WO1993014188A1 - Targeted virus - Google Patents

Targeted virus Download PDF

Info

Publication number
WO1993014188A1
WO1993014188A1 PCT/US1993/000072 US9300072W WO9314188A1 WO 1993014188 A1 WO1993014188 A1 WO 1993014188A1 US 9300072 W US9300072 W US 9300072W WO 9314188 A1 WO9314188 A1 WO 9314188A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
virus
human
envelope protein
gene
Prior art date
Application number
PCT/US1993/000072
Other languages
French (fr)
Inventor
Michael F. Clarke
Stephen G. Emerson
Bernhard O. Palsson
Original Assignee
The Regents Of The University Of Michigan
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 The Regents Of The University Of Michigan filed Critical The Regents Of The University Of Michigan
Publication of WO1993014188A1 publication Critical patent/WO1993014188A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C07K14/54Interleukins [IL]
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16045Special targeting system for viral vectors
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/60Vectors comprising as targeting moiety peptide derived from defined protein from viruses
    • C12N2810/6045RNA rev transcr viruses
    • C12N2810/6054Retroviridae
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates

Definitions

  • the present invention relates to a virus which has an chimeric envelope protein on its surface that effects the specific binding of the virus to a cell of interest.
  • the present invention also relates to a method of producing such a virus and methods of transducing cells and gene therapy utilizing such a virus.
  • the present invention further relates to stem and/or progenitor cells which have been transduced with such a virus and a packaging cell line which produces such a virus.
  • the principle underlying gene therapy is to, rather than deliver doses of pharmacologic molecules, deliver a functional gene whose RNA or protein product will produce the desired biochemical effect in the target cell or tissue.
  • gene therapy can in principle be permanent within an individual, as the gene will continue to function in the target cells and their progeny.
  • the hematopoietic system is an ideal choice as a delivery system for gene therapy.
  • Hematopoietic cells are readily accessible, simply by bone marrow aspiration or by peripheral blood mononuclear cell harvest. Once the genetic insertion is accomplished in vitro, the treated cells can be reinfused intravenously, after which the genetically transformed cells will home to and develop in the bone marrow. Since mature blood cells circulate throughout the body, the genetically modified cells would deliver the specific gene product to any desired tissue. Most importantly, hematopoietic tissues contain stem cells, which possess extensive (perhaps unlimited) capacities for self-renewal.
  • hematopoietic stem cell gene therapy has broad application, to both diseases specific to the hematopoietic system and to other organ system diseases.
  • both inherited and acquired diseases can be treated by stem cell gene therapy.
  • hemoglobin deficiencies such as ⁇ and ⁇ Thalessemias could be treated by the insertion of the gene coding for the globin ⁇ or ⁇ chain, together with regulatory sequences that confer high level tissue-specific expression of these genes in erythrocytes (Grosveld F, van Assendelft GB, Greaves DR, Kollias G, Cell.
  • sickle cell anemia could be corrected by the genetic insertion of the fetal globin gene into hematopoietic stem cells, as the regulated expression of high levels of fetal hemoglobin are sufficient to prevent sickling in red cells despite the copresence of sickle hemoglobin (Sunshine HR, Hofrichter J, et al. , J. Molec. Biol. 133:435; 1979).
  • lymphocyte immunodeficiency diseases such as severe combined immunodeficiency (SCID) due to adenosine deaminase deficiency.
  • SCID severe combined immunodeficiency
  • Retroviral gene therapy of circulating T cells with the ADA gene has been found to be successful at reducing the clinical immunodeficiency experienced by these patients, but the effects are only temporary, because the transfected T lymphocytes have a finite life span in vivo (Kasid A, Morecki S, Aebersold P, Cornetta K, Culver K, Freeman S, Director E, Lotze MT, Blaese RM, Anderson WF, et al., Proc. Natl. Acad. Sci. U.S.A.
  • the T cell ADA deficiency would be permanently treated by a single gene transfer stem cell treatment (Wilson JM, Danos 0, Grossman M, Raulet DH, Mulligan RC, Proc. Natl. Acad. Sci.. U.S.A.. 87:439-443;1990) .
  • stem cell gene therapy could be useful for protecting stem cells and their progeny from toxic exogenous agents such as viruses or chemotherapy.
  • gene transfer of DNA sequences encoding the TAR binding site of the HIV TAT transactivating factor have been shown to protect T cells from spreading infection by the HIV virus (Sullenger BA, Gallardo HF, Ungers GE, Gilboa E, Cell. 1990;63:601-8). Stable transfection of these sequences into hematopoietic stem cells would result in a pool of T cells, all arising from these stem cells, which would be relatively or absolutely resistant to the spread of HIV.
  • hematopoietic stem cell gene therapy will also be useful for acquired hematopoietic diseases such as leukemia, lymphoma and aplastic anemia. Once the genetic causes of these diseases are discovered, insertion of a gene whose product either overcomes that of the abnormal gene in the cell or corrects it directly (e.g., by splicing out and replacing the gene, via homologous recombination) would correct the abnormality.
  • hematopoietic stem cell gene therapy would be useful for the treatment of diseases outside the hematopoietic system as well.
  • Gene transfer of DNA sequences carrying therapeutic soluble proteins could give rise to mature blood cells which permanently secrete the desired amounts of a therapeutic molecule.
  • this approach could be useful for the treatment of, e.g., diabetes mellitus by the insertion of DNA sequences for insulin along with regulatory DNA sequences that control the proper expression of the transfected insulin gene, perhaps in response to elevated plasma glucose levels.
  • Systemic hypertension could be treated by genetic insertion of stem cells with DNA sequences encoding secretory peptides which act as competitive inhibitors to angiotensin converting enzyme, to vascular smooth muscle calcium channels, or to adrenergic receptors.
  • Alzheimer's disease could possibly be treated by genetic insertion into stem cells of DNA sequences encoding enzymes which break down amyloid plaques within the central nervous system.
  • the major impediments to achieving successful human hematopoietic stem cell gene therapy has been the inability to have cultured stem cells replicate to allow for the stable insertion of genes, and the existence of a retroviral packaging cell line which efficiently infects human stem cells.
  • Successful infection with a retroviral vector requires: binding of the virus to the target cell, penetration into the cell, uncoating of the virus, reverse transcription of viral RNA into DNA, integration of the viral DNA into the host chromosome (using a viral integrase) , and expression of the gene of interest.
  • a retrovirus is an RNA virus consisting of approximately 9,000 bases of RNA. After infection of the host cell, the RNA is converted into double stranded DNA which inserts into the host chromosome.
  • the integrated viral DNA is called the provirus.
  • the provirus consists of two repetitive sequences, called the long terminal repeat (LTR) , at the beginning (5') and the end (3') of the virus sequences.
  • LTR long terminal repeat
  • the 5' LTR directs transcription of virus mRNA's which encode at least two transcripts which encode the viral structural and replication proteins.
  • the first m NA encodes the GAG and POL/INT proteins
  • the second mRNA encodes the envelope proteins.
  • Some viruses also encode transcripts involved in other virus replication functions, for example the HIV REV gene.
  • the host and cell type specificity of the retrovirus is determined in some cases by the ability of the LTR to function in the specific cell and in other cases by the presence of the receptor for the viral envelope on the cell. In the latter case, the cell receptor binds to the viral envelope (ligand) , and the virus enters the cell. It is felt that lentiviruses, such as the HIV retrovirus, enter the cell via virus envelope mediated fusion after the viral envelope binds to the CD4 molecule on the cell surface (Grewe C, Beck A, and Gelderblom HR, J. AIDS. 3, 965-979, 1990).
  • amphotropic murine retroviruses which are used in current packaging cell lines, may enter the host via a pH-independent envelope fusion with endosomal particles (McClure MO, So merfelt MA, Marsh M, and Weiss RA, J. Gen. Virol.. 71, 767-773, 1990).
  • the amphotropic virus envelope binds to a cell surface receptor present on all cells.
  • the virus particle is assembled at the cell membrane.
  • the structural proteins, the GAG proteins in the case of MoMLV, and replication proteins (RT/INT) bind to ⁇ is-acting sequences present in viral RNA, and assemble at the cell surface with the virus envelope proteins and the virus particle buds off the cell surface.
  • the packaging virus In order to accomplish gene therapy, the packaging virus must be able to efficiently infect the host cell.
  • the majority of packaging lines developed to date are based on the mouse MoMLV amphotropic virus. Although this viral coat will bind to human cells, it is very inefficient at infecting human hematopoietic cells (Gruber DE, Finley KD, Hershberg RM, Katzman SS, Laikind PK, Siegmiller JE, Friedmann T, Vee JK and Jolley DJ, Science. 230, 1057-1061, 1985; Hogge DE & Humphries RK, Blood. 69, 611-617, 1987).
  • xenotropic virus a virus which infects species other than mouse packaging cell line
  • 3T3 xenotropic packaging cell lines a virus which infects species other than mouse packaging cell line
  • a Gibbon Ape Leukemia Virus envelope which recognizes a widely distributed cell receptor, has been used to make a 3T3 pseudotype packaging cell line (Miller D et al. , J. Virol.. 65, 2220-4, 1991) .
  • the ideal packaging virus would have an envelope (ligand) which recognizes a receptor present only on the subset of cells which are the intended target for gene therapy.
  • This cell type specificity would have multiple advantages over the present amphotropic and xenotropic packaging cell lines which recognize a receptor present on all cell types.
  • human hematopoietic stem cells probably only represent .01% or less of mononuclear bone marrow cells.
  • these cells appear to grow only when in contact with stromal cells, which represent a large proportion of bone marrow cultures.
  • extremely high viral titers are required to infect a stem cell. These high viral titers are very difficult to obtain.
  • the ideal packaging line would be constructed using a human retrovirus envelope, since these viruses have evolved to efficiently infect human cells.
  • the known human retroviruses HTLV and HIV
  • T-cell specificity is due to the fact that only T-cells express the viral receptor on their cell membranes.
  • studies have shown that the HIV envelope will form virions with the GAG and POL proteins of an amphotropic virus (Wilson K, Reitz MS, Okayama H, and Eiden MB, J. Virol.. 63, 2374-8, 1989).
  • U.S. patent no. 4,731,324 describes a viral lysis assay in which modified viral particles are utilized. Specifically, the virus particles contain on their surface non-viral anti- analyte molecules which are composed of an anti-analyte which is derivatized with a lipid moiety which anchors the molecule to the virus lipid bilayer surface.
  • U.S. patent no. 4,948,590 discloses avidin- or streptavidin-conjugated liposomes which can be utilized as a vehicle for site-specific targeting of encapsulated drugs or other macromolecules, including retrovirus vectors.
  • the conjugated liposomes are prepared by coupling, e.g., the carboxyl residues of streptavidin to the phospholipid amino groups of the liposome.
  • U.S. patent no. 4,786,590 describes the use of monoclonal antibodies which bind to one or more determinant sites of cell surface membrane receptors (CSMR) for directing agents to the site of transformed cells.
  • CSMR cell surface membrane receptors
  • agents include radionuclides, toxins, and factors involved in complement lysis.
  • U.S. patent no. 4,701,416 teaches the use of protein carriers to which is conjugated an amino acid sequence which is homologous to at least a portion of gp70 envelope protein of FeLV as a FeLV immunogen.
  • carrier proteins include keyhole limpet hemocyanin, ovalbumin, porcine thyroglobulin, and bovine serum albumin.
  • U.S. patent no. 4,861,588 describes hepatitis B vaccines containing an amino acid chain corresponding to a chain of amino acids in the pre-S region linked to a carrier. Lipid vesicles are described as the preferred carrier.
  • U.S. patent no. 4,859,769 teaches the use of liposomes which may be tagged with a viral component which recognizes the. second-step receptor on the surface of a cell.
  • the liposome may be used to carry a toxic drug.
  • U.S. patent no. 4,545,985 discloses a method for modifying Pseudomonas exotoxin for conjugating the exotoxin to a monoclonal antibody which binds to a cell surface membrane receptor to form an immunotoxin.
  • viruses which contain, on their surface, proteins specific for receptors found on the membrane of targeted cells. Further, there remains a need for methods and packaging cell lines to prepare such retroviruses.
  • viruses including retroviruses, which possess on their surface at least one chimeric envelope protein which binds specifically to a receptor found on targeted cells. It is another object of the present invention to provide viruses, including retroviruses, which contain on their surface at least one chimeric envelope protein which specifically binds to mammalian, including human, stem and/or progenitor cells.
  • viruses including retroviruses, which possess on their surface at least one chimeric envelope protein which specifically binds to human hematopoietic stem and/or progenitor cells.
  • a genetically engineered virus including a retrovirus, containing a therapeutic gene packaged in a virion with a chimeric envelope protein which specifically binds to a receptor found on the membrane of the stem and/or progenitor cell.
  • viruses including retroviruses
  • the viral membrane contains a chimeric envelope peptide which will specifically bind to a targeted cell and that such retroviruses may be used to transduce specific targeted cells.
  • the present invention relates to the use of a genetically engineered retroviral packaging cell line that has altered the viral envelope such that it contains a peptide which will bind to a molecule on the membrane of the target cell.
  • the present invention can be used for the transfer of any genetic information that can be engineered into a recombinant retrovirus, or any other gene transfer vector that requires a virus for delivery.
  • the present invention also relates to a stromal cell line which supports hematopoiesis and is transfected with the retrovirus packaging vectors.
  • Genetically modified human stem cells produced by the present invention can be applied to a wide variety of clinical diseases, as described in the preceding text. Further, the present invention also provides methodology that enhances the efficiency of genetic transfer into human progenitor cells.
  • the present invention includes a genetically modified human stem cell produced with the use of the present retroviruses and/or packaging cell line.
  • tissue specificity of retrovirus infection can be conferred by virus envelope (ligand) binding to a cell receptor, and both stem and progenitor cell specific ligands and receptors are known
  • virus envelope ligand
  • stem and progenitor cell specific ligands and receptors are known
  • the construction of chimeric viral envelope/stem cell ligand molecules will confer stem and progenitor cell tropism to virus particles.
  • the HIV viral membrane gene has been cloned, and the envelope amino acids which bind to the CD4 antigen on the T-cell membrane are known (Lasky LA, et al. Cell. 50, 975-985, 1987) .
  • This knowledge provides the information needed to substitute into the gene for the HIV envelope CD4 antigen binding site a sequence that encodes the peptide that is known to bind to the cell membrane of any specific targeted cell of interest.
  • virus particles that infect target cells efficiently and selectively, because the virus particles will now encode a chimeric envelope protein which will bind specifically to the stem and/or progenitor cell receptor molecule, and will not bind to cell types which do not have the receptor.
  • CF cystic fibrosis
  • the present invention includes a human stromal cell line derived from, e.g., fetal liver (which supports hematopoiesis) or adult bone marrow for use as the packaging cell line.
  • a retrovirus envelope mRNA usually encodes a protein which is cleaved into two peptides. These peptides are inserted into the cell membrane and are associated as a dimer or tetramer.
  • the viral envelope consists of an intracellular and extracellular domain.
  • the present invention is achieved by genetically engineering the extracellular domain of the retroviral envelope so that it consists of a chimeric molecule which will associate as a dimer with the small envelope peptide.
  • the viral envelope extracellular domain contains a ligand which will specifically bind to the cell membrane of interest.
  • the target cells are human hematopoietic stem cells
  • the genes for the c-kit ligand, the Leukemic Inhibitory Factor (LIF) , the CS-1 peptide of the alternatively spliced non-type III connecting segment (IIICS) of human plasma fibronectin, or any other ligand which recognizes human stem cells may be substituted for the CD4 binding amino acids in the HIV envelope.
  • LIF Leukemic Inhibitory Factor
  • IIICS alternatively spliced non-type III connecting segment
  • the appropriate ligand is selected and substituted for all or a portion of the ligand naturally occurring on the virus envelope. The selection of the appropriate ligand for the target cell of interest is well within the abilities of the artisan of ordinary skill.
  • substitution of a ligand specific for the target cell receptor it is meant that either the entire protein or a portion thereof may be substituted for a portion of the ligand naturally occurring on the virus envelope.
  • the selection of the particular portion of a ligand responsible for the binding to a target cell and the optimization of the size of the portion of both ligand being replaced and the ligand being inserted are within the capabilities of one of ordinary skill in the art.
  • the replacement of the portion of the ligand, naturally occurring on the virus envelope and responsible for binding to a cell surface, with the ligand or portion thereof responsible for binding to the target cell is achieved by altering the sequence of the gene encoding the ligand naturally occurring on the virus envelope using recombinant DNA technology.
  • a portion of the gene which encodes the sequence of the naturally occurring ligand necessary for binding to the natural receptor is excised and replaced with either the DNA sequence encoding the entire ligand which binds to the target cell or any portion of the ligand which results in binding to the target cell.
  • the present invention relates to retroviruses which have been engineered to specifically bind to human stem and/or progenitor cells.
  • Human hematopoietic cells may be isolated from bone marrow, peripheral blood, fetal liver, or umbilical cord blood. It is further preferred that these cells are cultured as described in U.S. Patent Applications Serial Nos. 07/737,024, 07/628,343, and 07/366,639, which are incorporated herein by reference, under conditions in which about 50% of the medium and serum is exchanged daily, and the replacing volume is supplemented with hematopoietic growth factors.
  • Such hematopoietic growth factors may be supplied exogenously by addition of the growth factors to the culture medium or may be present in the medium as a result of the presence of transformed stromal cells in the culture which endogenously produce the growth factors. It should be noted that these conditions result in formation of a stroma during the time of the retroviral infection.
  • the growth factors are GM-CSF (0.1 to 100 ng/ml/day, preferably about 5 ng/ml/day) plus IL-3 (0.1 to 100 ng/ml/day, preferably about 10 ng/ml/day) with or without IL-l ⁇ (10 to 100 U/ml per 3 to 5 day period, preferably about 50 U/ml/4 day period) , with or without c-kit ligand (Mast cell growth factor) (1 to 100 ng/ml/day, preferably about 10 ng/ml/day) , with or without IL-6 (1 to 100 ng/ml/day, preferably about 10 U/ml/day) .
  • GM-CSF 0.1 to 100 ng/ml/day, preferably about 5 ng/ml/day
  • IL-3 0.1 to 100 ng/ml/day, preferably about 10 ng/ml/day
  • IL-l ⁇ 10 to 100 U/ml per 3 to 5 day period, preferably about 50
  • the retroviral infection (transduction) of the cells is preferably carried out as described in U.S. Patent Application Serial No. 07/740,590, which is incorporated herein by reference.
  • transduction it is meant the delivery of genes into cells by means of a viral vector.
  • the retroviral infection is performed by including supernatants produced by retroviral packaging lines infected with recombinant retrovirus, during the first days of the culture, or by culturing the cells directly over the infected retroviral packaging lines themselves. In the preferred embodiment, retroviral supernatants are used, and the period of incubation in the presence of virus is about 14 days.
  • the retrovirus of the present invention used to infect the target cell should of course also contain within its genome the genetic information encoding the desired therapeutic gene and any required regulatory sequence.
  • the choice of therapeutic gene will depend on the particular disease or condition to be treated. Generally speaking, the therapeutic gene and any attendant regulatory sequences should be no more than approximately 6 to 8 kb in size. Examples of therapeutic genes have been discussed above, and the choice of the particular therapeutic gene is within the abilities of those skilled in the art.
  • the present retroviruses may be prepared by the following method, which is described in terms of the HIV envelope protein containing the CD4 binding domain. However, it is to be understood that other envelope proteins for which the binding domain is known may take the place of the exemplified HIV envelope protein.
  • Two plasmids which contain the necessary genes for retrovirus packaging may be constructed.
  • One plasmid may include the GAG, POL, and RT genes, with or without the HTLV TAT and/or REV genes, of either MoMLV, HTLVI, or HIV.
  • the second plasmid may include a genetically modified HIV envelope gene which has the CD4 binding domain (Olshensky U, Helseth E, Furma C, Li J, Haseltine W, Sodroski J, J. Virol.. 64, 5701-5707, 1990) replaced by either the c-kit ligand (Zsebo KM et al. Cell. 63, 195-201, 1990; Anderson KM et al. Cell.
  • human fetal liver stroma or bone marrow stroma may be immortalized with the SV40 large T antigen, and a transformed cell line which supports hematopoiesis may then be selected. These cells may then be transfected with the packaging vectors, and a virus producer cell line selected to act as the packaging line.
  • viruses of the present invention are characterized by the presence, on their envelope, of multiple copies of at least one type of chimeric envelope protein which results in the virus binding specifically to a receptor on a cell surface to which the natural virus would not specifically bind.
  • natural virus it is meant the virus which has on its surface the naturally occurring envelope protein.
  • a ligand (envelope protein) is said to specifically bind to a cell surface receptor, if the ligand binds to that cell surface receptor preferentially in the presence of other cell surface receptors to which it does not specifically bind.
  • ligands which specifically bind to cell surface receptors do so with an affinity constant of 10 ⁇ 6 to 10 ⁇ 12 M.
  • any gene which is inserted into a recombinant retrovirus vector together with suitable promoter and enhancer elements that permit its expression can be incorporated into human hematopoietic stem cells.
  • the achievement of the present invention derives from the conditions that permit stem cell infection through the use of a genetically altered retroviral envelope gene which confers binding specificity for human stem and/or progenitor calls, and the use of a human stromal cell line as the packaging cell line.
  • LIF LIF is known to be found on mouse embryonal stem cells and early hematopoietic cells but not on mature tissue cells. Thus, this protein should have specificity for stem and early progenitor cells. It should be noted that other ligands such as IL1 through 10, GM-CSF, the c-kit ligand, the ligand for Flk 2, etc. can also be used. In addition, just the receptor binding sequences of the ligand may be used, and these sequences may be substituted into different parts of the HIV ENV gene, or into the binding protein (ENV) genes of other retroviruses or other viruses. Furthermore, some or all of the extracellular domain, not including the portion of the extracellular domain which associates with the retrovirus small envelope protein, of the ENV gene may be deleted.
  • ENV binding protein
  • an HIV ENV, LIF chimeric retroviral ENV gene may be made as follows.
  • the HIV ENV coding region may be isolated in two parts by using the polymerase chain reaction (PCR) using specific primers designed to introduce useful restriction endonuclease sites into the ENV gene. Since amino acids 410-421 (Ratner L, et al, Nature. 313, 277-284, 1985), contain sequences essential for CD4 binding (Lasky LA, Cell. 50, 975-985. 1991), they may be deleted to abrogate CD4 binding.
  • the primers may be CCGTCGACATGAGAGTGAAGGAGAAA TATCAGCACTTG and the sequence
  • the HIV clone pBHIO may be used as a template for the PCR amplification of part of the ENV gene. Two more primers,
  • CCGGAATTCAGTGGACAAATTAGATGTTCATCAAATATTACAGGG and CCGTCGACTTATAGCAAAATCCTTTCCAAGCCCTGTCT may be used to amplify the HIV clone pBHIO ENV (Hahn B et al, Nature. 312, 166, 1984) .
  • the two PCR products may be digested with the restriction endonuclease Sal 1 and Eco Rl, and then included in a tripartite ligation reaction containing Sal 1 digested pSub 1, a eukaryotic expression vector (Castle VP, Varani J, Fligiel S, Prochownik EV, and Dixit VM, J. Clin. Inv..
  • the ligation reaction may be transfected into E. coli, and a recombinant plasmid containing the HIV envelope gene with a deletion of the bases encoding amino acids critical for CD4 binding may then be identified. There is now an Eco Rl restriction endonuclease site substituted for the deleted CD4 binding region.
  • a clone containing only the coding sequences of LIF may be isolated by PCR amplification of LIF (Moreau J-F et al. Nature. 336,690-692, 1988)) using the primers CCGAATTCCAGCCCCCTCCCCATCACCCCTGTCAACGCCACCTGT and CCGAATTCGAAGGCCTGGGCCAACACGGCGATGATCTG.
  • the PCR amplified LIF coding sequence is now flanked by an Eco Rl restriction endonuclease site.
  • the PCR product may then be digested with the restriction endonuclease Eco Rl and ligated into the Eco Rl site which was introduced into the HIV ENV gene.
  • a clone with an in-frame substitution of LIF for the CD4 binding region of the HIV ENV gene may be isolated and transfected into a cell line expressing the GAG POL genes of either an amphotropic MoMLV, or another retrovirus. This results in a packaging cell line which produces virions with a chimeric LIF/HIV envelope when transfected with a recombinant retroviral vector.

Abstract

Genetically engineered viruses which comprise a genetically engineered gene and having a chimeric envelope protein which specifically binds to a target cell receptor, to which the naturally occurring envelope protein does not bind, may be used for cell type-specific gene therapy. Transduction of human stem and/or progenitor cells with such viruses containing chimeric envelope proteins which specifically bind to human stem and/or progenitor cells proceeds with high specificity and efficiency, especially when such viruses are incubated with human stem and/or progenitor cells cultured under conditions of rapid medium exchange.

Description

Description
Targeted Virus
Technical Field
The present invention relates to a virus which has an chimeric envelope protein on its surface that effects the specific binding of the virus to a cell of interest. The present invention also relates to a method of producing such a virus and methods of transducing cells and gene therapy utilizing such a virus. The present invention further relates to stem and/or progenitor cells which have been transduced with such a virus and a packaging cell line which produces such a virus.
Background Art
Genetically transformed human stem cells have wide potential application in clinical medicine, as agents of gene therapy. Gene therapy is an emerging approach to clinical treatment which has evolved from earlier approaches in medical care. The earliest approaches to medical care, evolving over centuries, include gross surgical procedures and the administration of crude mixtures as medicinal agents. In the past century, biochemical pharmacology has supervened as the major method of medical treatment. Under this paradigm, pure pharmaceutical molecules are delivered to the patient. In general, such pharmacologic agents act either as poisons (such as antimicrobials or cancer chemotherapy agents) , physiologic imetics which stimulate endogenous receptors (e.g., opiates, adrenergic agonists) , or physiologic antagonists which block endogenous receptors (e.g. antihypertensives, anesthetics) .
The principle underlying gene therapy is to, rather than deliver doses of pharmacologic molecules, deliver a functional gene whose RNA or protein product will produce the desired biochemical effect in the target cell or tissue. There are several potential advantages of gene therapy over classical biochemical pharmacology. First, inserted genes can produce extremely complex molecules, including DNA, RNA and proteins, which can be extraordinarily difficult to administer and deliver themselves. Next, controlled insertion of the desired gene into specific target cells can control the production of gene product to defined tissues. Finally, gene therapy can in principle be permanent within an individual, as the gene will continue to function in the target cells and their progeny.
However, there are several problems that must be addressed for successful gene therapy. The first is to be able to insert the desired therapeutic gene into the chosen cells. Second, the gene must be adequately expressed in the target cell, resulting in the appropriate levels of gene product. Finally the DNA, RNA or protein produced must be properly processed by the target cell so that it is functional, i.e. so that gene therapy actually confers clinical therapy. Several methods of gene insertion into human cells in vitro have been accomplished and are listed in Table 1.
Table 1: Comparison of DNA transfer methods.
Figure imgf000004_0001
Other techniques, such as homologous recombination, are being developed as well in many laboratories. With these issues in mind, research on gene therapy has been on-going for several years. This research, which began in several types of cells in vitro, has progressed to animal studies, and has recently entered the first human clinical trials (Kasid A, Morecki S, Aebersold P, Cornetta K, Culver K, Freeman S, Director E, Lotze MT, Blaese RM, Anderson WF et al., Proc. Natl. Acad. Sci. U.S.A.. 1990; 87:473-7; Culver K, Cornetta K, Morgan R, Morecki S, Aebersold P, Kasid A, Lotze M, Rosenberg SA, Anderson WF, Blaese RM, Proc. Natl. Acad. Sci. U.S.A.. 1991; 88:3155-9.) Tissue-specific expression of a functionally rearranged λl Ig gene by transduction with a retrovirus vector has been observed (Cone RD, Reilly EB, Eisen HN, and Mulligan RC, Science. 236:954-957 (1987)).
The hematopoietic system is an ideal choice as a delivery system for gene therapy. Hematopoietic cells are readily accessible, simply by bone marrow aspiration or by peripheral blood mononuclear cell harvest. Once the genetic insertion is accomplished in vitro, the treated cells can be reinfused intravenously, after which the genetically transformed cells will home to and develop in the bone marrow. Since mature blood cells circulate throughout the body, the genetically modified cells would deliver the specific gene product to any desired tissue. Most importantly, hematopoietic tissues contain stem cells, which possess extensive (perhaps unlimited) capacities for self-renewal. This implies that if genetic material were stably transduced into these stem cells, then upon reinfusion of the hematopoietic tissue, these altered stem cells can expand and repopulate the marrow with cells that express the new gene. This would lead to longlasting, perhaps lifelong delivery of the desired gene product. Similarly, successful stable gene transfer into stem cells located in other tissues, or into embryonic stem cells, would likewise lead to longlasting gene product delivery.
Successful hematopoietic stem cell gene therapy has broad application, to both diseases specific to the hematopoietic system and to other organ system diseases. Within the hematopoietic system, both inherited and acquired diseases can be treated by stem cell gene therapy. For example, hemoglobin deficiencies such as α and β Thalessemias could be treated by the insertion of the gene coding for the globin α or β chain, together with regulatory sequences that confer high level tissue-specific expression of these genes in erythrocytes (Grosveld F, van Assendelft GB, Greaves DR, Kollias G, Cell. 51:975-985;1987; Soriano P, Cone RD, Mulligan c, Jaenisch R, Science, 234: 1409-1413; 1986). Similarly, sickle cell anemia could be corrected by the genetic insertion of the fetal globin gene into hematopoietic stem cells, as the regulated expression of high levels of fetal hemoglobin are sufficient to prevent sickling in red cells despite the copresence of sickle hemoglobin (Sunshine HR, Hofrichter J, et al. , J. Molec. Biol. 133:435; 1979). Genetic diseases of neutrophils caused by functional protein deficiencies, such as leukocyte adhesion deficiency (LAD) or chronic granulomatous disease (CGD) could be treated by the genetic insertion of the gene encoding the defective or absent gene, along with regulatory DNA sequences that confer high level, tissue specific expression into hematopoietic stem cells (Wilson M, Ping AJ, rauss JC, Mayo-Bond L, Rogers CE, Anderson DC, Todd RF, Science. 248:1413-1416; 1990). Genetic diseases involving platelets, such as von Willebrand's Disease, could be corrected by the genetic insertion of the gene encoding, e.g. von Willebrand's Factor, along with sequences which permit its expression and secretion.
The particular suitability of hematopoietic stem cell gene therapy for the replacement of congenitally deficient gene products is particularly evident in the treatment of lymphocyte immunodeficiency diseases, such as severe combined immunodeficiency (SCID) due to adenosine deaminase deficiency. Retroviral gene therapy of circulating T cells with the ADA gene has been found to be successful at reducing the clinical immunodeficiency experienced by these patients, but the effects are only temporary, because the transfected T lymphocytes have a finite life span in vivo (Kasid A, Morecki S, Aebersold P, Cornetta K, Culver K, Freeman S, Director E, Lotze MT, Blaese RM, Anderson WF, et al., Proc. Natl. Acad. Sci. U.S.A. 1990;87:473-7; Culver K, Cornetta K, Morgan R, Morecki S, Aebersold P, Kasid A, Lotze M, Rosenberg SA, Anderson WF, Blaese RM, Proc. Natl. Acad. Sci. U.S.A., 1991; 88:3155-9). If, however, the gene could be successfully transfected into hematopoietic stem cells, then all of the T cells which arose from these stem cells would contain and express the ADA gene. Therefore, since the transfected stem cells would persist and proliferate for the life of the patient, the T cell ADA deficiency would be permanently treated by a single gene transfer stem cell treatment (Wilson JM, Danos 0, Grossman M, Raulet DH, Mulligan RC, Proc. Natl. Acad. Sci.. U.S.A.. 87:439-443;1990) .
In addition to treating inherited enzymatic abnormalities of the hematopoietic system, stem cell gene therapy could be useful for protecting stem cells and their progeny from toxic exogenous agents such as viruses or chemotherapy. For example, gene transfer of DNA sequences encoding the TAR binding site of the HIV TAT transactivating factor have been shown to protect T cells from spreading infection by the HIV virus (Sullenger BA, Gallardo HF, Ungers GE, Gilboa E, Cell. 1990;63:601-8). Stable transfection of these sequences into hematopoietic stem cells would result in a pool of T cells, all arising from these stem cells, which would be relatively or absolutely resistant to the spread of HIV. Similarly, successful transfection of the genes encoding the multi-drug resistance gene (MDR) or the methotrexate resistance gene into human bone marrow stem cells would create stem cells which would be relatively resistant to the affects of cancer chemotherapy. Following autologous bone marrow transplantation with these genetically manipulated cells, patients would be able to tolerate chemotherapy with the agents to which their stem cells were protected without suffering the profound bone marrow suppression commonly caused by these anti-cancer drugs. This would enable patients to receive more effective doses of cancer chemotherapy with less toxicity.
One can readily envision that hematopoietic stem cell gene therapy will also be useful for acquired hematopoietic diseases such as leukemia, lymphoma and aplastic anemia. Once the genetic causes of these diseases are discovered, insertion of a gene whose product either overcomes that of the abnormal gene in the cell or corrects it directly (e.g., by splicing out and replacing the gene, via homologous recombination) would correct the abnormality.
On a broader level, however, hematopoietic stem cell gene therapy would be useful for the treatment of diseases outside the hematopoietic system as well. Gene transfer of DNA sequences carrying therapeutic soluble proteins could give rise to mature blood cells which permanently secrete the desired amounts of a therapeutic molecule. By way of examples, this approach could be useful for the treatment of, e.g., diabetes mellitus by the insertion of DNA sequences for insulin along with regulatory DNA sequences that control the proper expression of the transfected insulin gene, perhaps in response to elevated plasma glucose levels. Systemic hypertension could be treated by genetic insertion of stem cells with DNA sequences encoding secretory peptides which act as competitive inhibitors to angiotensin converting enzyme, to vascular smooth muscle calcium channels, or to adrenergic receptors. Alzheimer's disease could possibly be treated by genetic insertion into stem cells of DNA sequences encoding enzymes which break down amyloid plaques within the central nervous system.
The many applications of gene therapy, particularly via stem cell genetic insertion, are thus well known and have been extensively reviewed (Boggs SS, Int. J. Cell Cloning. 8.80-96; 1990; Kohn DB, Anderson WF, Blaese RM, Cancer Investigation. 7:179-192;1989; Lehn PM, Bone Marrow Transplantation. 5:287-293;1990, Verma IM, Scientif Amer.. pp. 68-84;1990; Weatherall DJ, Nature. 349:275-276; 1991; Feigner PL, Rhodes G, Nature, 349:351-352; 1991). There are indeed increasing examples of success in achieving therapeutic gene transfer into differentiated human cells, as described, for example, in T lymphocytes (Kasid A, Morecki S, Aebersold P, Cornetta K, Culver K, Freeman S, Director E, Lotze MT, Blaese RM, Anderson WF, et al., Proc. Natl. Acad. Sci. U.S.A. 1990;87:473-7; Culver K, Cornotia K, Morgan R, Morecki S, Aebersold P, Kasid A, Lotze M, Rosenberg SA, Anderson WF, Blaese RM, Proc. Natl. Acad. Sci. U.S.A.. 1991; 88:3155-9).
Unfortunately, achieving gene transfer into human stem cells has not until now been accomplished. While several groups have demonstrated the feasibility of retroviral mediated gene transfer into human hematopoietic cells, human primitive hematopoietic stem cells have not been successfully transfected. This is in sharp contrast to experiments in the mouse, in which some level of retrovirally mediated gene transfer into hematopoietic stem cells has been possible (Wilson JM, Danos 0, Grossman M, Raulet DH, Mulligan RC, Proc. Natl. Acad. Sci.. U.S.A.. 87:439-443;1990) . The major impediments to achieving successful human hematopoietic stem cell gene therapy has been the inability to have cultured stem cells replicate to allow for the stable insertion of genes, and the existence of a retroviral packaging cell line which efficiently infects human stem cells. Successful infection with a retroviral vector requires: binding of the virus to the target cell, penetration into the cell, uncoating of the virus, reverse transcription of viral RNA into DNA, integration of the viral DNA into the host chromosome (using a viral integrase) , and expression of the gene of interest. Successful stable gene insertion via retroviral vectors into a target cell requires that the target cell undergo at least one round of cell division (Springett GM, Moen RC, Anderson S, Blaese RM, Anderson WF, J. Virol.. 1989;63:3865-9). Thus, if stem cells are not dividing in the presence of the desired genetic material, the material will not be stably inserted into the stem cells. Part of the difficulty in infecting stem cells is the retroviral delivery system itself.
A retrovirus is an RNA virus consisting of approximately 9,000 bases of RNA. After infection of the host cell, the RNA is converted into double stranded DNA which inserts into the host chromosome. The integrated viral DNA is called the provirus. The provirus consists of two repetitive sequences, called the long terminal repeat (LTR) , at the beginning (5') and the end (3') of the virus sequences. The 5' LTR directs transcription of virus mRNA's which encode at least two transcripts which encode the viral structural and replication proteins. The first m NA encodes the GAG and POL/INT proteins, and the second mRNA encodes the envelope proteins. Some viruses also encode transcripts involved in other virus replication functions, for example the HIV REV gene. The host and cell type specificity of the retrovirus is determined in some cases by the ability of the LTR to function in the specific cell and in other cases by the presence of the receptor for the viral envelope on the cell. In the latter case, the cell receptor binds to the viral envelope (ligand) , and the virus enters the cell. It is felt that lentiviruses, such as the HIV retrovirus, enter the cell via virus envelope mediated fusion after the viral envelope binds to the CD4 molecule on the cell surface (Grewe C, Beck A, and Gelderblom HR, J. AIDS. 3, 965-979, 1990). On the other hand, the amphotropic murine retroviruses (MoMLV) , which are used in current packaging cell lines, may enter the host via a pH-independent envelope fusion with endosomal particles (McClure MO, So merfelt MA, Marsh M, and Weiss RA, J. Gen. Virol.. 71, 767-773, 1990). The amphotropic virus envelope binds to a cell surface receptor present on all cells.
The virus particle is assembled at the cell membrane. The structural proteins, the GAG proteins in the case of MoMLV, and replication proteins (RT/INT) bind to σis-acting sequences present in viral RNA, and assemble at the cell surface with the virus envelope proteins and the virus particle buds off the cell surface.
It has been shown that the envelope proteins from a different virus will assemble virus particles in conjunction with the GAG and POL proteins of a second virus (Page KA, Landau NR, and Littman DR, J. Virol.. 64, 5270-5276, 1990).
In order to accomplish gene therapy, the packaging virus must be able to efficiently infect the host cell. The majority of packaging lines developed to date are based on the mouse MoMLV amphotropic virus. Although this viral coat will bind to human cells, it is very inefficient at infecting human hematopoietic cells (Gruber DE, Finley KD, Hershberg RM, Katzman SS, Laikind PK, Siegmiller JE, Friedmann T, Vee JK and Jolley DJ, Science. 230, 1057-1061, 1985; Hogge DE & Humphries RK, Blood. 69, 611-617, 1987). Attempts to use a xenotropic virus (a virus which infects species other than mouse) packaging cell line have been hindered by very low viral titers of 3T3 xenotropic packaging cell lines. Finally, a Gibbon Ape Leukemia Virus envelope, which recognizes a widely distributed cell receptor, has been used to make a 3T3 pseudotype packaging cell line (Miller D et al. , J. Virol.. 65, 2220-4, 1991) .
The ideal packaging virus would have an envelope (ligand) which recognizes a receptor present only on the subset of cells which are the intended target for gene therapy. This cell type specificity would have multiple advantages over the present amphotropic and xenotropic packaging cell lines which recognize a receptor present on all cell types. For example, human hematopoietic stem cells probably only represent .01% or less of mononuclear bone marrow cells. In addition, these cells appear to grow only when in contact with stromal cells, which represent a large proportion of bone marrow cultures. Thus, when attempting gene transfer in in vitro hematopoietic cultures, extremely high viral titers are required to infect a stem cell. These high viral titers are very difficult to obtain. Alternatively, enrichment of the mononuclear bone marrow for stem cells is possible, but the best method is by FACS which is very expensive and time consuming. In addition, the FACS sorted cells probably also require culture on a stromal layer which will also bind virus particles and effectively lower the virus titer.
Additionally, the ideal packaging line would be constructed using a human retrovirus envelope, since these viruses have evolved to efficiently infect human cells. Unfortunately, the known human retroviruses (HTLV and HIV) only infect T-cells. In the case of HIV, T-cell specificity is due to the fact that only T-cells express the viral receptor on their cell membranes. However, studies have shown that the HIV envelope will form virions with the GAG and POL proteins of an amphotropic virus (Wilson K, Reitz MS, Okayama H, and Eiden MB, J. Virol.. 63, 2374-8, 1989).
U.S. patent no. 4,731,324 describes a viral lysis assay in which modified viral particles are utilized. Specifically, the virus particles contain on their surface non-viral anti- analyte molecules which are composed of an anti-analyte which is derivatized with a lipid moiety which anchors the molecule to the virus lipid bilayer surface.
U.S. patent no. 4,948,590 discloses avidin- or streptavidin-conjugated liposomes which can be utilized as a vehicle for site-specific targeting of encapsulated drugs or other macromolecules, including retrovirus vectors. The conjugated liposomes are prepared by coupling, e.g., the carboxyl residues of streptavidin to the phospholipid amino groups of the liposome.
U.S. patent no. 4,786,590 describes the use of monoclonal antibodies which bind to one or more determinant sites of cell surface membrane receptors (CSMR) for directing agents to the site of transformed cells. Disclosed examples of such agents include radionuclides, toxins, and factors involved in complement lysis.
U.S. patent no. 4,701,416 teaches the use of protein carriers to which is conjugated an amino acid sequence which is homologous to at least a portion of gp70 envelope protein of FeLV as a FeLV immunogen. Disclosed examples of such carrier proteins include keyhole limpet hemocyanin, ovalbumin, porcine thyroglobulin, and bovine serum albumin. U.S. patent no. 4,861,588 describes hepatitis B vaccines containing an amino acid chain corresponding to a chain of amino acids in the pre-S region linked to a carrier. Lipid vesicles are described as the preferred carrier.
U.S. patent no. 4,859,769 teaches the use of liposomes which may be tagged with a viral component which recognizes the. second-step receptor on the surface of a cell. The liposome may be used to carry a toxic drug.
U.S. patent no. 4,545,985 discloses a method for modifying Pseudomonas exotoxin for conjugating the exotoxin to a monoclonal antibody which binds to a cell surface membrane receptor to form an immunotoxin.
However, none of the above-described methods is completely satisfactory for preparing targeted retroviral vectors. For example, the methods of U.S. patents 4,731,324 and 4,948,590 rely upon bonding an already produced compound to an existing virus or carrier. Thus, the existence of sufficient amounts of the protein and preformed virus or carrier is required.
Thus, there remains a need for viruses which contain, on their surface, proteins specific for receptors found on the membrane of targeted cells. Further, there remains a need for methods and packaging cell lines to prepare such retroviruses.
Disclosure of the Invention
Accordingly, it is an object of the present invention to provide novel viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which binds specifically to a receptor found on targeted cells. It is another object of the present invention to provide viruses, including retroviruses, which contain on their surface at least one chimeric envelope protein which specifically binds to mammalian, including human, stem and/or progenitor cells.
It is another object of the present invention to provide viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which specifically binds to human hematopoietic stem and/or progenitor cells.
It is another object of the present invention to provide a method for preparing viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which specifically binds to a receptor found on the surface of targeted cells.
It is another object of the present invention to provide a method for preparing viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which specifically binds to mammalian, including human, stem and/or progenitor cells.
It is another object of the present invention to provide a method for preparing viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which specifically binds to human hematopoietic stem and/or progenitor cells.
It is another object of the present invention to provide human stem and/or progenitor cells which have been transduced with a genetically engineered virus, including a retrovirus, containing a therapeutic gene packaged in a virion with a chimeric envelope protein which specifically binds to a receptor found on the membrane of the stem and/or progenitor cell.
It is another object of the present invention to provide a method of gene therapy, in which mammalian, including human, cells are transduced with a virus, including a retrovirus, which possesses on its surface at least one chimeric envelope protein which specifically binds to the transduced cell.
It is another object of the present invention to provide a method of gene therapy, in which human stem and/or progenitor cells are transduced with a virus, including a retrovirus, which possesses on its surface at least one chimeric envelope protein which specifically binds to the human stem and/or progenitor cell.
It is another object of the present invention to provide a method of gene therapy in which human hematopoietic stem cells are transduced with a virus, including a retrovirus, possessing on its surface at least one chimeric envelope protein which specifically binds to the human hematopoietic stem and/or progenitor cells.
It is another object of the present invention to provide packaging cell lines for producing novel viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which binds specifically to a receptor found on targeted cells.
It is another object of the present invention to provide packaging cell lines for producing novel viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which binds specifically to mammalian, including human, stem and/or progenitor cells. It is another object of the present invention to provide packaging cell lines for producing novel viruses, including retroviruses, which possess on their surface at least one chimeric envelope protein which binds specifically to human hematopoietic stem and/or progenitor cells.
These and other objects, which will become apparent during the following detailed description, have been achieved by the inventors' discovery that viruses, including retroviruses, may be genetically engineered such that the viral membrane contains a chimeric envelope peptide which will specifically bind to a targeted cell and that such retroviruses may be used to transduce specific targeted cells.
Best Mode for Carrying Out the Invention
Thus, the present invention relates to the use of a genetically engineered retroviral packaging cell line that has altered the viral envelope such that it contains a peptide which will bind to a molecule on the membrane of the target cell. This results in the creation of stable genetically transformed mammalian, including human, cells. The present invention can be used for the transfer of any genetic information that can be engineered into a recombinant retrovirus, or any other gene transfer vector that requires a virus for delivery. The present invention also relates to a stromal cell line which supports hematopoiesis and is transfected with the retrovirus packaging vectors. Genetically modified human stem cells produced by the present invention can be applied to a wide variety of clinical diseases, as described in the preceding text. Further, the present invention also provides methodology that enhances the efficiency of genetic transfer into human progenitor cells. Additionally, the present invention includes a genetically modified human stem cell produced with the use of the present retroviruses and/or packaging cell line.
Since tissue specificity of retrovirus infection can be conferred by virus envelope (ligand) binding to a cell receptor, and both stem and progenitor cell specific ligands and receptors are known, the construction of chimeric viral envelope/stem cell ligand molecules will confer stem and progenitor cell tropism to virus particles. For example, the HIV viral membrane gene has been cloned, and the envelope amino acids which bind to the CD4 antigen on the T-cell membrane are known (Lasky LA, et al. Cell. 50, 975-985, 1987) . This knowledge provides the information needed to substitute into the gene for the HIV envelope CD4 antigen binding site a sequence that encodes the peptide that is known to bind to the cell membrane of any specific targeted cell of interest. This allows the packaging line to produce virus particles that infect target cells efficiently and selectively, because the virus particles will now encode a chimeric envelope protein which will bind specifically to the stem and/or progenitor cell receptor molecule, and will not bind to cell types which do not have the receptor.
It should be noted that by making these "designer" retroviral packaging cell lines, in vivo gene therapy may be achieved. This is especially true in view of the increased viral titers available using high density cell culture technology. As noted above, conventional packaging cell lines, both the amphotropic and GALV systems, recognize a ubiquitous cell surface protein. If the selective transduction of a target cell is the object, then any attempt to use these systems for in vivo gene therapy would be doomed to failure, because most of the infused virus would bind to irrelevant cells, and these would not be sufficient recombinant virus particles infused to infect a significant number of the target cells. Also, infection of irrelevant cells could lead to unwanted side effects.
However, a "designer" would overcome these deficiencies. Sufficient numbers of these designer virus particles could be infused to infect a large number of just the targeted cell type. It is reiterated that this method is not limited to the hematopoietic system. For example, a "designer" cystic fibrosis (CF) virus recognizing a receptor unique to bronchial epithelium could be delivered in an aerosol to the lung for correction of the CF genetic defect.
In addition, conventional packaging cell lines are all derivatives of the mouse NIH 3T3 cell line. These cells are murine in origin, and as such produce many factors which have either no effect or a deleterious effect on human hematopoietic cells. Furthermore, experiments show that co-culture of the hematopoietic cells with the packaging cell line produces optimal infection. We have evidence that co-culture of human hematopoietic cells with 3T3 cells inhibits stem cell division. Therefore, the present invention includes a human stromal cell line derived from, e.g., fetal liver (which supports hematopoiesis) or adult bone marrow for use as the packaging cell line.
A retrovirus envelope mRNA usually encodes a protein which is cleaved into two peptides. These peptides are inserted into the cell membrane and are associated as a dimer or tetramer. Thus, the viral envelope consists of an intracellular and extracellular domain. The present invention is achieved by genetically engineering the extracellular domain of the retroviral envelope so that it consists of a chimeric molecule which will associate as a dimer with the small envelope peptide. Hence, the viral envelope extracellular domain contains a ligand which will specifically bind to the cell membrane of interest. For instance, when the target cells are human hematopoietic stem cells, the genes for the c-kit ligand, the Leukemic Inhibitory Factor (LIF) , the CS-1 peptide of the alternatively spliced non-type III connecting segment (IIICS) of human plasma fibronectin, or any other ligand which recognizes human stem cells may be substituted for the CD4 binding amino acids in the HIV envelope. Other reports have suggested that the human hematopoietic stem cell most likely has receptors for some or all of these molecules. For other target cells, the appropriate ligand is selected and substituted for all or a portion of the ligand naturally occurring on the virus envelope. The selection of the appropriate ligand for the target cell of interest is well within the abilities of the artisan of ordinary skill.
It should be understood that by substitution of a ligand specific for the target cell receptor it is meant that either the entire protein or a portion thereof may be substituted for a portion of the ligand naturally occurring on the virus envelope. The selection of the particular portion of a ligand responsible for the binding to a target cell and the optimization of the size of the portion of both ligand being replaced and the ligand being inserted are within the capabilities of one of ordinary skill in the art.
The replacement of the portion of the ligand, naturally occurring on the virus envelope and responsible for binding to a cell surface, with the ligand or portion thereof responsible for binding to the target cell is achieved by altering the sequence of the gene encoding the ligand naturally occurring on the virus envelope using recombinant DNA technology. Thus, a portion of the gene which encodes the sequence of the naturally occurring ligand necessary for binding to the natural receptor is excised and replaced with either the DNA sequence encoding the entire ligand which binds to the target cell or any portion of the ligand which results in binding to the target cell.
In a preferred embodiment, the present invention relates to retroviruses which have been engineered to specifically bind to human stem and/or progenitor cells. Human hematopoietic cells, may be isolated from bone marrow, peripheral blood, fetal liver, or umbilical cord blood. It is further preferred that these cells are cultured as described in U.S. Patent Applications Serial Nos. 07/737,024, 07/628,343, and 07/366,639, which are incorporated herein by reference, under conditions in which about 50% of the medium and serum is exchanged daily, and the replacing volume is supplemented with hematopoietic growth factors. Such hematopoietic growth factors may be supplied exogenously by addition of the growth factors to the culture medium or may be present in the medium as a result of the presence of transformed stromal cells in the culture which endogenously produce the growth factors. It should be noted that these conditions result in formation of a stroma during the time of the retroviral infection. In a preferred embodiment, the growth factors are GM-CSF (0.1 to 100 ng/ml/day, preferably about 5 ng/ml/day) plus IL-3 (0.1 to 100 ng/ml/day, preferably about 10 ng/ml/day) with or without IL-lα (10 to 100 U/ml per 3 to 5 day period, preferably about 50 U/ml/4 day period) , with or without c-kit ligand (Mast cell growth factor) (1 to 100 ng/ml/day, preferably about 10 ng/ml/day) , with or without IL-6 (1 to 100 ng/ml/day, preferably about 10 U/ml/day) .
The retroviral infection (transduction) of the cells is preferably carried out as described in U.S. Patent Application Serial No. 07/740,590, which is incorporated herein by reference. By transduction, it is meant the delivery of genes into cells by means of a viral vector. The retroviral infection is performed by including supernatants produced by retroviral packaging lines infected with recombinant retrovirus, during the first days of the culture, or by culturing the cells directly over the infected retroviral packaging lines themselves. In the preferred embodiment, retroviral supernatants are used, and the period of incubation in the presence of virus is about 14 days.
It must be emphasized that infection of human stem and/or progenitor cells with the present retroviruses in conjunction with the rapid perfusion culturing techniques described above results in a high degree of stem and/or progenitor cell infection as a combined result of at least two effects. First, the altered envelope of the present retrovirus affords an enhanced specificity of viral binding to the stem and/or progenitor cells. Second, the rapid medium exchange culture conditions provide an environment in which stem cells remain viable and continue to replicate for extended periods of time (up to at least 5 months) . Both factors contribute to an enhanced efficiency of infection of the stem and/or progenitor cells by the retrovirus.
The retrovirus of the present invention used to infect the target cell should of course also contain within its genome the genetic information encoding the desired therapeutic gene and any required regulatory sequence. The choice of therapeutic gene will depend on the particular disease or condition to be treated. Generally speaking, the therapeutic gene and any attendant regulatory sequences should be no more than approximately 6 to 8 kb in size. Examples of therapeutic genes have been discussed above, and the choice of the particular therapeutic gene is within the abilities of those skilled in the art.
The present retroviruses may be prepared by the following method, which is described in terms of the HIV envelope protein containing the CD4 binding domain. However, it is to be understood that other envelope proteins for which the binding domain is known may take the place of the exemplified HIV envelope protein.
Two plasmids which contain the necessary genes for retrovirus packaging may be constructed. One plasmid may include the GAG, POL, and RT genes, with or without the HTLV TAT and/or REV genes, of either MoMLV, HTLVI, or HIV. The second plasmid may include a genetically modified HIV envelope gene which has the CD4 binding domain (Olshensky U, Helseth E, Furma C, Li J, Haseltine W, Sodroski J, J. Virol.. 64, 5701-5707, 1990) replaced by either the c-kit ligand (Zsebo KM et al. Cell. 63, 195-201, 1990; Anderson KM et al. Cell. 63, 235-243, 1990; Martin FH et al. Cell. 63, 203, 1990), LIF (Moreau J-F, Donaldson DD, Bennet F, Witek-Giannotti J, Clark SC & Wong GG, Nature, 336, 690-692, 1988), the stem cell binding domain of the alternatively spliced fibronectin (Williams DA, Rios M, Stephens C, and Patel VP, Nature, 352:438-441; 1991), or any other ligand of choice or portions thereof. This genetically altered envelope protein will convey the tissue specificity of choice to the retrovirus packaging cell line(s). These plasmids may be transfected into a cell line, for example NIH 3T3 cells or a human stroma cell line described below, to make the packaging cell line.
Furthermore, human fetal liver stroma or bone marrow stroma may be immortalized with the SV40 large T antigen, and a transformed cell line which supports hematopoiesis may then be selected. These cells may then be transfected with the packaging vectors, and a virus producer cell line selected to act as the packaging line.
Thus, the viruses of the present invention are characterized by the presence, on their envelope, of multiple copies of at least one type of chimeric envelope protein which results in the virus binding specifically to a receptor on a cell surface to which the natural virus would not specifically bind. By natural virus it is meant the virus which has on its surface the naturally occurring envelope protein.
In the context of the present invention a ligand (envelope protein) is said to specifically bind to a cell surface receptor, if the ligand binds to that cell surface receptor preferentially in the presence of other cell surface receptors to which it does not specifically bind. Typically, ligands which specifically bind to cell surface receptors do so with an affinity constant of 10~6 to 10~12M.
It should be noted that by the present invention any gene which is inserted into a recombinant retrovirus vector together with suitable promoter and enhancer elements that permit its expression can be incorporated into human hematopoietic stem cells. The achievement of the present invention derives from the conditions that permit stem cell infection through the use of a genetically altered retroviral envelope gene which confers binding specificity for human stem and/or progenitor calls, and the use of a human stromal cell line as the packaging cell line.
Having generally described the present invention, a further understanding can be obtained by reference to certain specific examples which are provided herein for purposes of illustration only and are not intended to be limiting unless otherwise specified.
EXAMPLES
METHODS: The following is a description of how a LIF may be inserted into the HIV envelope GP 120 gene. LIF is known to be found on mouse embryonal stem cells and early hematopoietic cells but not on mature tissue cells. Thus, this protein should have specificity for stem and early progenitor cells. It should be noted that other ligands such as IL1 through 10, GM-CSF, the c-kit ligand, the ligand for Flk 2, etc. can also be used. In addition, just the receptor binding sequences of the ligand may be used, and these sequences may be substituted into different parts of the HIV ENV gene, or into the binding protein (ENV) genes of other retroviruses or other viruses. Furthermore, some or all of the extracellular domain, not including the portion of the extracellular domain which associates with the retrovirus small envelope protein, of the ENV gene may be deleted.
For example, an HIV ENV, LIF chimeric retroviral ENV gene may be made as follows. The HIV ENV coding region may be isolated in two parts by using the polymerase chain reaction (PCR) using specific primers designed to introduce useful restriction endonuclease sites into the ENV gene. Since amino acids 410-421 (Ratner L, et al, Nature. 313, 277-284, 1985), contain sequences essential for CD4 binding (Lasky LA, Cell. 50, 975-985. 1991), they may be deleted to abrogate CD4 binding. The primers may be CCGTCGACATGAGAGTGAAGGAGAAA TATCAGCACTTG and the sequence
CCGGAATTCATGTTTATAATTTGTTTTATTCTGCATGG. The HIV clone pBHIO may be used as a template for the PCR amplification of part of the ENV gene. Two more primers,
CCGGAATTCAGTGGACAAATTAGATGTTCATCAAATATTACAGGG and CCGTCGACTTATAGCAAAATCCTTTCCAAGCCCTGTCT may be used to amplify the HIV clone pBHIO ENV (Hahn B et al, Nature. 312, 166, 1984) . The two PCR products may be digested with the restriction endonuclease Sal 1 and Eco Rl, and then included in a tripartite ligation reaction containing Sal 1 digested pSub 1, a eukaryotic expression vector (Castle VP, Varani J, Fligiel S, Prochownik EV, and Dixit VM, J. Clin. Inv.. 87, 1883-8, 1991), and T4 DNA ligase. The ligation reaction may be transfected into E. coli, and a recombinant plasmid containing the HIV envelope gene with a deletion of the bases encoding amino acids critical for CD4 binding may then be identified. There is now an Eco Rl restriction endonuclease site substituted for the deleted CD4 binding region.
A clone containing only the coding sequences of LIF may be isolated by PCR amplification of LIF (Moreau J-F et al. Nature. 336,690-692, 1988)) using the primers CCGAATTCCAGCCCCCTCCCCATCACCCCTGTCAACGCCACCTGT and CCGAATTCGAAGGCCTGGGCCAACACGGCGATGATCTG. The PCR amplified LIF coding sequence is now flanked by an Eco Rl restriction endonuclease site. The PCR product may then be digested with the restriction endonuclease Eco Rl and ligated into the Eco Rl site which was introduced into the HIV ENV gene. A clone with an in-frame substitution of LIF for the CD4 binding region of the HIV ENV gene may be isolated and transfected into a cell line expressing the GAG POL genes of either an amphotropic MoMLV, or another retrovirus. This results in a packaging cell line which produces virions with a chimeric LIF/HIV envelope when transfected with a recombinant retroviral vector.
Obviously, numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that, within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein.

Claims

Claims
1. A genetically engineered virus comprising a genetically engineered gene and having on its surface at least one chimeric envelope protein which contains a binding region specific for a cell receptor to which the naturally occurring envelope protein of the virus does not specifically bind.
2. The virus of Claim 1, which binds specifically to a single target cell receptor.
3. The virus of Claim 1, wherein said chimeric envelope protein binds specifically to mammalian stem cells, progenitor cells, or mixtures thereof.
4. The virus of Claim 3, wherein said mammalian cells are human cells.
5. The virus of Claim 1, wherein said chimeric envelope protein binds specifically to human hematopoietic stem cells, progenitor cells, or mixtures thereof.
6. A retrovirus packaging cell line, comprising packaging cells which contain the genetic information encoding for a genetically engineered virus which comprises a genetically engineered gene encoding at least one chimeric envelope protein which specifically binds to a target cell receptor to which the naturally occurring envelope protein of the virus does not bind.
7. The packaging cell line of Claim 6, wherein said chimeric envelope protein binds specifically to mammalian stem cells, progenitor cells or mixtures thereof.
8. The packaging cell line of Claim 7, wherein said mammalian cells are human cells.
9. The packaging cell line of Claim 6, wherein said chimeric envelope protein binds specifically to human hematopoietic stem cells, progenitor cells, or mixtures thereof.
10. The packaging cell line of Claim 6, wherein said packaging cells are human stromal cells.
11. In a method of gene therapy, comprising transducing mammalian cells with a retrovirus comprising the genetic information for a therapeutic gene, the improvement being that said retrovirus is packaged in a virion comprising a chimeric envelope protein which binds specifically to a cell receptor to which the naturally occurring envelope protein of the retrovirus does not bind.
12. The method of Claim 11, wherein said mammalian cells are human cells.
13. The method of Claim 12, wherein said human cells are stem cells, progenitor cells, or mixtures thereof, and said chimeric envelope protein binds specifically to human stem cells, progenitor cells, or mixtures thereof.
14. The method of Claim 12, wherein said human cells are human hematopoietic stem cells, progenitor cells, or mixtures thereof, and said chimeric envelope protein binds specifically to human hematopoietic stem cells, progenitor cells, or mixtures thereof.
15. The method of Claim 11, wherein said transducing is carried out by exposing said cells to said virus, while said cells are being cultured by a method comprising culturing said cells in a liquid culture medium which is replaced at a rate of about 1 ml of medium per 1 ml of culture per about 24 to about 48 hours, and removing metabolic products and replenishing depleted nutrients, while maintaining said culture under physiologically acceptable conditions.
16. The method of Claim 15, wherein said cells are human cells.
17. The method of Claim 11, wherein said transducing is carried out in vivo.
18. Human stem and/or progenitor cells which have been transduced with a genetically engineered virus comprising a genetically engineered gene packaged in virion with a chimeric envelope protein which contains a binding region specific for a cell receptor to which the naturally occurring envelope protein of the virus does not specifically bind.
PCT/US1993/000072 1992-01-17 1993-01-13 Targeted virus WO1993014188A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US82213692A 1992-01-17 1992-01-17
US822,136 1992-01-17

Publications (1)

Publication Number Publication Date
WO1993014188A1 true WO1993014188A1 (en) 1993-07-22

Family

ID=25235257

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/000072 WO1993014188A1 (en) 1992-01-17 1993-01-13 Targeted virus

Country Status (2)

Country Link
AU (1) AU3434393A (en)
WO (1) WO1993014188A1 (en)

Cited By (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994006920A1 (en) * 1992-09-22 1994-03-31 Medical Research Council Recombinant viruses displaying a nonviral polypeptide on their external surface
EP0633943A1 (en) * 1992-04-03 1995-01-18 Alexander T. YOUNG Gene therapy using targeted viral vectors
WO1996009400A1 (en) * 1994-09-19 1996-03-28 Systemix, Inc. Methods for genetically modifying hematopoietic stem cells
EP0746625A1 (en) * 1992-11-09 1996-12-11 UNITED STATES GOVERNMENT as represented by THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES Targetable vector particles
US5643756A (en) * 1992-08-28 1997-07-01 The Public Health Research Institute Of The City Of New York, Inc. Fusion glycoproteins
US5733732A (en) * 1996-01-03 1998-03-31 University Of Iowa Research Foundation Methods for detecting primary adhalinopathy
US5928638A (en) * 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
WO1999038008A1 (en) * 1998-01-23 1999-07-29 Prolifaron, Inc. Methods and compositions for the identification of growth factor mimetics, growth factors and inhibitors
WO1999051748A2 (en) 1998-04-07 1999-10-14 Corixa Corporation Fusion proteins of mycobacterium tuberculosis antigens and their uses
WO1999055893A1 (en) * 1998-04-29 1999-11-04 University Of Southern California Retroviral vectors including modified envelope escort proteins
WO1999061639A2 (en) * 1998-05-22 1999-12-02 Oxford Biomedica (Uk) Limited Retroviral delivery system
US6004798A (en) * 1997-05-14 1999-12-21 University Of Southern California Retroviral envelopes having modified hypervariable polyproline regions
US6074850A (en) * 1996-11-15 2000-06-13 Canji, Inc. Retinoblastoma fusion polypeptides
WO2002002641A1 (en) 2000-06-16 2002-01-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
US6379927B1 (en) 1996-11-15 2002-04-30 Canji, Inc. Retinoblastoma fusion proteins
US6392069B2 (en) 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
WO2002097033A2 (en) 2001-05-25 2002-12-05 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
WO2003086458A1 (en) 2002-04-12 2003-10-23 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
US6699656B2 (en) 1996-06-24 2004-03-02 University Of Maryland Biotechnology Institute Treatment and prevention of HIV infection by administration of derivatives of human chorionic gonadotropin
US6759393B1 (en) 1999-04-12 2004-07-06 Pfizer Inc. Growth hormone and growth hormone releasing hormone compositions
US6818209B1 (en) 1998-05-22 2004-11-16 Oxford Biomedica (Uk) Limited Retroviral delivery system
WO2005001038A2 (en) 2003-05-28 2005-01-06 Seattle Genetics, Inc. Recombinant anti-cd30 antibodies and uses thereof
US6849399B1 (en) 1996-05-23 2005-02-01 Bio-Rad Laboratories, Inc. Methods and compositions for diagnosis and treatment of iron misregulation diseases
US6864082B2 (en) 1997-04-10 2005-03-08 University Of Southern California Modified viral surface proteins for binding to extracellular matrix components
WO2005032572A2 (en) 2003-10-03 2005-04-14 Vib Vzw Means and methods for the recruitment and identification of stem cells
WO2005042708A2 (en) 2003-10-27 2005-05-12 Rosetta Inpharmatics Llc METHOD OF DESIGNING siRNAS FOR GENE SILENCING
US6974572B2 (en) 2001-05-22 2005-12-13 Pfizer, Inc. Non-anaphylactogenic IgE fusion proteins
WO2006001888A2 (en) 2004-04-16 2006-01-05 Acuity Pharmaceuticals Inc Compositions and methods for inhibiting angiogenesis
US7002027B1 (en) 1996-01-08 2006-02-21 Canji, Inc. Compositions and methods for therapeutic use
US7026116B1 (en) 1996-04-04 2006-04-11 Bio-Rad Laboratories, Inc. Polymorphisms in the region of the human hemochromatosis gene
WO2006046994A2 (en) 2004-07-30 2006-05-04 Mount Sinai School Of Medicine Of New York University Klf6 alternative splice forms and a germline klf6 dna polymorphism associated with increased cancer risk
US7067255B2 (en) 1996-04-04 2006-06-27 Bio-Rad Laboratories, Inc. Hereditary hemochromatosis gene
WO2006069253A2 (en) 2004-12-22 2006-06-29 Auckland Uniservices Limited Trefoil factors and methods of treating proliferation disorders using same
US7078483B2 (en) 1998-04-29 2006-07-18 University Of Southern California Retroviral vectors including modified envelope escort proteins
WO2006091871A1 (en) 2005-02-23 2006-08-31 Halozyme Therapeutics, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2006102095A2 (en) 2005-03-18 2006-09-28 Medimmune, Inc. Framework-shuffling of antibodies
US7276364B1 (en) 1999-11-18 2007-10-02 Dendreon Corporation Nucleic acids encoding endotheliases, endotheliases and uses thereof
US7393534B2 (en) 2003-07-15 2008-07-01 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
WO2008105797A2 (en) 2006-06-30 2008-09-04 Bristol-Myers Squibb Company Polynucleotides encoding novel pcsk9 variants
EP1967205A2 (en) 2000-08-30 2008-09-10 Pfizer Products Inc. Anti-IgE vaccines
EP1978098A2 (en) 1999-12-10 2008-10-08 Invitrogen Corporation Use of multiple recombination sites with unique specificity in recombinational cloning
US7449562B2 (en) 2001-06-29 2008-11-11 Novartis Ag PERV screening method and use thereof
WO2008157379A2 (en) 2007-06-21 2008-12-24 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2009002193A1 (en) 2007-06-27 2008-12-31 Auckland Uniservices Limited Polypeptides and polynucleotides for artemin and related ligands, and methods of use thereof
EP2014674A1 (en) 2001-11-26 2009-01-14 Cellvir Protein-protein interactions in human immunodeficiency virus
EP2027874A2 (en) 2000-11-28 2009-02-25 Medimmune, Inc. Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
US7498314B2 (en) 2001-05-03 2009-03-03 Fit Biotech Oyj Plc Expression vectors and uses thereof
US7566452B1 (en) 1999-05-04 2009-07-28 New York University Cancer treatment with endothelin receptor antagonists
WO2009102488A2 (en) 2008-02-15 2009-08-20 Tufts University A humanized model of membrane attack complex (mac) formation on murine retina and compositions, kits and methods for treatment of macular degeneration
WO2009123894A2 (en) 2008-04-02 2009-10-08 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
EP2128270A1 (en) 2003-08-08 2009-12-02 Genenews Inc. Osteoarthritis biomarkers and uses thereof
WO2009151717A2 (en) 2008-04-02 2009-12-17 Macrogenics, Inc. Bcr-complex-specific antibodies and methods of using same
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
EP2163643A1 (en) 2003-03-05 2010-03-17 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
WO2010033279A2 (en) 2008-06-04 2010-03-25 Macrogenics, Inc. Antibodies with altered binding to fcrn and methods of using same
US7700341B2 (en) 2000-02-03 2010-04-20 Dendreon Corporation Nucleic acid molecules encoding transmembrane serine proteases, the encoded proteins and methods based thereon
WO2010072684A1 (en) 2008-12-22 2010-07-01 Universität Regensburg Norrin in the treatment of diseases associated with an increased tgf-beta activity
WO2010080538A1 (en) 2008-12-19 2010-07-15 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2218779A1 (en) 2002-12-16 2010-08-18 Halozyme, Inc. Human chondroitinase glycoprotein (chasegp), process for preparing the same, and pharmaceutical compositions comprising thereof
EP2228389A2 (en) 2001-04-13 2010-09-15 Human Genome Sciences, Inc. Antibodies against vascular endothelial growth factor 2
WO2010124365A1 (en) 2009-04-27 2010-11-04 Ottawa Hospital Research Institute Compositions and methods for modulating stem cells and uses thereof
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
EP2272566A2 (en) 2003-08-18 2011-01-12 MedImmune, LLC Humanisation of antibodies
WO2011020079A1 (en) 2009-08-13 2011-02-17 Calmune Corporation Antibodies against human respiratory syncytial virus (rsv) and methods of use
EP2292663A2 (en) 2006-08-28 2011-03-09 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2298869A1 (en) 2003-06-13 2011-03-23 University Of Medicine And Dentistry Of New Jersey Recombinant protein production in the presence of mRNA interferase
WO2011035205A2 (en) 2009-09-18 2011-03-24 Calmune Corporation Antibodies against candida, collections thereof and methods of use
EP2308996A1 (en) 1998-03-30 2011-04-13 NorthWest Biotherapeutics, Inc. Therapeutic and diagnostic applications based on the role of the CXCR-4 and SDF-1 genes in tumourigenesis
US7928189B2 (en) 2008-05-05 2011-04-19 Ottawa Health Research Institute PCSK9 polypeptide fragment
WO2011046457A1 (en) 2009-10-16 2011-04-21 Auckland Uniservices Limited Anti-neoplastic uses of artemin antagonists
EP2316487A1 (en) 2003-04-11 2011-05-04 MedImmune, LLC Recombinant IL-9 antibodies & uses thereof
EP2319941A2 (en) 2005-10-21 2011-05-11 GeneNews Inc. Method and apparatus for correlating levels of biomarker products with disease
WO2011057188A1 (en) 2009-11-06 2011-05-12 Idexx Laboratories, Inc. Canine anti-cd20 antibodies
US7964708B2 (en) 2006-11-15 2011-06-21 Limin Li Anti-TSG101 antibodies and their uses for treatment of viral infections
US7973139B2 (en) 2004-03-26 2011-07-05 Human Genome Sciences, Inc. Antibodies against nogo receptor
EP2341060A1 (en) 2000-12-12 2011-07-06 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
EP2351584A1 (en) 2003-12-23 2011-08-03 Genentech, Inc. Novel anti-IL 13 antibodies and uses thereof
EP2357192A1 (en) 1999-02-26 2011-08-17 Human Genome Sciences, Inc. Human endokine alpha and methods of use
EP2360476A1 (en) 2002-02-13 2011-08-24 American Diagnostica Inc. Methods for selecting treatment regimens and predicting outcomes in cancer patients
WO2011101031A1 (en) 2010-02-19 2011-08-25 Université de Liège A polynucleotide for use in treatment of influenza a virus induced diseases, encoding modified mx protein, said modified mx protein, and a transgenic animal expressing gene encoding modified mx protein
US8021833B2 (en) 2003-02-12 2011-09-20 Functional Genetics, Inc. Method for reducing HIV viral budding by administering a VPS28-specfic antibody that disrupts Gag-TSG101-VPS28 binding interactions
US8022189B2 (en) 1998-08-21 2011-09-20 Albany Medical College Isolated antibodies against biologically active leptin-related peptides
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2371389A2 (en) 2002-08-14 2011-10-05 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
EP2383350A1 (en) 2004-05-07 2011-11-02 Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Methods of diagnosing or treating prostate cancer using the erg gene, alone or in combination with other over or under expressed genes in prostate cancer
EP2385124A2 (en) 1999-05-14 2011-11-09 Arbor Vita Corporation Peptides or peptide analogues for modulating the binding of a PDZ protein and a PL protein
EP2387995A1 (en) 2006-03-30 2011-11-23 PTC Therapeutics, Inc. Methods for the production of functional protein from DNA having a nonsense mutation and the treatment of disorders associated therewith
US8088382B2 (en) 2005-07-05 2012-01-03 Cornell Research Foundation, Inc. Methods of inhibiting transendothelial migration of neutrophils and monocytes with anti-CD99L2 antibodies
WO2012006596A2 (en) 2010-07-09 2012-01-12 Calmune Corporation Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
WO2012013249A1 (en) 2010-07-30 2012-02-02 Université de Liège Dentin matrix protein 1 (dmp1) for use in pharmaceutical compositions
WO2012018687A1 (en) 2010-08-02 2012-02-09 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2422811A2 (en) 2004-10-27 2012-02-29 MedImmune, LLC Modulation of antibody specificity by tailoring the affinity to cognate antigens
EP2431054A2 (en) 2000-06-15 2012-03-21 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
EP2453024A2 (en) 2004-06-21 2012-05-16 The Board of Trustees of The Leland Stanford Junior University Genes and pathways differentially expressed in bipolar disorder and/or major depressive disorder
US8211858B2 (en) 2007-04-27 2012-07-03 The University Of Toledo Modified plasminogen activator inhibitor type-1 molecule and methods based thereon
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
EP2500030A2 (en) 2005-11-04 2012-09-19 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP2505209A1 (en) 2006-06-26 2012-10-03 MacroGenics, Inc. Fcgamma-RIIB-specific antibodies and methods of the use thereof
EP2518163A2 (en) 2006-10-10 2012-10-31 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Prostate cancer specific alterations in erg gene expression and detection and treatment methods based on those alterations
EP2520669A2 (en) 2005-02-07 2012-11-07 GeneNews Inc. Mild osteoathritis biomarkers and uses thereof
WO2012162068A2 (en) 2011-05-21 2012-11-29 Macrogenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
EP2564864A2 (en) 2005-11-12 2013-03-06 The Board of Trustees of the Leland FGF2-related methods for diagnosing and treating depression
WO2013040341A2 (en) 2011-09-16 2013-03-21 Ottawa Hospital Research Institute Wnt7a compositions and methods of using the same
EP2573114A1 (en) 2005-08-10 2013-03-27 MacroGenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2610267A1 (en) 2006-12-18 2013-07-03 Genentech, Inc. Antagonist anti-Notch3 antibodies and their use in the prevention and treatment of Notch3-related diseases
WO2014006063A2 (en) 2012-07-02 2014-01-09 Medizinische Universität Wien Complement split product c4d for the treatment of inflammatory conditions
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US8809287B2 (en) 2004-11-15 2014-08-19 Icahn School Of Medicine At Mount Sinai Compositions and methods for altering Wnt autocrine signaling
US8877896B2 (en) 2008-02-15 2014-11-04 Tufts University Compositions, methods and kits for modeling, diagnosing, and treating complement disorders
US8916517B2 (en) 2009-11-02 2014-12-23 The Administrators Of The Tulane Educational Fund Analogs of pituitary adenylate cyclase-activating polypeptide (PACAP) and methods for their use
US8937169B2 (en) 1996-01-11 2015-01-20 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9006181B2 (en) 2004-07-21 2015-04-14 The Administrators Of The Tulane Educational Fund Treatment of renal dysfunction and multiple myeloma using PACAP compounds
EP2932982A1 (en) 2005-05-17 2015-10-21 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
US9211315B2 (en) 2004-03-05 2015-12-15 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
US9447454B2 (en) 2003-10-23 2016-09-20 The Rockefeller University Method of purifying RNA binding protein-RNA complexes
US9458464B2 (en) 2014-06-23 2016-10-04 The Johns Hopkins University Treatment of neuropathic pain
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
WO2017182981A1 (en) 2016-04-20 2017-10-26 Washington University Ppar agonist or lxr agonist for use in the treatment of systemic lupus erythematosus by modulation of lap activity
US9873722B2 (en) 2011-09-16 2018-01-23 Fate Therapeutics, Inc. Wnt compositions and therapeutic uses of such compositions
US9920100B2 (en) 2015-06-05 2018-03-20 The Chinese University Of Hong Kong Mimotopes of tropomyosin for use in immunotherapy for shellfish and/or arthropod allergy
EP3320906A1 (en) 2012-10-26 2018-05-16 The Chinese University of Hong Kong Treatment of melanoma and lung carcinoma using a smad3 inhibitor
EP3375868A1 (en) 2007-11-08 2018-09-19 Dana Farber Cancer Institute, Inc. Stimulation of anti-tumor immunity using dendritic cell/tumor cell fusions and anti-cd3/cd28
US10130687B2 (en) 2011-01-05 2018-11-20 Rhode Island Hospital Compositions and methods for the treatment of orthopedic disease or injury
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
EP3450571A1 (en) 2014-02-24 2019-03-06 Celgene Corporation Methods of using an activator of cereblon for neural cell expansion and the treatment of central nervous system disorders
EP3479844A1 (en) 2005-04-15 2019-05-08 MacroGenics, Inc. Covalent diabodies and uses thereof
US10351617B2 (en) 2010-08-13 2019-07-16 Trustees Of Tufts College Compositions, kits and methods for treatment of complement-related disorders
WO2019157447A1 (en) 2018-02-12 2019-08-15 Trustees Of Tufts College Cd59 for inhibiting inflammasome activation
US10570200B2 (en) 2013-02-01 2020-02-25 California Institute Of Technology Antibody-mediated immunocontraception
WO2020097261A1 (en) 2018-11-06 2020-05-14 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services New compositions and methods for treating beta-globinopathies
US10729790B2 (en) 2015-05-26 2020-08-04 Salk Institute For Biological Studies Motor neuron-specific expression vectors
US10813977B2 (en) 2014-08-28 2020-10-27 Trustees Of Tufts College Compositions, methods and kits for treating complement related disorders
WO2021138581A1 (en) 2020-01-03 2021-07-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tnap locally administered for promoting periodontal health
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
WO2022157548A1 (en) 2021-01-24 2022-07-28 Forrest Michael David Inhibitors of atp synthase - cosmetic and therapeutic uses
WO2022240824A1 (en) 2021-05-13 2022-11-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for treating sickle cell diseases
WO2023004332A2 (en) 2021-07-19 2023-01-26 New York University Adeno-associated viral vector compositions and methods of promoting muscle regeneration
WO2023064255A2 (en) 2021-10-15 2023-04-20 The Regents Of The University Of California Diagnosis and treatment of anti-pf4 induced thrombocytopenia
WO2023081167A2 (en) 2021-11-02 2023-05-11 The Regents Of The University Of California P-selectin mutants and modulation of integrin-mediated signaling
WO2023089564A1 (en) 2021-11-19 2023-05-25 Janssen Biotech, Inc. Method of treating geographic atrophy with a gene therapy vector expressing soluble cd59
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023146807A1 (en) 2022-01-25 2023-08-03 The Regents Of The University Of California Vegf mutants and modulation of integrin-mediated signaling
WO2023200897A1 (en) 2022-04-13 2023-10-19 The Regents Of The University Of California Use of viral il-6 in cancer therapy
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BLOOD, Volume 69, No. 2, issued February 1987, HOGGE et al., "Gene Transfer to Primary Normal and Malignant Human Hemotopoietic Progenitors Using Recombinant Retroviruses", pages 611-617. *
CELL, Volume 63, issued 05 October 1990, F.H. MARTIN et al., "Primary Structure and Functional Expression of Rat and Human Stem Cell Factor cDNAs", pages 203-211. *
JOURNAL OF VIROLOGY, Volume 63, No. 5, issued May 1989, "Formation of Infectious Hybrid Virions with Gibbon Ape Leukemia Virus and Human T-Cell Leukemia Virus Retroviral Envelope Glycoproteins and the gag and pol Proteins of Moloney Murine Leukemia Virus", pages 2374-2378. *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, Volume 87, issued January 1990, J.M. WILSON et al., "Expression of Human Adenosine Deaminase in Mice Reconstituted with Retrovirus-Transduced Hematopoietic Stem Cells", pages 439-443. *

Cited By (261)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0633943A4 (en) * 1992-04-03 1997-05-02 Alexander T Young Gene therapy using targeted viral vectors.
EP0633943A1 (en) * 1992-04-03 1995-01-18 Alexander T. YOUNG Gene therapy using targeted viral vectors
US5952474A (en) * 1992-08-28 1999-09-14 The Public Health Research Institute Of The City Of New York, Inc. Fusion glycoproteins
US5643756A (en) * 1992-08-28 1997-07-01 The Public Health Research Institute Of The City Of New York, Inc. Fusion glycoproteins
WO1994006920A1 (en) * 1992-09-22 1994-03-31 Medical Research Council Recombinant viruses displaying a nonviral polypeptide on their external surface
US5723287A (en) * 1992-09-22 1998-03-03 Medical Research Council Recombinant viruses displaying a nonviral polypeptide on their external surface
US6297004B1 (en) 1992-09-22 2001-10-02 Cambridge Drug Discovery Holding, Ltd Recombinant viruses displaying a nonviral polypeptide on their external surface
EP0746625A4 (en) * 1992-11-09 1997-05-02 Us Health Targetable vector particles
EP0746625A1 (en) * 1992-11-09 1996-12-11 UNITED STATES GOVERNMENT as represented by THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES Targetable vector particles
US5985655A (en) * 1992-11-09 1999-11-16 The United States Of America As Represented By The Department Of Health And Human Sevices Targetable vector particles
US6503501B1 (en) 1992-11-09 2003-01-07 W. French Anderson Targetable vector particles
WO1996009400A1 (en) * 1994-09-19 1996-03-28 Systemix, Inc. Methods for genetically modifying hematopoietic stem cells
US5733732A (en) * 1996-01-03 1998-03-31 University Of Iowa Research Foundation Methods for detecting primary adhalinopathy
US7534769B2 (en) 1996-01-08 2009-05-19 Canji, Inc. Compositions and methods for enhancing delivery of therapeutic agents to cells
US6392069B2 (en) 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
US7002027B1 (en) 1996-01-08 2006-02-21 Canji, Inc. Compositions and methods for therapeutic use
US8937169B2 (en) 1996-01-11 2015-01-20 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US7579169B2 (en) 1996-04-04 2009-08-25 Bio-Rad Laboratories, Inc. Hereditary hemochromatosis gene
US7067255B2 (en) 1996-04-04 2006-06-27 Bio-Rad Laboratories, Inc. Hereditary hemochromatosis gene
US7052845B2 (en) 1996-04-04 2006-05-30 Bio-Rad Laboratories, Inc. Polymorphisms in the region of the human hemochromatosis gene
US7595385B2 (en) 1996-04-04 2009-09-29 Bio-Rad Laboratories, Inc. Polymorphisms in the region of the human hemochromatosis gene
US8257927B2 (en) 1996-04-04 2012-09-04 Bio-Rad Laboratories, Inc. Hereditary hemochromatosis gene
US7026116B1 (en) 1996-04-04 2006-04-11 Bio-Rad Laboratories, Inc. Polymorphisms in the region of the human hemochromatosis gene
US7998680B2 (en) 1996-04-04 2011-08-16 Bio-Rad Laboratories, Inc. Determining genotype of a polymorphic site in the hereditary hemochromatosis gene
US6849399B1 (en) 1996-05-23 2005-02-01 Bio-Rad Laboratories, Inc. Methods and compositions for diagnosis and treatment of iron misregulation diseases
US5928638A (en) * 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
US6699656B2 (en) 1996-06-24 2004-03-02 University Of Maryland Biotechnology Institute Treatment and prevention of HIV infection by administration of derivatives of human chorionic gonadotropin
US6379927B1 (en) 1996-11-15 2002-04-30 Canji, Inc. Retinoblastoma fusion proteins
US6074850A (en) * 1996-11-15 2000-06-13 Canji, Inc. Retinoblastoma fusion polypeptides
US6902731B1 (en) 1996-11-15 2005-06-07 Canji, Inc. Methods of treating hyperproliferative disorders using retinoblastoma fusion proteins
US7347998B2 (en) 1997-04-10 2008-03-25 University Of Southern California Method of delivering therapeutic agents to site of tissue injury
US6864082B2 (en) 1997-04-10 2005-03-08 University Of Southern California Modified viral surface proteins for binding to extracellular matrix components
US8530441B2 (en) 1997-04-10 2013-09-10 University Of Southern California Transgene delivering retrovirus targeting collagen exposed at site of tissue injury
US8148509B2 (en) 1997-04-10 2012-04-03 University Of Southern California Transgene delivering retrovirus targeting collagen exposed at site of tissue injury
US8871734B2 (en) 1997-04-10 2014-10-28 The University Of Southern California Transgene delivering retrovirus targeting collagen exposed at site of tissue injury
US7820157B2 (en) 1997-04-10 2010-10-26 University Of Southern California Transgene delivering retrovirus targeting collagen exposed at site of tissue injury
US6004798A (en) * 1997-05-14 1999-12-21 University Of Southern California Retroviral envelopes having modified hypervariable polyproline regions
EP2106807A1 (en) 1997-07-08 2009-10-07 CANJI, Inc. Compositions and kits for enhancing delivery of therapeutic agents to cells
WO1999038008A1 (en) * 1998-01-23 1999-07-29 Prolifaron, Inc. Methods and compositions for the identification of growth factor mimetics, growth factors and inhibitors
EP2308996A1 (en) 1998-03-30 2011-04-13 NorthWest Biotherapeutics, Inc. Therapeutic and diagnostic applications based on the role of the CXCR-4 and SDF-1 genes in tumourigenesis
WO1999051748A2 (en) 1998-04-07 1999-10-14 Corixa Corporation Fusion proteins of mycobacterium tuberculosis antigens and their uses
US7078483B2 (en) 1998-04-29 2006-07-18 University Of Southern California Retroviral vectors including modified envelope escort proteins
WO1999055893A1 (en) * 1998-04-29 1999-11-04 University Of Southern California Retroviral vectors including modified envelope escort proteins
GB2351290A (en) * 1998-05-22 2000-12-27 Oxford Biomedica Ltd Retroviral delivery sytem
US6818209B1 (en) 1998-05-22 2004-11-16 Oxford Biomedica (Uk) Limited Retroviral delivery system
WO1999061639A2 (en) * 1998-05-22 1999-12-02 Oxford Biomedica (Uk) Limited Retroviral delivery system
WO1999061639A3 (en) * 1998-05-22 2000-01-27 Oxford Biomedica Ltd Retroviral delivery system
US8022189B2 (en) 1998-08-21 2011-09-20 Albany Medical College Isolated antibodies against biologically active leptin-related peptides
US8067545B2 (en) 1998-08-21 2011-11-29 Albany Medical College Isolated antibodies against biologically active leptin-related peptides
EP2357192A1 (en) 1999-02-26 2011-08-17 Human Genome Sciences, Inc. Human endokine alpha and methods of use
US6759393B1 (en) 1999-04-12 2004-07-06 Pfizer Inc. Growth hormone and growth hormone releasing hormone compositions
US8597645B2 (en) 1999-05-04 2013-12-03 New York University Cancer treatment with endothelin receptor antagonists
US8067000B2 (en) 1999-05-04 2011-11-29 New York University Cancer treatment with endothelin receptor antagonists
US9125897B2 (en) 1999-05-04 2015-09-08 New York University Cancer treatment with endothelin receptor antagonists
US7566452B1 (en) 1999-05-04 2009-07-28 New York University Cancer treatment with endothelin receptor antagonists
EP2385124A2 (en) 1999-05-14 2011-11-09 Arbor Vita Corporation Peptides or peptide analogues for modulating the binding of a PDZ protein and a PL protein
US7276364B1 (en) 1999-11-18 2007-10-02 Dendreon Corporation Nucleic acids encoding endotheliases, endotheliases and uses thereof
EP1978098A2 (en) 1999-12-10 2008-10-08 Invitrogen Corporation Use of multiple recombination sites with unique specificity in recombinational cloning
EP2210948A2 (en) 1999-12-10 2010-07-28 Life Technologies Corporation Use of multiple recombination sites with unique specificity in recombinational cloning
US7700341B2 (en) 2000-02-03 2010-04-20 Dendreon Corporation Nucleic acid molecules encoding transmembrane serine proteases, the encoded proteins and methods based thereon
EP2431054A2 (en) 2000-06-15 2012-03-21 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
EP2281843A1 (en) 2000-06-16 2011-02-09 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP2281842A1 (en) 2000-06-16 2011-02-09 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to BLyS
WO2002002641A1 (en) 2000-06-16 2002-01-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP2275449A1 (en) 2000-06-16 2011-01-19 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP1967206A2 (en) 2000-08-30 2008-09-10 Pfizer Products Inc. Anti-IgE vaccines
EP1967205A2 (en) 2000-08-30 2008-09-10 Pfizer Products Inc. Anti-IgE vaccines
US8273356B2 (en) 2000-08-30 2012-09-25 Pfizer Inc. Anti-IgE vaccines
EP2065052A2 (en) 2000-08-30 2009-06-03 Pfizer Products Inc. Anti-IgE vaccines
US7897151B2 (en) 2000-08-30 2011-03-01 Pharmacia & Upjohn Company, Llc Anti-IgE vaccines
EP2361635A2 (en) 2000-08-30 2011-08-31 Pfizer Products Inc. Anti IgE vaccines
EP2338512A1 (en) 2000-11-28 2011-06-29 MedImmune, LLC Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
EP2027874A2 (en) 2000-11-28 2009-02-25 Medimmune, Inc. Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
EP2412384A1 (en) 2000-11-28 2012-02-01 MedImmune, LLC Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
EP2341060A1 (en) 2000-12-12 2011-07-06 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
EP2354149A1 (en) 2000-12-12 2011-08-10 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
EP3569610A2 (en) 2000-12-12 2019-11-20 Medlmmune, LLC Molecules with extended half lives, compositions and uses thereof
EP2357187A1 (en) 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US8231878B2 (en) 2001-03-20 2012-07-31 Cosmo Research & Development S.P.A. Receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
EP2228389A2 (en) 2001-04-13 2010-09-15 Human Genome Sciences, Inc. Antibodies against vascular endothelial growth factor 2
US9725486B2 (en) 2001-05-03 2017-08-08 Fit Biotech Oy Methods of treating HIV diseases using novel expression vectors
US7510718B2 (en) 2001-05-03 2009-03-31 Fit Biotech Oyj Plc Expression vectors and uses thereof
US7498314B2 (en) 2001-05-03 2009-03-03 Fit Biotech Oyj Plc Expression vectors and uses thereof
US7420047B2 (en) 2001-05-22 2008-09-02 Pfizer Inc. Non-anaphylactogenic IgE fusion proteins
US6974572B2 (en) 2001-05-22 2005-12-13 Pfizer, Inc. Non-anaphylactogenic IgE fusion proteins
WO2002097033A2 (en) 2001-05-25 2002-12-05 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
US7449562B2 (en) 2001-06-29 2008-11-11 Novartis Ag PERV screening method and use thereof
EP2014674A1 (en) 2001-11-26 2009-01-14 Cellvir Protein-protein interactions in human immunodeficiency virus
EP2360476A1 (en) 2002-02-13 2011-08-24 American Diagnostica Inc. Methods for selecting treatment regimens and predicting outcomes in cancer patients
EP2270049A2 (en) 2002-04-12 2011-01-05 Medimmune, Inc. Recombinant anti-interleukin-9-antibody
WO2003086458A1 (en) 2002-04-12 2003-10-23 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
EP2371389A2 (en) 2002-08-14 2011-10-05 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
EP2218779A1 (en) 2002-12-16 2010-08-18 Halozyme, Inc. Human chondroitinase glycoprotein (chasegp), process for preparing the same, and pharmaceutical compositions comprising thereof
EP2298874A1 (en) 2002-12-16 2011-03-23 Halozyme, Inc. Human chondroitinase glycoprotein (CHASEGP), process for preparing the same, and pharmaceutical compositions comprising thereof
US8815558B2 (en) 2002-12-16 2014-08-26 Halozyme, Inc. Human chondroitinase glycoprotein (CHASEGP), process for preparing the same, and pharmaceutical compositions comprising thereof
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8021833B2 (en) 2003-02-12 2011-09-20 Functional Genetics, Inc. Method for reducing HIV viral budding by administering a VPS28-specfic antibody that disrupts Gag-TSG101-VPS28 binding interactions
EP2177620A1 (en) 2003-03-05 2010-04-21 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
EP2311973A1 (en) 2003-03-05 2011-04-20 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US9677061B2 (en) 2003-03-05 2017-06-13 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US11723959B2 (en) 2003-03-05 2023-08-15 Halozyme, Inc. Preparation of mammalian oocyte for fertilization via a soluble human PH20 hyaluronidase polypeptide
EP2405015A2 (en) 2003-03-05 2012-01-11 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US9677062B2 (en) 2003-03-05 2017-06-13 Halozyme, Inc. Hyaluronidase and factor VIII compositions
EP2330213A1 (en) 2003-03-05 2011-06-08 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US9562223B2 (en) 2003-03-05 2017-02-07 Halozyme, Inc. Methods for reducing intraocular pressure by administering a soluble hyaluronidase glycoprotein (sHASEGP)
EP2163643A1 (en) 2003-03-05 2010-03-17 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
EP3009517A1 (en) 2003-03-05 2016-04-20 Halozyme, Inc. Soluble hyaluronidase glycoprotein (shasegp), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US10898551B2 (en) 2003-03-05 2021-01-26 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
US10286044B2 (en) 2003-03-05 2019-05-14 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
EP2316487A1 (en) 2003-04-11 2011-05-04 MedImmune, LLC Recombinant IL-9 antibodies & uses thereof
WO2005001038A2 (en) 2003-05-28 2005-01-06 Seattle Genetics, Inc. Recombinant anti-cd30 antibodies and uses thereof
EP2302040A1 (en) 2003-06-13 2011-03-30 University Of Medicine And Dentistry Of New Jersey Medical use of mRNA interferase
EP2298869A1 (en) 2003-06-13 2011-03-23 University Of Medicine And Dentistry Of New Jersey Recombinant protein production in the presence of mRNA interferase
US8257714B2 (en) 2003-07-15 2012-09-04 Michigan State University Compositions and methods for immunotherapy of cancer and infectious diseases
US7393534B2 (en) 2003-07-15 2008-07-01 Barros Research Institute Compositions and methods for immunotherapy of cancer and infectious diseases
EP2128270A1 (en) 2003-08-08 2009-12-02 Genenews Inc. Osteoarthritis biomarkers and uses thereof
EP2272566A2 (en) 2003-08-18 2011-01-12 MedImmune, LLC Humanisation of antibodies
WO2005032572A2 (en) 2003-10-03 2005-04-14 Vib Vzw Means and methods for the recruitment and identification of stem cells
US8003096B2 (en) 2003-10-03 2011-08-23 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw Means and methods for the recruitment and identification of stem cells
US9447454B2 (en) 2003-10-23 2016-09-20 The Rockefeller University Method of purifying RNA binding protein-RNA complexes
WO2005042708A2 (en) 2003-10-27 2005-05-12 Rosetta Inpharmatics Llc METHOD OF DESIGNING siRNAS FOR GENE SILENCING
EP2805728A1 (en) 2003-12-23 2014-11-26 Genentech, Inc. Novel anti-IL 13 antibodies and uses thereof
EP2351584A1 (en) 2003-12-23 2011-08-03 Genentech, Inc. Novel anti-IL 13 antibodies and uses thereof
EP3718564A1 (en) 2003-12-23 2020-10-07 Genentech, Inc. Novel anti-il 13 antibodies and uses thereof
US10016491B2 (en) 2004-03-05 2018-07-10 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US9211315B2 (en) 2004-03-05 2015-12-15 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US7973139B2 (en) 2004-03-26 2011-07-05 Human Genome Sciences, Inc. Antibodies against nogo receptor
WO2006001888A2 (en) 2004-04-16 2006-01-05 Acuity Pharmaceuticals Inc Compositions and methods for inhibiting angiogenesis
EP2383350A1 (en) 2004-05-07 2011-11-02 Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Methods of diagnosing or treating prostate cancer using the erg gene, alone or in combination with other over or under expressed genes in prostate cancer
EP2453024A2 (en) 2004-06-21 2012-05-16 The Board of Trustees of The Leland Stanford Junior University Genes and pathways differentially expressed in bipolar disorder and/or major depressive disorder
US9006181B2 (en) 2004-07-21 2015-04-14 The Administrators Of The Tulane Educational Fund Treatment of renal dysfunction and multiple myeloma using PACAP compounds
WO2006046994A2 (en) 2004-07-30 2006-05-04 Mount Sinai School Of Medicine Of New York University Klf6 alternative splice forms and a germline klf6 dna polymorphism associated with increased cancer risk
EP2422811A2 (en) 2004-10-27 2012-02-29 MedImmune, LLC Modulation of antibody specificity by tailoring the affinity to cognate antigens
US8809287B2 (en) 2004-11-15 2014-08-19 Icahn School Of Medicine At Mount Sinai Compositions and methods for altering Wnt autocrine signaling
US10603357B2 (en) 2004-11-29 2020-03-31 Bristol-Myers Squibb Company Therapeutic TREM-1 peptides
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
WO2006069253A2 (en) 2004-12-22 2006-06-29 Auckland Uniservices Limited Trefoil factors and methods of treating proliferation disorders using same
EP2520669A2 (en) 2005-02-07 2012-11-07 GeneNews Inc. Mild osteoathritis biomarkers and uses thereof
EP3943501A1 (en) 2005-02-23 2022-01-26 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2006091871A1 (en) 2005-02-23 2006-08-31 Halozyme Therapeutics, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US10588983B2 (en) 2005-02-23 2020-03-17 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
EP3045472A1 (en) 2005-02-23 2016-07-20 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2006102095A2 (en) 2005-03-18 2006-09-28 Medimmune, Inc. Framework-shuffling of antibodies
EP3479844A1 (en) 2005-04-15 2019-05-08 MacroGenics, Inc. Covalent diabodies and uses thereof
EP2932982A1 (en) 2005-05-17 2015-10-21 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
EP3782655A1 (en) 2005-05-17 2021-02-24 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
EP3441090A1 (en) 2005-05-17 2019-02-13 Amicus Therapeutics, Inc. A method for the treatment of pompe disease using 1-deoxynojirimycin and derivatives
US8088382B2 (en) 2005-07-05 2012-01-03 Cornell Research Foundation, Inc. Methods of inhibiting transendothelial migration of neutrophils and monocytes with anti-CD99L2 antibodies
EP2573114A1 (en) 2005-08-10 2013-03-27 MacroGenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2319941A2 (en) 2005-10-21 2011-05-11 GeneNews Inc. Method and apparatus for correlating levels of biomarker products with disease
EP2500030A2 (en) 2005-11-04 2012-09-19 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP2998318A1 (en) 2005-11-04 2016-03-23 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP3299027A1 (en) 2005-11-04 2018-03-28 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP2564864A2 (en) 2005-11-12 2013-03-06 The Board of Trustees of the Leland FGF2-related methods for diagnosing and treating depression
EP2387995A1 (en) 2006-03-30 2011-11-23 PTC Therapeutics, Inc. Methods for the production of functional protein from DNA having a nonsense mutation and the treatment of disorders associated therewith
EP2505209A1 (en) 2006-06-26 2012-10-03 MacroGenics, Inc. Fcgamma-RIIB-specific antibodies and methods of the use thereof
EP2639301A2 (en) 2006-06-30 2013-09-18 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
WO2008105797A2 (en) 2006-06-30 2008-09-04 Bristol-Myers Squibb Company Polynucleotides encoding novel pcsk9 variants
EP2671946A1 (en) 2006-06-30 2013-12-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
EP2484696A1 (en) 2006-08-28 2012-08-08 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2292663A2 (en) 2006-08-28 2011-03-09 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2837697A2 (en) 2006-10-10 2015-02-18 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Prostate cancer-specific alterations in ERG gene expression and detection and treatment methods based on those alternations
EP2518163A2 (en) 2006-10-10 2012-10-31 The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc. Prostate cancer specific alterations in erg gene expression and detection and treatment methods based on those alterations
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
US8796423B2 (en) 2006-11-15 2014-08-05 Eli Lilly And Company Anti-TSG101 antibodies and their uses for treatment of viral infections
US7964708B2 (en) 2006-11-15 2011-06-21 Limin Li Anti-TSG101 antibodies and their uses for treatment of viral infections
EP2514439A1 (en) 2006-11-15 2012-10-24 Functional Genetics, Inc. Anti-TSG101antibodies and their uses for treatment of viral infections
EP2610267A1 (en) 2006-12-18 2013-07-03 Genentech, Inc. Antagonist anti-Notch3 antibodies and their use in the prevention and treatment of Notch3-related diseases
US8211858B2 (en) 2007-04-27 2012-07-03 The University Of Toledo Modified plasminogen activator inhibitor type-1 molecule and methods based thereon
EP3424951A1 (en) 2007-06-21 2019-01-09 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2008157379A2 (en) 2007-06-21 2008-12-24 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2009002193A1 (en) 2007-06-27 2008-12-31 Auckland Uniservices Limited Polypeptides and polynucleotides for artemin and related ligands, and methods of use thereof
US8980262B2 (en) 2007-08-29 2015-03-17 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9175087B2 (en) 2007-08-29 2015-11-03 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9243067B2 (en) 2007-08-29 2016-01-26 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9815902B2 (en) 2007-08-29 2017-11-14 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US9228019B2 (en) 2007-08-29 2016-01-05 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
EP3375868A1 (en) 2007-11-08 2018-09-19 Dana Farber Cancer Institute, Inc. Stimulation of anti-tumor immunity using dendritic cell/tumor cell fusions and anti-cd3/cd28
US8877896B2 (en) 2008-02-15 2014-11-04 Tufts University Compositions, methods and kits for modeling, diagnosing, and treating complement disorders
US8324182B2 (en) 2008-02-15 2012-12-04 Tufts University Humanized model of membrane attack complex (MAC) formation on murine retina and compositions, kits and methods for treatment of macular degeneration
WO2009102488A2 (en) 2008-02-15 2009-08-20 Tufts University A humanized model of membrane attack complex (mac) formation on murine retina and compositions, kits and methods for treatment of macular degeneration
EP3045475A1 (en) 2008-04-02 2016-07-20 MacroGenics, Inc. Bcr-complex-specific antibodies and methods of using same
WO2009123894A2 (en) 2008-04-02 2009-10-08 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
EP3067063A1 (en) 2008-04-02 2016-09-14 MacroGenics, Inc. Her2/neu-specific antibodies and methods of using same
WO2009151717A2 (en) 2008-04-02 2009-12-17 Macrogenics, Inc. Bcr-complex-specific antibodies and methods of using same
US7928189B2 (en) 2008-05-05 2011-04-19 Ottawa Health Research Institute PCSK9 polypeptide fragment
WO2010033279A2 (en) 2008-06-04 2010-03-25 Macrogenics, Inc. Antibodies with altered binding to fcrn and methods of using same
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
EP3482769A1 (en) 2008-12-19 2019-05-15 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2010080538A1 (en) 2008-12-19 2010-07-15 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2786762A2 (en) 2008-12-19 2014-10-08 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2010072684A1 (en) 2008-12-22 2010-07-01 Universität Regensburg Norrin in the treatment of diseases associated with an increased tgf-beta activity
WO2010124365A1 (en) 2009-04-27 2010-11-04 Ottawa Hospital Research Institute Compositions and methods for modulating stem cells and uses thereof
US10828346B2 (en) 2009-04-27 2020-11-10 Ottawa Hospital Research Institute Compositions and methods for modulating stem cells and uses thereof
US10071138B2 (en) 2009-04-27 2018-09-11 Ottawa Hospital Research Institute Compositions and methods for modulating stem cells and uses thereof
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
US8568719B2 (en) 2009-08-13 2013-10-29 Crucell Holland B.V. Antibodies against human respiratory syncytial virus (RSV) and methods of use
WO2011020079A1 (en) 2009-08-13 2011-02-17 Calmune Corporation Antibodies against human respiratory syncytial virus (rsv) and methods of use
US9403900B2 (en) 2009-08-13 2016-08-02 Crucell Holland B.V. Anti-human respiratory syncytial virus (RSV) antibodies and methods of use
US9365638B2 (en) 2009-08-13 2016-06-14 Crucell Holland B. V. Antibodies against human respiratory syncytial virus (RSV) and methods of use
US9988437B2 (en) 2009-08-13 2018-06-05 Janssen Vaccines & Prevention B.V. Anti-human Respiratory Syncytial Virus (RSV) antibodies and methods of use
WO2011035205A2 (en) 2009-09-18 2011-03-24 Calmune Corporation Antibodies against candida, collections thereof and methods of use
WO2011046457A1 (en) 2009-10-16 2011-04-21 Auckland Uniservices Limited Anti-neoplastic uses of artemin antagonists
US8916517B2 (en) 2009-11-02 2014-12-23 The Administrators Of The Tulane Educational Fund Analogs of pituitary adenylate cyclase-activating polypeptide (PACAP) and methods for their use
WO2011057188A1 (en) 2009-11-06 2011-05-12 Idexx Laboratories, Inc. Canine anti-cd20 antibodies
US9149025B2 (en) 2010-02-19 2015-10-06 Universite De Liege Polynucleotide for use in treatment of influenza A virus induced diseases, encoding modified Mx protein, said modified Mx protein, and a transgenic animal expressing gene encoding modified Mx protein
WO2011101031A1 (en) 2010-02-19 2011-08-25 Université de Liège A polynucleotide for use in treatment of influenza a virus induced diseases, encoding modified mx protein, said modified mx protein, and a transgenic animal expressing gene encoding modified mx protein
US9139642B2 (en) 2010-07-09 2015-09-22 Crucell Holland B.V. Anti-human respiratory syncytial virus (RSV) antibodies and methods of use
WO2012006596A2 (en) 2010-07-09 2012-01-12 Calmune Corporation Anti-human respiratory syncytial virus (rsv) antibodies and methods of use
WO2012013249A1 (en) 2010-07-30 2012-02-02 Université de Liège Dentin matrix protein 1 (dmp1) for use in pharmaceutical compositions
WO2012018687A1 (en) 2010-08-02 2012-02-09 Macrogenics, Inc. Covalent diabodies and uses thereof
US10351617B2 (en) 2010-08-13 2019-07-16 Trustees Of Tufts College Compositions, kits and methods for treatment of complement-related disorders
US10130687B2 (en) 2011-01-05 2018-11-20 Rhode Island Hospital Compositions and methods for the treatment of orthopedic disease or injury
WO2012162068A2 (en) 2011-05-21 2012-11-29 Macrogenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
WO2013040341A2 (en) 2011-09-16 2013-03-21 Ottawa Hospital Research Institute Wnt7a compositions and methods of using the same
US9873722B2 (en) 2011-09-16 2018-01-23 Fate Therapeutics, Inc. Wnt compositions and therapeutic uses of such compositions
US9732130B2 (en) 2011-09-16 2017-08-15 Ottawa Hospital Research Institute WNT7A compositions and method of using the same
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US10189904B2 (en) 2012-02-15 2019-01-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10150809B2 (en) 2012-02-15 2018-12-11 Bristol-Myers Squibb Company Antibodies that bind peptidoglycan recognition protein 1
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906975B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation with antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906965B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation wtih antibodies that bind peptidoglycan recognition protein 1
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10525130B2 (en) 2012-03-26 2020-01-07 Sanofi Stable IGG4 based binding agent formulations
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
WO2014006063A2 (en) 2012-07-02 2014-01-09 Medizinische Universität Wien Complement split product c4d for the treatment of inflammatory conditions
EP3527213A1 (en) 2012-10-26 2019-08-21 The Chinese University Of Hong Kong Treatment of cancer using a smad3 inhibitor
EP3320906A1 (en) 2012-10-26 2018-05-16 The Chinese University of Hong Kong Treatment of melanoma and lung carcinoma using a smad3 inhibitor
US10570200B2 (en) 2013-02-01 2020-02-25 California Institute Of Technology Antibody-mediated immunocontraception
EP3450571A1 (en) 2014-02-24 2019-03-06 Celgene Corporation Methods of using an activator of cereblon for neural cell expansion and the treatment of central nervous system disorders
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US9458464B2 (en) 2014-06-23 2016-10-04 The Johns Hopkins University Treatment of neuropathic pain
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US11072654B2 (en) 2014-07-17 2021-07-27 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US10813977B2 (en) 2014-08-28 2020-10-27 Trustees Of Tufts College Compositions, methods and kits for treating complement related disorders
EP3892291A1 (en) 2014-08-28 2021-10-13 Tufts University Compositions, methods and kits for treating complement related disorders
US11654179B2 (en) 2014-08-28 2023-05-23 Trustees Of Tufts College Compositions, methods and kits for treating complement related disorders
US10729790B2 (en) 2015-05-26 2020-08-04 Salk Institute For Biological Studies Motor neuron-specific expression vectors
US11642423B2 (en) 2015-05-26 2023-05-09 Salk Institute For Biological Studies Motor neuron-specific expression vectors
US9920100B2 (en) 2015-06-05 2018-03-20 The Chinese University Of Hong Kong Mimotopes of tropomyosin for use in immunotherapy for shellfish and/or arthropod allergy
WO2017182981A1 (en) 2016-04-20 2017-10-26 Washington University Ppar agonist or lxr agonist for use in the treatment of systemic lupus erythematosus by modulation of lap activity
WO2019157447A1 (en) 2018-02-12 2019-08-15 Trustees Of Tufts College Cd59 for inhibiting inflammasome activation
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
US11952420B2 (en) 2018-04-02 2024-04-09 Bristol-Myers Squibb Company Nucleic acids encoding anti-TREM-1 antibodies
US11919954B2 (en) 2018-04-02 2024-03-05 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
WO2020097261A1 (en) 2018-11-06 2020-05-14 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services New compositions and methods for treating beta-globinopathies
US11325978B2 (en) 2018-11-06 2022-05-10 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Compositions and methods for treating beta-globinopathies
WO2021138581A1 (en) 2020-01-03 2021-07-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Tnap locally administered for promoting periodontal health
WO2022157548A1 (en) 2021-01-24 2022-07-28 Forrest Michael David Inhibitors of atp synthase - cosmetic and therapeutic uses
WO2022240824A1 (en) 2021-05-13 2022-11-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions and methods for treating sickle cell diseases
WO2023004332A2 (en) 2021-07-19 2023-01-26 New York University Adeno-associated viral vector compositions and methods of promoting muscle regeneration
WO2023064255A2 (en) 2021-10-15 2023-04-20 The Regents Of The University Of California Diagnosis and treatment of anti-pf4 induced thrombocytopenia
WO2023081167A2 (en) 2021-11-02 2023-05-11 The Regents Of The University Of California P-selectin mutants and modulation of integrin-mediated signaling
WO2023089564A1 (en) 2021-11-19 2023-05-25 Janssen Biotech, Inc. Method of treating geographic atrophy with a gene therapy vector expressing soluble cd59
WO2023131901A1 (en) 2022-01-07 2023-07-13 Johnson & Johnson Enterprise Innovation Inc. Materials and methods of il-1beta binding proteins
WO2023146807A1 (en) 2022-01-25 2023-08-03 The Regents Of The University Of California Vegf mutants and modulation of integrin-mediated signaling
WO2023200897A1 (en) 2022-04-13 2023-10-19 The Regents Of The University Of California Use of viral il-6 in cancer therapy
WO2024013727A1 (en) 2022-07-15 2024-01-18 Janssen Biotech, Inc. Material and methods for improved bioengineered pairing of antigen-binding variable regions

Also Published As

Publication number Publication date
AU3434393A (en) 1993-08-03

Similar Documents

Publication Publication Date Title
WO1993014188A1 (en) Targeted virus
AU690667B2 (en) Enhanced virus-mediated DNA transfer
US8889419B2 (en) Methods for enhanced virus-mediated DNA transfer using molecules with virus- and cell-binding domains
Hanawa et al. Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus–based lentiviral vector system
US5736387A (en) Envelope fusion vectors for use in gene delivery
US7790849B2 (en) Enhancement of retroviral gene transduction employing polypeptides comprising the fibronectin heparin II binding domain
WO1997011604A9 (en) Methods for enhanced virus-mediated dna transfer using molecules with virus- and cell-binding domains
US5576201A (en) Retroviral vector particles for transducing non-proliferating cells
WO2005116225A1 (en) Method for introducing and expressing rna in cells
US7759467B2 (en) Enhanced mediated DNA transfer
US20030157070A1 (en) High efficiency ex vivo transduction of cells by high titer recombinant retroviral preparations
US6033907A (en) Enhanced virus-mediated DNA transfer
US5580766A (en) Retroviral vector particles for transducing non-proliferating cells
JPH10501140A (en) Retroviral gene therapy vector and therapeutic method based thereon
JPH11514209A (en) Highly efficient ex vivo transduction of hematopoietic stem cells with recombinant retrovirus preparations
WO1990001870A1 (en) Retroviral vectors expressing soluble cd4; a gene therapy for aids
US20040229361A1 (en) Use of human serum resistant vector particles and cell lines for human gene therapy
US20060257381A1 (en) Method for transplanting lymphohematopoietic cells into mammal
EP0871756A2 (en) High efficiency ex vivo transduction of cells by high titer recombinant retroviral preparations
US6984379B1 (en) Gene therapy by administration of genetically engineered CD34+ cells obtained from cord blood
Bierhuizen et al. Efficient detection and selection of immature rhesus monkey and human CD34+ hematopoietic cells expressing the enhanced green fluorescent protein (EGFP)
MXPA96004240A (en) Improved transfer of dna mediated by vi

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BB BG BR CA FI HU JP KP KR LK MG MN MW NO NZ PL RO RU SD UA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA