocytes (red blood cells) in the blood.
  • a number of these hematopoietic lineage cells are secretory cells upon activation.
  • platelets the smallest corpuscular components of human blood, are characterized by a diameter of about 2-4 micrometers, the absence of a nucleus, and a physiological number varying from 150,000 to 300,000 per cubic millimeter of blood. Platelets contribute to the complex, multistep, and highly regulated process of thrombus formation and arterial occlusive disorders, a leading cause of human morbidity. Platelets target and adhere to sites of vascular injury. At the sites of vascular injury, the platelets are activated and form aggregates that provide a provisional seal.
  • Platelets preferentially release their granular contents at the site of injury, e.g., contributing to the subsequent growth and stability of thrombi in part through the release of von Willebrand factor (vWF), fibrinogen, and other coagulation proteins such as Factor V (Holt J. C., and Niewiarowski, S. 1985 Sem. Hematol. 22:151-163) from their alpha-granules.
  • vWF von Willebrand factor
  • fibrinogen fibrinogen
  • Other coagulation proteins such as Factor V (Holt J. C., and Niewiarowski, S. 1985 Sem. Hematol. 22:151-163) from their alpha-granules.
  • Activated platelets also release proteins that inhibit thrombolysis, chief among which is plasminogen activator inhibitor-1 (PAI-1). Over 90% of the circulating PAI-1 is stored in platelet alpha-granules (Booth, N. A et al, 1988 Brit. J. Hae
  • platelets also contain or can bind small amounts of plasma-derived profibrinolytic proteins, including urokinase-type plasminogen activator (u-PA) and plasminogen (Fay, W. P et al, 1994 cited above; Lenich, C et al, 1997 Blood 90:3579-3586; Jiang, Y et al. 1996 Blood 87:2775-2781; Holt, J. C., and Niewiarowski, S. 1980 Circulation 62:342a).
  • u-PA urokinase-type plasminogen activator
  • plasminogen plasminogen
  • Quebec Platelet Disorder is a rare bleeding disorder not responsive to platelet transfusion, but responsive to anti-fibrinolytic agents, such as tranexamic acid (Hayward, C. P. et al, 1997 Blood 89:1243-1253; Hayward, C. P. et al, 1996 Blood 87:4967-4978; Hayward, C. P. et al, 1997 Brit. J. Haematol. 97:497-503).
  • the etiology of QPD has been ascribed recently to ectopic expression of an excess of u-PA in megakaryocytes and platelets (Kahr, W.
  • QPD platelets contain predominantly activated two-chain urokinase (tcu-PA).
  • tcu-PA activated two-chain urokinase
  • the etiology for the bleeding diathesis may in part be due to local release of activated u-PA within thrombi leading to premature lysis.
  • degradation of multiple platelet alpha-granular proteins, including vWF and Factor V, presumably by plasmin generated as a result of urokinase, may interfere with thrombus development as well.
  • the invention provides a recombinant nucleic acid molecule comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of the transgene product in a hematopoietic progenitor cell or a cell differentiated therefrom
  • the cell that expresses the transgene is a secretory cell.
  • the cell is a lymphoid progenitor cell, a myeloid progenitor cell or a cell further differentiated therefrom, such as a platelet.
  • the invention provides a hematopoietic progenitor, lymphoid progenitor, or nyeloid progenitor host cell transformed, transduced, infected or transfected with an above-described nucleic acid molecule.
  • the invention provides a host cell differentiated from a progenitor cell that is transformed, transduced, infected or transfected with an above-described nucleic acid molecule.
  • the invention provides a platelet transformed, transduced, infected or transfected with a nucleic acid molecule comprising a first sequence encoding a transgene, which is optionally a fibrinolytic protein, under the control of regulatory sequences that direct expression of the transgene in the platelet.
  • the invention provides a method for generating a modified hematopoietic stem cell, a progenitor cell, or a modified cell differentiated from the hematopoietic stem cell and/or progenitor cell.
  • the method involves transferring a nucleic acid molecule as described above into the cell via transformation, transduction, infection or transfection.
  • the invention provides methods for treating or preventing certain disorders, diseases, symptoms or injuries in which cells of the hematopoietic lineage are involved, by delivering to a mammalian patient a recombinant nucleic acid molecule described above or a suitable host cell described above, that contain a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in the host cell.
  • a differentiated cell produces the product at a suitable in the mammal.
  • the invention provides a method for preventing unwanted thrombus formation in a mammal by administering an above-described molecule or platelet containing same, in which the transgene is a fibrinolytic protein and the regulatory sequences direct expression of the product of the transgene in a platelet.
  • the platelet produces the product at the site of the thrombus formation.
  • FIG. 1 is a schematic representation of the 10.2 kb Sac II insert obtained from the transgenic mice expressing u-PA in their platelets.
  • the insert contains the 1.2 kb murine Xba I/Kpn I PF4 promoter (open box) plus a 5′ untranslated region (5′-UTR; light gray box) followed by the 11-exon murine u-PA gene (Heckel, J. L. et al, 1990 Cell 62:447-456, black boxes) and ending with the hGH 3′-UTR and polyadenylation sequence (dark gray box).
  • the 2.8 kb Bgl II fragment containing the transgene was detected in a genomic Southern blot (not shown).
  • the present invention provides novel nucleic acid molecules, host cells containing the molecules, and methods for delivery of recombinant transgenes using modified cells of the hematopoietic stem cell lineage. Methods for treatment and prophylaxis using these cells are disclosed for a variety of disorders involving or mediated by the cells of the hematopoietic lineage. Such disorders can include inflammations, infections, tissue injuries, vascular injuries, and the like.
  • One aspect of this invention includes isolated, synthetic or recombinant nucleic acid molecules and sequences comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of the transgene in a cell of the hematopoietic lineage.
  • nucleic acid molecules are used to express the transgene product in vitro or ex vivo, or to permit expression of the transgene product in vivo in a mammal.
  • isolated nucleotide molecule or sequence refers to a nucleic acid segment or fragment which is free from contamination with other biological components that are associated with the molecule or sequence in its natural environment.
  • isolated nucleotide molecule or sequence of this invention is a sequence separated from sequences which flank it in a naturally occurring state, e.g., a DNA fragment which has been removed from the sequences which are normally adjacent to the fragment, such as the sequences adjacent to the fragment in a genome in which it naturally occurs.
  • the nucleotide sequences and molecules of this invention have been altered to encode a selected transgene product.
  • isolated nucleic acid molecule or sequence also applies to nucleic acid sequences or molecules that have been substantially purified from other components that naturally accompany the unmutagenized nucleic acid, e.g., RNA or DNA, or proteins, in the cell.
  • An isolated nucleotide molecule or sequence of this invention also encompasses sequences and molecules that have been prepared by other conventional methods, such as recombinant methods, synthetic methods, e.g., mutagenesis, or combinations of such methods.
  • nucleotide sequences or molecules of this invention should not be construed as being limited solely to the specific nucleotide sequences presented herein, but rather should be construed to include any and all nucleotide sequences which share homology (i.e., have sequence identity) with the nucleotide sequences presented herein.
  • promoter or “regulatory sequence” is meant a DNA sequence required to direct expression of a nucleic acid operably linked thereto in a cell of hematopoietic lineage.
  • the promoter/regulatory sequence is positioned at the 5′ end of the transgene coding sequence such that it drives expression of the transgene-encoded protein in a cell.
  • the promoter/regulatory sequence may also include an enhancer sequence and other regulatory elements that are required for expression in these hematopoietic cells.
  • Suitable regulatory sequences/promoters for use in the present invention are readily selected from among regulatory sequences that express the selected transgene in the hematopoietic stem cell itself, or in one of the progenitor cells of the hematopoietic lineage, including the common lymphoid progenitor, the common myeloid progenitor, the megakaryotic/erythrocyte progenitor or the granulocyte/macrophage progenitor.
  • the regulatory sequences are able to direct expression of the transgene in a cell that is further differentiated from these progenitor cells.
  • suitable cells differentiated from the megakaryotic/erythrocyte progenitor cells are platelets, megakaryocytes or erythrocytes.
  • Suitable cells differentiated from granulocyte/macrophage progenitor cells are neutrophils, eosinophils, monocytes, basophils and immature dendritic cells.
  • Still other suitable cells differentiated from monocytes are mast cells, macrophages and dendritic cells.
  • Suitable cells differentiated from said lymphoid progenitors are natural killer cells. Promoters capable of directing expression of a transgene in any of these cells of the hematopoietic lineage are useful as regulatory sequences in the nucleic acid molecules of this invention.
  • the promoter expresses the gene in secretory cells of the hematopoietic lineage, i.e., cells that release the contents of their granules upon activation, e.g., platelets, mast cells, neutrophils, eosinophils, etc. See, e.g., IMMUNOBIOLOGY, THE IMMUNE SYSTEM IN HEALTH AND DISEASE, 5 th edit., C. Janeway et al., Ed., Garland Publishing, New York, N.Y.: 2001.
  • a platelet-specific promoter is used as a regulatory sequence to direct expression of the transgene in a plasmid.
  • suitable promoters are, without limitation, the Platelet factor 4 (PF4) promoter, the glycoprotein IIb promoter, the glycoprotein IIIa promoter, and the glycoprotein VI promoter.
  • the regulatory sequence is a neutrophil-specific promoter, such as one or more of the human neutrophil alpha defensin promoters DEFA1, DEFA2, DEFA3, and DEFA4, among others.
  • the regulatory sequence is a natural killer cell-specific promoter, for example, the human perform gene promoter.
  • the regulatory sequence useful in the nucleic acid molecule of this invention is an eosinophil-specific promoter.
  • eosinophil-specific promoters include, without limitation, the human eotaxin gene promoter and the eosinophil peroxidase gene promoter.
  • a suitable regulatory sequence is an erythrocyte-specific promoter, such as the human RhD gene promoter.
  • the transgene is any peptide, polypeptide or protein useful for the treatment of a disorder or reduction or prevention of a symptom in which cells of the hematopoietic system are involved.
  • a non-exclusive list of products includes those encoded by therapeutic transgenes for the treatment of a variety of inflammatory conditions, microbial or parasitic infections, injuries, such as vascular injuries and other hematopoietic cell-involved disorders.
  • such products include fibrinolytic proteins, including without limitation, urokinase-type plasminogen activator, plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor, fibrinogen, Factor V, and plasminogen for use in altering the hemostatic balance at sites of thrombosis.
  • fibrinolytic proteins including without limitation, urokinase-type plasminogen activator, plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor, fibrinogen, Factor V, and plasminogen for use in altering the hemostatic balance at sites of thrombosis.
  • Suitable products also include, without limitation, hormones and growth and differentiation factors including, without limitation, insulin, glucagon, growth hormone (GH), parathyroid hormone (PTH), growth hormone releasing factor (GHRF), follicle stimulating hormone (FSH), luteinizing hormone (LH), human chorionic gonadotropin (hCG), vascular endothelial growth factor (VEGF), angiopoietins, angiostatin, granulocyte colony stimulating factor (GCSF), erythropoietin (EPO), connective tissue growth factor (CTGF), basic fibroblast growth factor (bFGF), acidic fibroblast growth factor (aFGF), epidermal growth factor (EGF), transforming growth factor a (TGF ⁇ ), platelet-derived growth factor (PDGF), insulin growth factors I and II (IGF-I and IGF-II), any one of the transforming growth factor ⁇ superfamily, including TGF ⁇ , activins, inhibins, or any of the bone morphogenic proteins (B
  • transgene products include proteins that regulate the immune system including, without limitation, cytokines and lymphokines such as thrombopoietin (TPO), interleukins (IL) IL-1 through IL-25 (including, IL-2, IL-4, IL-12, and IL-18), monocyte chemoattractant protein, leukemia inhibitory factor, granulocyte-macrophage colony stimulating factor, Fas ligand, tumor necrosis factors ⁇ and ⁇ , interferons ⁇ , ⁇ , and ⁇ , stem cell factor, flk-2/flt3 ligand.
  • TPO thrombopoietin
  • IL interleukins
  • IL-1 through IL-25 including, IL-2, IL-4, IL-12, and IL-18
  • monocyte chemoattractant protein including, IL-2, IL-4, IL-12, and IL-18
  • Fas ligand granulocyte-macrophage colony stimulating factor
  • immunoglobulins IgG, IgM, IgA, IgD and IgE include, without limitation, immunoglobulins IgG, IgM, IgA, IgD and IgE, chimeric immunoglobulins, humanized antibodies, single chain antibodies, T cell receptors, chimeric T cell receptors, single chain T cell receptors, class I and class II MHC molecules, as well as engineered immunoglobulins and MHC molecules.
  • Useful gene products also include complement regulatory proteins, membrane cofactor protein (MCP), decay accelerating factor (DAF), CR1, CF2 and CD59.
  • Still other useful gene products include any one of the receptors for the hormones, growth factors, cytokines, lymphokines, regulatory proteins and immune system proteins.
  • the invention encompasses receptors for cholesterol regulation, including the low density lipoprotein (LDL) receptor, high density lipoprotein (HDL) receptor, the very low density lipoprotein (VLDL) receptor, and the scavenger receptor.
  • the invention also encompasses gene products such as members of the steroid hormone receptor superfamily including glucocorticoid receptors and estrogen receptors, Vitamin D receptors and other nuclear receptors.
  • useful gene products include transcription factors such as jun, fos, max, mad, serum response factor (SRF), AP-1, AP2, myb, MyoD, myogenin, ETS-box containing proteins, TFE3, E2F, ATF1, ATF2, ATF3, ATF4, ZF5, NFAT, CREB, HNF-4, C/EBP, SP1, CCAAT-box binding proteins, interferon regulation factor (IRF-1), Wilms tumor protein, ETS-binding protein, STAT, GATA-box binding proteins, e.g., GATA-3, and the forkhead family of winged helix proteins.
  • SRF serum response factor
  • AP-1 AP-1
  • AP2F myb
  • MyoD myogenin
  • ETS-box containing proteins TFE3, E2F, ATF1, ATF2, ATF3, ATF4, ZF5, NFAT, CREB, HNF-4, C/EBP, SP1, CCAAT-box binding proteins
  • IRF-1 interferon regulation
  • Other useful gene products include, carbamoyl synthetase I, ornithine transcarbamylase, arginosuccinate synthetase, arginosuccinate lyase, arginase, fumarylacetacetate hydrolase, phenylalanine hydroxylase, alpha-1 antitrypsin, glucose-6-phosphatase, porphobilinogen deaminase, factor VIII, factor IX, cystathione beta-synthase, branched chain ketoacid decarboxylase, albumin, isovaleryl-coA dehydrogenase, propionyl CoA carboxylase, methyl malonyl CoA mutase, glutaryl CoA dehydrogenase, insulin, beta-glucosidase, pyruvate carboxylate, hepatic phosphorylase, phosphorylase kinase, glycine decarboxylase, H-protein, T-protein
  • Still other useful gene products include enzymes such as are useful in enzyme replacement therapy, which is useful in a variety of conditions resulting from deficient activity of enzyme.
  • Non-naturally occurring polypeptides such as chimeric or hybrid polypeptides having a non-naturally occurring amino acid sequence containing insertions, deletions or amino acid substitutions.
  • non-naturally occurring polypeptides such as chimeric or hybrid polypeptides having a non-naturally occurring amino acid sequence containing insertions, deletions or amino acid substitutions.
  • single-chain engineered immunoglobulins could be useful in certain immunocompromised patients.
  • Other types of non-naturally occurring gene sequences include antisense molecules and catalytic nucleic acids, such as ribozymes, which could be used to reduce overexpression of a target.
  • compositions of this invention are employed to express as transgene products polypeptides that can reduce the activity or inactivate oncogenes such as myb, myc, fyn, and the translocation gene bcr/abl, ras, src, P53, neu, trk and EGRF.
  • Anti-cancer treatments and protective regimens include transgene products directed to inactivate or reduce the activity of variable regions of antibodies made by B cell lymphomas and variable regions of T cell receptors of T cell lymphomas which, in some embodiments, are also used as target antigens for autoimmune disease.
  • Other tumor-associated polypeptides can be used as target polypeptides such as polypeptides, which are found at higher levels in tumor cells including the polypeptide recognized by monoclonal antibody 17-1A and folate binding polypeptides.
  • Suitable therapeutic polypeptides and proteins include those which are useful for treating individuals suffering from autoimmune diseases and disorders by conferring a broad based protective immune response against targets that are associated with autoimmunity including cell receptors and cells which produce “self”-directed antibodies.
  • T cell mediated autoimmune diseases include Rheumatoid arthritis (RA), multiple sclerosis (MS), Sjögren's syndrome, sarcoidosis, insulin dependent diabetes mellitus (IDDM), autoimmune thyroiditis, reactive arthritis, ankylosing spondylitis, scleroderma, polymyositis, dermatomyositis, psoriasis, vasculitis, Wegener's granulomatosis, Crohn's disease and ulcerative colitis.
  • RA Rheumatoid arthritis
  • MS multiple sclerosis
  • Sjögren's syndrome sarcoidosis
  • IDM insulin dependent diabetes mellitus
  • transgenes encode fibrinolytic proteins and peptides suitable for delivery by transgene expression in platelets, such as illustrated in the below-noted examples.
  • the selection of the transgene sequence, or other molecule carried by the nucleic acid molecule, is not a limitation of this invention. Choice of a transgene sequence is within the skill of the artisan in accordance with the teachings of this application.
  • nucleotide or amino acid position in both of the two molecules When a nucleotide or amino acid position in both of the two molecules is occupied by the same monomeric nucleotide or amino acid, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions, e.g. if half (e.g., five positions in a polymer ten subunits in length) of the positions in two compound sequences are homologous, then the two sequences are 50% homologous. If 90% of the positions, e.g, 9 of 10, are matched or homologous, the two sequences share 90% homology.
  • the DNA sequences 3′ATTGCC5′ and 3′TATGCG5′ share 50% homology.
  • substantially homologous is meant DNA or RNA which is about 70% homologous, more preferably about 80% homologous, and most preferably about 90% homologous to the desired nucleic acid.
  • the invention is also directed to an isolated nucleotide molecule comprising a nucleic acid sequence that is at least 70%, 80% or 90% homologous to a nucleic acid sequence encoding a naturally occurring transgene-encoded protein or a polypeptide that has similar biological effect as the native transgene product.
  • any three-nucleotide codon that encodes a mutant or substituted amino acid residue of a transgene-encoded protein, described herein is within the scope of the invention.
  • proteins, and/or DNA sequences encoding them, or other sequences useful in nucleic acid molecules or compositions described herein are defined by their percent homologies or identities to identified sequences
  • the algorithms used to calculate the percent homologies or percent identities include the following: the Smith-Waterman algorithm (J. F. Collins et al, 1988, Comput. Appl. Biosci., 4:67-72; J. F. Collins et al, Molecular Sequence Comparison and Alignment, (M. J.
  • a single-stranded or double-stranded DNA comprises two DNA sequences and that the two DNA sequences are arranged within the molecule or sequence in such a manner that at least one of the DNA sequences is able to exert a physiological effect by which it is characterized upon the other.
  • each protein encoding transgene sequence and necessary regulatory sequences of this invention are present in a separate viral or non-viral recombinant vector (including non-viral methods of delivery of a nucleic acid molecule into a cell).
  • a separate viral or non-viral recombinant vector including non-viral methods of delivery of a nucleic acid molecule into a cell.
  • two or more of these nucleic acid sequences encoding duplicate copies of a the transgene-encoded protein or encoding multiple different therapeutic proteins of this invention are contained in a polycistronic transcript, i.e., a single molecule designed to express multiple gene products.
  • the isolated nucleic acid of this invention is desirably a recombinant vector, particularly a plasmid, containing isolated and purified DNA sequences comprising DNA sequences that encode a selected therapeutic protein.
  • vector as used herein, is meant a DNA molecule derived from viral or non-viral, e.g., bacterial, species that has been designed to encode an exogenous or heterologous nucleic acid sequence.
  • the term includes conventional bacterial plasmids.
  • Such plasmids or vectors can include plasmid sequences from viruses or phages.
  • Such vectors include chromosomal, episomal and virus-derived vectors, e.g., vectors derived from bacterial plasmids, bacteriophages, yeast episomes, yeast chromosomal elements, and viruses.
  • Vectors may also be derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, cosmids, and phagemids.
  • the term also includes non-replicating viruses that transfer a gene from one cell to another.
  • the term should also be construed to include non-plasmid and non-viral compounds which facilitate transfer of nucleic acid into cells, such as, for example, polylysine compounds and the like.
  • the nucleic acid molecules of the invention include non-viral vectors or methods for delivery of the sequences encoding the therapeutic protein to a host cell according to this invention.
  • a variety of non-viral vectors are known in the art and may include, without limitation, plasmids, bacterial vectors, bacteriophage vectors, “naked” DNA and DNA condensed with cationic lipids or polymers.
  • bacterial vectors include, but are not limited to, sequences derived from bacille Calmette Guérin (BCG), Salmonella, Shigella, E. coli, and Listeria, among others.
  • Suitable plasmid vectors include, for example, pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pK37, pKC101, pAC105, pVA51, pKH47, pUB110, pMB9, pBR325, Co1 E1, pSC101, pBR313, pML21, RSF2124, pCR1, RP4, pBAD18, and pBR328.
  • Suitable inducible Escherichia coli expression vectors include pTrc (Amann et al., 1988 Gene, 69:301-315), the arabinose expression vectors (e.g., pBAD18, Guzman et al, 1995 J. Bacteriol., 177:4121-4130), and pETIId (Studier et al., 1990 Methods in Enzymology, 185:60-89).
  • a suitable cloning vector includes, but is not limited to the vectors such as bacteriophage ⁇ vector system, ⁇ gt11, ⁇ gt ⁇ WES.tB, Charon 4, ⁇ gt-WES- ⁇ B, Charon 28, Charon 4A, ⁇ gt-1- ⁇ BC, ⁇ gt-1- ⁇ B, M13mp7, M13mp8, or M13mp9, among others.
  • a mammalian expression vector is used for expression of the selected transgene in mammalian cells of the hematopoietic lineage.
  • mammalian expression vectors include pCDM8 (Seed, 1987 Nature, 329:840-842) and pMT2PC (Kaufman et al., 1987 EMBO J., 6(1):187-93).
  • the expression vector's control functions are often provided by viral regulatory elements other than the promoters specified above.
  • recombinant vector is a recombinant single or double-stranded RNA or DNA viral vector.
  • viral vector systems are known in the art. Examples of such vectors include, without limitation, recombinant adenoviral vectors, herpes simplex virus (HSV)-based vectors, adeno-associated viral (AAV) vectors, hybrid adenoviral/AAV vectors, recombinant retroviruses or lentiviruses, recombinant poxvirus vectors, recombinant vaccinia virus vectors, SV-40 vectors, insect viruses such as baculoviruses, and the like that are constructed to carry or express a selected nucleic acid composition of interest.
  • HSV herpes simplex virus
  • AAV adeno-associated viral
  • retroviruses or lentiviruses recombinant retroviruses or lentiviruses
  • poxvirus vectors recombinant poxvirus vectors
  • Retrovirus vectors that can be employed include those described in EP 0 415 731; International Patent Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; and WO 93/25234; U.S. Pat. No. 5,219,740; International Patent Publication Nos. WO 93/11230 and WO 93/10218; Vile and Hart, 1993 Cancer Res. 53:3860-3864; Vile and Hart, 1993 Cancer Res. 53:962-967; Ram et al., 1993 Cancer Res. 53:83-88; Takamiya et al., 1992 J. Neurosci. Res. 33:493-503; Baba et al., 1993 J.
  • Suitable recombinant retroviruses include those described in International Patent Publication No. WO 91/02805. See also, D. A. Wilcox et al, 2000 Blood, 95 (12):3645-3652.
  • Alphavirus-based vectors may also be used as the nucleic acid molecule encoding the therapeutic protein.
  • Such vectors can be constructed from a wide variety of alphaviruses, including, for example, Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532).
  • Representative examples of such vector systems include those described in U.S. Pat. Nos. 5,091,309; 5,217,879; and 5,185,440; and International Patent Publication Nos. WO 92/10578; WO 94/21792; WO 95/27069; WO 95/27044; and WO 95/07994.
  • adenoviral vectors include those described by Berkner, 1988 Biotechniques 6:616-627; Rosenfeld et al., 1991 Science 252:431-434; International Patent Publication No. WO 93/19191; Kolls et al., 1994 PNAS 91:215-219; Kass-Eisler et al., 1993 PNAS 90:11498-11502; Guzman et al., 1993 Circulation 88:2838-2848; Guzmanetal., 1993 Cir. Res. 73:1202-1207;Zabner et al., 1993 Cell 75:207-216; Li et al, 1993 Hum. Gene Ther.
  • adenoviral vectors include those described in International Patent Publication Nos. WO 94/12649; WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655.
  • Other adenoviral vectors include those derived from chimpanzee adenoviruses, such as those described in U.S. Pat. No. 6,083,716.
  • Another viral vector is based on a parvovirus such as an adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • Representative examples include the AAV vectors described in International Patent Publication No. WO 93/09239, Samulski et al., 1989 J. Virol. 63:3822-3828; Mendelson et al., 1988 Virol. 166:154-165; and Flotte et al., 1993 PNAS 90:10613-10617.
  • Other particularly desirable AAV vectors include those based upon AAV1; see, International Patent Publication No. WO 00/28061, published May 18, 2000.
  • AAV vectors include those which are pseudotyped, i.e., contain a minigene composed of AAV 5′ ITRs, a transgene, and AAV 3′ ITRs packaged in a capsid of an AAV serotype heterologous to the AAV ITRs. Methods of producing such pseudotyped AAV vectors are described in detail in International Patent Publication No. WOO1/83692.
  • the nucleic acid molecule of the invention is “naked DNA”, it is combined with polymers including traditional polymers and non-traditional polymers such as cyclodextrin-containing polymers and protective, interactive noncondensing polymers, among others.
  • the “naked” DNA and DNA condensed with cationic lipids or polymers are typically delivered to the cells using chemical methods.
  • a number of chemical methods are known in the art for cell delivery and include using lipids, polymers, or proteins to complex with DNA, optionally condensing the same into particles, and delivering to the cells.
  • Another non-viral chemical method includes using cations to condense DNA, which is then placed in a liposome and used according to the present invention. See, C. Henry, 2001 Chemical and Engineering News, 79(48):35-41.
  • the nucleic acid molecule encoding the selected therapeutic protein is introduced directly into the cells of the hematopoietic lineage either as “naked” DNA (U.S. Pat. No. 5,580,859) or formulated in compositions with agents that facilitate direct immunization, such as bupivicaine and other local anesthetics (U.S. Pat. No. 6,127,170).
  • Each nucleic acid sequence encoding a protein according to this invention is preferably under the control of regulatory sequences that direct the replication and generation of the product of each nucleic acid sequence, preferably ectopically, in a mammalian hematopoietic lineage, progenitor cell or differentiated cell.
  • Additional regulatory sequences for inclusion in a nucleic acid sequence, molecule or vector of this invention include, without limitation, one or more of an enhancer sequence, a polyadenylation sequence, a splice donor sequence and a splice acceptor sequence, a site for transcription initiation and termination positioned at the beginning and end, respectively, of the polypeptide to be translated, a ribosome binding site for translation in the transcribed region, an epitope tag, a nuclear localization sequence, an IRES element, a Goldberg-Hogness “TATA” element, a restriction enzyme cleavage site, a selectable marker and the like.
  • Enhancer sequences include, e.g., the 72 bp tandem repeat of SV40 DNA or the retroviral long terminal repeats or LTRs, etc. and are employed to increase transcriptional efficiency. Selection of other non-promoter common vector elements is conventional and many such sequences are available with which to design the nucleotide molecules and vectors useful in this invention. See, e.g., Sambrook et al, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, New York, (1989) and references cited therein at, for example, pages 3.18-3.26 and 16.17-16.27 and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York (1989). One of skill in the art may readily select from among such known non-promoter sequences to prepare nucleic acid molecules of this invention. The selection of such non-promoter sequences is not a limitation of this invention.
  • nucleotide sequences and nucleotide molecules of this invention are well within the ability of the person having ordinary skill in the art using available material.
  • the synthesis methods are not a limitation of this invention.
  • the examples below detail presently preferred embodiments of synthesis of sequences encoding an exemplary transgene useful in this invention for the construction of a transgenic animal model.
  • similar methods are employed to produce nucleic acid molecules for the generation of therapeutic or prophylactic compositions of this invention.
  • nucleotide molecules and sequences of this invention are produced by chemical synthesis methods.
  • the nucleotide sequences useful in the invention are prepared conventionally by resort to known chemical synthesis techniques, e.g., solid-phase chemical synthesis, such as described by Merrifield, 1963 J. Amer. Chem. Soc., 85:2149-2154; J. Stuart and J. Young, Solid Phase Peptide Synthesis, Pierce Chemical Company, Rockford, Ill. (1984); Matteucci et al., 1981 J. Am. Chem. Soc., 103:3185; Alvarado-Urbina et al., 1980 Science, 214:270; and Sinha, N. D. et al., 1984 Nucl.
  • the nucleic acid molecules of this invention are constructed recombinantly using conventional molecular biology techniques, site-directed mutagenesis, genetic engineering or polymerase chain reaction, such as, by cloning and expressing a nucleotide molecule encoding a desired therapeutic protein with optional other proteins within a host cell of the hematopoietic lineage, etc. utilizing the information provided herein (See, e.g., Sambrook et al., cited above; Ausubel et al. cited above). Coding sequences for the transgenes and the regulatory sequences can be prepared synthetically (W. P. C. Stemmer et al, 1995 Gene, 164:49).
  • recombinant DNA techniques involve obtaining by synthesis or isolation a DNA sequence that encodes the desired therapeutic protein as described above, and introducing it into an appropriate vector where it is expressed preferably under the control of the selected promoter that can direct expression in a cell of the hematopoietic lineage.
  • any of the methods described for the insertion of DNA into a vector is used to ligate the selected promoter and other regulatory control elements into specific sites within the selected recombinant vector to generate the nucleic acid molecule.
  • a cell of the hematopoietic lineage is manipulated to contain a nucleic acid molecule described above.
  • the nucleic acid molecule generated as described above is transferred into an isolated hematopoietic stem cell, a common lymphoid progenitor cell, a common myeloid progenitor cell, a megakaryotic/erythrocyte progenitor cell or a granulocyte/macrophage progenitor cell.
  • the hematopoietic stem cells or progenitor cells of the hematopoietic lineage are isolated from bone marrow of a suitable human or non-human mammal.
  • the cells are isolated from a mammalian patient into whom the manipulated cells are re-introduced.
  • the mammal providing the host cells is a different mammal, for either introduction into another mammal or for research or laboratory use.
  • Methods for isolating such cells from bone marrow are well known. See, for example, the Stem Cell Database of Princeton University; Phillips, R L et al, 2000 Science, 288:1635-1640 and references cited therein.
  • the host cells of the hematopoietic lineage are those cells found in the peripheral blood or tissue, such as platelets, megakaryocytes, neutrophils, eosinophils, monocytes, basophils, dendritic cells, mast cells, macrophages, dendritic cells, erythrocytes, and natural killer cells.
  • These cells must be isolated from the peripheral blood or tissue of a suitable human or non-human mammal.
  • the cells are isolated from a mammalian patient into whom the manipulated cells would be re-introduced.
  • the mammal providing the host cells is a different mammal, for either introduction into another mammal or for research or laboratory use. Methods for isolating such cells from peripheral blood or tissue are well known.
  • the introduction of a platelet expressing a suitable transgene under the platelet specific PF4 regulatory sequence is exemplified in the examples below.
  • the nucleic acid molecule or vector is transferred therein in vitro or ex vivo by a conventional technique such as transformation, transduction, infection or transfection.
  • a conventional technique such as transformation, transduction, infection or transfection.
  • the selection of other suitable methods for transferring the nucleic acid molecule or the vector into an isolated cell of the hematopoietic lineage can be performed by one of skill in the art by reference to known techniques. See, e.g., Gething and Sambrook, 1981 Nature, 293:620-625, among others.
  • Such host cells are cultured.
  • Culture conditions are well known in the art and include ionic composition and concentration, temperature, pH and the like.
  • transfected cells are maintained under culture conditions in a culture medium. Suitable media for various cell types are well known in the art.
  • the temperature is from about 20° C. to about 50° C., more preferably from about 30° C. to about 40° C. and, even more preferably about 37° C.
  • the pH is preferably from about a value of 6.0 to a value of about 8.0, more preferably from about a value of about 6.8 to a value of about 7.8 and, most preferably about 7.4.
  • Osmolality is preferably from about 200 milliosmols per liter (mosm/L) to about 400 mosm/l and, more preferably from about 290 mosm/L to about 310 mosm/L.
  • Other biological conditions needed for transfection of a vector are well known in the art.
  • the host cells of the hematopoietic lineage are employed in pharmaceutical or prophylactic compositions and methods for the treatment of a variety of disorders in human or non-human mammals. Such treatment may include enhancement of a biological activity or reduction or a disadvantageous biological activity occurring in the body.
  • the nucleic acid molecules themselves are used in direct treatment of disorders such as inflammatory disorders, microbial or parasitic infection, vascular or hemorrhagic disorders, and the like in which the hematopoietic cells, their progenitors or differentiated cells are implicated.
  • the host cells preferably platelets
  • disorders include coagulation disorders (either an insufficiency or excess thereof), acute lung injury and sepsis, helminth infection, asthma or other allergic reactions, viral infections, bacterial infections, etc.
  • compositions of this invention are useful in the prevention and/or treatment of disease(s) caused by microbial infections including, without limitation, Haemophilus influenzae (both typable and nontypable), Haemophilus somnus, Moraxella catarrhalis, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Streptococcus faecalis, Helicobacter pylori, Neisseria meningitidis, Neisseria gonorrhoeae, Chlamydia trachomatis, Chlamydia pneumoniae, Chlamydia psittaci, Bordetella pertussis, Alloiococcus otiditis, Salmonella typhi, Salmonella typhimurium, Salmonella choleraesuis, Escherichia coli, Shigella, Vibrio cholerae, Cor
  • compositions of this invention are useful in the prevention and/or treatment of disease caused by, without limitation, Respiratory syncytial virus, Parainfluenza virus types 1-3, Human metapneumovirus, Influenza virus, Herpes simplex virus, Human cytomegalovirus, Human immunodeficiency virus, Simian immunodeficiency virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C.
  • virus Human papillomavirus, Poliovirus, rotavirus, caliciviruses, Measles virus, Mumps virus, Rubella virus, adenovirus, rabies virus, canine distemper virus, rinderpest virus, avian pneumovirus (formerly turkey rhinotracheitis virus), Hendra virus, Nipah virus, coronavirus, parvovirus, infectious rhinotracheitis viruses, feline leukemia virus, feline infectious peritonitis virus, avian infectious bursal disease virus, Newcastle disease virus, Marek's disease virus, porcine respiratory and reproductive syndrome virus, equine arteritis virus and various Encephalitis viruses.
  • avian pneumovirus now turkey rhinotracheitis virus
  • Hendra virus Nipah virus
  • coronavirus coronavirus
  • parvovirus infectious rhinotracheitis viruses
  • feline leukemia virus feline infectious peritonitis virus
  • avian infectious bursal disease virus Newcastle disease virus
  • compositions of this invention are useful in enhancing response against fungal pathogens such as Aspergillis, Blastomyces, Candida, Coccidiodes, Cryptococcus and Histoplasma or against parasites including Leishmania major, Ascaris, Trichuris, Giardia, Schistosoma, Cryptosporidium, Trichomonas, Toxoplasma gondii and Pneumocystis carinii.
  • fungal pathogens such as Aspergillis, Blastomyces, Candida, Coccidiodes, Cryptococcus and Histoplasma
  • parasites including Leishmania major, Ascaris, Trichuris, Giardia, Schistosoma, Cryptosporidium, Trichomonas, Toxoplasma gondii and Pneumocystis carinii.
  • compositions of this invention may also be useful for the prevention and/or treatment of disease(s), without limitation, such as autoimmune disease, such as multiple sclerosis, lupus and rheumatoid arthritis and others, asthma, atherosclerosis, Alzheimer disease, amyloidosis or amyloidogenic disease, and cancers.
  • autoimmune disease such as multiple sclerosis, lupus and rheumatoid arthritis and others
  • asthma atherosclerosis
  • Alzheimer disease amyloidosis or amyloidogenic disease
  • cancers Clotting disorders and other vascular injuries caused by other infections, injury, aging, thrombocytopenia, inappropriate thrombus formation, myelodysplasia, AML, and the like may also be treated according to the methods of this invention.
  • These compositions and methods can be useful to treat allergic reactions to allergens such as pollen, insect venoms, animal dander, fungal spores and drugs (such as penicillin).
  • compositions and methods of this invention are not a limitation of this invention.
  • One of skill in the art may readily include other disorders suitable for the treatment described herein.
  • One method for treating a disorder in a mammal includes the step of delivering to a mammal a recombinant nucleic acid molecule comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a hematopoietic stem cell, a progenitor cell of the hematopoietic lineage, or a cell differentiated therefrom.
  • the method permits a differentiated cell of a hematopoietic lineage to express the product of the selected transgene at a suitable site in the mammal.
  • the cell may express the transgene produce ectopically and target the expression to a particular site.
  • a plasmid expressing a transgene will target to the site of vascular injury or thrombus formation.
  • the method of the invention involves harvesting stem cells or progenitor cells from bone marrow of a patent, transferring the nucleic acid molecule into the cells ex vivo and reinfusing the cells into the bone marrow or peripheral blood of the mammalian patient.
  • the method of treatment can involve infusing or injecting into the patients bone marrow or blood a non-self transfected host cell.
  • the method of the invention involves harvesting differentiated cells of the hematopoietic lineage from peripheral blood of a mammalian patient; transferring the nucleic acid molecule into the differentiated cells; and
  • the method of treatment can involve infusing or injecting into the patient's blood a non-self transfected differentiated host cell.
  • a method of treatment can involve directly administering to a mammalian patient, simply the nucleic acid molecule, i.e., as naked DNA.
  • the regulatory sequences should aid in the uptake of the molecule by the appropriate differentiated cells of the hematopoietic lineage.
  • the invention provides a method for enhancing coagulation in a patient by delivering to the mammalian patient with an insufficient clotting mechanism a recombinant nucleic acid molecule (or a platelet containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet.
  • the regulatory sequence would be a platelet-specific sequence mentioned above.
  • suitable transgenes for this method are transgenes encoding one or more of Factor VIIa, Factor VIII, Factor IX or fibrinogen.
  • Another specific embodiment involves a method for preventing or reducing coagulation in a mammalian patient, where needed.
  • the patient is administered a recombinant nucleic acid molecule (or a platelet containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet.
  • the regulatory sequence would be a platelet-specific sequence mentioned above.
  • suitable transgenes for this method are transgenes encoding one or more of urokinase plasminogen activator, plasminogen, tissue plasminogen activator, and tissue factor pathway inhibitor.
  • Another example of a method of this invention is a method for enhancing coagulation in a mammalian patient by delivering to the patient a recombinant nucleic acid molecule (or an erythrocyte containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in an erythrocyte.
  • the regulatory sequence would be an erythrocyte-specific sequence mentioned above.
  • suitable transgenes for this method are transgenes encoding a urokinase plasminogen activator receptor, preferably expressed on the cell surface.
  • Still another example of this invention is a method for treating acute lung injury and sepsis in a mammalian patient.
  • This method includes delivering to the patient a recombinant nucleic acid molecule (or neutrophils containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a neutrophil.
  • the regulatory sequence would be a neutrophil-specific sequence mentioned above.
  • An example of a suitable transgene for this method is a transgene encoding activated Protein C.
  • compositions of this invention may also be employed in a method for treating parasitic helminth infection of a mammalian human or non-human patient.
  • This method involves delivering to a mammal a recombinant nucleic acid molecule (or eosinophils containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in eosinophils.
  • the regulatory sequence would be an eosinophil-specific sequence mentioned above.
  • An example of a suitable transgene for this method is a transgene encoding a protein toxic to a helminth.
  • Another method according to this invention involves treating asthma or allergic responses in a mammalian patient.
  • This method involves delivering to a mammal a recombinant nucleic acid molecule (or an eosinophils containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in an eosinophil.
  • the regulatory sequence would be an eosinophil-specific sequence mentioned above.
  • Examples of a suitable transgene for this method are transgenes encoding one or more of human TSG6, an antibody to IL-1 receptor alpha, and an anti-inflammatory protein.
  • the invention also includes a method for treating a viral infection in a mammal comprising delivering to a mammal a recombinant nucleic acid molecule (or NK cell containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a natural killer cell.
  • the regulatory sequence would be an NK cell-specific sequence mentioned-above.
  • Examples of a suitable transgene for this method is a transgene encoding a neutralizing antibody against a viral coat protein, e.g., anti-HIV gag protein, anti-HPV proteins, anti-HIV proteins, etc.
  • the present invention provides a method for the treatment and prevention of undesirable thrombus development in a mammalian patient by administering to the patient a recombinant nucleic acid molecule (or platelet containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet.
  • a platelet that ectopically expressed u-PA under the control of the platelet specific PF4 promoter was able to control thrombus formation in a patient. See the Examples below.
  • the nucleic acid molecules or host cells are employed in compositions containing a physiologically acceptable diluent or a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline.
  • a physiologically acceptable diluent or a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline.
  • the compositions may also be mixed with such diluents or carriers in a conventional manner.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with administration to humans or other vertebrate hosts.
  • the appropriate carrier will be evident to those skilled in the art and will depend in large part upon the route of administration.
  • compositions may also include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
  • compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • Still additional components that are present are adjuvants, preservatives, chemical stabilizers, or other proteins.
  • stabilizers, adjuvants, and preservatives are optimized to determine the best formulation for efficacy in the target human or animal.
  • Suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and parachlorophenol.
  • Suitable stabilizing ingredients that are used include, for example, casanino acids, sucrose, gelatin, phenol red, N-Z amine, monopotassium diphosphate, lactose, lactalbumin hydrolysate, and dried milk.
  • cytokine granulocyte-macrophage colony stimulating factor (GM-CSF), which has a nucleotide sequence as described in U.S. Pat. No. 5,078,996, which is hereby incorporated by reference.
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • a plasmid containing GM-CSF cDNA has been transformed into E coli and has been deposited with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, under Accession Number 39900.
  • ATCC American Type Culture Collection
  • IL-12 cytokine Interleukin-12
  • cytokines or lymphokines include but are not limited to, the interleukins 1- ⁇ , 1- ⁇ , through 24, the interferons- ⁇ , ⁇ and ⁇ , granulocyte colony stimulating factor, and the tumor necrosis factors ⁇ and ⁇ .
  • compositions of this invention include, but are not limited to: surface active substances (e.g., hexadecylamine, octadecylamine, octadecyl amino acid esters, lysolecithin, dimethyl-dioctadecylammonium bromide), methoxyhexadecylgylcerol, and pluronic polyols; polyamines, e.g., pyran, dextransulfate, poly IC, carbopol; peptides, e.g., muramyl dipeptide, dimethylglycine, tuftsin; oil emulsions; and mineral gels, e.g., aluminum phosphate, etc. and immune stimulating complexes, liposomes, polysaccharides, lipopolysaccharides and/or other polymers.
  • surface active substances e.g., hexadecylamine, octadec
  • compositions composed of polynucleotide molecules desirably contain optional polynucleotide facilitating agents or “co-agents”, such as a local anesthetic, a peptide, a lipid including cationic lipids, a liposome or lipidic particle, a polycation such as polylysine, a branched, three-dimensional polycation such as a dendrimer, a carbohydrate, a cationic amphiphile, a detergent, a benzylammonium surfactant, or another compound that facilitates polynucleotide transfer to cells.
  • a facilitating agent includes bupivicaine (see U.S.
  • the local anesthetic is present in an amount that forms one or more complexes with the nucleic acid molecules.
  • the local anesthetic is mixed with nucleic acid molecules or plasmids of this invention, it forms a variety of small complexes or particles that pack the DNA and are homogeneous.
  • the complexes are formed by mixing the local anesthetic and at least one plasmid of this invention. Any single complex resulting from this mixture may contain a variety of combinations of the different plasmids.
  • the local anesthetic is pre-mixed with each plasmid separately, and then the separate mixtures combined in a single composition to ensure the desired ratio of the plasmids is present in a single composition, if all plasmids are to be administered in a single bolus administration.
  • the local anesthetic and each plasmid are mixed separately and administered separately to obtain the desired ratio.
  • the facilitating agent used is a local anesthetic, preferably bupivacaine, an amount of from about 0.1 weight percent to about 1.0 weight percent based on the total weight of the polynucleotide composition is preferred. See, also, International Patent Publication No.
  • the amount of local anesthetic is present in a ratio to the nucleic acid molecules of 0.01-2.5% w/v local anesthetic to 1-10 ⁇ g/ml nucleic acid. Another such range is 0.05-1.25% w/v local anesthetic to 100 ⁇ g/ml to 1 ml/ml nucleic acid.
  • such a polynucleotide composition may express the transgene product on a transient basis in vivo; no genetic material is inserted or integrated into the chromosomes of the host.
  • compositions may also contain other additives suitable for the selected mode of administration of the composition.
  • the composition of the invention may also involve lyophilized polynucleotides, which can be used with other pharmaceutically acceptable excipients for developing powder, liquid or suspension dosage forms. See, e.g., Remington: The Science and Practice of Pharmacy, Vol. 2, 19 th edition (1995), e.g., Chapter 95 Aerosols; and International Patent Publication No. W099/45966, the teachings of which are hereby incorporated by reference. Routes of administration for these compositions are combined, if desired, or adjusted.
  • nucleic acid molecule-containing compositions can contain additives suitable for administration via any conventional route of administration.
  • administration is intravenous or directly into the bone marrow.
  • the composition of the invention is prepared for administration to human subjects in the form of, for example, liquids, powders, aerosols, tablets, etc.
  • compositions of the present invention are not limited by the selection of the conventional, physiologically acceptable, carriers, adjuvants, or other ingredients useful in pharmaceutical preparations of the types described above.
  • the preparation of these pharmaceutically acceptable compositions, from the above-described components, having appropriate pH isotonicity, stability and other conventional characteristics is within the skill of the art.
  • Suitable routes of administration include, but are not limited to, intranasal, oral, vaginal, rectal, parenteral, intradermal, transdermal, intramuscular, intraperitoneal, subcutaneous, intravenous and intraarterial.
  • the appropriate route is selected depending on the nature of the composition used, and an evaluation of the age, weight, sex and general health of the patient and the components present in the immunogenic composition, and similar factors by an attending physician.
  • compositions of this invention are readily determined by one of skill in the art, depending upon the condition being treated, the health, age and weight of the veterinary or human patient, and other related factors, and the other characteristics of the composition, e.g., nucleotide molecule, vector or host cell.
  • selection of the appropriate “effective amount” or dosage for the composition(s) of the present invention will also be based upon the form of the composition, the identity of the transgene and host cell, as well as the physical condition of the subject.
  • the method and routes of administration and the presence of additional components in the compositions may also affect the dosages and amounts of the compositions. Such selection and upward or downward adjustment of the effective dose is within the skill of the art.
  • the amount of composition required to produce a suitable response in the patient without significant adverse side effects varies depending upon these factors. Suitable doses are readily determined by persons skilled in the art.
  • each dose will comprise between about 50 ⁇ g to about 1 mg of nucleic acid molecule, e.g., DNA plasmid, per mL of a sterile solution.
  • a suitable dose where the vector is a viral vector is in the range of 10 3 to 10 18 , preferably about 10 5 to 10 16 transducing units (TU) per dose, and most preferably, about 10 7 to 10 9 TU for an adult human having a weight of about 80 kg.
  • Transducing Units (TU) represents the number of infectious particles and is determined by evaluation of transgene expression upon infection of host cells.
  • the host cells when used for ex vivo therapy, are infected with 10 5 TU to 10 10 TU viral vectors for each 10 1 to 10 10 cells in a population.
  • 10 5 TU to 10 10 TU viral vectors for each 10 1 to 10 10 cells in a population.
  • other suitable ex vivo dosing levels are readily selected by one of skill in the art.
  • This dose is formulated in a pharmaceutical composition, as described above (e.g., suspended in about 0.01 mL to about 1 mL of a physiologically compatible carrier) and delivered by any suitable means.
  • the number of doses and the dosage regimen for the composition are also readily determined by persons skilled in the art.
  • the intended therapeutic or prophylactic effect is conferred by a single dose of the composition or may require the administration of several doses, in addition to booster doses at later times.
  • the dose is repeated, as needed or desired, daily, weekly, monthly, or at other selected intervals.
  • the methods of this invention were employed to modify the biological behavior of platelets by causing these hematopoietic lineage cells to ectopically express a protein of interest at a site of vascular injury.
  • a murine urokinase-type plasminogen activator (u-PA) transgene was expressed transgenically in mice under the regulatory control of a platelet factor 4 promoter that directed expression to platelets.
  • the resultant transgenic animals have altered platelet biology; i.e., they express and store u-PA selectively in their platelet alpha-granules, in the absence of systemic fibrinolysis and bleeding characteristic of transgenic over-expression of u-PA in the liver (Heckel, J. L. et al, 1990 Cell 62:447-456).
  • These transgenic mice also had a characteristic bleeding diathesis similar to that seen in patients with Quebec Platelet Disorder (QPD) that is predominantly at the time of parturition and that can be controlled by tranexamic acid.
  • QPD Quebec Platelet Disorder
  • mice were resistant to the development of occlusive carotid arterial thrombi in the absence of systemic fibrinolysis.
  • the mice rapidly lysed pulmonary venous thrombi or emboli.
  • transfusion of small numbers of urokinase-expressing platelets into wildtype recipients prevented formation of stable, occlusive carotid arterial thrombi.
  • this animal model confirms that ectopic expression of u-PA in platelets is the etiology of inherited QPD, provides new insights into the contribution of activated platelets to thrombus stability, and provides a new method for preventing inopportune thrombus development.
  • the transgenic model shows that developing megakaryocytes are genetically altered in such a way that platelet function is effectively tipped from pro-thrombotic to anti-thrombotic.
  • the present invention involves the discovery that ectopic expression of proteins in platelets are useful to favorably alter the hemostatic balance at sites of thrombosis.
  • Fibrinolytic agents can be delivered to sites of incipient thrombus formation through selective storage in platelets, a new method to prevent thrombosis and hemorrhage.
  • ectopically expressed proteins carrying a signal peptide are stored within platelets and released specifically at sites of injury. By use of this method, one may alter the prothrombotic role of platelets as a means to modify pathological thrombus development.
  • the method of this invention has clinical application in situations where re-thrombosis or the prevention of extension of a previous thrombosis is desirable for an extended period of time.
  • transient expression of u-PA in megakaryocyte progenitors harvested from peripheral blood and treated with present-day retroviral gene-transfer vectors are a potent mechanism to prevent post-angioplasty restenosis for several weeks to months with low risk of long-term alteration of the recipient's stem cell genome.
  • Untoward bleeding associated with the therapy can be controlled by treatment with an anti-fibrinolytic agent such as tranexamic acid.
  • the methods of this invention enable delivery of pro-hemostatic proteins to sites of vascular interruption in patients with diverse hemorrhagic disorders.
  • the u-PA was partially counter-balanced by PAI-1, which is present in platelet alpha-granules of both human and mice platelets (Ngo, T. H., and Declerck, P. J. 1999 Thromb. Haemost. 82:1510-1515). This method tipped the balance sufficiently to lessen the resistance of clots, especially in the arterial circulation where platelet activation is more intense, to commonly employed agents.
  • the “line #19” transgenic animals had u-PA mRNA, u-PA-like protease activity and u-PA protein detectable in their platelets.
  • these animals did not demonstrate systemic fibrinolysis. Few of the transgenic adults developed spontaneous hemorrhage despite life-long expression of elevated levels of platelet-uPA. The platelet counts were normal, and the red cells had normal morphology. Plasma fibrinogen was intact and D-dimers were not detected in the plasma of these mice.
  • u-PA Megakaryocyte-expressed u-PA was not secreted to a considerable extent, but rather was preferentially stored in the circulating platelets, where it led to the digestion of alpha-granular proteins, as in the human disorder termed QPD.
  • the u-PA within the alpha-granules was predominantly in the form of enzymatically active tcu-PA, as determined by immunoblotting. Platelets are believed to endocytose sufficient plasminogen from plasma (Holt, J. C., and Niewiarowski, S. cited above) into their alpha-granules to form plasmin, as suggested by the capacity of u-PA platelets to digest exogenously added Factor V.
  • Plasmin formation is initiated by the low level of intrinsic activity of scu-PA (Lenich, C. et al, 1997 Blood 90:3579-3586) or by the activation of scu-PA by u-PAR (Higazi, A. et al, 1995 J. Biol. Chem. 270:17375-17380) or, less likely, by another platelet granular enzyme. It is likely that plasmin, once formed, then converts additional scu-PA to tcu-PA within the granules themselves, although the role of other proteases again cannot be excluded. Little tcu-PA was found as high molecular weight complexes with PAI-1.
  • Murine platelets may contain less PAI-1 than human platelets, or the latent state of PAI-1 within the granules (Booth, N. A. et al, 1988 Brit. J. Haematol. 70:327-333; Lang, I. M. et al, 1992 Blood 80:2269-2274) may limit its binding to u-PA, or complex formation is inhibited by u-PAR (Higazi, A. A.-R et al, 1996 Blood 87:3545-3549), the pH or other components of the alpha-granules, or, more likely, most of the PAI-1 was degraded by the u-PA.
  • the ectopic expression of u-PA is essentially confined to the platelets, and insufficient u-PA is secreted into the plasma to cause disseminated fibrinolysis.
  • the phenotype of neither is improved by platelet infusions, but is improved by use of an anti-fibrinolytic agent, such as tranexamic acid.
  • a transgene was designed as illustrated schematically in FIG. 1.
  • 1.2 kb of the 129 SV murine platelet factor 4 (PF4) proximal promoter region plus its 5′-untranslated region (UTR) (Zhang, C. et al, 2001 Blood 98:610-607) was PCR amplified with an artificial upstream Xba I site and a downstream Kpn I site added. The inventors found that this promoter could also drive megakaryocyte-specific expression of LacZ in transgenic mice (data not shown).
  • This promoter was inserted in place of the albumin enhancer/promoter immediately upstream of a mouse u-PA minigene construct that contained a 3′-UTR and poly-adenylation sequence from the human growth hormone (hGH) gene.
  • hGH human growth hormone
  • a 10.2 kb Sac II fragment containing this construct was used to create transgenic mice by pronuclear injections following standard methods at the University of Pennsylvania Transgenic Mice Core Facility. Seven transgenic founders were obtained with copy numbers ranging from 1 to >20 copy per haploid genome.
  • Genomic DNA was isolated from mouse tails using a QIAampTM DNA Mini Kit (Qiagen, Valencia, Calif.). Positive founder lines were detected by genomic Southern blot analysis (Tunnacliffe, A. et al, 1992 Blood 79: 2896-2900). Genomic Southern blot analysis was made of Bgl II-digested DNA from a wildtype animal and the three lines that had offspring with the fewest copy numbers of the transgene (Lines #13, #19 and #49). After digestion, genomic DNA was separated on a 1% (wt/vol) agarose gel. The probe used was the mouse PF4 proximal promoter from ⁇ 680 to ⁇ 360 bp upstream of the transcriptional start site (Zhang, C.
  • the transgene copy number per haploid genome was determined by phosphorimaging (not shown). The intensity of bands on film was analyzed using the Imagequant PhosphorImager software (Amersham Biosciences). The copy number was determined by comparing the intensity of the 2.8 kb transgene PF4 band to the native genomic 1.2 kb PF4 band.
  • Founder animals and their offspring were also characterized by genomic PCR analysis using a mouse PF4 5′-UTR (5′-CACTTAAGAGCCCTAGACCCATTTCC-3′; SEQ ID NO: 1) sense primer and a mouse u-PA exon 2 (5′-TTCAGAGTTTTT CACCACCAA-3′; SEQ ID NO: 2) antisense primer, which generates a 479 bp genomic and a 114 bp cDNA band.
  • RT-PCR analysis of total platelet RNA was performed on lines #13, #19 and #49 for the 114 bp transgenic u-PA message that extended from the 5′-UTR of PF4 into the murine u-PA 2 nd exon in their platelets.
  • the 185 bp PF4 message was used as a positive control of the platelet nature of the RNA. PCR was performed to confirm founder lines using primers complementary to the mouse PF4 5′-UTR and exon 2 of the murine u-PA gene (data not shown).
  • Example 1 The animals of Example 1 were then examined to determine whether they expressed u-PA in their platelets.
  • Murine platelet RNA was isolated using RNA STAT-60TM (Tel-Test, Friendswood, Tex.) as previously described (Zhang, C. et al, cited above). Tissues ( ⁇ 100 mg each) from these animals were collected, rinsed vigorously with saline, disaggregated in 500 ⁇ l of RNA STAT-60TM, and RNA was isolated (Zhang, C. et al, cited above).
  • Some platelet RNA samples were pretreated with DNase free RNase (1 U/10 ⁇ l reaction, Sigma, St. Louis, Mo.) or RNase free DNase (1 U/10 ⁇ l reaction, Life Technologies, Gaithersburg, Md.) for 1 hour at 37° C.
  • Reverse transcription was performed using the SuperScript II Reverse Transcriptase KitTM (Life Technologies) as per the manufacturer's instructions.
  • PCR amplification of the transgenic u-PA cDNA was accomplished using the two primers discussed above.
  • Platelet-specific control RT-PCR primers for PF4 message were sense 5′-AATTCTCGGGATCTGGGT-3′ SEQ ID NO: 3 and antisense 5′-CTGGGCTCTAGACAGCAGT-3 SEQ ID NO: 4 (Eisman, R. et al, 1990 Blood 76:336-344), with an expected cDNA product of 185 bp.
  • RT-PCR for the housekeeping gene human phosphoribosyltransferase used primers 5′-GCTGGTGAAAAGGACCT CT-3′ SEQ ID NO: 5 and 5′-CACAGGACTAGAACACCTGC-3′ SEQ ID NO: 6, with an expected cDNA product of 249 bp (Jackson, C. L. et al, 1984 Proc. Nat. Acad. Sci. U.S.A. 81:2945-2949).
  • Platelet murine transgenic u-PA cDNA band was isolated using a QIAkwikTM Gel Extraction Kit (Qiagen) and directly sequenced using an ABI 373A automated sequencer (ABI Instruments, Foster City, Calif.).
  • Platelet-rich plasma was obtained as described in Zhang, cited above. Platelet counts were determined using a HemaVet counter (Triad Associates, Concord, Calif.). The platelets were pelleted at 800 g for 5 minutes and resuspended immediately in 1 ⁇ NuPage Sample Buffer (Invitrogen, Carlsbad, Calif.).
  • Total platelet protein and platelet releasates (0.5 to 10 ⁇ g/lane) were separated by size under non-reducing conditions on a 12% SDS-polyacrylamide gel (SDS-PAGE) with 0.4% nonfat dry milk, e.g., casein (Carnation Instant Skim Milk Powder, Nestle, Fulton, N.Y.) with or without supplemental 20 ⁇ g/ml human plasminogen (American Diagnostica, Greenwich, Conn.), and then renatured with 0.5% Triton X-100 in PBS, pH 7.4, for 1 hour as previously described (Heckel, J. L. et al, cited above). The gel was incubated at 37° C. for 3 hours. A control lane of 0.1 ng of human 2-chain urokinase (American Diagnostica) was included in each gel.
  • SDS-PAGE SDS-polyacrylamide gel
  • Mouse u-PA was detected with a murine anti-mouse u-PA monoclonal antibody (A10, Molecular Innovations, Southfield, Mich.) added at a 1:75 dilution and detected with a biotinylated anti-mouse monoclonal antibody (Molecular Innovations) followed by peroxidase-conjugated streptavidin (StreptABComplex/HRP, DAKO, Carpinteria, Calif.).
  • Murine vWF was detected using a 1:200 dilution of a horse radish peroxidase (HRP)-conjugated rabbit anti-human vWF polyclonal antibody (DAKO), and murine fibrinogen was detected using a 1:100 dilution of an HRP-conjugated goat anti-human fibrinogen polyclonal antibody (Rockland Immunochemicals, Gilbertsville, Pa.).
  • HRP horse radish peroxidase
  • DAKO horse radish peroxidase-conjugated rabbit anti-human vWF polyclonal antibody
  • HRP-conjugated goat anti-human fibrinogen polyclonal antibody Rockland Immunochemicals, Gilbertsville, Pa.
  • Platelet lysate from line #19 digested the Factor V in a rapid fashion, with degradation nearly complete by 4 hours, giving a similar pattern to that seen with exogenously added human u-PA (data not shown). Platelets from wildtype littermates also degraded Factor V, but at a much slower rate.
  • the addition of exogenous plasminogen to the platelet releasate enhanced Factor V digestion by line #19 platelet lysate, but had little effect on the rate of digestion by wildtype platelets. This suggests that the amount of plasminogen available in alpha-granules in the transgenic line #19 platelets is rate limiting for maximal protein degradation.
  • line #19 and #49 transgenic mice were born than expected. Assuming that 50% of the offspring from a cross between a hemizygous animal and a wildtype animal should be transgenic, line #19 showed a 68% mortality rate (57 transgenic animals vs. 175 wildtype littermates at weaning) and line #49 showed an 85% mortality rate (2 transgenic and 12 wildtype). Line #13 had a very low mortality rate, consistent with little platelet u-PA expression (71 transgenic and 80 wildtype).
  • line #19 embryos looked normal and appeared at the expected frequency (10 transgenic and 11 wildtype), indicating that transgenic animals were lost peripartum Surviving transgenic animals were normal for weight and growth, although occasional spontaneous deaths occurred among the adults secondary to subcutaneous or internal hemorrhage. Autopsies on two such adult line #19 mice expressing u-PA in their platelets showed examples of spontaneous hemorrhage: one mouse had free blood in the opened peritoneum and the other showed free blood filling a small intestinal loop.
  • Results of the counts showed nearly identical platelet counts and hemoglobin levels for the line #19 and wildtype controls (data not shown).
  • Peripheral blood smears were normal, including normal platelet numbers and appearance and no evidence of red cell schistocytes or spherocytes (data not shown).
  • Ferric chloride-induced arterial injury was performed according to published procedures (Fay, W. P et al, 1994 cited above; Fay, W. P. et al, 1999 Blood 93:1825-1830) in 6-8 week old animals. Briefly, the right common carotid artery was exposed by blunt dissection, and a miniature Doppler flow probe (Model 0.5VB, Transonic Systems, Ithaca, N.Y.) was positioned around the artery. A 1 ⁇ 2 mm 2 strip of Number 1 Whatman filter paper (Fisher Scientific, Pittsburgh, Pa.) soaked in 10% ferric chloride was then applied to the adventitial surface of the artery for 2 min. The field was flushed with saline, and blood flow was continuously monitored for 30 minutes. The time to the initial complete occlusion and the presence or absence of arterial occlusion at 30 min was recorded.
  • a miniature Doppler flow probe Model 0.5VB, Transonic Systems, Ithaca, N.Y.
  • 125 I-labeled human microemboli 1.5-3.5 ⁇ 3 in size, were prepared as previously described (Bdeir, K. et al, 2000 Blood 96:1820-1826). Transgenic and wildtype animals were injected with these particles within 48 hours of preparation. At various time points (2-60 minutes) the animals were euthanized, the lungs removed, washed free of blood, and the amount of 125 I activity measured using a ZM Coulter Counter (Coulter Electronics, Hialeah, Fla.). In other experiments, autoradiograms of lungs were taken from wildtype and transgenic mice 30 minutes after injection of the microemboli by exposing the lungs to X-OMAT film (Kodak, Rochester N.Y.).

Abstract

Recombinant nucleic acid molecules are constructed with a first sequence encoding a transgene under the control of regulatory sequences that direct expression of the transgene product in a hematopoietic stem cell, or a progenitor cell therefrom or cell differentiated therefrom. In one embodiment, the cell which expresses the transgene is a secretory cell. The cell is a megakaryotic progenitor cell, or a cell further differentiated therefrom, such as a platelet. The cell is a granulocyte/macrophage progenitor cell or a cell further differentiated therefrom, such as a mast cell or neutrophils. Such host cells containing the molecule or the molecule itself are employed in methods for treating or preventing infection, inflammation or vascular injuries or any disorders involving or mediated by cells of the hematopoietic lineage.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of the priority of U.S. Provisional Patent Application No. 60/424,234, filed Nov. 5, 2002.[0001]
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • [0002] The present invention was supported, in part, by the National Institutes of Health, Grant Nos. HL54749, HL64190, HL60169, HL66442, HL47826 and HL63194. The United States government has an interest in this invention.
  • BACKGROUND OF THE INVENTION
  • Hematopoietic stem cells are pluripotent cells present in the bone marrow, which divide to produce more specialized progenitor stem cells, i.e., lymphoid progenitors and myeloid progenitors. Cells that are differentiated from the lymphoid progenitors in the bone marrow and that are found in the peripheral blood include B cells and T cells. From B cells are generated plasma cells; from T cells are generated activated T cells. Similarly, the common myeloid progenitor stem cells produce the granulocytes/macrophage progenitor cells and the megakaryocyte/erthyrocyte progenitor cells in the bone marrow. Cells differentiated from the granulocyte/macrophage progenitors that are present in the blood include neutrophils, eosinophils, basophils, monocytes and immature dendritic cells. Monocytes further give rise to mast cells, macrophages and dendritic cells that are present in tissue and lymph nodes. Cells differentiated from the megakaryocyte/erthrocyte progenitors include megakaryocytes and erythroblasts, which further differentiate into platelets and erythrocytes (red blood cells) in the blood. [0003]
  • A number of these hematopoietic lineage cells are secretory cells upon activation. For example, platelets, the smallest corpuscular components of human blood, are characterized by a diameter of about 2-4 micrometers, the absence of a nucleus, and a physiological number varying from 150,000 to 300,000 per cubic millimeter of blood. Platelets contribute to the complex, multistep, and highly regulated process of thrombus formation and arterial occlusive disorders, a leading cause of human morbidity. Platelets target and adhere to sites of vascular injury. At the sites of vascular injury, the platelets are activated and form aggregates that provide a provisional seal. Platelets preferentially release their granular contents at the site of injury, e.g., contributing to the subsequent growth and stability of thrombi in part through the release of von Willebrand factor (vWF), fibrinogen, and other coagulation proteins such as Factor V (Holt J. C., and Niewiarowski, S. 1985 [0004] Sem. Hematol. 22:151-163) from their alpha-granules. Activated platelets also release proteins that inhibit thrombolysis, chief among which is plasminogen activator inhibitor-1 (PAI-1). Over 90% of the circulating PAI-1 is stored in platelet alpha-granules (Booth, N. A et al, 1988 Brit. J. Haematol. 70:327-333), although much of this is in an inactive form (Declerck, P. J et al, 1988 Blood 71:220-225; Kruithof, E. K et al, 1987 Blood 70:1645-1653). Nonetheless, this pool of PAI-1 is thought to be one of the main reasons why platelet-rich thrombi are especially resistant to thrombolytic therapy (Booth, N. A et al, 1992 Ann. N. Y. Acad. Sci. 667:70-80; Fay, W. P et al, 1994 Blood 83:351-356).
  • Paradoxically, platelets also contain or can bind small amounts of plasma-derived profibrinolytic proteins, including urokinase-type plasminogen activator (u-PA) and plasminogen (Fay, W. P et al, 1994 cited above; Lenich, C et al, 1997 [0005] Blood 90:3579-3586; Jiang, Y et al. 1996 Blood 87:2775-2781; Holt, J. C., and Niewiarowski, S. 1980 Circulation 62:342a). However, these proteins are found at very low levels, and their activity is overwhelmed by the large amounts of PAI-1, which helps to stabilize nascent thrombi.
  • Recently, the effect of changing this balance in platelet fibrinolytic proteins has been described. Quebec Platelet Disorder (QPD) is a rare bleeding disorder not responsive to platelet transfusion, but responsive to anti-fibrinolytic agents, such as tranexamic acid (Hayward, C. P. et al, 1997 [0006] Blood 89:1243-1253; Hayward, C. P. et al, 1996 Blood 87:4967-4978; Hayward, C. P. et al, 1997 Brit. J. Haematol. 97:497-503). The etiology of QPD has been ascribed recently to ectopic expression of an excess of u-PA in megakaryocytes and platelets (Kahr, W. H. et al., 2001 Blood 98:257-265). QPD platelets contain predominantly activated two-chain urokinase (tcu-PA). The etiology for the bleeding diathesis may in part be due to local release of activated u-PA within thrombi leading to premature lysis. However, degradation of multiple platelet alpha-granular proteins, including vWF and Factor V, presumably by plasmin generated as a result of urokinase, may interfere with thrombus development as well.
  • There remains a need in the art for methods for harnessing the cellular mechanisms of platelets as well as other cells differentiated from hematopoietic progenitor cells for therapeutic, diagnostic and research purposes. [0007]
  • SUMMARY OF THE INVENTION
  • In one aspect, the invention provides a recombinant nucleic acid molecule comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of the transgene product in a hematopoietic progenitor cell or a cell differentiated therefrom In one embodiment, the cell that expresses the transgene is a secretory cell. In another embodiment, the cell is a lymphoid progenitor cell, a myeloid progenitor cell or a cell further differentiated therefrom, such as a platelet. [0008]
  • In another aspect, the invention provides a hematopoietic progenitor, lymphoid progenitor, or nyeloid progenitor host cell transformed, transduced, infected or transfected with an above-described nucleic acid molecule. [0009]
  • In still another aspect, the invention provides a host cell differentiated from a progenitor cell that is transformed, transduced, infected or transfected with an above-described nucleic acid molecule. [0010]
  • In yet a further aspect, the invention provides a platelet transformed, transduced, infected or transfected with a nucleic acid molecule comprising a first sequence encoding a transgene, which is optionally a fibrinolytic protein, under the control of regulatory sequences that direct expression of the transgene in the platelet. [0011]
  • In a further aspect, the invention provides a method for generating a modified hematopoietic stem cell, a progenitor cell, or a modified cell differentiated from the hematopoietic stem cell and/or progenitor cell. The method involves transferring a nucleic acid molecule as described above into the cell via transformation, transduction, infection or transfection. [0012]
  • In yet another aspect, the invention provides methods for treating or preventing certain disorders, diseases, symptoms or injuries in which cells of the hematopoietic lineage are involved, by delivering to a mammalian patient a recombinant nucleic acid molecule described above or a suitable host cell described above, that contain a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in the host cell. A differentiated cell produces the product at a suitable in the mammal. [0013]
  • In still a further aspect, the invention provides a method for preventing unwanted thrombus formation in a mammal by administering an above-described molecule or platelet containing same, in which the transgene is a fibrinolytic protein and the regulatory sequences direct expression of the product of the transgene in a platelet. The platelet produces the product at the site of the thrombus formation.[0014]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a schematic representation of the 10.2 kb Sac II insert obtained from the transgenic mice expressing u-PA in their platelets. The insert contains the 1.2 kb murine Xba I/Kpn I PF4 promoter (open box) plus a 5′ untranslated region (5′-UTR; light gray box) followed by the 11-exon murine u-PA gene (Heckel, J. L. et al, 1990 [0015] Cell 62:447-456, black boxes) and ending with the hGH 3′-UTR and polyadenylation sequence (dark gray box). The 2.8 kb Bgl II fragment containing the transgene was detected in a genomic Southern blot (not shown).
  • FIG. 2 is a graph of a pulmonary microemboli lysis study. Solid circles show residual labeled microemboli remaining in the lungs of wildtype mice at the indicated times (n=6). Open squares show the residual radioactivity in the lungs of [0016] line #19 mice studied in parallel (n=6). Mean±1 SD is shown.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides novel nucleic acid molecules, host cells containing the molecules, and methods for delivery of recombinant transgenes using modified cells of the hematopoietic stem cell lineage. Methods for treatment and prophylaxis using these cells are disclosed for a variety of disorders involving or mediated by the cells of the hematopoietic lineage. Such disorders can include inflammations, infections, tissue injuries, vascular injuries, and the like. [0017]
  • I. NUCLEIC ACID MOLECULES OF THE INVENTION
  • One aspect of this invention includes isolated, synthetic or recombinant nucleic acid molecules and sequences comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of the transgene in a cell of the hematopoietic lineage. Such nucleic acid molecules are used to express the transgene product in vitro or ex vivo, or to permit expression of the transgene product in vivo in a mammal. [0018]
  • As used herein, the term “isolated nucleotide molecule or sequence” refers to a nucleic acid segment or fragment which is free from contamination with other biological components that are associated with the molecule or sequence in its natural environment. For example, one embodiment of an isolated nucleotide molecule or sequence of this invention is a sequence separated from sequences which flank it in a naturally occurring state, e.g., a DNA fragment which has been removed from the sequences which are normally adjacent to the fragment, such as the sequences adjacent to the fragment in a genome in which it naturally occurs. Further, the nucleotide sequences and molecules of this invention have been altered to encode a selected transgene product. Thus, the term “isolated nucleic acid molecule or sequence” also applies to nucleic acid sequences or molecules that have been substantially purified from other components that naturally accompany the unmutagenized nucleic acid, e.g., RNA or DNA, or proteins, in the cell. An isolated nucleotide molecule or sequence of this invention also encompasses sequences and molecules that have been prepared by other conventional methods, such as recombinant methods, synthetic methods, e.g., mutagenesis, or combinations of such methods. The nucleotide sequences or molecules of this invention should not be construed as being limited solely to the specific nucleotide sequences presented herein, but rather should be construed to include any and all nucleotide sequences which share homology (i.e., have sequence identity) with the nucleotide sequences presented herein. [0019]
  • By the term “promoter” or “regulatory sequence” is meant a DNA sequence required to direct expression of a nucleic acid operably linked thereto in a cell of hematopoietic lineage. Preferably the promoter/regulatory sequence is positioned at the 5′ end of the transgene coding sequence such that it drives expression of the transgene-encoded protein in a cell. In the molecules of this invention, the promoter/regulatory sequence may also include an enhancer sequence and other regulatory elements that are required for expression in these hematopoietic cells. [0020]
  • Suitable regulatory sequences/promoters for use in the present invention are readily selected from among regulatory sequences that express the selected transgene in the hematopoietic stem cell itself, or in one of the progenitor cells of the hematopoietic lineage, including the common lymphoid progenitor, the common myeloid progenitor, the megakaryotic/erythrocyte progenitor or the granulocyte/macrophage progenitor. In one embodiment the regulatory sequences are able to direct expression of the transgene in a cell that is further differentiated from these progenitor cells. For example, suitable cells differentiated from the megakaryotic/erythrocyte progenitor cells are platelets, megakaryocytes or erythrocytes. Suitable cells differentiated from granulocyte/macrophage progenitor cells are neutrophils, eosinophils, monocytes, basophils and immature dendritic cells. Still other suitable cells differentiated from monocytes are mast cells, macrophages and dendritic cells. Suitable cells differentiated from said lymphoid progenitors are natural killer cells. Promoters capable of directing expression of a transgene in any of these cells of the hematopoietic lineage are useful as regulatory sequences in the nucleic acid molecules of this invention. In one embodiment, the promoter expresses the gene in secretory cells of the hematopoietic lineage, i.e., cells that release the contents of their granules upon activation, e.g., platelets, mast cells, neutrophils, eosinophils, etc. See, e.g., IMMUNOBIOLOGY, THE IMMUNE SYSTEM IN HEALTH AND DISEASE, 5[0021] th edit., C. Janeway et al., Ed., Garland Publishing, New York, N.Y.: 2001.
  • In one embodiment, a platelet-specific promoter is used as a regulatory sequence to direct expression of the transgene in a plasmid. Among suitable promoters are, without limitation, the Platelet factor 4 (PF4) promoter, the glycoprotein IIb promoter, the glycoprotein IIIa promoter, and the glycoprotein VI promoter. In another embodiment, the regulatory sequence is a neutrophil-specific promoter, such as one or more of the human neutrophil alpha defensin promoters DEFA1, DEFA2, DEFA3, and DEFA4, among others. Where the transgene is desirably expressed in a NK cell, the regulatory sequence is a natural killer cell-specific promoter, for example, the human perform gene promoter. In still another embodiment, the regulatory sequence useful in the nucleic acid molecule of this invention is an eosinophil-specific promoter. Examples of eosinophil-specific promoters include, without limitation, the human eotaxin gene promoter and the eosinophil peroxidase gene promoter. In another embodiment, a suitable regulatory sequence is an erythrocyte-specific promoter, such as the human RhD gene promoter. [0022]
  • Depending upon the use for which the nucleic acid molecule is constructed, the transgene is any peptide, polypeptide or protein useful for the treatment of a disorder or reduction or prevention of a symptom in which cells of the hematopoietic system are involved. For example, a non-exclusive list of products includes those encoded by therapeutic transgenes for the treatment of a variety of inflammatory conditions, microbial or parasitic infections, injuries, such as vascular injuries and other hematopoietic cell-involved disorders. In one embodiment, such products include fibrinolytic proteins, including without limitation, urokinase-type plasminogen activator, plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor, fibrinogen, Factor V, and plasminogen for use in altering the hemostatic balance at sites of thrombosis. Suitable products also include, without limitation, hormones and growth and differentiation factors including, without limitation, insulin, glucagon, growth hormone (GH), parathyroid hormone (PTH), growth hormone releasing factor (GHRF), follicle stimulating hormone (FSH), luteinizing hormone (LH), human chorionic gonadotropin (hCG), vascular endothelial growth factor (VEGF), angiopoietins, angiostatin, granulocyte colony stimulating factor (GCSF), erythropoietin (EPO), connective tissue growth factor (CTGF), basic fibroblast growth factor (bFGF), acidic fibroblast growth factor (aFGF), epidermal growth factor (EGF), transforming growth factor a (TGFα), platelet-derived growth factor (PDGF), insulin growth factors I and II (IGF-I and IGF-II), any one of the transforming growth factor β superfamily, including TGF β, activins, inhibins, or any of the bone morphogenic proteins (BMP) including BMPs 1-15, any one of the heregluin/neuregulin/ARIA/neu differentiation factor (NDF) family of growth factors, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophins NT-3 and NT-4/5, ciliary neurotrophic factor (CNTF), glial cell line derived neurotrophic factor (GDNF), neurturin, agrin, any one of the family of semaphorins/collapsins, netrin-1 and netrin-2, hepatocyte growth factor (HGF), ephrins, noggin, sonic hedgehog and tyrosine hydroxylase. [0023]
  • Other useful transgene products include proteins that regulate the immune system including, without limitation, cytokines and lymphokines such as thrombopoietin (TPO), interleukins (IL) IL-1 through IL-25 (including, IL-2, IL-4, IL-12, and IL-18), monocyte chemoattractant protein, leukemia inhibitory factor, granulocyte-macrophage colony stimulating factor, Fas ligand, tumor necrosis factors α and β, interferons α, β, and γ, stem cell factor, flk-2/flt3 ligand. Gene products produced by the immune system are also useful in the invention. These include, without limitation, immunoglobulins IgG, IgM, IgA, IgD and IgE, chimeric immunoglobulins, humanized antibodies, single chain antibodies, T cell receptors, chimeric T cell receptors, single chain T cell receptors, class I and class II MHC molecules, as well as engineered immunoglobulins and MHC molecules. Useful gene products also include complement regulatory proteins, membrane cofactor protein (MCP), decay accelerating factor (DAF), CR1, CF2 and CD59. [0024]
  • Still other useful gene products include any one of the receptors for the hormones, growth factors, cytokines, lymphokines, regulatory proteins and immune system proteins. The invention encompasses receptors for cholesterol regulation, including the low density lipoprotein (LDL) receptor, high density lipoprotein (HDL) receptor, the very low density lipoprotein (VLDL) receptor, and the scavenger receptor. The invention also encompasses gene products such as members of the steroid hormone receptor superfamily including glucocorticoid receptors and estrogen receptors, Vitamin D receptors and other nuclear receptors. In addition, useful gene products include transcription factors such as jun, fos, max, mad, serum response factor (SRF), AP-1, AP2, myb, MyoD, myogenin, ETS-box containing proteins, TFE3, E2F, ATF1, ATF2, ATF3, ATF4, ZF5, NFAT, CREB, HNF-4, C/EBP, SP1, CCAAT-box binding proteins, interferon regulation factor (IRF-1), Wilms tumor protein, ETS-binding protein, STAT, GATA-box binding proteins, e.g., GATA-3, and the forkhead family of winged helix proteins. [0025]
  • Other useful gene products include, carbamoyl synthetase I, ornithine transcarbamylase, arginosuccinate synthetase, arginosuccinate lyase, arginase, fumarylacetacetate hydrolase, phenylalanine hydroxylase, alpha-1 antitrypsin, glucose-6-phosphatase, porphobilinogen deaminase, factor VIII, factor IX, cystathione beta-synthase, branched chain ketoacid decarboxylase, albumin, isovaleryl-coA dehydrogenase, propionyl CoA carboxylase, methyl malonyl CoA mutase, glutaryl CoA dehydrogenase, insulin, beta-glucosidase, pyruvate carboxylate, hepatic phosphorylase, phosphorylase kinase, glycine decarboxylase, H-protein, T-protein, a cystic fibrosis transmembrane regulator (CFTR) sequence, and a dystrophin cDNA sequence. [0026]
  • Still other useful gene products include enzymes such as are useful in enzyme replacement therapy, which is useful in a variety of conditions resulting from deficient activity of enzyme. [0027]
  • Further useful gene products include non-naturally occurring polypeptides, such as chimeric or hybrid polypeptides having a non-naturally occurring amino acid sequence containing insertions, deletions or amino acid substitutions. For example, single-chain engineered immunoglobulins could be useful in certain immunocompromised patients. Other types of non-naturally occurring gene sequences include antisense molecules and catalytic nucleic acids, such as ribozymes, which could be used to reduce overexpression of a target. [0028]
  • Reduction and/or modulation of expression of a gene are particularly desirable for treatment of hyperproliferative conditions characterized by hyperproliferating cells, as are cancers and psoriasis. Some polypeptides, which are produced exclusively or at higher levels in hyperproliferative cells as compared to normal cells. The compositions of this invention are employed to express as transgene products polypeptides that can reduce the activity or inactivate oncogenes such as myb, myc, fyn, and the translocation gene bcr/abl, ras, src, P53, neu, trk and EGRF. Anti-cancer treatments and protective regimens include transgene products directed to inactivate or reduce the activity of variable regions of antibodies made by B cell lymphomas and variable regions of T cell receptors of T cell lymphomas which, in some embodiments, are also used as target antigens for autoimmune disease. Other tumor-associated polypeptides can be used as target polypeptides such as polypeptides, which are found at higher levels in tumor cells including the polypeptide recognized by monoclonal antibody 17-1A and folate binding polypeptides. [0029]
  • Other suitable therapeutic polypeptides and proteins include those which are useful for treating individuals suffering from autoimmune diseases and disorders by conferring a broad based protective immune response against targets that are associated with autoimmunity including cell receptors and cells which produce “self”-directed antibodies. T cell mediated autoimmune diseases include Rheumatoid arthritis (RA), multiple sclerosis (MS), Sjögren's syndrome, sarcoidosis, insulin dependent diabetes mellitus (IDDM), autoimmune thyroiditis, reactive arthritis, ankylosing spondylitis, scleroderma, polymyositis, dermatomyositis, psoriasis, vasculitis, Wegener's granulomatosis, Crohn's disease and ulcerative colitis. Each of these diseases is characterized by T cell receptors (TCRs) that bind to endogenous antigens and initiate the inflammatory cascade associated with autoimmune diseases. [0030]
  • Still other suitable transgenes encode fibrinolytic proteins and peptides suitable for delivery by transgene expression in platelets, such as illustrated in the below-noted examples. The selection of the transgene sequence, or other molecule carried by the nucleic acid molecule, is not a limitation of this invention. Choice of a transgene sequence is within the skill of the artisan in accordance with the teachings of this application. [0031]
  • The terms “homology” or “similarity,” when referring to a nucleic acid or fragment thereof, indicate that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 70% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, a program in GCG Version 6.1. The term “homologous” as used herein, refers to the sequence similarity between two polymeric molecules, e.g., between two nucleic acid molecules, e.g., two DNA molecules, two RNA molecules, or two polypeptide molecules. When a nucleotide or amino acid position in both of the two molecules is occupied by the same monomeric nucleotide or amino acid, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position. The homology between two sequences is a direct function of the number of matching or homologous positions, e.g. if half (e.g., five positions in a polymer ten subunits in length) of the positions in two compound sequences are homologous, then the two sequences are 50% homologous. If 90% of the positions, e.g, 9 of 10, are matched or homologous, the two sequences share 90% homology. By way of example, the DNA sequences 3′ATTGCC5′ and 3′TATGCG5′ share 50% homology. By the term “substantially homologous” as used herein, is meant DNA or RNA which is about 70% homologous, more preferably about 80% homologous, and most preferably about 90% homologous to the desired nucleic acid. [0032]
  • When referring to the transgenes or the regulatory sequences disclosed above, the invention is also directed to an isolated nucleotide molecule comprising a nucleic acid sequence that is at least 70%, 80% or 90% homologous to a nucleic acid sequence encoding a naturally occurring transgene-encoded protein or a polypeptide that has similar biological effect as the native transgene product. Furthermore, due to the degeneracy of the genetic code, any three-nucleotide codon that encodes a mutant or substituted amino acid residue of a transgene-encoded protein, described herein is within the scope of the invention. [0033]
  • Where, as discussed herein, proteins, and/or DNA sequences encoding them, or other sequences useful in nucleic acid molecules or compositions described herein are defined by their percent homologies or identities to identified sequences, the algorithms used to calculate the percent homologies or percent identities include the following: the Smith-Waterman algorithm (J. F. Collins et al, 1988, [0034] Comput. Appl. Biosci., 4:67-72; J. F. Collins et al, Molecular Sequence Comparison and Alignment, (M. J. Bishop et al, eds.) In Practical Approach Series: Nucleic Acid and Protein Sequence Analysis XVIII, IRL Press: Oxford, England, UK (1987) pp.417), and the BLAST and FASTA programs (E. G. Shpaer et al, 1996, Genomics, 38:179-191). These references are incorporated herein by reference.
  • By describing two DNA sequences as being “operably linked” as in the relationship between the transgene and the regulatory sequences used herein, is meant that a single-stranded or double-stranded DNA comprises two DNA sequences and that the two DNA sequences are arranged within the molecule or sequence in such a manner that at least one of the DNA sequences is able to exert a physiological effect by which it is characterized upon the other. [0035]
  • Preferably, each protein encoding transgene sequence and necessary regulatory sequences of this invention are present in a separate viral or non-viral recombinant vector (including non-viral methods of delivery of a nucleic acid molecule into a cell). Alternatively, two or more of these nucleic acid sequences encoding duplicate copies of a the transgene-encoded protein or encoding multiple different therapeutic proteins of this invention are contained in a polycistronic transcript, i.e., a single molecule designed to express multiple gene products. [0036]
  • The isolated nucleic acid of this invention is desirably a recombinant vector, particularly a plasmid, containing isolated and purified DNA sequences comprising DNA sequences that encode a selected therapeutic protein. By the term “vector” as used herein, is meant a DNA molecule derived from viral or non-viral, e.g., bacterial, species that has been designed to encode an exogenous or heterologous nucleic acid sequence. Thus, the term includes conventional bacterial plasmids. Such plasmids or vectors can include plasmid sequences from viruses or phages. Such vectors include chromosomal, episomal and virus-derived vectors, e.g., vectors derived from bacterial plasmids, bacteriophages, yeast episomes, yeast chromosomal elements, and viruses. Vectors may also be derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, cosmids, and phagemids. The term also includes non-replicating viruses that transfer a gene from one cell to another. The term should also be construed to include non-plasmid and non-viral compounds which facilitate transfer of nucleic acid into cells, such as, for example, polylysine compounds and the like. [0037]
  • The nucleic acid molecules of the invention include non-viral vectors or methods for delivery of the sequences encoding the therapeutic protein to a host cell according to this invention. A variety of non-viral vectors are known in the art and may include, without limitation, plasmids, bacterial vectors, bacteriophage vectors, “naked” DNA and DNA condensed with cationic lipids or polymers. [0038]
  • Examples of bacterial vectors include, but are not limited to, sequences derived from bacille Calmette Guérin (BCG), Salmonella, Shigella, [0039] E. coli, and Listeria, among others. Suitable plasmid vectors include, for example, pBR322, pBR325, pACYC177, pACYC184, pUC8, pUC9, pUC18, pUC19, pLG339, pR290, pK37, pKC101, pAC105, pVA51, pKH47, pUB110, pMB9, pBR325, Co1 E1, pSC101, pBR313, pML21, RSF2124, pCR1, RP4, pBAD18, and pBR328. Examples of suitable inducible Escherichia coli expression vectors include pTrc (Amann et al., 1988 Gene, 69:301-315), the arabinose expression vectors (e.g., pBAD18, Guzman et al, 1995 J. Bacteriol., 177:4121-4130), and pETIId (Studier et al., 1990 Methods in Enzymology, 185:60-89).
  • Another type of useful vector is a single or double-stranded bacteriophage vector. For example, a suitable cloning vector includes, but is not limited to the vectors such as bacteriophage λ vector system, λgt11, μgt μWES.tB, Charon 4, λgt-WES-λB, Charon 28, Charon 4A, λgt-1-λBC, λgt-1-λB, M13mp7, M13mp8, or M13mp9, among others. [0040]
  • In yet another embodiment, a mammalian expression vector is used for expression of the selected transgene in mammalian cells of the hematopoietic lineage. Examples of mammalian expression vectors include pCDM8 (Seed, 1987 [0041] Nature, 329:840-842) and pMT2PC (Kaufman et al., 1987 EMBO J., 6(1):187-93). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements other than the promoters specified above.
  • One type of recombinant vector is a recombinant single or double-stranded RNA or DNA viral vector. A variety of viral vector systems are known in the art. Examples of such vectors include, without limitation, recombinant adenoviral vectors, herpes simplex virus (HSV)-based vectors, adeno-associated viral (AAV) vectors, hybrid adenoviral/AAV vectors, recombinant retroviruses or lentiviruses, recombinant poxvirus vectors, recombinant vaccinia virus vectors, SV-40 vectors, insect viruses such as baculoviruses, and the like that are constructed to carry or express a selected nucleic acid composition of interest. [0042]
  • Retrovirus vectors that can be employed include those described in [0043] EP 0 415 731; International Patent Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; and WO 93/25234; U.S. Pat. No. 5,219,740; International Patent Publication Nos. WO 93/11230 and WO 93/10218; Vile and Hart, 1993 Cancer Res. 53:3860-3864; Vile and Hart, 1993 Cancer Res. 53:962-967; Ram et al., 1993 Cancer Res. 53:83-88; Takamiya et al., 1992 J. Neurosci. Res. 33:493-503; Baba et al., 1993 J. Neurosurg. 79:729-735; U.S. Pat. No. 4,777,127; GB Pat. No. 2,200,651; and EP 0 345 242. Examples of suitable recombinant retroviruses include those described in International Patent Publication No. WO 91/02805. See also, D. A. Wilcox et al, 2000 Blood, 95 (12):3645-3652.
  • Alphavirus-based vectors may also be used as the nucleic acid molecule encoding the therapeutic protein. Such vectors can be constructed from a wide variety of alphaviruses, including, for example, Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532). Representative examples of such vector systems include those described in U.S. Pat. Nos. 5,091,309; 5,217,879; and 5,185,440; and International Patent Publication Nos. WO 92/10578; WO 94/21792; WO 95/27069; WO 95/27044; and WO 95/07994. [0044]
  • Examples of adenoviral vectors include those described by Berkner, 1988 [0045] Biotechniques 6:616-627; Rosenfeld et al., 1991 Science 252:431-434; International Patent Publication No. WO 93/19191; Kolls et al., 1994 PNAS 91:215-219; Kass-Eisler et al., 1993 PNAS 90:11498-11502; Guzman et al., 1993 Circulation 88:2838-2848; Guzmanetal., 1993 Cir. Res. 73:1202-1207;Zabner et al., 1993 Cell 75:207-216; Li et al, 1993 Hum. Gene Ther. 4:403-409; Cailaud et al., 1993 Eur. J. Neurosci. 5:1287-1291; Vincent et al., 1993 Nat. Genet. 5:130-134; Jaffe et al., 1992 Nat. Genet. 1:372-378; and Levrero et al., 1991 Gene 101: 195-202. Exemplary adenoviral vectors include those described in International Patent Publication Nos. WO 94/12649; WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655. Other adenoviral vectors include those derived from chimpanzee adenoviruses, such as those described in U.S. Pat. No. 6,083,716.
  • Another viral vector is based on a parvovirus such as an adeno-associated virus (AAV). Representative examples include the AAV vectors described in International Patent Publication No. WO 93/09239, Samulski et al., 1989 [0046] J. Virol. 63:3822-3828; Mendelson et al., 1988 Virol. 166:154-165; and Flotte et al., 1993 PNAS 90:10613-10617. Other particularly desirable AAV vectors include those based upon AAV1; see, International Patent Publication No. WO 00/28061, published May 18, 2000. Other desirable AAV vectors include those which are pseudotyped, i.e., contain a minigene composed of AAV 5′ ITRs, a transgene, and AAV 3′ ITRs packaged in a capsid of an AAV serotype heterologous to the AAV ITRs. Methods of producing such pseudotyped AAV vectors are described in detail in International Patent Publication No. WOO1/83692.
  • In an embodiment in which the nucleic acid molecule of the invention is “naked DNA”, it is combined with polymers including traditional polymers and non-traditional polymers such as cyclodextrin-containing polymers and protective, interactive noncondensing polymers, among others. The “naked” DNA and DNA condensed with cationic lipids or polymers are typically delivered to the cells using chemical methods. A number of chemical methods are known in the art for cell delivery and include using lipids, polymers, or proteins to complex with DNA, optionally condensing the same into particles, and delivering to the cells. Another non-viral chemical method includes using cations to condense DNA, which is then placed in a liposome and used according to the present invention. See, C. Henry, 2001 [0047] Chemical and Engineering News, 79(48):35-41.
  • The nucleic acid molecule encoding the selected therapeutic protein is introduced directly into the cells of the hematopoietic lineage either as “naked” DNA (U.S. Pat. No. 5,580,859) or formulated in compositions with agents that facilitate direct immunization, such as bupivicaine and other local anesthetics (U.S. Pat. No. 6,127,170). [0048]
  • All components of the viral and non-viral vectors above are readily selected from among known materials in the art and available from the pharmaceutical industry. Selection of the vector components other than the regulatory sequences are not considered a limitation on this invention. Each nucleic acid sequence encoding a protein according to this invention is preferably under the control of regulatory sequences that direct the replication and generation of the product of each nucleic acid sequence, preferably ectopically, in a mammalian hematopoietic lineage, progenitor cell or differentiated cell. [0049]
  • Additional regulatory sequences for inclusion in a nucleic acid sequence, molecule or vector of this invention include, without limitation, one or more of an enhancer sequence, a polyadenylation sequence, a splice donor sequence and a splice acceptor sequence, a site for transcription initiation and termination positioned at the beginning and end, respectively, of the polypeptide to be translated, a ribosome binding site for translation in the transcribed region, an epitope tag, a nuclear localization sequence, an IRES element, a Goldberg-Hogness “TATA” element, a restriction enzyme cleavage site, a selectable marker and the like. Enhancer sequences include, e.g., the 72 bp tandem repeat of SV40 DNA or the retroviral long terminal repeats or LTRs, etc. and are employed to increase transcriptional efficiency. Selection of other non-promoter common vector elements is conventional and many such sequences are available with which to design the nucleotide molecules and vectors useful in this invention. See, e.g., Sambrook et al, [0050] Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, New York, (1989) and references cited therein at, for example, pages 3.18-3.26 and 16.17-16.27 and Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York (1989). One of skill in the art may readily select from among such known non-promoter sequences to prepare nucleic acid molecules of this invention. The selection of such non-promoter sequences is not a limitation of this invention.
  • II. METHODS OF PRODUCING THE NUCLEIC ACID MOLECULES
  • The preparation or synthesis of the nucleotide sequences and nucleotide molecules of this invention is well within the ability of the person having ordinary skill in the art using available material. The synthesis methods are not a limitation of this invention. The examples below detail presently preferred embodiments of synthesis of sequences encoding an exemplary transgene useful in this invention for the construction of a transgenic animal model. However, similar methods are employed to produce nucleic acid molecules for the generation of therapeutic or prophylactic compositions of this invention. [0051]
  • The nucleotide molecules and sequences of this invention are produced by chemical synthesis methods. For example, the nucleotide sequences useful in the invention are prepared conventionally by resort to known chemical synthesis techniques, e.g., solid-phase chemical synthesis, such as described by Merrifield, 1963 [0052] J. Amer. Chem. Soc., 85:2149-2154; J. Stuart and J. Young, Solid Phase Peptide Synthesis, Pierce Chemical Company, Rockford, Ill. (1984); Matteucci et al., 1981 J. Am. Chem. Soc., 103:3185; Alvarado-Urbina et al., 1980 Science, 214:270; and Sinha, N. D. et al., 1984 Nucl. Acids Res., 13:4539, among others. See, also, e.g., PROTEINS—STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York, 1993; Wold, F., “Posttranslational Protein Modifications: Perspectives and Prospects”, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, 1983; Seifter et al., 1990 Meth. Enzymol., 182:626-646, and Rattan et al., 1992 Ann. N.Y Acad. Sci., 663:48-62.
  • Alternatively, the nucleic acid molecules of this invention are constructed recombinantly using conventional molecular biology techniques, site-directed mutagenesis, genetic engineering or polymerase chain reaction, such as, by cloning and expressing a nucleotide molecule encoding a desired therapeutic protein with optional other proteins within a host cell of the hematopoietic lineage, etc. utilizing the information provided herein (See, e.g., Sambrook et al., cited above; Ausubel et al. cited above). Coding sequences for the transgenes and the regulatory sequences can be prepared synthetically (W. P. C. Stemmer et al, 1995 [0053] Gene, 164:49).
  • In general, recombinant DNA techniques involve obtaining by synthesis or isolation a DNA sequence that encodes the desired therapeutic protein as described above, and introducing it into an appropriate vector where it is expressed preferably under the control of the selected promoter that can direct expression in a cell of the hematopoietic lineage. [0054]
  • Any of the methods described for the insertion of DNA into a vector is used to ligate the selected promoter and other regulatory control elements into specific sites within the selected recombinant vector to generate the nucleic acid molecule. [0055]
  • III. HOST CELLS OF THE INVENTION
  • In another aspect of this invention, a cell of the hematopoietic lineage is manipulated to contain a nucleic acid molecule described above. [0056]
  • In one embodiment, the nucleic acid molecule generated as described above is transferred into an isolated hematopoietic stem cell, a common lymphoid progenitor cell, a common myeloid progenitor cell, a megakaryotic/erythrocyte progenitor cell or a granulocyte/macrophage progenitor cell. The hematopoietic stem cells or progenitor cells of the hematopoietic lineage are isolated from bone marrow of a suitable human or non-human mammal. For example, the cells are isolated from a mammalian patient into whom the manipulated cells are re-introduced. Alternatively, the mammal providing the host cells is a different mammal, for either introduction into another mammal or for research or laboratory use. Methods for isolating such cells from bone marrow are well known. See, for example, the Stem Cell Database of Princeton University; Phillips, R L et al, 2000 Science, 288:1635-1640 and references cited therein. [0057]
  • In another embodiment, the host cells of the hematopoietic lineage are those cells found in the peripheral blood or tissue, such as platelets, megakaryocytes, neutrophils, eosinophils, monocytes, basophils, dendritic cells, mast cells, macrophages, dendritic cells, erythrocytes, and natural killer cells. These cells must be isolated from the peripheral blood or tissue of a suitable human or non-human mammal. For example, the cells are isolated from a mammalian patient into whom the manipulated cells would be re-introduced. Alternatively, the mammal providing the host cells is a different mammal, for either introduction into another mammal or for research or laboratory use. Methods for isolating such cells from peripheral blood or tissue are well known. The introduction of a platelet expressing a suitable transgene under the platelet specific PF4 regulatory sequence is exemplified in the examples below. [0058]
  • Once the suitable host cell is isolated, the nucleic acid molecule or vector is transferred therein in vitro or ex vivo by a conventional technique such as transformation, transduction, infection or transfection. The selection of other suitable methods for transferring the nucleic acid molecule or the vector into an isolated cell of the hematopoietic lineage can be performed by one of skill in the art by reference to known techniques. See, e.g., Gething and Sambrook, 1981 [0059] Nature, 293:620-625, among others.
  • If necessary, such host cells are cultured. Culture conditions are well known in the art and include ionic composition and concentration, temperature, pH and the like. Typically, transfected cells are maintained under culture conditions in a culture medium. Suitable media for various cell types are well known in the art. In a preferred embodiment, the temperature is from about 20° C. to about 50° C., more preferably from about 30° C. to about 40° C. and, even more preferably about 37° C. [0060]
  • The pH is preferably from about a value of 6.0 to a value of about 8.0, more preferably from about a value of about 6.8 to a value of about 7.8 and, most preferably about 7.4. Osmolality is preferably from about 200 milliosmols per liter (mosm/L) to about 400 mosm/l and, more preferably from about 290 mosm/L to about 310 mosm/L. Other biological conditions needed for transfection of a vector are well known in the art. [0061]
  • IV. METHODS OF U.S.E. OF THE NUCLEIC ACID COMPOSITION AND HOST CELLS OF THE INVENTION
  • Once transfected, the host cells of the hematopoietic lineage are employed in pharmaceutical or prophylactic compositions and methods for the treatment of a variety of disorders in human or non-human mammals. Such treatment may include enhancement of a biological activity or reduction or a disadvantageous biological activity occurring in the body. Similarly, the nucleic acid molecules themselves are used in direct treatment of disorders such as inflammatory disorders, microbial or parasitic infection, vascular or hemorrhagic disorders, and the like in which the hematopoietic cells, their progenitors or differentiated cells are implicated. In other embodiments, the host cells (preferably platelets) are employed to prevent formation of stable, occlusive thrombus development. [0062]
  • One of skill in the art may readily identify a number of such disorders. Among such disorders are included without limitation, coagulation disorders (either an insufficiency or excess thereof), acute lung injury and sepsis, helminth infection, asthma or other allergic reactions, viral infections, bacterial infections, etc. For example, the compositions of this invention are useful in the prevention and/or treatment of disease(s) caused by microbial infections including, without limitation, [0063] Haemophilus influenzae (both typable and nontypable), Haemophilus somnus, Moraxella catarrhalis, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Streptococcus faecalis, Helicobacter pylori, Neisseria meningitidis, Neisseria gonorrhoeae, Chlamydia trachomatis, Chlamydia pneumoniae, Chlamydia psittaci, Bordetella pertussis, Alloiococcus otiditis, Salmonella typhi, Salmonella typhimurium, Salmonella choleraesuis, Escherichia coli, Shigella, Vibrio cholerae, Corynebacterium diphtheriae, Mycobacterium tuberculosis, Mycobacterium avium-Mycobacterium intracellulare complex, Proteus mirabilis, Proteus vulgaris, Staphylococcus aureus, Staphylococcus epidermidis, Clostridium tetani, Leptospira interrogans, Borrelia burgdorferi, Pasteurella haemolytica, Pasteurella multocida, Actinobacillus pleuropneumoniae and Mycoplasma gallisepticum.
  • The compositions of this invention are useful in the prevention and/or treatment of disease caused by, without limitation, Respiratory syncytial virus, Parainfluenza virus types 1-3, Human metapneumovirus, Influenza virus, Herpes simplex virus, Human cytomegalovirus, Human immunodeficiency virus, Simian immunodeficiency virus, Hepatitis A virus, Hepatitis B virus, Hepatitis C. virus, Human papillomavirus, Poliovirus, rotavirus, caliciviruses, Measles virus, Mumps virus, Rubella virus, adenovirus, rabies virus, canine distemper virus, rinderpest virus, avian pneumovirus (formerly turkey rhinotracheitis virus), Hendra virus, Nipah virus, coronavirus, parvovirus, infectious rhinotracheitis viruses, feline leukemia virus, feline infectious peritonitis virus, avian infectious bursal disease virus, Newcastle disease virus, Marek's disease virus, porcine respiratory and reproductive syndrome virus, equine arteritis virus and various Encephalitis viruses. [0064]
  • The compositions of this invention are useful in enhancing response against fungal pathogens such as Aspergillis, Blastomyces, Candida, Coccidiodes, Cryptococcus and Histoplasma or against parasites including [0065] Leishmania major, Ascaris, Trichuris, Giardia, Schistosoma, Cryptosporidium, Trichomonas, Toxoplasma gondii and Pneumocystis carinii.
  • Compositions of this invention may also be useful for the prevention and/or treatment of disease(s), without limitation, such as autoimmune disease, such as multiple sclerosis, lupus and rheumatoid arthritis and others, asthma, atherosclerosis, Alzheimer disease, amyloidosis or amyloidogenic disease, and cancers. Clotting disorders and other vascular injuries caused by other infections, injury, aging, thrombocytopenia, inappropriate thrombus formation, myelodysplasia, AML, and the like may also be treated according to the methods of this invention. These compositions and methods can be useful to treat allergic reactions to allergens such as pollen, insect venoms, animal dander, fungal spores and drugs (such as penicillin). Other conditions that are treated by the methods of this invention included disease characterized by unwanted thrombus formation, amyloid deposition, diabetes, and gastroesophageal reflux disease, among others. The methods of this invention may also be useful in the enhancement of wound healing. The selection of the disorder to be treated by the compositions and methods of this invention is not a limitation of this invention. One of skill in the art may readily include other disorders suitable for the treatment described herein. [0066]
  • One method for treating a disorder in a mammal according to this invention includes the step of delivering to a mammal a recombinant nucleic acid molecule comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a hematopoietic stem cell, a progenitor cell of the hematopoietic lineage, or a cell differentiated therefrom. Depending upon the identity of the regulatory sequence and the transgene, the method permits a differentiated cell of a hematopoietic lineage to express the product of the selected transgene at a suitable site in the mammal. Where the differentiated cell is secretory, the cell may express the transgene produce ectopically and target the expression to a particular site. For example, a plasmid expressing a transgene will target to the site of vascular injury or thrombus formation. [0067]
  • In one embodiment, therefore, the method of the invention involves harvesting stem cells or progenitor cells from bone marrow of a patent, transferring the nucleic acid molecule into the cells ex vivo and reinfusing the cells into the bone marrow or peripheral blood of the mammalian patient. Alternatively, the method of treatment can involve infusing or injecting into the patients bone marrow or blood a non-self transfected host cell. [0068]
  • In another embodiment, the method of the invention involves harvesting differentiated cells of the hematopoietic lineage from peripheral blood of a mammalian patient; transferring the nucleic acid molecule into the differentiated cells; and [0069]
  • returning these cells into the blood of the patient. Alternatively, the method of treatment can involve infusing or injecting into the patient's blood a non-self transfected differentiated host cell. [0070]
  • In still another embodiment of this invention, a method of treatment can involve directly administering to a mammalian patient, simply the nucleic acid molecule, i.e., as naked DNA. The regulatory sequences should aid in the uptake of the molecule by the appropriate differentiated cells of the hematopoietic lineage. [0071]
  • Thus, as one specific embodiment, the invention provides a method for enhancing coagulation in a patient by delivering to the mammalian patient with an insufficient clotting mechanism a recombinant nucleic acid molecule (or a platelet containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet. Preferably the regulatory sequence would be a platelet-specific sequence mentioned above. Examples of suitable transgenes for this method are transgenes encoding one or more of Factor VIIa, Factor VIII, Factor IX or fibrinogen. [0072]
  • Another specific embodiment involves a method for preventing or reducing coagulation in a mammalian patient, where needed. According to this method, the patient is administered a recombinant nucleic acid molecule (or a platelet containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet. Preferably the regulatory sequence would be a platelet-specific sequence mentioned above. Examples of suitable transgenes for this method are transgenes encoding one or more of urokinase plasminogen activator, plasminogen, tissue plasminogen activator, and tissue factor pathway inhibitor. [0073]
  • Another example of a method of this invention is a method for enhancing coagulation in a mammalian patient by delivering to the patient a recombinant nucleic acid molecule (or an erythrocyte containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in an erythrocyte. Preferably the regulatory sequence would be an erythrocyte-specific sequence mentioned above. Examples of suitable transgenes for this method are transgenes encoding a urokinase plasminogen activator receptor, preferably expressed on the cell surface. [0074]
  • Still another example of this invention is a method for treating acute lung injury and sepsis in a mammalian patient. This method includes delivering to the patient a recombinant nucleic acid molecule (or neutrophils containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a neutrophil. Preferably the regulatory sequence would be a neutrophil-specific sequence mentioned above. An example of a suitable transgene for this method is a transgene encoding activated Protein C. [0075]
  • The methods and compositions of this invention may also be employed in a method for treating parasitic helminth infection of a mammalian human or non-human patient. This method involves delivering to a mammal a recombinant nucleic acid molecule (or eosinophils containing the molecule) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in eosinophils. Preferably the regulatory sequence would be an eosinophil-specific sequence mentioned above. An example of a suitable transgene for this method is a transgene encoding a protein toxic to a helminth. [0076]
  • Another method according to this invention involves treating asthma or allergic responses in a mammalian patient. This method involves delivering to a mammal a recombinant nucleic acid molecule (or an eosinophils containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in an eosinophil. Preferably the regulatory sequence would be an eosinophil-specific sequence mentioned above. Examples of a suitable transgene for this method are transgenes encoding one or more of human TSG6, an antibody to IL-1 receptor alpha, and an anti-inflammatory protein. [0077]
  • The invention also includes a method for treating a viral infection in a mammal comprising delivering to a mammal a recombinant nucleic acid molecule (or NK cell containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a natural killer cell. Preferably the regulatory sequence would be an NK cell-specific sequence mentioned-above. Examples of a suitable transgene for this method is a transgene encoding a neutralizing antibody against a viral coat protein, e.g., anti-HIV gag protein, anti-HPV proteins, anti-HIV proteins, etc. [0078]
  • Finally, as demonstrated specifically by the examples below, the present invention provides a method for the treatment and prevention of undesirable thrombus development in a mammalian patient by administering to the patient a recombinant nucleic acid molecule (or platelet containing same) comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of the transgene in a platelet. As illustrated below, a platelet that ectopically expressed u-PA under the control of the platelet specific PF4 promoter was able to control thrombus formation in a patient. See the Examples below. [0079]
  • The nucleic acid molecules or host cells are employed in compositions containing a physiologically acceptable diluent or a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. The compositions may also be mixed with such diluents or carriers in a conventional manner. As used herein the language “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with administration to humans or other vertebrate hosts. The appropriate carrier will be evident to those skilled in the art and will depend in large part upon the route of administration. [0080]
  • The compositions may also include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and implantable sustained-release or biodegradable formulations. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents. In one embodiment of a formulation for parenteral administration, the active ingredient is provided in dry (i.e., powder or granular) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition. Other formulations may optionally include a liposomal preparation, or as a component of a biodegradable polymer system Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt. [0081]
  • Still additional components that are present are adjuvants, preservatives, chemical stabilizers, or other proteins. Typically, stabilizers, adjuvants, and preservatives are optimized to determine the best formulation for efficacy in the target human or animal. Suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and parachlorophenol. Suitable stabilizing ingredients that are used include, for example, casanino acids, sucrose, gelatin, phenol red, N-Z amine, monopotassium diphosphate, lactose, lactalbumin hydrolysate, and dried milk. [0082]
  • Various cytokines and lymphokines are also suitable for inclusion in the compositions of this invention or administration therewith. One such cytokine is granulocyte-macrophage colony stimulating factor (GM-CSF), which has a nucleotide sequence as described in U.S. Pat. No. 5,078,996, which is hereby incorporated by reference. A plasmid containing GM-CSF cDNA has been transformed into E coli and has been deposited with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, Va. 20110-2209, under Accession Number 39900. The cytokine Interleukin-12 (IL-12) is described in U.S. Pat. No. 5,723,127, which is hereby incorporated by reference (available from Genetics Institute, Inc., Cambridge, Ma.). Other cytokines or lymphokines include but are not limited to, the interleukins 1-α, 1-β, through 24, the interferons-α, β and γ, granulocyte colony stimulating factor, and the tumor necrosis factors α and β. [0083]
  • Still other suitable optional components of the compositions of this invention include, but are not limited to: surface active substances (e.g., hexadecylamine, octadecylamine, octadecyl amino acid esters, lysolecithin, dimethyl-dioctadecylammonium bromide), methoxyhexadecylgylcerol, and pluronic polyols; polyamines, e.g., pyran, dextransulfate, poly IC, carbopol; peptides, e.g., muramyl dipeptide, dimethylglycine, tuftsin; oil emulsions; and mineral gels, e.g., aluminum phosphate, etc. and immune stimulating complexes, liposomes, polysaccharides, lipopolysaccharides and/or other polymers. [0084]
  • In addition to a carrier as described above, compositions composed of polynucleotide molecules desirably contain optional polynucleotide facilitating agents or “co-agents”, such as a local anesthetic, a peptide, a lipid including cationic lipids, a liposome or lipidic particle, a polycation such as polylysine, a branched, three-dimensional polycation such as a dendrimer, a carbohydrate, a cationic amphiphile, a detergent, a benzylammonium surfactant, or another compound that facilitates polynucleotide transfer to cells. Such a facilitating agent includes bupivicaine (see U.S. Pat. No. 5,593,972, which is hereby incorporated by reference). Other non-exclusive examples of such facilitating agents or co-agents useful in this invention are described in U.S. Pat. Nos. 5,703,055; 5,739,118; 5,837,533; International Patent Publication No. W096/10038, published Apr. 4, 1996; and International Patent Publication No W094/16737, published Aug. 8, 1994, which are each incorporated herein by reference. [0085]
  • Most preferably, the local anesthetic is present in an amount that forms one or more complexes with the nucleic acid molecules. When the local anesthetic is mixed with nucleic acid molecules or plasmids of this invention, it forms a variety of small complexes or particles that pack the DNA and are homogeneous. Thus, in one embodiment of the compositions of this invention, the complexes are formed by mixing the local anesthetic and at least one plasmid of this invention. Any single complex resulting from this mixture may contain a variety of combinations of the different plasmids. Alternatively, in another embodiment of the compositions of this invention, the local anesthetic is pre-mixed with each plasmid separately, and then the separate mixtures combined in a single composition to ensure the desired ratio of the plasmids is present in a single composition, if all plasmids are to be administered in a single bolus administration. Alternatively, the local anesthetic and each plasmid are mixed separately and administered separately to obtain the desired ratio. When the facilitating agent used is a local anesthetic, preferably bupivacaine, an amount of from about 0.1 weight percent to about 1.0 weight percent based on the total weight of the polynucleotide composition is preferred. See, also, International Patent Publication No. W099/21591, which is hereby incorporated by reference, and which teaches the incorporation of benzylammonium surfactants as co-agents, preferably administered in an amount of between about 0.001-0.03 weight %. According to the present invention, the amount of local anesthetic is present in a ratio to the nucleic acid molecules of 0.01-2.5% w/v local anesthetic to 1-10 μg/ml nucleic acid. Another such range is 0.05-1.25% w/v local anesthetic to 100 μg/ml to 1 ml/ml nucleic acid. [0086]
  • As used, such a polynucleotide composition may express the transgene product on a transient basis in vivo; no genetic material is inserted or integrated into the chromosomes of the host. [0087]
  • The compositions may also contain other additives suitable for the selected mode of administration of the composition. The composition of the invention may also involve lyophilized polynucleotides, which can be used with other pharmaceutically acceptable excipients for developing powder, liquid or suspension dosage forms. See, e.g., Remington: The Science and Practice of Pharmacy, Vol. 2, 19[0088] th edition (1995), e.g., Chapter 95 Aerosols; and International Patent Publication No. W099/45966, the teachings of which are hereby incorporated by reference. Routes of administration for these compositions are combined, if desired, or adjusted.
  • These nucleic acid molecule-containing compositions can contain additives suitable for administration via any conventional route of administration. In some embodiments, administration is intravenous or directly into the bone marrow. In some embodiments, the composition of the invention is prepared for administration to human subjects in the form of, for example, liquids, powders, aerosols, tablets, etc. [0089]
  • The compositions of the present invention (whether host cell-containing or nucleic acid molecule-containing compositions), as described above, are not limited by the selection of the conventional, physiologically acceptable, carriers, adjuvants, or other ingredients useful in pharmaceutical preparations of the types described above. The preparation of these pharmaceutically acceptable compositions, from the above-described components, having appropriate pH isotonicity, stability and other conventional characteristics is within the skill of the art. [0090]
  • Suitable routes of administration include, but are not limited to, intranasal, oral, vaginal, rectal, parenteral, intradermal, transdermal, intramuscular, intraperitoneal, subcutaneous, intravenous and intraarterial. The appropriate route is selected depending on the nature of the composition used, and an evaluation of the age, weight, sex and general health of the patient and the components present in the immunogenic composition, and similar factors by an attending physician. [0091]
  • Similarly, suitable doses of compositions of this invention are readily determined by one of skill in the art, depending upon the condition being treated, the health, age and weight of the veterinary or human patient, and other related factors, and the other characteristics of the composition, e.g., nucleotide molecule, vector or host cell. In general, selection of the appropriate “effective amount” or dosage for the composition(s) of the present invention will also be based upon the form of the composition, the identity of the transgene and host cell, as well as the physical condition of the subject. The method and routes of administration and the presence of additional components in the compositions may also affect the dosages and amounts of the compositions. Such selection and upward or downward adjustment of the effective dose is within the skill of the art. The amount of composition required to produce a suitable response in the patient without significant adverse side effects varies depending upon these factors. Suitable doses are readily determined by persons skilled in the art. [0092]
  • For example, the amounts of nucleotide molecules in the DNA and vector compositions are selected and adjusted by one of skill in the art. In one embodiment, each dose will comprise between about 50 μg to about 1 mg of nucleic acid molecule, e.g., DNA plasmid, per mL of a sterile solution. Generally, a suitable dose where the vector is a viral vector is in the range of 10[0093] 3 to 1018, preferably about 105 to 1016 transducing units (TU) per dose, and most preferably, about 107 to 109 TU for an adult human having a weight of about 80 kg. Transducing Units (TU) represents the number of infectious particles and is determined by evaluation of transgene expression upon infection of host cells. Generally, when used for ex vivo therapy, the host cells are infected with 105 TU to 1010 TU viral vectors for each 101 to 1010 cells in a population. However, other suitable ex vivo dosing levels are readily selected by one of skill in the art. This dose is formulated in a pharmaceutical composition, as described above (e.g., suspended in about 0.01 mL to about 1 mL of a physiologically compatible carrier) and delivered by any suitable means.
  • The number of doses and the dosage regimen for the composition are also readily determined by persons skilled in the art. The intended therapeutic or prophylactic effect is conferred by a single dose of the composition or may require the administration of several doses, in addition to booster doses at later times. The dose is repeated, as needed or desired, daily, weekly, monthly, or at other selected intervals. [0094]
  • V. EXAMPLES OF THE INVENITON
  • As illustrated in the examples below, the methods of this invention were employed to modify the biological behavior of platelets by causing these hematopoietic lineage cells to ectopically express a protein of interest at a site of vascular injury. [0095]
  • A. Transgenic Animal Model [0096]
  • A murine urokinase-type plasminogen activator (u-PA) transgene was expressed transgenically in mice under the regulatory control of a platelet factor 4 promoter that directed expression to platelets. The resultant transgenic animals have altered platelet biology; i.e., they express and store u-PA selectively in their platelet alpha-granules, in the absence of systemic fibrinolysis and bleeding characteristic of transgenic over-expression of u-PA in the liver (Heckel, J. L. et al, 1990 [0097] Cell 62:447-456). These transgenic mice also had a characteristic bleeding diathesis similar to that seen in patients with Quebec Platelet Disorder (QPD) that is predominantly at the time of parturition and that can be controlled by tranexamic acid.
  • More importantly, these mice were resistant to the development of occlusive carotid arterial thrombi in the absence of systemic fibrinolysis. The mice rapidly lysed pulmonary venous thrombi or emboli. Moreover, transfusion of small numbers of urokinase-expressing platelets into wildtype recipients prevented formation of stable, occlusive carotid arterial thrombi. [0098]
  • Thus, this animal model confirms that ectopic expression of u-PA in platelets is the etiology of inherited QPD, provides new insights into the contribution of activated platelets to thrombus stability, and provides a new method for preventing inopportune thrombus development. The transgenic model shows that developing megakaryocytes are genetically altered in such a way that platelet function is effectively tipped from pro-thrombotic to anti-thrombotic. These examples provide a evidence that ectopic u-PA PA expression in platelets can be achieved. [0099]
  • B. Summary of the Examples [0100]
  • The present invention involves the discovery that ectopic expression of proteins in platelets are useful to favorably alter the hemostatic balance at sites of thrombosis. Fibrinolytic agents can be delivered to sites of incipient thrombus formation through selective storage in platelets, a new method to prevent thrombosis and hemorrhage. As demonstrated in the Examples below, ectopically expressed proteins carrying a signal peptide are stored within platelets and released specifically at sites of injury. By use of this method, one may alter the prothrombotic role of platelets as a means to modify pathological thrombus development. [0101]
  • The method of this invention has clinical application in situations where re-thrombosis or the prevention of extension of a previous thrombosis is desirable for an extended period of time. For example, transient expression of u-PA in megakaryocyte progenitors harvested from peripheral blood and treated with present-day retroviral gene-transfer vectors are a potent mechanism to prevent post-angioplasty restenosis for several weeks to months with low risk of long-term alteration of the recipient's stem cell genome. Untoward bleeding associated with the therapy can be controlled by treatment with an anti-fibrinolytic agent such as tranexamic acid. Lastly, the methods of this invention enable delivery of pro-hemostatic proteins to sites of vascular interruption in patients with diverse hemorrhagic disorders. [0102]
  • To determine whether developing megakaryocytes could be altered to express either anti-thrombotic or pro-hemostatic proteins, which would be stored in platelet and be released in a concentrated fashion directly at a site of injury, a transgenic model was developed. In this model, the expression of u-PA was directed to platelets of transgenic mice using the PF4 promoter, which is known to drive megakaryocyte-specific expression (Ravid, K. et al, 1991 [0103] Proc. Nat. Acad. Sci. U.S.A. 88:1521-1525). It was found that platelet u-PA was stored in platelets and was released within developing thrombi when platelets were activated. The u-PA was partially counter-balanced by PAI-1, which is present in platelet alpha-granules of both human and mice platelets (Ngo, T. H., and Declerck, P. J. 1999 Thromb. Haemost. 82:1510-1515). This method tipped the balance sufficiently to lessen the resistance of clots, especially in the arterial circulation where platelet activation is more intense, to commonly employed agents.
  • The “[0104] line #19” transgenic animals, described in the examples below, had u-PA mRNA, u-PA-like protease activity and u-PA protein detectable in their platelets. Several of the constituent platelet alpha-granular proteins, including vWF and fibrinogen, were partially degraded, probably due to plasmin-mediated proteolysis. However, unlike the previously described murine u-PA over-expressors driven by a liver-specific promoter (Heckel, J. L. et al, cited above), these animals did not demonstrate systemic fibrinolysis. Few of the transgenic adults developed spontaneous hemorrhage despite life-long expression of elevated levels of platelet-uPA. The platelet counts were normal, and the red cells had normal morphology. Plasma fibrinogen was intact and D-dimers were not detected in the plasma of these mice.
  • Megakaryocyte-expressed u-PA was not secreted to a considerable extent, but rather was preferentially stored in the circulating platelets, where it led to the digestion of alpha-granular proteins, as in the human disorder termed QPD. The u-PA within the alpha-granules was predominantly in the form of enzymatically active tcu-PA, as determined by immunoblotting. Platelets are believed to endocytose sufficient plasminogen from plasma (Holt, J. C., and Niewiarowski, S. cited above) into their alpha-granules to form plasmin, as suggested by the capacity of u-PA platelets to digest exogenously added Factor V. Plasmin formation is initiated by the low level of intrinsic activity of scu-PA (Lenich, C. et al, 1997 [0105] Blood 90:3579-3586) or by the activation of scu-PA by u-PAR (Higazi, A. et al, 1995 J. Biol. Chem. 270:17375-17380) or, less likely, by another platelet granular enzyme. It is likely that plasmin, once formed, then converts additional scu-PA to tcu-PA within the granules themselves, although the role of other proteases again cannot be excluded. Little tcu-PA was found as high molecular weight complexes with PAI-1. Murine platelets may contain less PAI-1 than human platelets, or the latent state of PAI-1 within the granules (Booth, N. A. et al, 1988 Brit. J. Haematol. 70:327-333; Lang, I. M. et al, 1992 Blood 80:2269-2274) may limit its binding to u-PA, or complex formation is inhibited by u-PAR (Higazi, A. A.-R et al, 1996 Blood 87:3545-3549), the pH or other components of the alpha-granules, or, more likely, most of the PAI-1 was degraded by the u-PA.
  • Aside from peripartum deaths, adult transgenic mice had few spontaneous mucosal bleeding episodes, had normal bleeding times, and exhibited normal platelet aggregation in vitro. However, these animals clearly were resistant to developing occlusive thrombi in a ferric chloride-induced carotid artery thrombosis model. Only 5% of the u-PA expressing animals developed complete arterial occlusion by the end of the study (60 minutes) as opposed to 85% of their wildtype littermates (Table 1). It appears that the thrombi that did form were far more friable and transitory in nature. Moreover, lysis of preformed, fibrin microemboli targeted to the lungs occurred far more rapidly in the u-PA expressing mice than in their wildtype counterparts. These data suggest that platelet activation initiated by fibrin contributes to thrombus growth or stability on the venous side of the circulation (Bdeir, K. et al, cited above) and that local release of u-PA causes rapid lysis of the nascent thrombi. [0106]
  • Recent studies using combined platelet glycoprotein IIb/IIIa inhibitors and agents (Murciano, J. C. et al, 2002 [0107] Am. J. Physiol. Lung Cell. Mol. Physiol. 282:L529-539), and studies using pharmacological inhibition of PAI-1 (Rupin, A. et al, 2001 Thromb. Haemost. 86:1528-1531) are consistent with these conclusions.
  • Transfusion of u-PA platelets equivalent to 10% of the recipient's platelet mass potently inhibited thrombus development in wildtype recipients. The ability of the transgenic platelets to disrupt thrombus development is more likely to be due to the released u-PA than to degradation of hemostatic alpha-granular proteins for several reasons. First, the near total loss of granular proteins, as in the Gray Platelet Disorder, is associated with little or no bleeding (Rao, A. K, 1998 [0108] Am. J. Med Sci. 316:69-76, 1998). Second, arterial thrombi are enriched with PAI-1. The absence of PAI-1 in mice results in a failure of thrombus formation in a chemically-induced carotid artery thrombosis model (Eitzman, D. T. et al, 2000 Blood 95:577-580). Third, transfusion of transgenic platelets into wildtype recipients that had been drinking water containing tranexamic acid reversed the effect of platelet-associated u-PA on thrombus formation (Table 2), notwithstanding the persistence of degraded alpha-granular proteins in the transfused platelets. These results suggest that it is the u-PA that is the major cause for the defective hemostasis in these mice, although a contribution from degraded alpha-granular proteins is by no means excluded.
  • Based on an analysis of the data generated by these examples similar to that of (Kahr, W. H. et al., cited above), [0109] line #19 platelets were estimated to contain ˜20 pg of u-PA per μg of total platelet protein. Moreover, there are striking similarities between the phenotype of the QPD patients and these transgenic mice. This provides additional support for the conclusion that this syndrome results from the ectopic expression of u-PA in platelets. Both are dominantly inherited disorders. Both QPD and the transgenic mice ectopically express u-PA in the platelets, which also contain many degraded alpha-granular proteins. In both QPD and the transgenic mice, the ectopic expression of u-PA is essentially confined to the platelets, and insufficient u-PA is secreted into the plasma to cause disseminated fibrinolysis. The phenotype of neither is improved by platelet infusions, but is improved by use of an anti-fibrinolytic agent, such as tranexamic acid.
  • In summary, these studies support the etiology of QPD as being due to ectopic expression of u-PA in platelets. Second, they highlight the importance of the balance between platelet-dependent coagulation and fibrinolysis during thrombus growth on venous as well as on the arterial side of the circulation. Third, they show that the ectopic expression of u-PA in platelets is a valid method for preventing untoward thrombus development. [0110]
  • The following examples are provided to illustrate construction and use of the recombinant vectors and compositions of the invention and do not limit the scope thereof. One skilled in the art will appreciate that although specific elements, reagents and conditions are outlined in the following examples, modifications can be made which are meant to be encompassed by the spirit and scope of the invention. [0111]
  • Example 1 Establishment of Transgenic Mice Expressing u-PA Message
  • A transgene was designed as illustrated schematically in FIG. 1. 1.2 kb of the 129 SV murine platelet factor 4 (PF4) proximal promoter region plus its 5′-untranslated region (UTR) (Zhang, C. et al, 2001 [0112] Blood 98:610-607) was PCR amplified with an artificial upstream Xba I site and a downstream Kpn I site added. The inventors found that this promoter could also drive megakaryocyte-specific expression of LacZ in transgenic mice (data not shown). This promoter was inserted in place of the albumin enhancer/promoter immediately upstream of a mouse u-PA minigene construct that contained a 3′-UTR and poly-adenylation sequence from the human growth hormone (hGH) gene. This construct was previously described in Heckel, J. L. et al, 1990 Cell 62:447-456, which is incorporated by reference herein.
  • A 10.2 kb Sac II fragment containing this construct was used to create transgenic mice by pronuclear injections following standard methods at the University of Pennsylvania Transgenic Mice Core Facility. Seven transgenic founders were obtained with copy numbers ranging from 1 to >20 copy per haploid genome. [0113]
  • Genomic DNA was isolated from mouse tails using a QIAamp™ DNA Mini Kit (Qiagen, Valencia, Calif.). Positive founder lines were detected by genomic Southern blot analysis (Tunnacliffe, A. et al, 1992 [0114] Blood 79: 2896-2900). Genomic Southern blot analysis was made of Bgl II-digested DNA from a wildtype animal and the three lines that had offspring with the fewest copy numbers of the transgene (Lines #13, #19 and #49). After digestion, genomic DNA was separated on a 1% (wt/vol) agarose gel. The probe used was the mouse PF4 proximal promoter from −680 to −360 bp upstream of the transcriptional start site (Zhang, C. et al, cited above), and the final probed membrane was exposed on a PhosphorImaging screen (Amersham Biosciences, Sunnyvale, Calif.). The 2.8 kb Bgl II fragment was detected for the transgene in the genomic Southern blot.
  • The transgene copy number per haploid genome was determined by phosphorimaging (not shown). The intensity of bands on film was analyzed using the Imagequant PhosphorImager software (Amersham Biosciences). The copy number was determined by comparing the intensity of the 2.8 kb transgene PF4 band to the native genomic 1.2 kb PF4 band. [0115]
  • Three male founders had offspring with the fewest copy number of transgene (i.e., line #13, [0116] copy number 1, line #19, copy number 2 and line #49, copy number 3), although the line #49 transgene did not transmit well (see below), and these mice were not available for most studies. All three female founders had high transgene copy number, and when-pregnant, died peripartum from uterine hemorrhage (see below). The final male founder was high copy number (>20) and never produced offspring.
  • Founder animals and their offspring were also characterized by genomic PCR analysis using a mouse PF4 5′-UTR (5′-CACTTAAGAGCCCTAGACCCATTTCC-3′; SEQ ID NO: 1) sense primer and a mouse u-PA exon 2 (5′-TTCAGAGTTTTT CACCACCAA-3′; SEQ ID NO: 2) antisense primer, which generates a 479 bp genomic and a 114 bp cDNA band. RT-PCR analysis of total platelet RNA was performed on lines #13, #19 and #49 for the 114 bp transgenic u-PA message that extended from the 5′-UTR of PF4 into the murine u-PA 2[0117] nd exon in their platelets. The 185 bp PF4 message was used as a positive control of the platelet nature of the RNA. PCR was performed to confirm founder lines using primers complementary to the mouse PF4 5′-UTR and exon 2 of the murine u-PA gene (data not shown).
  • Direct sequence analysis of this RT-PCR product for all three lines confirmed that the 1[0118] st intron was appropriately spliced out (data not shown). When RT-PCR was performed on multiple tissues, e.g., spleen, liver, lung, heart, kidney, adrenal, tongue, brain, and bone marrow, only bone marrow tested positive for the 114 bp transgenic-derived u-PA mRNA band. All samples showed the 249 bp HPRT positive control cDNA band. Immunohistochemistry of the bone marrow showed detectable u-PA only in morphologically recognizable megakaryocytes in the transgenic animals, but not in wild-type marrow.
  • All biological studies described below were done with transgenic animals that had been backcrossed at least 4 generations onto a C57BL6 background. Wildtype littermates served as controls. Cardiac or portal vein blood was drawn from mice into 1/10[0119] th volume of 3.8% sodium citrate. All studies were approved by the Animal Care and Use Committee of the Children's Hospital of Philadelphia.
  • Example 2 Transgenic u-PA Message Detection
  • The animals of Example 1 were then examined to determine whether they expressed u-PA in their platelets. Murine platelet RNA was isolated using RNA STAT-60™ (Tel-Test, Friendswood, Tex.) as previously described (Zhang, C. et al, cited above). Tissues (˜100 mg each) from these animals were collected, rinsed vigorously with saline, disaggregated in 500 μl of RNA STAT-60™, and RNA was isolated (Zhang, C. et al, cited above). Some platelet RNA samples were pretreated with DNase free RNase (1 U/10 μl reaction, Sigma, St. Louis, Mo.) or RNase free DNase (1 U/10 μl reaction, Life Technologies, Gaithersburg, Md.) for 1 hour at 37° C. [0120]
  • Reverse transcription was performed using the SuperScript II Reverse Transcriptase Kit™ (Life Technologies) as per the manufacturer's instructions. PCR amplification of the transgenic u-PA cDNA was accomplished using the two primers discussed above. Platelet-specific control RT-PCR primers for PF4 message were sense 5′-AATTCTCGGGATCTGGGT-3′ SEQ ID NO: 3 and antisense 5′-CTGGGCTCTAGACAGCAGT-3 SEQ ID NO: 4 (Eisman, R. et al, 1990 [0121] Blood 76:336-344), with an expected cDNA product of 185 bp. RT-PCR for the housekeeping gene human phosphoribosyltransferase (HPRT) used primers 5′-GCTGGTGAAAAGGACCT CT-3′ SEQ ID NO: 5 and 5′-CACAGGACTAGAACACCTGC-3′ SEQ ID NO: 6, with an expected cDNA product of 249 bp (Jackson, C. L. et al, 1984 Proc. Nat. Acad. Sci. U.S.A. 81:2945-2949).
  • Platelet murine transgenic u-PA cDNA band was isolated using a QIAkwik™ Gel Extraction Kit (Qiagen) and directly sequenced using an ABI 373A automated sequencer (ABI Instruments, Foster City, Calif.). [0122]
  • Example 3 Immunohistochemical Staining for Murine Urokinase
  • Spleen and bone marrow aspirates from wildtype and murine u-PA transgenic mice were stained for murine u-PA expression using a mouse monoclonal anti-murine u-PA antibody (A10, Molecular Innovations, Inc., Southfield, Mich.) as the primary antibody and a biotinylated anti-mouse immunoglobulin (ARK detections system, DAKO, Caprinteria, Calif.) as the secondary antibody. Specifically, formalin-fixed, paraffin-embedded 5 μm sections were deparaffinized in xylene and rehydrated. Endogenous peroxidase activity was quenched with 0. 9% peroxide in methanol for a total of 20 minutes. Slides were then treated with trypsin (1 mg/ml in PBS) for 10 minutes at 37° C. After incubation of the monoclonal anti-murine u-PA antibody (0.5 μg/ml) with biotinylated ARK reagent (according to manufacturer's instructions) for 15 minutes at room temperature, the slides were stained at room temperature for 2 hours. Slides were washed and incubated with Streptavidin-HRP (DAKO) for 15 minutes at room temperature. Slides were again washed DAB reagent (DAKO) was applied for 5 minutes at room temperature. Slides were counterstained with dilute (1:10) hematoxylin for 30 seconds. [0123]
  • Immunohistochemistry of the bone marrow in wildtype and [0124] line #19 mice showed detectable u-PA only in morphologically recognizable megakaryocytes m the transgenic animals, but not in wildtype marrow.
  • Example 4 u-PA Activity in the Platelets—Zymogram and Immunoblot Studies
  • A. Zymogram Studies [0125]
  • Platelet-rich plasma (PRP) was obtained as described in Zhang, cited above. Platelet counts were determined using a HemaVet counter (Triad Associates, Concord, Calif.). The platelets were pelleted at 800 g for 5 minutes and resuspended immediately in 1× NuPage Sample Buffer (Invitrogen, Carlsbad, Calif.). Total platelet protein and platelet releasates (0.5 to 10 μg/lane) were separated by size under non-reducing conditions on a 12% SDS-polyacrylamide gel (SDS-PAGE) with 0.4% nonfat dry milk, e.g., casein (Carnation Instant Skim Milk Powder, Nestle, Fulton, N.Y.) with or without supplemental 20 μg/ml human plasminogen (American Diagnostica, Greenwich, Conn.), and then renatured with 0.5% Triton X-100 in PBS, pH 7.4, for 1 hour as previously described (Heckel, J. L. et al, cited above). The gel was incubated at 37° C. for 3 hours. A control lane of 0.1 ng of human 2-chain urokinase (American Diagnostica) was included in each gel. [0126]
  • Zymograms of two separate preparations of lysates from [0127] line #19 and #13 platelets and wildtype platelets (10 μg/lane) reveal a prominent band likely representing tcu- at ˜45 kDa was seen with platelets from line #19 (data not shown). This is similar to what has been previously described for murine u-PA (Heckel, J. L. et al, cited above), consistent with the fact that mouse u-PA is not glycosylated. Line #13 and wildtype lanes had no detectable zones of lysis, although twice as much platelet protein was loaded compared to the #19 line lanes. Omission of plasminogen from the zymogram or the inclusion of the selective u-PA inhibitor amiloride in the zymogram (data not shown) abolished lysis induced by transgenic platelet lysates and the human tcu-PA control. These results are similar to those reported for QPD platelets, although the lower band in the human disorder was ˜10% as intense as the upper band, rather than ˜30% as seen in lysates from the transgenic mouse platelets.
  • B. Immunoblots [0128]
  • To confirm the presence of u-PA in the transgenic platelets, total platelet proteins were separated by size on a non-reducing Western gel and then immunoblotted with A10, an anti-murine u-PA mAb. Platelet immunoblots were performed as previously described (Zhang, C. et al, cited above), except that 4-8% gradient gels were used (NuPAGE Novex Bis-Tris Gels, Invitrogen, Carlsbad, Calif.). Gels were run under non-reducing conditions, except when studying platelet fibrinogen, where 1 μl of reducing agent (NuPAGE Sample Reducing Agent) was added per sample. Mouse u-PA was detected with a murine anti-mouse u-PA monoclonal antibody (A10, Molecular Innovations, Southfield, Mich.) added at a 1:75 dilution and detected with a biotinylated anti-mouse monoclonal antibody (Molecular Innovations) followed by peroxidase-conjugated streptavidin (StreptABComplex/HRP, DAKO, Carpinteria, Calif.). Murine vWF was detected using a 1:200 dilution of a horse radish peroxidase (HRP)-conjugated rabbit anti-human vWF polyclonal antibody (DAKO), and murine fibrinogen was detected using a 1:100 dilution of an HRP-conjugated goat anti-human fibrinogen polyclonal antibody (Rockland Immunochemicals, Gilbertsville, Pa.). [0129]
  • The Western blot of platelet lysates (10 μg/lane) for [0130] lines #19 and #13, wildtype, and renal extract (30 μg/lane) revealed a major doublet at ˜45 kDa in line #19; total platelet lysate that was not seen in wildtype littermate platelet lysate (data not shown). The major component in the doublet appears to be tcu-PA and co-migrates with the renal extract control. The higher, less intense band is likely to be single chain uPA (scu-PA) or an otherwise modified tcu-PA as previously described (Franco, P. et al, 1997 J. Cell Biol. 137:779-791) and migrates similar to a band in renal extract. Thus, as with QPD platelet lysates, the major form of u-PA in the transgenic platelets appears to be tcu-PA (Kahr, W. H. et al., cited above).
  • Also, as with QPD platelet lysates, a few high molecular weight species were observed that likely include covalent complexes between PAI-1 and both tcu-PA and low molecular weight proteolytic derivatives of u-PA (Jiang, Y et al. 1996 [0131] Blood 87:2775-2781). The typical low molecular weight u-PA species commonly observed in biological specimens (˜30 kD), was not a feature of platelet lysates. Rather, as observed with QPD platelets, a somewhat smaller low molecular weight u-PA species was observed in transgenic mouse platelet lysates. None of these mouse u-PA bands was detected when a comparable amount of platelet lysate from wildtype and line #13 mice were studied. This lack of detectable u-PA in Line #13 is consistent with the lack of u-PA activity seen in the above-described zymogram and in the clinical course of these animals (discussed below).
  • Example 5 Alpha-Granular Proteins
  • Several alpha-granular proteins, including vWF, fibrinogen and Factor V undergo proteolysis in QPD platelets. Proteolysis of these proteins has been attributed to the ectopically expressed u-PA (Kahr, W. H. et al., cited above). To determine whether the presence of mouse u-PA in platelets led to a similar expression of proteolytic activity and study the in vitro digestion of Factor V by platelet lysate, 10 ng of plasma-derived human Factor V (Rodney Camire, Children's Hospital of Philadelphia) was digested with either human tcu-PA (10 pg, American Diagnostica) or with 2.5 μg of total murine platelet lysate prepared from 1.3×10[0132] 6/μl platelets in the presence or absence of supplemental plasminogen (1 μg). The resultant digest was then separated by SDS-PAGE. Factor V was detected using a 1:100 dilution of an HRP-conjugated sheep anti-human Factor V polyclonal antibody (Affinity Biologicals, Inc., Hamilton, ON, Canada) as the primary antibody.
  • In a Western blot of platelet lysate (2.5 μg/lane) from lines #13 and #19 and wildtype mice immunoblotted with a rabbit anti-human vWF polyclonal antibody (not shown), vWF underwent extensive digestion in the [0133] line #19 platelets, compared with the intact high molecular weight vWF observed in the littermate wildtype sample. Platelet fibrinogen immunoblots of line #19 lysate consistently contained high molecular weight complexes that entered the gel poorly. These high molecular weight complexes are absent on a reduced gel, suggesting that they represent disulfide bond, cross-linked fibrinogen-derived products generated by u-PA. Consistent with the immunoblot for mouse u-PA, no degradation of vWF was seen in platelets from line #13 mice.
  • In a Western blot of reduced gel of platelet lysates as above or plasma (3 μg/lane for mouse and 6 μg/lane for human) proteins (not shown) both murine and human fibrinogen were detected using a goat anti-human fibrinogen polyclonal antibody. Platelet alpha-granular fibrinogen was also degraded in [0134] transgenic line #19, but not in line #13 and in wildtype platelets, as assessed on the non-reducing SDS-PAGE gel. In contrast, plasma fibrinogen was not degraded in line #19 mice.
  • This finding clearly contrasts with the systemic fibrinolysis that develops in the previously described u-PA over-expressing transgenic mice that were generated using a liver-specific promoter (Heckel, J. L. et al, cited above). Platelet fibrinogen immunoblots of [0135] line #19 lysate consistently contained high molecular weight complexes that entered the gel poorly. These high molecular weight complexes were absent on a reduced gel, suggesting that they represent disulfide-bonded, cross-linked, fibrinogen-derived products generated by u-PA and plasmin.
  • In another test to determine whether platelet releasate from the transgenic mice could proteolyze human Factor V in a manner similar to that seen in QPD, platelet lysate was incubated with human Factor V for up to 4 hours. Factor V (FV) (10 ng/lane) was digested with 2.5 μg/lane of the indicated platelet lysate or human u-PA (5 μg/lane) at room temperature, in the absence or presence of 100 μg/lane of plasminogen per lane. [0136]
  • Platelet lysate from [0137] line #19 digested the Factor V in a rapid fashion, with degradation nearly complete by 4 hours, giving a similar pattern to that seen with exogenously added human u-PA (data not shown). Platelets from wildtype littermates also degraded Factor V, but at a much slower rate. The addition of exogenous plasminogen to the platelet releasate enhanced Factor V digestion by line #19 platelet lysate, but had little effect on the rate of digestion by wildtype platelets. This suggests that the amount of plasminogen available in alpha-granules in the transgenic line #19 platelets is rate limiting for maximal protein degradation.
  • Example 6 Clinical Course and Hematologic Studies
  • A. Clinical Course [0138]
  • [0139] Fewer line #19 and #49 transgenic mice were born than expected. Assuming that 50% of the offspring from a cross between a hemizygous animal and a wildtype animal should be transgenic, line #19 showed a 68% mortality rate (57 transgenic animals vs. 175 wildtype littermates at weaning) and line #49 showed an 85% mortality rate (2 transgenic and 12 wildtype). Line #13 had a very low mortality rate, consistent with little platelet u-PA expression (71 transgenic and 80 wildtype). At day 16.5 of gestation, line #19 embryos looked normal and appeared at the expected frequency (10 transgenic and 11 wildtype), indicating that transgenic animals were lost peripartum Surviving transgenic animals were normal for weight and growth, although occasional spontaneous deaths occurred among the adults secondary to subcutaneous or internal hemorrhage. Autopsies on two such adult line #19 mice expressing u-PA in their platelets showed examples of spontaneous hemorrhage: one mouse had free blood in the opened peritoneum and the other showed free blood filling a small intestinal loop.
  • However, when ten [0140] line #19 animals and ten littermate wildtype controls were specifically observed for >11 months, no deaths were observed in either group, suggesting a low rate of death in the line #19 animals.
  • As noted above, none of the female founder lines survived birthing. This was also true for [0141] pregnant line #19 and #43 transgenic females. In line #19, none of nine pregnant females survived giving birth, exsanguinating from uterine bleeding at birth (data not shown). Litter sizes were normal, with embryos containing equal numbers of wildtype and transgenic animals, and all embryos appeared normal in size (data not shown).
  • Three [0142] pregnant line #19 females were given tranexamic acid (20 mg/ml), an inhibitor of plasminogen activator, in their drinking water (Hattori, N. et al, 2000 J. Clin. Invest. 106:1341-50) during the last week of pregnancy and all survived. One lost her pups at birth, and the other two had small litters, consisting of 3 pups each (2 of the 6 were transgenic).
  • B. Blood Counts [0143]
  • Complete blood counts were performed on [0144] line #19 adult mice and wildtype littermate controls (n=10, each). Blood counts were measured using a Hemavet 850 (CDC Technologies, Inc, Oxford, Conn.) calibrated for murine blood. Dried blood smears were stained using Wright-Giemsa reagent (EM Science, Gibson, N.J.) and the red cell and platelet morphology was examined. Fibrin D-dimers were measured in plasma samples obtained according to manufacturer's instructions (Asserachrom D-Di, American Bioproducts Co., Diagnostica Stago, Asnieres, France).
  • Results of the counts showed nearly identical platelet counts and hemoglobin levels for the [0145] line #19 and wildtype controls (data not shown). Peripheral blood smears were normal, including normal platelet numbers and appearance and no evidence of red cell schistocytes or spherocytes (data not shown).
  • C. Platelet Aggregation [0146]
  • Platelet aggregation studies were performed as described previously (Kowalska, M. A. et al, 2000 [0147] Blood 96:50-57). Aggregation and dense granule ATP release were measured in a lumi-aggregometer (Chrono-Log, Havertown, Pa.). Agonists studied included collagen (1-5 μg/ml), ADP (1-5 μM), epinephrine (50 μM), and thrombin (0.1-1 U/ml) (Bachem Torrance, Calif.). Marrow samples from wildtype and transgenic mice were stained for mouse u-PA expression using the murine anti-mouse u-PA A10 primary antibody as described previously (Zhang, C. et al, cited above).
  • Platelet aggregation in response to collagen (1-5 μg/ml), ADP (1-5 μM), epinephrine (50 μM), and thrombin (0.1-1 U/ml) were also normal (data not shown). D-dimer measurements were negative in the [0148] line #19 mice (data not shown). Together with the measurements of plasma fibrinogen discussed above, these studies confirm that line #19 mice do not exhibit systemic fibrinolysis.
  • Example 7 Carotid Artery Thrombosis Model
  • Bleeding times were normal in [0149] line #19 mice when compared with wildtype littermate controls (5.8±3.2 mins versus 5.1±3.4 (n=7, each), respectively), but this test has previously proven to be an insensitive measurement of thrombotic tendency (Mayadas, T. N. et al, 1993 Cell 74:541-554). Therefore, the carotid artery injury thrombosis model was employed. This model has been used successfully to demonstrate a bleeding diathesis in diverse mouse backgrounds. This approach also permits study of the effect of ectopic expression of u-PA in platelets on thrombus development and stability.
  • Ferric chloride-induced arterial injury was performed according to published procedures (Fay, W. P et al, 1994 cited above; Fay, W. P. et al, 1999 [0150] Blood 93:1825-1830) in 6-8 week old animals. Briefly, the right common carotid artery was exposed by blunt dissection, and a miniature Doppler flow probe (Model 0.5VB, Transonic Systems, Ithaca, N.Y.) was positioned around the artery. A 1×2 mm2 strip of Number 1 Whatman filter paper (Fisher Scientific, Pittsburgh, Pa.) soaked in 10% ferric chloride was then applied to the adventitial surface of the artery for 2 min. The field was flushed with saline, and blood flow was continuously monitored for 30 minutes. The time to the initial complete occlusion and the presence or absence of arterial occlusion at 30 min was recorded.
  • To study the effects of a platelet transfusion, 1.2-1.5×10[0151] 8 gel-filtered platelets in 300 μl of gel-filtering buffer (4 mM NaH2PO4; 5mM Hepes; 137 mM NaCl; 2.6 mM KCl; 5 mM glucose; 1 mM MgCl2) was prepared as previously described (Kowalska, M. A. et al, cited above) and infused into the left jugular vein immediately before the ferric chloride patch was applied. Platelets were used within 2 hours of collection. Total blood counts were measured immediately before and 2 minutes after platelet infusion.
  • Unlike their wildtype littermates, few transgenic animals expressing platelet u-PA formed completely occlusive coronary artery thrombi after ferric chloride-induced injury (Table 1), and those that occluded tended to reopen rapidly. At 30 minutes, only 5% of occlusive thrombi formed in the u-PA mice remained as opposed to the >85% of thrombi formed in controls (Table 1). Supplementing the transgenic animals with 20 mg/ml tranexamic acid, a small molecule inhibitor of plasminogen activation, reversed this protection from occlusive coronary artery thrombosis. Line #13 mice occluded normally. [0152]
    TABLE 1
    Ferric chloride thrombosis model.
    Time to initial Occluded at
    Founder line occlusion (mins) Initial occlusion (5) 30 mins (%)
    WT  8.8 ± 4.3 31/34 (91%) 29/34 (85%)
    #13  6.8 ± 3.3  9/10 (90%)  9/10 (90%)
    #19 10.3 ± 4.8  4/20 (20%)  1/20 (5%)
    WT + TA  8.3 ± 1.1  5/5 (100%)  4/5 (80%)
    #19 + TA  9.4 ± 1.6  5/5 (100%)  5/5 (100%)
  • Example 8 Pulmonary Microemboli Model
  • The following experiment was performed to determine whether the u-PA containing platelets are also effective on the venous side of the circulation and would lead to rapid dissolution of pulmonary microemboli. Previously, the lysis of [0153] 125I-labeled, fibrin microparticles was shown to depend on u-PA expression in the recipient mice as an intravenous infusion of u-PA into u-PA null mice normalized the rate of fibrinolysis (Bdeir, K. et al, cited above). This study determined whether u-PA-containing platelets similarly enhance fibrin clot breakdown.
  • [0154] 125I-labeled human microemboli, 1.5-3.5μ3 in size, were prepared as previously described (Bdeir, K. et al, 2000 Blood 96:1820-1826). Transgenic and wildtype animals were injected with these particles within 48 hours of preparation. At various time points (2-60 minutes) the animals were euthanized, the lungs removed, washed free of blood, and the amount of 125I activity measured using a ZM Coulter Counter (Coulter Electronics, Hialeah, Fla.). In other experiments, autoradiograms of lungs were taken from wildtype and transgenic mice 30 minutes after injection of the microemboli by exposing the lungs to X-OMAT film (Kodak, Rochester N.Y.).
  • FIG. 2 shows residual labeled microemboli remaining in the lungs of wildtype mice over time and residual radioactivity in the lungs of [0155] line #19 mice studied in parallel (n=6). Fibrinolysis was dramatically accelerated in line #19 mice compared to wildtype controls. Differences were already evident at two minutes. By 10 minutes, at a time when there was minimal or no fibrinolysis in wildtype mice, >80% of the clot burden had already been cleared from the lungs of the line #19 mice.
  • Example 9 Platelet Transfusion/Carotid Artery Studies
  • Based on the prior experiments that showed that platelet-expressed u-PA both prevented untoward arterial thrombosis and accelerated lysis of venous emboli, this experiment investigated whether transfused u-PA-containing platelets would interfere with thrombus development and stability in wildtype animals. [0156]
  • Platelet transfusions equivalent to ˜10% of the total circulating platelets in an animal were given from [0157] line #19 animals to wildtype littermate controls prior to exposing the carotid artery to a ferric chloride injury. Surprisingly, wildtype animals that received u-PA-containing platelets were protected against the development of thrombosis to the same extent as line #19 mice themselves (Table 2). Transfusion of buffer only or wildtype platelets into wildtype animals did not prevent the development of occlusive thrombi. As anticipated from the above data, giving wildtype platelets to line #19 mice did not confer resistance to arterial thrombosis.
    TABLE 2
    Platelet transfusion ferric chloride thrombosis model.
    Founder Line Time to initial Initial occlusion Occluded at
    Recipient Donor occlusion (mins) (%) 30 mins (%)
    WT Buffer  9.3 ± 1.2 5/5 (100%) 5/5 (100%)
    WT WT  8.2 ± 1.4 5/5 (100%) 5/5 (100%)
    WT # 19 12.5 1/10 (10%) 1/10 (0%)
    WT/TA # 19  8.3 ± 1.9 5/5 (100%) 4/5 (80%)
    WT # 19/TA 10.5, 22.5 2/3 (67%) 0/3 (0%)
    #19 WT 0/3 (0%) 0/3 (0)%
  • To test whether protection from thrombosis seen in wildtype animals receiving [0158] line #19 platelets was attributable to the released u-PA or to the released degraded alpha-granular proteins, wildtype animals were placed on drinking water containing tranexamic acid. A week later, carotid artery injury was performed on these wildtype mice after they had received an infusion of platelets from the line #19 animals. It was anticipated that platelet-associated plasminogen activation would be inhibited in tranexamic acid-treated recipients leading to loss of protection from thrombosis if released u-PA is the primary mechanism by which line #19 platelets interfere with normal thrombus development. As hypothesized, recipient wildtype animals that had received tranexamic acid lost their resistance to arterial occlusion notwithstanding transfusion of the transgenic platelets, suggesting that the released u-PA contributes significantly to the observed anti-thrombotic effect of the line #19 platelets. The converse experiment, infusing platelets from a line #19 animal that had been on tranexamic acid for one week to wildtype littermates showed that the infused platelets conferred resistance to arterial thrombosis. These data support the conclusion that line #19 platelets interfere with thrombosis in wildtype animals primarily because ectopically expressed u-PA is released from the donor platelets.
  • Applicants specifically incorporate by reference D. Kufrin et al, 2003 [0159] Blood, 102(3):926-933. All publications cited in this specification are incorporated herein by reference. While the invention has been described with reference to a particularly preferred embodiment, it will be appreciated that modifications can be made without departing from the spirit of the invention. Such modifications are intended to fall within the scope of the appended claims.
  • 1 6 1 26 DNA Artificial mouse PF4 5′UTR sense primer 1 cacttaagag ccctagaccc atttcc 26 2 21 DNA Artificial mouse u-PA exon 2 antisense primer 2 ttcagagttt ttcaccacca a 21 3 18 DNA Artificial mouse PF4 5′UTR sense primer 3 aattctcggg atctgggt 18 4 19 DNA Artificial mouse u-PA exon 2 antisense primer 4 ctgggctcta gacagcagt 19 5 17 DNA Artificial primer 5 gctggtgaaa aggacct 17 6 20 DNA Artificial primer 6 cacaggacta gaacacctgc 20

Claims (35)

1. A recombinant nucleic acid molecule comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of said transgene in a hematopoietic stem cell, a progenitor cell or cell differentiated therefrom.
2. The molecule according to claim 1, wherein said differentiated cell is a secretory cell.
3. The molecule according to claim 1, wherein said progenitor cell is selected from the group consisting of common lymphoid progenitor, common myeloid progenitor, megakaryotic/erythrocyte progenitor and granulocytes/macrophage progenitor.
4. The molecule according to claim 3, wherein said cells differentiated from megakaryotic/erythrocyte progenitor cells are selected from the group consisting of platelets, megakaryocytes and erythrocytes.
5. The molecule according to claim 3, wherein said cells differentiated from granulocyte/macrophage progenitor cells are selected from the group consisting of neutrophils, eosinophils, monocytes, basophils and immature dendritic cells.
6. The molecule according to claim 3, wherein said cells differentiated from said lymphoid progenitors are natural killer cells.
7. The molecule according to claim 5, wherein said cells differentiated from said monocytes are selected from the group consisting of mast cells, macrophages and dendritic cells.
8. The molecule according to claim 1, wherein said nucleic acid molecule is a viral or non-viral vector.
9. The molecule according to claim 1, wherein said regulatory sequence is a platelet-specific promoter.
10. The molecule according to claim 9, wherein said promoter is selected from the group consisting of the Platelet factor 4 promoter, the glycoprotein IIb promoter, the glycoprotein IIIa promoter, and the glycoprotein VI promoter.
11. The molecule according to claim 1, wherein said regulatory sequence is a neutrophil-specific promoter.
12. The molecule according to claim 11, wherein said promoter is selected from the group consisting of DEFA1 human neutrophil alpha defensin promoter, the DEFA2 human neutrophil alpha defensin promoter, DEFA3 human neutrophil alpha defensin promoter, and the DEFA4 human neutrophil alpha defensin promoter.
13. The molecule according to claim 1, wherein said regulatory sequence is a natural killer cell-specific promoter.
14. The molecule according to claim 13, wherein said promoter is the human perforin gene promoter.
15. The molecule according to claim 1, wherein said regulatory sequence is an eosinophil-specific promoter.
16. The molecule according to claim 15, wherein said promoter is selected from the group consisting of the human eotaxin gene promoter and the eosinophil peroxidase gene promoter.
17. The molecule according to claim 1, wherein said regulatory sequence is an erythrocyte-specific promoter.
18. The molecule according to claim 17 wherein said promoter is the human RhD gene promoter.
19. A hematopoietic stem cell transformed, transduced, infected or transfected with a nucleic acid molecule of claim 1.
20. A host cell differentiated from a hematopoietic stem cell transformed, transduced, infected or transfected with a nucleic acid molecule of claim 1.
21. The host cell according to claim 20, selected from the group consisting of a common lymphoid progenitor cell, a common myeloid progenitor cell, a megakaryotic/erythrocyte progenitor cell, a granulocyte/macrophage progenitor cell, platelets, megakaryocytes, neutrophils, eosinophils, monocytes, basophils, dendritic cells, mast cells, macrophages, dendritic cells, erythrocytes, and natural killer cells.
22. A platelet transformed, transduced, infected or transfected with a nucleic acid molecule comprising a first sequence encoding a transgene under the control of regulatory sequences that direct expression of said transgene in said platelet.
23. A method for generating a modified hematopoietic stem cell, modified progenitor cell or a modified cell differentiated from said stem cell or progenitor cell comprising the step of transferring a nucleic acid molecule of claim 1 into said cell via transformation, transduction, infection or transfection.
24. The method according to claim 23, further comprising the step of harvesting said stem cells or progenitor cells of the hematopoietic lineage from bone marrow of a mammal prior to said transferring step, wherein said transferring step occurs in vitro or ex vivo.
25. The method according to claim 23, further comprising the step of harvesting said cells differentiated from said hematopoietic stem cells or progenitor cells from peripheral blood of a mammal prior to said transferring step, wherein said transferring step occurs in vitro or ex vivo.
26. A method for treating a disorder in a mammal comprising the steps of
delivering to said mammal a recombinant nucleic acid molecule comprising a first sequence comprising a transgene encoding a product under the control of regulatory sequences that direct expression of the product of said transgene in a hematopoietic stem cell, a progenitor cell of the hematopoietic lineage, or a cell differentiated therefrom.
27. The method according to claim 26, wherein a differentiated cell produces said product at a suitable site in said mammal.
28. The method according to claim 26, wherein delivering step comprises
(a) harvesting said stem cells or progenitor cells from bone marrow of said mammal; and
(b) transferring said nucleic acid molecule into said cells.
29. The method according to claim 28 further comprising reinfusing said cells into the bone marrow of said mammal.
30. The method according to claim 26, wherein delivering step comprises
(a) harvesting said differentiated cells from peripheral blood of said mammal; and
(b) transferring said nucleic acid molecule into said differentiated cells.
31. The method according to claim 30, further comprising reinfusing said cells into the blood of said mammal.
32. The method according to claim 26, wherein said delivering step comprises administering said nucleic acid molecule directly into the mammal.
33. A method for treating or preventing thrombus formation in a mammal comprising delivering to a mammalian patient a recombinant nucleic acid molecule comprising a first sequence comprising a transgene encoding a fibrinolytic protein under the control of regulatory sequences that direct expression of the product of said transgene in a platelet.
34. The method according to claim 33, wherein said fibrinolytic protein is selected from the group consisting of u-PA, Factor VIIa, Factor VIII, Factor IX and fibrinogen.
35. The method according to claim 33, wherein said nucleic acid molecule is present in a platelet and said delivering step comprises administering to said patient said platelet.
US10/701,331 2002-11-05 2003-11-04 Delivery vehicle for recombinant proteins Abandoned US20040126885A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/701,331 US20040126885A1 (en) 2002-11-05 2003-11-04 Delivery vehicle for recombinant proteins
US11/581,559 US7939063B2 (en) 2002-11-05 2006-10-16 Delivery vehicle for recombinant proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42423402P 2002-11-05 2002-11-05
US10/701,331 US20040126885A1 (en) 2002-11-05 2003-11-04 Delivery vehicle for recombinant proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/581,559 Division US7939063B2 (en) 2002-11-05 2006-10-16 Delivery vehicle for recombinant proteins

Publications (1)

Publication Number Publication Date
US20040126885A1 true US20040126885A1 (en) 2004-07-01

Family

ID=32659305

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/701,331 Abandoned US20040126885A1 (en) 2002-11-05 2003-11-04 Delivery vehicle for recombinant proteins
US11/581,559 Active 2026-05-18 US7939063B2 (en) 2002-11-05 2006-10-16 Delivery vehicle for recombinant proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/581,559 Active 2026-05-18 US7939063B2 (en) 2002-11-05 2006-10-16 Delivery vehicle for recombinant proteins

Country Status (1)

Country Link
US (2) US20040126885A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050100536A1 (en) * 2002-04-13 2005-05-12 Allan Mishra Compositions and minimally invasive methods for treating incomplete tissue repair
WO2005065242A2 (en) * 2003-12-29 2005-07-21 Am Biosolutions Method of treating cancer using platelet releasate
US20050186193A1 (en) * 2002-04-13 2005-08-25 Allan Mishra Method and kit for treatment of tissue injury
US20060127382A1 (en) * 2004-08-20 2006-06-15 Allan Mishra Particle/cell separation device and compositions
WO2006103569A2 (en) * 2005-03-18 2006-10-05 Innate Pharma Expression vectors and methods for obtaining nk cell specific expression
US20070110737A1 (en) * 2003-12-29 2007-05-17 Allan Mishra Compositions and method for decreasing the appearance of skin wrinkles
US20070122906A1 (en) * 2003-12-29 2007-05-31 Allan Mishra Method of culturing cells
EP1958648A1 (en) * 2005-10-28 2008-08-20 Dnavec Corporation Therapeutic method for blood coagulation disorder
US20080317761A1 (en) * 2004-04-28 2008-12-25 The Trustees Of The University Of Pennsylvania Peptide-Mediated Protein Transduction Into Cells of the Hematopoietic Lineage
US20090226830A1 (en) * 2004-11-22 2009-09-10 Hodogaya Chemical Co., Ltd. Electrophotographic photosensitive body
US20110070154A1 (en) * 2008-08-13 2011-03-24 Hyde Roderick A Artificial cells
US8119128B2 (en) 2004-10-29 2012-02-21 The Regents Of The University Of Colorado, A Body Corporate Antibodies that bind urokinase-type plasminogen activator and epitopes therefor
US9351999B2 (en) 2008-10-07 2016-05-31 Bioparadox, Llc Use of platelet rich plasma composition in the treatment of cardiac conduction abnormalities
US10214727B2 (en) 2013-06-04 2019-02-26 Allan Mishra Platelet-rich plasma compositions and methods of preparation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010048625A1 (en) * 2008-10-24 2010-04-29 University Of Utah Research Foundation Active agents, compositions, and methods for inhibiting and reversing platelet function

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135736A (en) * 1988-08-15 1992-08-04 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5489743A (en) * 1993-01-19 1996-02-06 Amgen Inc. Transgenic animal models for thrombocytopenia
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5804604A (en) * 1989-12-21 1998-09-08 Biogen, Inc. Tat-derived transport polypeptides and fusion proteins
US6348185B1 (en) * 1998-06-20 2002-02-19 Washington University School Of Medicine Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992006190A1 (en) 1990-10-01 1992-04-16 Massachusetts Institute Of Technology Transgenic model for selective targeting of gene products with a megakaryocyte promoter
US7060673B2 (en) 2001-09-27 2006-06-13 Lg Household And Health Care Ltd. Fusion peptide of human parathyroid hormone derived peptide and tat peptide, preparation thereof, and skin slimming cosmetic composition comprising the same
AU2002356824A1 (en) 2001-10-19 2003-05-06 The Scripps Research Institute Novel methods for introducing molecules into cells and vectors and compositions for use in such methods

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135736A (en) * 1988-08-15 1992-08-04 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5670617A (en) * 1989-12-21 1997-09-23 Biogen Inc Nucleic acid conjugates of tat-derived transport polypeptides
US5674980A (en) * 1989-12-21 1997-10-07 Biogen Inc Fusion protein comprising tat-derived transport moiety
US5747641A (en) * 1989-12-21 1998-05-05 Biogen Inc Tat-derived transport polypeptide conjugates
US5804604A (en) * 1989-12-21 1998-09-08 Biogen, Inc. Tat-derived transport polypeptides and fusion proteins
US5489743A (en) * 1993-01-19 1996-02-06 Amgen Inc. Transgenic animal models for thrombocytopenia
US6348185B1 (en) * 1998-06-20 2002-02-19 Washington University School Of Medicine Membrane-permeant peptide complexes for medical imaging, diagnostics, and pharmaceutical therapy

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070264245A1 (en) * 2002-04-13 2007-11-15 Allan Mishra Compositions and minimally invasive methods for treating incomplete tissue repair
US9320762B2 (en) 2002-04-13 2016-04-26 Allan Mishra Compositions and minimally invasive methods for treating incomplete tissue repair
US20050186193A1 (en) * 2002-04-13 2005-08-25 Allan Mishra Method and kit for treatment of tissue injury
US8163277B2 (en) 2002-04-13 2012-04-24 Allan Mishra Kits for treating dysfunction of cardiac muscle
US20050100536A1 (en) * 2002-04-13 2005-05-12 Allan Mishra Compositions and minimally invasive methods for treating incomplete tissue repair
US7608258B2 (en) 2002-04-13 2009-10-27 Allan Mishra Method for treatment of tendinosis using platelet rich plasma
US7314617B2 (en) 2002-04-13 2008-01-01 Allan Mishra PRP composition and minimally invasive method for treating myocardial infarction
US20100135969A1 (en) * 2003-12-29 2010-06-03 Allan Mishra Method of treating cancer using platelet releasate
US7678780B2 (en) 2003-12-29 2010-03-16 Allan Mishra Method of treating cancer using platelet releasate
US20070184029A1 (en) * 2003-12-29 2007-08-09 Am Biosolutions Method of treating cancer using platelet releasate
US20070110737A1 (en) * 2003-12-29 2007-05-17 Allan Mishra Compositions and method for decreasing the appearance of skin wrinkles
WO2005065242A2 (en) * 2003-12-29 2005-07-21 Am Biosolutions Method of treating cancer using platelet releasate
US20070122906A1 (en) * 2003-12-29 2007-05-31 Allan Mishra Method of culturing cells
WO2005065242A3 (en) * 2003-12-29 2006-06-22 Am Biosolutions Method of treating cancer using platelet releasate
US20080317761A1 (en) * 2004-04-28 2008-12-25 The Trustees Of The University Of Pennsylvania Peptide-Mediated Protein Transduction Into Cells of the Hematopoietic Lineage
US20060127382A1 (en) * 2004-08-20 2006-06-15 Allan Mishra Particle/cell separation device and compositions
US7462268B2 (en) 2004-08-20 2008-12-09 Allan Mishra Particle/cell separation device and compositions
US20090092679A1 (en) * 2004-08-20 2009-04-09 Allan Mishra Particle/cell separation device and compositions
US8142993B1 (en) 2004-08-20 2012-03-27 Allan Mishra Method of preparing neutrophil-depleted platelet-rich plasma
US8119128B2 (en) 2004-10-29 2012-02-21 The Regents Of The University Of Colorado, A Body Corporate Antibodies that bind urokinase-type plasminogen activator and epitopes therefor
US20090226830A1 (en) * 2004-11-22 2009-09-10 Hodogaya Chemical Co., Ltd. Electrophotographic photosensitive body
WO2006103569A2 (en) * 2005-03-18 2006-10-05 Innate Pharma Expression vectors and methods for obtaining nk cell specific expression
WO2006103569A3 (en) * 2005-03-18 2007-03-15 Innate Pharma Sa Expression vectors and methods for obtaining nk cell specific expression
EP1958648A4 (en) * 2005-10-28 2009-12-30 Dnavec Corp Therapeutic method for blood coagulation disorder
US20090148425A1 (en) * 2005-10-28 2009-06-11 Tsukasa Ohmori Therapeutic method for blood coagulation disorder
EP1958648A1 (en) * 2005-10-28 2008-08-20 Dnavec Corporation Therapeutic method for blood coagulation disorder
US20110070154A1 (en) * 2008-08-13 2011-03-24 Hyde Roderick A Artificial cells
US9351999B2 (en) 2008-10-07 2016-05-31 Bioparadox, Llc Use of platelet rich plasma composition in the treatment of cardiac conduction abnormalities
US11638548B2 (en) 2008-10-07 2023-05-02 Blue Engine Biologies, LLC Use of platelet rich plasma composition in the treatment of cardiac conduction abnormalities
US10214727B2 (en) 2013-06-04 2019-02-26 Allan Mishra Platelet-rich plasma compositions and methods of preparation

Also Published As

Publication number Publication date
US20070031392A1 (en) 2007-02-08
US7939063B2 (en) 2011-05-10

Similar Documents

Publication Publication Date Title
US7939063B2 (en) Delivery vehicle for recombinant proteins
US7351813B2 (en) Liver-specific gene expression cassettes, and methods of use
EP0644944B1 (en) Adeno-associated virus with inverted terminal repeat sequences as promoter for the in vivo transfer of a functional cftr gene
Miao et al. High-level factor VIII gene expression in vivo achieved by nonviral liver-specific gene therapy vectors
JP4854519B2 (en) Use of aerosolized antithrombin in the treatment of acute lung injury
Lieber et al. Adenovirus-mediated urokinase gene transfer induces liver regeneration and allows for efficient retrovirus transduction of hepatocytes in vivo.
JPH09503740A (en) Inhibition of selected tumor growth by recombinant viral vectors
US5980886A (en) Recombinant vectors for reconstitution of liver
US20120073002A1 (en) Prevention and treatment of blood coagulation-related disases
JP2023002657A (en) Genetically modified mesenchymal stem cells expressing α1-antitrypsin (AAT)
JP2011162555A (en) Method for preventing formation of fibrin clot in lung tissue using aerosolized anticoagulant
ES2256266T3 (en) THROMBOMODULIN ANALOGS AND THEIR PHARMACEUTICAL USE.
MXPA01006741A (en) Methods for reducing adverse side effects associated with cellular transplantation.
US5955441A (en) Genetic inhibition of complement mediated inflammatory response
US20040009158A1 (en) Promotion of neovascularization using bone marrow-derived endothelial-progenitor cells
US20100144620A1 (en) Complementation of factor xi deficeincy by factor v mutants
US9808505B2 (en) Suppression of malignant mesothelioma by overexpression or stimulation of endothelial protein C receptors (EPCR)
Almus et al. Regulation of factor VIIa/tissue factor functional activity in an umbilical vein model.
JPH10508187A (en) Transplant and gene therapy for inflammation or thrombosis
MXPA01012049A (en) Gene therapy of pulmonary disease.
JP2001522601A (en) Nuclear targeting using bacterial proteins
JP2001522808A (en) Use of protein H as a cytostatic
Martin et al. Thrombin does not alter vascular hyporeactivity in models of endotoxin-induced septic shock in rats
WO2000063369A2 (en) Gene therapy
Chen et al. Muscle injection of rAAV/mFIX to secrete clotting factor IX corrects the hemorrhagic tendencies in hemophilia B mice

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:CHILDREN'S HOSPITAL OF PHILADELPHIA;REEL/FRAME:021121/0170

Effective date: 20040116

AS Assignment

Owner name: THE CHILDREN'S HOSPITAL OF PHILADELPHIA, PENNSYLVA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PONCZ, MORTIMER;REEL/FRAME:025877/0199

Effective date: 20110201

Owner name: THE TRUSTEES OF THE UNIVERSITY OF PENNSYLVANIA, PE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CINES, DOUGLAS B;REEL/FRAME:025877/0148

Effective date: 20070510

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MA

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:CHILDREN'S HOSPITAL OF PHILADELPHIA;REEL/FRAME:051321/0568

Effective date: 20191111