US20040023911A1 - Expression of exogenous polynucleotide sequences in a vertebrate - Google Patents

Expression of exogenous polynucleotide sequences in a vertebrate Download PDF

Info

Publication number
US20040023911A1
US20040023911A1 US10/387,525 US38752503A US2004023911A1 US 20040023911 A1 US20040023911 A1 US 20040023911A1 US 38752503 A US38752503 A US 38752503A US 2004023911 A1 US2004023911 A1 US 2004023911A1
Authority
US
United States
Prior art keywords
polynucleotide
cells
polypeptide
dna
tissue
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/387,525
Inventor
Philip Felgner
Jon Wolff
Gary Rhodes
Robert Malone
Dennis Carson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wisconsin Alumni Research Foundation
Fresh Tracks Therapeutics Inc
Original Assignee
Vical Inc
Wisconsin Alumni Research Foundation
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
Priority claimed from US08/481,919 external-priority patent/US6214804B1/en
Application filed by Vical Inc, Wisconsin Alumni Research Foundation filed Critical Vical Inc
Priority to US10/387,525 priority Critical patent/US20040023911A1/en
Publication of US20040023911A1 publication Critical patent/US20040023911A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • 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
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0069Oxidoreductases (1.) acting on single donors with incorporation of molecular oxygen, i.e. oxygenases (1.13)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/1029Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • C12N9/1033Chloramphenicol O-acetyltransferase (2.3.1.28)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1247DNA-directed RNA polymerase (2.7.7.6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y113/00Oxidoreductases acting on single donors with incorporation of molecular oxygen (oxygenases) (1.13)
    • C12Y113/12Oxidoreductases acting on single donors with incorporation of molecular oxygen (oxygenases) (1.13) with incorporation of one atom of oxygen (internal monooxygenases or internal mixed function oxidases)(1.13.12)
    • C12Y113/12007Photinus-luciferin 4-monooxygenase (ATP-hydrolysing) (1.13.12.7), i.e. firefly-luciferase
    • 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
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates to introduction of naked DNA and RNA sequences into a vertebrate to achieve controlled expression of a polypeptide. It is useful in gene therapy, vaccination, and any therapeutic situation in which a polypeptide should be administered to cells in vivo.
  • Vaccination with immunogenic proteins has eliminated or reduced the incidence of many diseases; however there are major difficulties in using proteins associated with other pathogens and disease states as immunogens. Many protein antigens are not intrinsically immunogenic. More often, they are not effective as vaccines because of the manner in which the immune system operates.
  • Humoral immunity involves antibodies, proteins which are secreted into the body fluids and which directly recognize an antigen.
  • the cellular system in contrast, relies on special cells which recognize and kill other cells which are producing foreign antigens. This basic functional division reflects two different strategies of immune defense.
  • Humoral immunity is mainly directed at antigens which are exogenous to the animal whereas the cellular system responds to antigens which are actively synthesized within the animal.
  • Antibody molecules the effectors of humoral immunity, are secreted by special B lymphoid cells, B cells, in response to antigen.
  • Antibodies can bind to and inactivate antigen directly (neutralizing antibodies) or activate other cells of the immune system to destroy the antigen.
  • Cellular immune recognition is mediated by a special class of lymphoid cells, the cytotoxic T cells. These cells do not recognize whole antigens but instead they respond to degraded peptide fragments thereof which appear on the surface of the target cell bound to proteins called class I major histocompatibility complex (MHC) molecules. Essentially all nucleated cells have class I molecules. It is believed that proteins produced within the cell are continually degraded to peptides as part of normal cellular metabolism. These fragments are bound to the MHC molecules and are transported to the cell surface. Thus the cellular immune system is constantly monitoring the spectra of proteins produced in all cells in the body and is poised to eliminate any cells producing foreign antigens.
  • MHC major histocompatibility complex
  • Vaccination is the process of preparing an animal to respond to an antigen. Vaccination is more complex than immune recognition and involves not only B cells and cytotoxic T cells but other types of lymphoid cells as well. During vaccination, cells which recognize the antigen (B cells or cytotoxic T cells) are clonally expanded. In addition, the population of ancillary cells (helper T cells) specific for the antigen also increase. Vaccination also involves specialized antigen presenting cells which can process the antigen and display it in a form which can stimulate one of the two pathways.
  • Vaccination has changed little since the time of Louis Pasteur.
  • a foreign antigen is introduced into an animal where it activates specific B cells by binding to surface immunoglobulins. It is also taken up by antigen processing cells, wherein it is degraded, and appears in fragments on the surface of these cells bound to Class II MHC molecules.
  • Peptides bound to class II molecules are capable of stimulating the helper class of T cells. Both helper T cells and activated B cells are required to produce active humoral immunization. Cellular immunity is thought to be stimulated by a similar but poorly understood mechanism.
  • Normal vaccination schemes will always produce a humoral immune response. They may also provide cytotoxic immunity.
  • the humoral system protects a vaccinated individual from subsequent challenge from a pathogen and can prevent the spread of an intracellular infection if the pathogen goes through an extracellular phase during its life cycle; however, it can do relatively little to eliminate intracellular pathogens.
  • Cytotoxic immunity complements the humoral system by eliminating the infected cells. Thus effective vaccination should activate both types of immunity.
  • a cytotoxic T cell response is necessary to remove intracellular pathogens such as viruses as well as malignant cells. It has proven difficult to present an exogenously administered antigen in adequate concentrations in conjunction with Class I molecules to assure an adequate response. This has severely hindered the development of vaccines against tumor-specific antigens (e.g., on breast or colon cancer cells), and against weakly immunogenic viral proteins (e.g., HIV, Herpes, non-A, non-B hepatitis, CMV and EBV).
  • tumor-specific antigens e.g., on breast or colon cancer cells
  • weakly immunogenic viral proteins e.g., HIV, Herpes, non-A, non-B hepatitis, CMV and EBV.
  • Another major problem with protein or peptide vaccines is anaphylactic reaction which can occur when injections of antigen are repeated in efforts to produce a potent immune response.
  • IgE antibodies formed in response to the antigen cause severe and sometimes fatal allergic reactions.
  • a large number of disease states can benefit from the administration of therapeutic peptides.
  • Such peptides include lymphokines, such as interleukin-2, tumor necrosis factor, and the interferons; growth factors, such as nerve growth factor, epidermal growth factor, and human growth hormone; tissue plasminogen activator; factor VIII:C; granulocyte-macrophage colony-stimulating factor; erythropoietin; insulin; calcitonin; thymidine kinase; and the like.
  • lymphokines such as interleukin-2, tumor necrosis factor, and the interferons
  • growth factors such as nerve growth factor, epidermal growth factor, and human growth hormone
  • tissue plasminogen activator such as tissue plasminogen activator
  • factor VIII:C granulocyte-macrophage colony-stimulating factor
  • erythropoietin insulin
  • calcitonin thymidine kinase
  • the present invention provides a method for delivering a pharmaceutical or immunogenic polypeptide to the interior of a cell of a vertebrate in vivo, comprising the step of introducing a preparation comprising a pharmaceutically acceptable injectable carrier and a naked polynucleotide operatively coding for the polypeptide into the interstitial space of a tissue comprising the cell, whereby the naked polynucleotide is taken up into the interior of the cell and has an immunogenic or pharmacological effect on the vertebrate.
  • a method for introducing a polynucleotide into muscle cells in vivo comprising the steps of providing a composition comprising a naked polynucleotide in a pharmaceutically acceptable carrier, and contacting the composition with muscle tissue of a vertebrate in vivo, whereby the polynucleotide is introduced into muscle cells of the tissue.
  • the polynucleotide may be an antisense polynucleotide.
  • the polynucleotide may code for a therapeutic peptide that is expressed by the muscle cells after the contacting step to provide therapy to the vertebrate.
  • it may code for an immunogenic peptide that is expressed by the muscle cells after the contacting step and which generates an immune response, thereby immunizing the vertebrate.
  • One particularly attractive aspect of the invention is a method for obtaining long term administration of a polypeptide to a vertebrate, comprising the step of introducing a naked DNA sequence operatively coding for the polypeptide interstitially into tissue of the vertebrate, whereby cells of the tissue produce the polypeptide for at least one month or at least 3 months, more preferably at least 6 months.
  • the cells producing the polypeptide are nonproliferating cells, such as muscle cells.
  • Another method according to the invention is a method for obtaining transitory expression of a polypeptide in a vertebrate, comprising the step of introducing a naked mRNA sequence operatively coding for the polypeptide interstitially into tissue of the vertebrate, whereby cells of the tissue produce the polypeptide for less than about 20 days, usually less than about 10 days, and often less than 3 or 5 days.
  • administration into solid tissue is preferred.
  • One important aspect of the invention is a method for treatment of muscular dystrophy, comprising the steps of introducing a therapeutic amount of a composition comprising a polynucleotide operatively coding for dystrophin in a pharmaceutically acceptable injectable carrier in vivo into muscle tissue of an animal suffering from muscular dystrophy, whereby the polynucleotide is taken up into the cells and dystrophin is produced in vivo.
  • the polynucleotide is a naked polynucleotide and the composition is introduced interstitially into the muscle tissue.
  • the present invention also includes pharmaceutical products for all of the uses contemplated in the methods described herein.
  • a pharmaceutical product comprising naked polynucleotide, operatively coding for a biologically active polypeptide, in physiologically acceptable administrable form, in a container, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the polynucleotide for human or veterinary administration.
  • Such notice for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert.
  • the invention provides a pharmaceutical product, comprising naked polynucleotide, operatively coding for a biologically active peptide, in solution in a physiologically acceptable injectable carrier and suitable for introduction interstitially into a tissue to cause cells of the tissue to express the polypeptide, a container enclosing the solution, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of manufacture, use, or sale of the solution of polynucleotide for human or veterinary administration.
  • the peptide may be immunogenic and administration of the solution to a human may serve to vaccinate the human, or an animal.
  • the peptide may be therapeutic and administration of the solution to a vertebrate in need of therapy relating to the polypeptide will have a therapeutic effect.
  • a pharmaceutical product comprising naked antisense polynucleotide, in solution in a physiologically acceptable injectable carrier and suitable for introduction interstitially into a tissue to cause cells of the tissue to take up the polynucleotide and provide a therapeutic effect, a container enclosing the solution, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of manufacture, use, or sale of the solution of polynucleotide for human or veterinary administration.
  • One particularly important aspect of the invention relates to a pharmaceutical product for treatment of muscular dystrophy, comprising a sterile, pharmaceutically acceptable carrier, a pharmaceutically effective amount of a naked polynucleotide operatively coding for dystrophin in the carrier, and a container enclosing the carrier and the polynucleotide in sterile fashion.
  • the polynucleotide is DNA.
  • the invention includes a pharmaceutical product for use in supplying a biologically active polypeptide to a vertebrate, comprising a pharmaceutically effective amount of a naked polynucleotide operatively coding for the polypeptide, a container enclosing the carrier and the polynucleotide in a sterile fashion, and means associated with the container for permitting transfer of the polynucleotide from the container to the interstitial space of a tissue, whereby cells of the tissue can take up and express the polynucleotide.
  • the means for permitting such transfer can include a conventional septum that can be penetrated, e.g., by a needle.
  • the means may be considered to comprise the plunger of the syringe or a needle attached to the syringe.
  • Containers used in the present invention will usually have at least 1, preferably at least 5 or 10, and more preferably at least 50 or 100 micrograms of polynucleotide, to provide one or more unit dosages.
  • the container will have at least 500 micrograms or 1 milligram, and often will contain at least 50 or 100 milligrams of polynucleotide.
  • Another aspect of the invention provides a pharmaceutical product for use in immunizing a vertebrate, comprising a pharmaceutically effective amount of a naked polynucleotide operatively coding for an immunogenic polypeptide, a sealed container enclosing the polynucleotide in a sterile fashion, and means associated with the container for permitting transfer of the polynucleotide from the container to the interstitial space of a tissue, whereby cells of the tissue can take up and express the polynucleotide.
  • Still another aspect of the present invention is the use of naked polynucleotide operatively coding for a physiologically active polypeptide in the preparation of a pharmaceutical for introduction interstitially into tissue to cause cells comprising the tissue to produce the polypeptide.
  • the pharmaceutical for example, may be for introduction into muscle tissue whereby muscle cells produce the polypeptide.
  • the peptide is dystrophin and the pharmaceutical is for treatment of muscular dystrophy.
  • Another use according to the invention is use of naked antisense polynucleotide in the preparation of a pharmaceutical for introduction interstitially into tissue of a vertebrate to inhibit translation of polynucleotide in cells of the vertebrate.
  • the tissue into which the polynucleotide is introduced can be a persistent, non-dividing cell.
  • the polynucleotide may be either a DNA or RNA sequence.
  • DNA When the polynucleotide is DNA, it can also be a DNA sequence which is itself non-replicating, but is inserted into a plasmid, and the plasmid further comprises a replicator.
  • the DNA may be a sequence engineered so as not to integrate into the host cell genome.
  • the polynucleotide sequences may code for a polypeptide which is either contained within the cells or secreted therefrom, or may comprise a sequence which directs the secretion of the peptide.
  • the DNA sequence may also include a promoter sequence.
  • the DNA sequence includes a cell-specific promoter that permits substantial transcription of the DNA only in predetermined cells.
  • the DNA may also code for a polymerase for transcribing the DNA, and may comprise recognition sites for the polymerase and the injectable preparation may include an initial quantity of the polymerase.
  • the polynucleotide is translated for a limited period of time so that the polypeptide delivery is transitory.
  • the polypeptide may advantageously be a therapeutic polypeptide, and may comprise an enzyme, a hormone, a lymphokine, a receptor, particularly a cell surface receptor, a regulatory protein, such as a growth factor or other regulatory agent, or any other protein or peptide that one desires to deliver to a cell in a living vertebrate and for which corresponding DNA or mRNA can be obtained.
  • the polynucleotide is introduced into muscle tissue; in other embodiments the polynucleotide is incorporated into tissues of skin, brain, lung, liver, spleen or blood.
  • the preparation is injected into the vertebrate by a variety of routes, which may be intradermally, subdermally, intrathecally, or intravenously, or it may be placed within cavities of the body.
  • the polynucleotide is injected intramuscularly.
  • the preparation comprising the polynucleotide is impressed into the skin. Transdermal administration is also contemplated, as is inhalation.
  • the polynucleotide is DNA coding for both a polypeptide and a polymerase for transcribing the DNA
  • the DNA includes recognition sites for the polymerase and the injectable preparation further includes a means for providing an initial quantity of the polymerase in the cell.
  • the initial quantity of polymerase may be physically present together with the DNA.
  • the DNA is preferably a plasmid.
  • the polymerase is phage T7 polymerase and the recognition site is a T7 origin of replication sequence.
  • a method for treating a disease associated with the deficiency or absence of a specific polypeptide in a vertebrate comprising the steps of obtaining an injectable preparation comprising a pharmaceutically acceptable injectable carrier containing a naked polynucleotide coding for the specific polypeptide; introducing the injectable preparation into a vertebrate and permitting the polynucleotide to be incorporated into a cell, wherein the polypeptide is formed as the translation product of the polynucleotide, and whereby the deficiency or absence of the polypeptide is compensated for.
  • the preparation is introduced into muscle tissue and the method is applied repetitively.
  • the method is advantageously applied where the deficiency or absence is due to a genetic defect.
  • the polynucleotide is preferably a non-replicating DNA sequence; the DNA sequence may also be incorporated into a plasmid vector which comprises an origin of replication.
  • the polynucleotide codes for a non-secreted polypeptide, and the polypeptide remains in situ.
  • the method when the polynucleotide codes for the polypeptide dystrophin, the method provides a therapy for Duchenne's syndrome; alternatively, when the polynucleotide codes for the polypeptide phenylalanine hydroxylase, the method comprises a therapy for phenylketonuria.
  • the polynucleotide codes for a polypeptide which is secreted by the cell and released into the circulation of the vertebrate; in a particularly preferred embodiment the polynucleotide codes for human growth hormone.
  • a therapy for hypercholesterolemia wherein a polynucleotide coding for a receptor associated with cholesterol homeostasis is introduced into a liver cell, and the receptor is expressed by the cell.
  • a method for immunizing a vertebrate comprising the steps of obtaining a preparation comprising an expressible polynucleotide coding for an immunogenic translation product, and introducing the preparation into a vertebrate wherein the translation product of the polynucleotide is formed by a cell of the vertebrate, which elicits an immune response against the immunogen.
  • the injectable preparation comprises a pharmaceutically acceptable carrier containing an expressible polynucleotide coding for an immunogenic peptide, and on the introduction of the preparation into the vertebrate, the polynucleotide is incorporated into a cell of the vertebrate wherein an immunogenic translation product of the polynucleotide is formed, which elicits an immune response against the immunogen.
  • the preparation comprises one or more cells obtained from the vertebrate and transfected in vitro with the polynucleotide, whereby the polynucleotide is incorporated into said cells, where an immunogenic translation product of the polynucleotide is formed, and whereby on the introduction of the preparation into the vertebrate, an immune response against the immunogen is elicited.
  • the immunogenic product may be secreted by the cells, or it may be presented by a cell of the vertebrate in the context of the major histocompatibility antigens, thereby eliciting an immune response against the immunogen.
  • the method may be practiced using non-dividing, differentiated cells from the vertebrates, which cells may be lymphocytes, obtained from a blood sample; alternatively, it may be practiced using partially differentiated skin fibroblasts which are capable of dividing.
  • the method is practiced by incorporating the polynucleotide coding for an immunogenic translation product into muscle tissue.
  • the polynucleotide used for immunization is preferably an mRNA sequence, although a non-replicating DNA sequence may be used.
  • the polynucleotide may be introduced into tissues of the body using the injectable carrier alone; liposomal preparations are preferred for methods in which in vitro transfections of cells obtained from the vertebrate are carried out.
  • the carrier preferably is isotonic, hypotonic, or weakly hypertonic, and has a relatively low ionic strength, such as provided by a sucrose solution.
  • the preparation may further advantageously comprise a source of a cytokine which is incorporated into liposomes in the form of a polypeptide or as a polynucleotide.
  • the method may be used to selectively elicit a humoral immune response, a cellular immune response, or a mixture of these.
  • the immune response is cellular and comprises the production of cytotoxic T-cells.
  • the immunogenic peptide is associated with a virus, is presented in the context of Class I antigens, and stimulates cytotoxic T-cells which are capable of destroying cells infected with the virus.
  • a cytotoxic T-cell response may also be produced according the method where the polynucleotide codes for a truncated viral antigen lacking humoral epitopes.
  • the immunogenic peptide is associated with a tumor, is presented in the context of Class I antigens, and stimulates cytotoxic T cells which are capable of destroying tumor cells.
  • the injectable preparation comprises cells taken from the animal and transfected in vitro, the cells expressing major histocompatibility antigen of class I and class II, and the immune response is both humoral and cellular and comprises the production of both antibody and cytotoxic T-cells.
  • a method of immunizing a vertebrate comprising the steps of obtaining a positively charged liposome containing an expressible polynucleotide coding for an immunogenic peptide, and introducing the liposome into a vertebrate, whereby the liposome is incorporated into a monocyte, a macrophage, or another cell, where an immunogenic translation product of the polynucleotide is formed, and the product is processed and presented by the cell in the context of the major histocompatibility complex, thereby eliciting an immune response against the immunogen.
  • the polynucleotide is preferably mRNA, although DNA may also be used.
  • the present invention also encompasses the use of DNA coding for a polypeptide and for a polymerase for transcribing the DNA, and wherein the DNA includes recognition sites for the polymerase.
  • the initial quantity of polymerase is provided by including mRNA coding therefor in the preparation, which mRNA is translated by the cell.
  • the mRNA preferably is provided with means for retarding its degradation in the cell. This can include capping the mRNA, circularizing the mRNA, or chemically blocking the 5′ end of the mRNA.
  • the DNA used in the invention may be in the form of linear DNA or may be a plasmid. Episomal DNA is also contemplated.
  • One preferred polymerase is phage T7 RNA polymerase and a preferred recognition site is a T7 RNA polymerase promoter.
  • FIG. 1 comprises autoradiograms of chromatographic studies showing the expression of the CAT gene in mouse muscle.
  • FIGS. 2A to 2 F comprise photomicrographs of muscle tissue stained for ⁇ -galactosidase activity following injection with the pRSVLac-Z DNA vector.
  • FIGS. 3A, 3B, and 3 C present data for luciferase activity in muscle following the injection of ⁇ gLuc ⁇ gA n into muscle.
  • FIG. 4 presents an autoradiogram of a Southern blot after analysis of extracts from pRSVL-injected muscle.
  • FIGS. 5A and 5B comprise graphs showing antibody production in mice following the injection of a gene for an immunogenic peptide.
  • FIGS. 6A and 6B comprise graphs showing antibody production in mice following the injection of mouse cells transfected with a gene for an immunogenic peptide.
  • the practice of the present invention requires obtaining naked polynucleotide operatively coding for a polypeptide for incorporation into vertebrate cells.
  • a polynucleotide operatively codes for a polypeptide when it has all the genetic information necessary for expression by a target cell, such as promoters and the like.
  • These polynucleotides can be administered to the vertebrate by any method that delivers injectable materials to cells of the vertebrate, such as by injection into the interstitial space of tissues such as muscles or skin, introduction into the circulation or into body cavities or by inhalation or insufflation.
  • a naked polynucleotide is injected or otherwise delivered to the animal with a pharmaceutically acceptable liquid carrier.
  • the liquid carrier is aqueous or partly aqueous, comprising sterile, pyrogen-free water.
  • the pH of the preparation is suitably adjusted and buffered.
  • the polynucleotide can comprise a complete gene, a fragment of a gene, or several genes, together with recognition and other sequences necessary for expression.
  • the polynucleotide when it is to be associated with a liposome, it requires a material for forming liposomes, preferably cationic or positively charged liposomes, and requires that liposomal preparations be made from these materials.
  • the polynucleotide may advantageously be used to transfect cells in vitro for use as immunizing agents, or to administer polynucleotides into bodily sites where liposomes may be taken up by phagocytic cells.
  • the naked polynucleotide materials used according to the methods of the invention comprise DNA and RNA sequences or DNA and RNA sequences coding for polypeptides that have useful therapeutic applications.
  • These polynucleotide sequences are naked in the sense that they are free from any delivery vehicle that can act to facilitate entry into the cell, for example, the polynucleotide sequences are free of viral sequences, particularly any viral particles which may carry genetic information. They are similarly free from, or naked with respect to, any material which promotes transfection, such as liposomal formulations, charged lipids such as LipofectinTM or precipitating agents such as CaPO 4 .
  • the DNA sequences used in these methods can be those sequences which do not integrate into the genome of the host cell. These may be non-replicating DNA sequences, or specific replicating sequences genetically engineered to lack the genome-integration ability.
  • the polynucleotide sequences of the invention are DNA or RNA sequences having a therapeutic effect after being taken up by a cell.
  • Examples of polynucleotides that are themselves therapeutic are anti-sense DNA and RNA; DNA coding for an anti-sense RNA; or DNA coding for tRNA or rRNA to replace defective or deficient endogenous molecules.
  • the polynucleotides of the invention can also code for therapeutic polypeptides.
  • a polypeptide is understood to be any translation product of a polynucleotide regardless of size, and whether glycosylated or not.
  • Therapeutic polypeptides include as a primary example, those polypeptides that can compensate for defective or deficient species in an animal, or those that act through toxic effects to limit or remove harmful cells from the body.
  • Therapeutic polynucleotides provided by the invention can also code for immunity-conferring polypeptides, which can act as endogenous immunogens to provoke a humoral or cellular response, or both.
  • the polynucleotides employed according to the present invention can also code for an antibody.
  • the term “antibody” encompasses whole immunoglobulin of any class, chimeric antibodies and hybrid antibodies with dual or multiple antigen or epitope specificities, and fragments, such as F(ab) 2 , Fab′, Fab and the like, including hybrid fragments. Also included within the meaning of “antibody” are conjugates of such fragments, and so-called antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference.
  • an isolated polynucleotide coding for variable regions of an antibody can be introduced, in accordance with the present invention, to enable the treated subject to produce antibody in situ.
  • an antibody-encoding polynucleotides see Ward et al. Nature, 341:544-546 (1989); Gillies et al., Biotechnol. 7:799-804 (1989); and Nakatani et al., loc. cit., 805-810 (1989).
  • the antibody in turn would exert a therapeutic effect, for example, by binding a surface antigen associated with a pathogen.
  • the encoded antibodies can be anti-idiotypic antibodies (antibodies that bind other antibodies) as described, for example, in U.S. Pat. No. 4,699,880.
  • anti-idiotypic antibodies could bind endogenous or foreign antibodies in a treated individual, thereby to ameliorate or prevent pathological conditions associated with an immune response, e.g., in the context of an autoimmune disease.
  • Polynucleotide sequences of the invention preferably code for therapeutic or immunogenic polypeptides, and these sequences may be used in association with other polynucleotide sequences coding for regulatory proteins that control the expression of these polypeptides.
  • the regulatory protein can act by binding to genomic DNA so as to regulate its transcription;
  • RNA can act by binding to messenger RNA to increase or decrease its stability or translation efficiency.
  • the polynucleotide material delivered to the cells in vivo can take any number of forms, and the present invention is not limited to any particular polynucleotide coding for any particular polypeptide. Plasmids containing genes coding for a large number of physiologically active peptides and antigens or immunogens have been reported in the literature and can be readily obtained by those of skill in the art.
  • promoters suitable for use in various vertebrate systems are well known.
  • suitable strong promoters include RSV LTR, MPSV LTR, SV40 IEP, and metallothionein promoter.
  • promoters such as CMV IEP may advantageously be used. All forms of DNA, whether replicating or non-replicating, which do not become integrated into the genome, and which are expressible, are within the methods contemplated by the invention.
  • both DNA and RNA can be synthesized directly when the nucleotide sequence is known or by a combination of PCR cloning and fermentation. Moreover, when the sequence of the desired polypeptide is known, a suitable coding sequence for the polynucleotide can be inferred.
  • the polynucleotide when the polynucleotide is mRNA, it can be readily prepared from the corresponding DNA in vitro.
  • conventional techniques utilize phage RNA polymerases SP6, T3, or T7 to prepare mRNA from DNA templates in the presence of the individual ribonucleoside triphosphates.
  • An appropriate phage promoter, such as a T7 origin of replication site is placed in the template DNA immediately upstream of the gene to be transcribed.
  • Systems utilizing T7 in this manner are well known, and are described in the literature, e.g., in Current Protocols in Molecular Biology, ⁇ 3.8 (Vol. 1 1988).
  • plasmids may advantageously comprise a promoter for a desired RNA polymerase, followed by a 5′ untranslated region, a 3′ untranslated region, and a template for a poly A tract. There should be a unique restriction site between these 5′ and 3′ regions to facilitate the insertion of any desired cDNA into the plasmid.
  • the plasmid is linearized by cutting in the polyadenylation region and is transcribed in vitro to form mRNA transcripts.
  • These transcripts are preferably provided with a 5′ cap, as demonstrated in Example 5.
  • a 5′ untranslated sequence such as EMC can be used which does not require a 5′ cap.
  • the mRNA can be prepared in commercially-available nucleotide synthesis apparatus.
  • mRNA in circular form can be prepared.
  • Exonuclease-resistant RNAs such as circular mRNA, chemically blocked mRNA, and mRNA with a 5′ cap are preferred, because of their greater half-life in vivo.
  • one preferred mRNA is a self-circularizing mRNA having the gene of interest preceded by the 5′ untranslated region of polio virus. It has been demonstrated that circular mRNA has an extremely long half-life (Harland & Misher, Development 102: 837-852 (1988)) and that the polio virus 5′ untranslated region can promote translation of mRNA without the usual 5′ cap (Pelletier & Finberg, Nature 334:320-325 (1988), hereby incorporated by reference).
  • This material may be prepared from a DNA template that is self-splicing and generates circular “lariat” mRNAs, using the method of Been & Cech, Cell 47:206-216 (1986)(hereby incorporated by reference). We modify that template by including the 5′ untranslated region of the polio virus immediately upstream of the gene of interest, following the procedure of Maniatis, T. et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y. (1982).
  • the present invention includes the use of mRNA that is chemically blocked at the 5′ and/or 3′ end to prevent access by RNAse.
  • This enzyme is an exonuclease and therefore does not cleave RNA in the middle of the chain.
  • Such chemical blockage can substantially lengthen the half life of the RNA in vivo.
  • Two agents which may be used to modify RNA are available from Clonetech Laboratories, Inc., Palo Alto, Calif.: C2 AminoModifier (Catalog # 5204-1) and Amino-7-dUTP (Catalog # K1022-1). These materials add reactive groups to the RNA. After introduction of either of these agents onto an RNA molecule of interest, an appropriate reactive substituent can be linked to the RNA according to the manufacturer's instructions. By adding a group with sufficient bulk, access to the chemically modified RNA by RNAse can be prevented.
  • one major advantage of the present invention is the transitory nature of the polynucleotide synthesis in the cells. (We refer to this as reversible gene therapy, transient gene therapy or TGT.) With mRNA introduced according to the present invention, the effect will generally last about one day. Also, in marked contrast to gene therapies proposed in the past, mRNA does not have to penetrate the nucleus to direct protein synthesis; therefore, it should have no genetic liability.
  • a preferred embodiment of the invention provides introducing a DNA sequence coding for a specific polypeptide into the cell.
  • non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of about up to six months, and we have observed no evidence of integration of the DNA sequences into the genome of the cells.
  • an even more prolonged effect can be achieved by introducing the DNA sequence into the cell by means of a vector plasmid having the DNA sequence inserted therein.
  • the plasmid further comprises a replicator.
  • plasmids are well known to those skilled in the art, for example, plasmid pBR322, with replicator pMB1, or plasmid pMK16, with replicator ColE1 (Ausubel, Current Protocols in Molecular Biology, John Wiley and Sons, New York (1988) ⁇ II:1.5.2.
  • results of studies of the time course of expression of DNA and mRNA introduced into muscle cells as described in Examples 1 and 13 indicate that mRNA expression is more rapid, although shorter in duration than DNA expression.
  • An immediate and long lived gene expression can be achieved by administering to the cell a liposomal preparation comprising both DNA and an RNA polymerase, such as the phage polymerases T7, T3, and SP6.
  • the liposome also includes an initial source of the appropriate RNA polymerase, by either including the actual enzyme itself, or alternatively, an mRNA coding for that enzyme. When the liposome is introduced into the organism, it delivers the DNA and the initial source of RNA polymerase to the cell.
  • RNA polymerase recognizing the promoters on the introduced DNA, transcribes both genes, resulting in translation products comprising more RNA polymerase and the desired polypeptide. Production of these materials continues until the introduced DNA (which is usually in the form of a plasmid) is degraded. In this manner, production of the desired polypeptide in vivo can be achieved in a few hours and be extended for one month or more.
  • the methods of the invention can accordingly be appropriately applied to treatment strategies requiring delivery and functional expression of missing or defective genes.
  • the polynucleotides may be delivered to the interstitial space of tissues of the animal body, including those of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue.
  • Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels.
  • Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts.
  • in vivo muscle cells are particularly competent in their ability to take up and express polynucleotides. This ability may be due to the singular tissue architecture of muscle, comprising multinucleated cells, sarcoplasmic reticulum, and transverse tubular system. Polynucleotides may enter the muscle through the transverse tubular system, which contains extracellular fluid and extends deep into the muscle cell. It is also possible that the polynucleotides enter damaged muscle cells which then recover.
  • Muscle is also advantageously used as a site for the delivery and expression of polynucleotides in a number of therapeutic applications because animals have a proportionately large muscle mass which is conveniently accessed by direct injection through the skin; for this reason, a comparatively large dose of polynucleotides can be deposited in muscle by multiple injections, and repetitive injections, to extend therapy over long periods of time, are easily performed and can be carried out safely and without special skill or devices.
  • Muscle tissue can be used as a site for injection and expression of polynucleotides in a set of general strategies, which are exemplary and not exhaustive.
  • muscle disorders related to defective or absent gene products can be treated by introducing polynucleotides coding for a non-secreted gene product into the diseased muscle tissue.
  • disorders of other organs or tissues due to the absence of a gene product, and which results in the build-up of a circulating toxic metabolite can be treated by introducing the specific therapeutic polypeptide into muscle tissue where the non-secreted gene product is expressed and clears the circulating metabolite.
  • a polynucleotide coding for an secretable therapeutic polypeptide can be injected into muscle tissue from where the polypeptide is released into the circulation to seek a metabolic target. This use is demonstrated in the expression of growth hormone gene injected into muscle, Example 18. Certain DNA segments, are known to serve as “signals” to direct secretion (Wickner, W. T. and H. F. Lodish, Science 230:400-407 (1985), and these may be advantageously employed.
  • muscle cells may be injected with polynucleotides coding for immunogenic peptides, and these peptides will be presented by muscle cells in the context of antigens of the major histocompatibility complex to provoke a selected immune response against the immunogen.
  • Tissues other than those of muscle, and having a less efficient uptake and expression of injected polynucleotides, may nonetheless be advantageously used as injection sites to produce therapeutic polypeptides or polynucleotides under certain conditions.
  • One such condition is the use of a polynucleotide to provide a polypeptide which to be effective must be present in association with cells of a specific type; for example, the cell surface receptors of liver cells associated with cholesterol homeostasis. (Brown and Goldstein, Science 232:34-47 (1986)).
  • an enzyme or hormone is the gene product, it is not necessary to achieve high levels of expression in order to effect a valuable therapeutic result.
  • TGT muscular dystrophy
  • the genetic basis of the muscular dystrophies is just beginning to be unraveled.
  • the gene related to Duchenne/Becker muscular dystrophy has recently been cloned and encodes a rather large protein, termed dystrophin.
  • Retroviral vectors are unlikely to be useful, because they could not accommodate the rather large size of the cDNA (about 13 kb) for dystrophin.
  • Very recently reported work is centered on transplanting myoblasts, but the utility of this approach remains to be determined.
  • an attractive approach would be to directly express the dystrophin gene within the muscle of patients with Duchennes. Since most patients die from respiratory failure, the muscles involved with respiration would be a primary target.
  • cystic fibrosis Another application is in the treatment of cystic fibrosis.
  • the gene for cystic fibrosis was recently identified (Goodfellow, P. Nature, 341(6238):102-3 (Sep. 14, 1989); Rommens, J. et al. Science, 245(4922):1059-1065 (Sep. 8, 1989); Beardsley, T. et al., Scientific American, 261(5):28-30 (1989).
  • Significant amelioration of the symptoms should be attainable by the expression of the dysfunctional protein within the appropriate lung cells.
  • the bronchial epithelial cells are postulated to be appropriate target lung cells and they could be accessible to gene transfer following instillation of genes into the lung. Since cystic fibrosis is an autosomal recessive disorder one would need to achieve only about 5% of normal levels of the cystic fibrosis gene product in order to significantly ameliorate the pulmonary symptoms.
  • Biochemical genetic defects of intermediary metabolism can also be treated by TGT.
  • These diseases include phenylketonuria, galactosemia, maple-syrup urine disease, homocystinuria, propionic acidemia, methylmalonic acidemia, and adenosine deaminase deficiency.
  • PKU phenylketonuria
  • the transferred gene could most often be expressed in a variety of tissues and still be able to clear the toxic biochemical.
  • Reversible gene therapy can also be used in treatment strategies requiring intracytoplasmic or intranuclear protein expression.
  • Some proteins are known that are capable of regulating transcription by binding to specific promoter regions on nuclear DNA. Other proteins bind to RNA, regulating its degradation, transport from the nucleus, or translation efficiency. Proteins of this class must be delivered intracellularly for activity. Extracellular delivery of recombinant transcriptional or translational regulatory proteins would not be expected to have biological activity, but functional delivery of the DNA or RNA by TGT would be active.
  • Representative proteins of this type that would benefit from TGT would include NEF, TAT, steroid receptor and the retinoid receptor.
  • Gene therapy can be used in a strategy to increase the resistance of an AIDS patient to HIV infection.
  • Introducing an AIDS resistance gene, such as, for example, the NEF gene or the soluble CD4 gene to prevent budding, into an AIDS patient's T cells will render his T cells less capable of producing active AIDS virus, thus sparing the cells of the immune system and improving his ability to mount a T cell dependent immune response.
  • a population of the AIDS patient's own T cells is isolated from the patient's blood. These cells are then transfected in vitro and then reintroduced back into the patient's blood.
  • the virus-resistant cells will have a selective advantage over the normal cells, and eventually repopulate the patient's lymphatic system.
  • DNA systemic delivery to macrophages or other target cells can be used in addition to the extracorporeal treatment strategy. Although this strategy would not be expected to eradicate virus in the macrophage reservoir, it will increase the level of T cells and improve the patient's immune response.
  • an effective DNA or mRNA dosage will generally be in the range of from about 0.05 ⁇ g/kg to about 50 mg/kg, usually about 0.005-5 mg/kg. However, as will be appreciated, this dosage will vary in a manner apparent to those of skill in the art according to the activity of the peptide coded for by the DNA or mRNA and the particular peptide used. For delivery of adenosine deaminase to mice or humans, for example, adequate levels of translation are achieved with a DNA or mRNA dosage of about 0.5 to 5 mg/kg. See Example 10. From this information, dosages for other peptides of known activity can be readily determined.
  • the present invention treats this disease by intracranial injection of from about 10 ⁇ g to about 100 ⁇ g of DNA or mRNA into the parenchyma through use of a stereotaxic apparatus. Specifically, the injection is targeted to the cholinergic neurons in the medial septum.
  • the DNA or mRNA injection is repeated every 1-3 days for 5′ capped, 3′ polyadenylated mRNA, and every week to 21 days for circular mRNA, and every 30 to 60 days for DNA.
  • DNA would be injected in corresponding amounts; however, frequency of injection would be greatly reduced.
  • Episomal DNA for example, could be active for a number of months, and reinjection would only be necessary upon notable regression by the patient.
  • the enzymes responsible for neurotransmitter synthesis could be expressed from transduced genes.
  • the gene for choline acetyl transferase could be expressed within the brain cells (neurons or glial) of specific areas to increase acetylcholine levels and improve brain function.
  • the critical enzymes involved in the synthesis of other neurotransmitters such as dopamine, norepinephrine, and GABA have been cloned and available.
  • the critical enzymes could be locally increased by gene transfer into a localized area of the brain.
  • the increased productions of these and other neurotransmitters would have broad relevance to manipulation of localized neurotransmitter function and thus to a broad range of brain disease in which disturbed neurotransmitter function plays a crucial role.
  • these diseases could include schizophrenia and manic-depressive illnesses and Parkinson's Disease. It is well established that patients with Parkinson's suffer from progressively disabled motor control due to the lack of dopamine synthesis within the basal ganglia.
  • the rate limiting step for dopamine synthesis is the conversion of tyrosine to L-DOPA by the enzyme, tyrosine hydroxylase.
  • L-DOPA is then converted to dopamine by the ubiquitous enzyme, DOPA decarboxylase. That is why the well-established therapy with L-DOPA is effective (at least for the first few years of treatment).
  • Gene therapy could accomplish the similar pharmacologic objective by expressing the genes for tyrosine hydroxylase and possibly DOPA decarboxylase as well.
  • Tyrosine is readily available within the CNS.
  • liver disease which is less common, is caused by the accumulation of an abnormal protein and would be less amenable to gene therapy.
  • the pulmonary complications would be amenable to the increased expression of alpha-1-antitrypsin within the lung. This should prevent the disabling and eventually lethal emphysema from developing.
  • Alpha-1-antitrypsin deficiency also occurs in tobacco smokers since tobacco smoke decreases alpha-1-antitrypsin activity and thus serine protease activity that leads to emphysema.
  • tobacco smoke's anti-trypsin effect to aneurysms of the aorta. Aneurysms would also be preventable by raising blood levels of anti-1-antitrypsin since this would decrease protease activity that leads to aneurysms.
  • TGT can be used in treatment strategies requiring the delivery of cell surface receptors. It could be argued that there is no need to decipher methodology for functional in vivo delivery of genes. There is, after all, an established technology for the synthesis and large scale production of proteins, and proteins are the end product of gene expression. This logic applies for many protein molecules which act extracellularly or interact with cell surface receptors, such as tissue plasminogen activator (TPA), growth hormone, insulin, interferon, granulocyte-macrophage colony stimulating factor (GMCSF), erythropoietin (EPO), etc.
  • TPA tissue plasminogen activator
  • GMCSF granulocyte-macrophage colony stimulating factor
  • EPO erythropoietin
  • Elevated levels of cholesterol in the blood may be reduced in accordance with the present invention by supplying mRNA coding for the LDL surface receptor to hepatocytes.
  • a slight elevation in the production of this receptor in the liver of patients with elevated LDL will have significant therapeutic benefits.
  • Therapies based on systemic administration of recombinant proteins are not able to compete with the present invention, because simply administering the recombinant protein could not get the receptor into the plasma membrane of the target cells.
  • the receptor must be properly inserted into the membrane in order to exert its biological effect. It is not usually necessary to regulate the level of receptor expression; the more expression the better. This simplifies the molecular biology involved in preparation of the mRNA for use in the present invention.
  • lipid/DNA or RNA complexes containing the LDL receptor gene may be prepared and supplied to the patient by repetitive I.V. injections.
  • the lipid complexes will be taken up largely by the liver. Some of the complexes will be taken up by hepatocytes.
  • the level of LDL receptor in the liver will increase gradually as the number of injections increases. Higher liver LDL receptor levels will lead to therapeutic lowering of LDL and cholesterol.
  • An effective mRNA dose will generally be from about 0.1 to about 5 mg/kg.
  • TGT beneficial applications include the introduction of the thymidine kinase gene into macrophages of patients infected with the HIV virus.
  • Introduction of the thymidine kinase gene into the macrophage reservoir will render those cells more capable of phosphorylating AZT. This tends to overcome their resistance to AZT therapy, making AZT capable of eradicating the HIV reservoir in macrophages.
  • Lipid/DNA complexes containing the thymidine kinase gene can be prepared and administered to the patient through repetitive intravenous injections. The lipid complexes will be taken up largely by the macrophage reservoir leading to elevated levels of thymidine kinase in the macrophages.
  • the thymidine kinase therapy can also be focused by putting the thymidine kinase gene under the control of the HTLV III promoter. According to this strategy, the thymidine kinase would only be synthesized on infection of the cell by HIV virus, and the production of the Tat protein which activates the promoter. An analogous therapy would supply cells with the gene for diphtheria toxin under the control of the same HTLV III promoter, with the lethal result occurring in cells only after HIV infection.
  • AIDS patients could also be treated by supplying the interferon gene to the macrophages according to the TGT method.
  • Increased levels of localized interferon production in macrophages could render them more resistant to the consequences of HIV infection. While local levels of interferon would be high, the overall systemic levels would remain low, thereby avoiding the systemic toxic effects like those observed after recombinant interferon administration.
  • Lipid/DNA or RNA complexes containing the interferon gene can be prepared and administered to the patient by repetitive intravenous injections. The lipid complexes will be taken up largely by the macrophage reservoir leading to elevated localized levels of interferon in the macrophages. This will render them less susceptible to HIV infection.
  • Various cancers may be treated using TGT by supplying a diphtheria toxin gene on a DNA template with a tissue specific enhancer to focus expression of the gene in the cancer cells.
  • Intracellular expression of diphtheria toxin kills cells.
  • These promoters could be tissue-specific such as using a pancreas-specific promoter for the pancreatic cancer.
  • a functional diphtheria toxin gene delivered to pancreatic cells could eradicate the entire pancreas. This strategy could be used as a treatment for pancreatic cancer.
  • the patients would have no insurmountable difficulty surviving without a pancreas.
  • the tissue specific enhancer would ensure that expression of diphtheria toxin would only occur in pancreatic cells.
  • DNA/lipid complexes containing the diphtheria toxin gene under the control of a tissue specific enhancer would be introduced directly into a cannulated artery feeding the pancreas. The infusion would occur on some dosing schedule for as long as necessary to eradicate the pancreatic tissue.
  • Other lethal genes besides diphtheria toxin could be used with similar effect, such as genes for ricin or cobra venom factor or enterotoxin.
  • cell-cycle specific promoter that would only kill cells that are rapidly cycling (dividing) such as cancer cells.
  • Cell-cycle specific killing could also be accomplished by designing mRNA encoding killer proteins that are stable only in cycling cells (i.e. histone mRNA that is only stable during S phase).
  • mRNA encoding killer proteins that are stable only in cycling cells (i.e. histone mRNA that is only stable during S phase).
  • developmental-specific promoters such as the use of alpha-fetoprotein that is only expressed in fetal liver cells and in hepatoblastoma cells that have dedifferentiated into a more fetal state.
  • the TGT strategy can be used to provide a controlled, sustained delivery of peptides.
  • Conventional drugs, as well as recombinant protein drugs, can benefit from controlled release devices.
  • the purpose of the controlled release device is to deliver drugs over a longer time period, so that the number of doses required is reduced. This results in improvements in patient convenience and compliance.
  • TGT can be used to obtain controlled delivery of therapeutic peptides. Regulated expression can be obtained by using suitable promoters, including cell-specific promoters. Suitable peptides delivered by the present invention include, for example, growth hormone, insulin, interleukins, interferons, GMCSF, EPO, and the like. Depending on the specific application, the DNA or an RNA construct selected can be designed to result in a gene product that is secreted from the injected cells and into the systemic circulation.
  • TGT can also comprise the controlled delivery of therapeutic polypeptides or peptides which is achieved by including with the polynucleotide to be expressed in the cell, an additional polynucleotide which codes for a regulatory protein which controls processes of transcription and translation.
  • These polynucleotides comprise those which operate either to up regulate or down regulate polypeptide expression, and exert their effects either within the nucleus or by controlling protein translation events in the cytoplasm.
  • the T7 polymerase gene can be used in conjunction with a gene of interest to obtain longer duration of effect of TGT.
  • Episomal DNA such as that obtained from the origin of replication region for the Epstein Barr virus can be used, as well as that from other origins of replication which are functionally active in mammalian cells, and preferably those that are active in human cells. This is a way to obtain expression from cells after many cell divisions, without risking unfavorable integration events that are common to retrovirus vectors. Controlled release of calcitonin could be obtained if a calcitonin gene under the control of its own promoter could be functionally introduced into some site, such as liver or skin. Cancer patients with hypercalcemia would be a group to whom this therapy could be applied.
  • TGT can be used in the delivery of anti-sense polynucleotides for turning off the expression of specific genes.
  • Conventional anti-sense methodology suffers from poor efficacy, in part, because the oligonucleotide sequences delivered are too short. With TGT, however, full length anti-sense sequences can be delivered as easily as short oligomers.
  • Anti-sense polynucleotides can be DNA or RNA molecules that themselves hybridize to (and, thereby, prevent transcription or translation of) an endogenous nucleotide sequence.
  • an anti-sense DNA may encode an RNA that hybridizes to an endogenous sequence, interfering with translation.
  • TGT uses of TGT in this vein include delivering a polynucleotide that encodes a tRNA or rRNA to replace a defective or deficient endogenous tRNA or rRNA, the presence of which causes the pathological condition.
  • Cell-specific promoters can also be used to permit expression of the gene only in the target cell. For example, certain genes are highly promoted in adults only in particular types of tumors. Similarly, tissue-specific promoters for specialized tissue, e.g., lens tissue of the eye, have also been identified and used in heterologous expression systems.
  • the method of the invention can be used to deliver polynucleotides to animal stock to increase production of milk in dairy cattle or muscle mass in animals that are raised for meat.
  • both expressible DNA and mRNA can be delivered to cells to form therein a polypeptide translation product. If the nucleic acids contain the proper control sequences, they will direct the synthesis of relatively large amounts of the encoded protein.
  • the methods can be applied to achieve improved and more effective immunity against infectious agents, including intracellular viruses, and also against tumor cells.
  • the methods of the invention may be applied by direct injection of the polynucleotide into cells of the animal in vivo, or by in vitro transfection of some of the animal cells which are then re-introduced into the animal body.
  • the polynucleotides may be delivered to various cells of the animal body, including muscle, skin, brain, lung, liver, spleen, or to the cells of the blood. Delivery of the polynucleotides directly in vivo is preferably to the cells of muscle or skin.
  • the polynucleotides may be injected into muscle or skin using an injection syringe. They may also be delivered into muscle or skin using a vaccine gun.
  • cationic lipids can be used to facilitate the transfection of cells in certain applications, particularly in vitro transfection.
  • Cationic lipid based transfection technology is preferred over other methods; it is more efficient and convenient than calcium phosphate, DEAE dextran or electroporation methods, and retrovirus mediated transfection, as discussed previously, can lead to integration events in the host cell genome that result in oncogene activation or other undesirable consequences.
  • the knowledge that cationic lipid technology works with messenger RNA is a further advantage to this approach because RNA is turned over rapidly by intracellular nucleases and is not integrated into the host genome.
  • a transfection system that results in high levels of reversible expression is preferred to alternative methodology requiring selection and expansion of stably transformed clones because many of the desired primary target cells do not rapidly divide in culture.
  • the ability to transfect cells at high efficiency with cationic liposomes provides an alternative method for immunization.
  • the gene for an antigen is introduced in to cells which have been removed from an animal.
  • the transfected cells, now expressing the antigen are reinjected into the animal where the immune system can respond to the (now) endogenous antigen.
  • the process can possibly be enhanced by coinjection of either an adjuvant or lymphokines to further stimulate the lymphoid cells.
  • Vaccination with nucleic acids containing a gene for an antigen may also provide a way to specifically target the cellular immune response.
  • Cells expressing proteins which are secreted will enter the normal antigen processing pathways and produce both a humoral and cytotoxic response. The response to proteins which are not secreted is more selective.
  • Non-secreted proteins synthesized in cells expressing only class I MHC molecules are expected to produce only a cytotoxic vaccination.
  • Expression of the same antigen in cells bearing both class I and class II molecules may produce a more vigorous response by stimulating both cytotoxic and helper T cells. Enhancement of the immune response may also be possible by injecting the gene for the antigen along with a peptide fragment of the antigen.
  • the antigen is presented via class I MHC molecules to the cellular immune system while the peptide is presented via class II MHC molecules to stimulate helper T cells.
  • this method provides a way to stimulate and modulate the immune response in a way which has not previously been possible.
  • glycoprotein antigens are seldom modified correctly in the recombinant expression systems used to make the antigens. Introducing the gene for a glycoprotein antigen will insure that the protein product is synthesized, modified and processed in the same species and cells that the pathogen protein would be. Thus, the expression of a gene for a human viral glycoprotein will contain the correct complement of sugar residues. This is important because it has been demonstrated that a substantial component of the neutralizing antibodies in some viral systems are directed at carbohydrate epitopes.
  • any appropriate antigen which is a candidate for an immune response can be used in its nucleic acid form.
  • the source of the cells could be fibroblasts taken from an individual which provide a convenient source of cells expressing only class I MHC molecules.
  • peripheral blood cells can be rapidly isolated from whole blood to provide a source of cells containing both class I and class II MHC proteins. They could be further fractionated into B cells, helper T cells, cytotoxic T cells or macrophage/monocyte cells if desired.
  • Bone marrow cells can provide a source of less differentiated lymphoid cells.
  • the cell will be transfected either with DNA containing a gene for the antigen or by the appropriate capped and polyadenylated mRNA transcribed from that gene or a circular RNA, chemically modified RNA, or an RNA which does not require 5′ capping.
  • the choice of the transfecting nucleotide may depend on the duration of expression desired. For vaccination purposes, a reversible expression of the immunogenic peptide, as occurs on mRNA transfection, is preferred. Transfected cells are injected into the animal and the expressed proteins will be processed and presented to the immune system by the normal cellular pathways.
  • the first is vaccination against viruses in which antibodies are known to be required or to enhanced viral infection.
  • DNA or mRNA vaccine therapy could similarly provide a means to provoke an effective cytotoxic T-cell response to weakly antigenic tumors.
  • a tumor-specific antigen were expressed by mRNA inside a cell in an already processed form, and incorporated directly into the Class I molecules on the cell surface, a cytotoxic T cell response would be elicited.
  • a second application is that this approach provides a method to treat latent viral infections.
  • viruses for example, Hepatitis B, HIV and members of the Herpes virus group
  • latent infections in which the virus is maintained intracellularly in an inactive or partially active form.
  • by inducing a cytolytic immunity against a latent viral protein the latently infected cells will be targeted and eliminated.
  • a related application of this approach is to the treatment of chronic pathogen infections.
  • pathogens which replicate slowly and spread directly from cell to cell. These infections are chronic, in some cases lasting years or decades. Examples of these are the slow viruses (e.g. Visna), the Scrapie agent and HIV.
  • this approach may also be applicable to the treatment of malignant disease.
  • Vaccination to mount a cellular immune response to a protein specific to the malignant state, be it an activated oncogene, a fetal antigen or an activation marker, will result in the elimination of these cells.
  • DNA/mRNA vaccines could in this way greatly enhance the immunogenicity of certain viral proteins, and cancer-specific antigens, that normally elicit a poor immune response.
  • the mRNA vaccine technique should be applicable to the induction of cytotoxic T cell immunity against poorly immunogenic viral proteins from the Herpes viruses, non-A, non-B hepatitis, and HIV, and it would avoid the hazards and difficulties associated with in vitro propagation of these viruses.
  • cell surface antigens such as viral coat proteins (e.g., HIV gp120)
  • MHC major histocompatibility complex
  • TGT tumor necrosis virus
  • One of the difficulties with vaccine development is the requirement to screen different structural variants of the antigen, for the optimal immune response. If the variant is derived from a recombinant source, the protein usually must be expressed and purified before it can be tested for antigenicity. This is a laborious and time consuming process. With in vitro mutagenesis, it is possible to obtain and sequence numerous clones of a given antigen. If these antigens can be screened for antigenicity at the DNA or RNA level by TGT, the vaccine development program could be made to proceed much faster.
  • the protein antigen is never exposed directly to serum antibody, but is always produced by the transfected cells themselves following translation of the mRNA. Hence, anaphylaxis should not be a problem.
  • the present invention permits the patient to be immunized repeatedly without the fear of allergic reactions.
  • the use of the DNA/mRNA vaccines of the present invention makes such immunization possible.
  • T cell immunization can be augmented by increasing the density of Class I and Class II histocompatibility antigens on the macrophage or other cell surface and/or by inducing the transfected cell to release cytokines that promote lymphocyte proliferation.
  • cytokines that promote lymphocyte proliferation.
  • cytokines are known to enhance macrophage activation. Their systemic use has been hampered because of side effects. However, when encapsulated in mRNA, along with mRNA for antigen, they should be expressed only by those cells that co-express antigen. In this situation, the induction of T cell immunity can be enhanced greatly.
  • Polynucleotide salts Administration of pharmaceutically acceptable salts of the polynucleotides described herein is included within the scope of the invention.
  • Such salts may be prepared from pharmaceutically acceptable non-toxic bases including organic bases and inorganic bases.
  • Salts derived from inorganic bases include sodium, potassium, lithium, ammonium, calcium, magnesium, and the like.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, basic amino acids, and the like.
  • Polynucleotides for injection may be prepared in unit dosage form in ampules, or in multidose containers.
  • the polynucleotides may be present in such forms as suspensions, solutions, or emulsions in oily or preferably aqueous vehicles.
  • the polynucleotide salt may be in lyophilized form for reconstitution, at the time of delivery, with a suitable vehicle, such as sterile pyrogen-free water.
  • a suitable vehicle such as sterile pyrogen-free water.
  • Both liquid as well as lyophilized forms that are to be reconstituted will comprise agents, preferably buffers, in amounts necessary to suitably adjust the pH of the injected solution.
  • the total concentration of solutes should be controlled to make the preparation isotonic, hypotonic, or weakly hypertonic.
  • Nonionic materials such as sugars, are preferred for adjusting tonicity, and sucrose is particularly preferred. Any of these forms may further comprise suitable formulatory agents, such as starch or sugar, glycerol or saline.
  • suitable formulatory agents such as starch or sugar, glycerol or saline.
  • the compositions per unit dosage, whether liquid or solid, may contain from 0.1% to 99% of polynucleotide material.
  • the units dosage ampules or multidose containers in which the polynucleotides are packaged prior to use, may comprise an hermetically sealed container enclosing an amount of polynucleotide or solution containing a polynucleotide suitable for a pharmaceutically effective dose thereof, or multiples of an effective dose.
  • the polynucleotide is packaged as a sterile formulation, and the hermetically sealed container is designed to preserve sterility of the formulation until use.
  • the container in which the polynucleotide is packaged is labeled, and the label bears a notice in the form prescribed by a governmental agency, for example the Food and Drug Administration, which notice is reflective of approval by the agency under Federal law, of the manufacture, use, or sale of the polynucleotide material therein for human administration.
  • a governmental agency for example the Food and Drug Administration, which notice is reflective of approval by the agency under Federal law, of the manufacture, use, or sale of the polynucleotide material therein for human administration.
  • the dosage to be administered depends to a large extent on the condition and size of the subject being treated as well as the frequency of treatment and the route of administration. Regimens for continuing therapy, including dose and frequency may be guided by the initial response and clinical judgment.
  • the parenteral route of injection into the interstitial space of tissues is preferred, although other parenteral routes, such as inhalation of an aerosol formulation, may be required in specific administration, as for example to the mucous membranes of the nose, throat, bronchial tissues or lungs.
  • a formulation comprising the naked polynucleotide in an aqueous carrier is injected into tissue in amounts of from 10 ⁇ l per site to about 1 ml per site.
  • concentration of polynucleotide in the formulation is from about 0.1 ⁇ g/ml to about 20 mg/ml.
  • mRNA based TGT requires the appropriate structural and sequence elements for efficient and correct translation, together with those elements which will enhance the stability of the transfected mRNA.
  • Negative elements include stable intramolecular 5′ UTR stem-loop structures (Muesing et al., Cell 48:691(1987)) and AUG sequences or short open reading frames preceded by an appropriate AUG in the 5′ UTR (Kozak, Supra, Rao et al., Mol. and Cell. Biol. 8:284(1988)).
  • certain sequence motifs such as the beta globin 5′ UTR may act to enhance translation (when placed adjacent to a heterologous 5′ UTR) by an unknown mechanism.
  • specific 5′ UTR sequences which regulate eukaryotic translational efficiency in response to environmental signals. These include the human ferritin 5′ UTR (Hentze et al., Proc. Natl.
  • mRNA stability In addition to translational concerns, mRNA stability must be considered during the development of mRNA based TGT protocols. As a general statement, capping and 3′ polyadenylation are the major positive determinants of eukaryotic mRNA stability (Drummond, supra; Ross, Mol. Biol. Med. 5:1(1988)) and function to protect the 5′ and 3′ ends of the mRNA from degradation. However, regulatory elements which affect the stability of eukaryotic mRNAs have also been defined, and therefore must be considered in the development of mRNA TGT protocols.
  • 3′ UTR uridine rich 3′ untranslated region
  • Liposomes are unilamellar or multilamellar vesicles, having a membrane portion formed of lipophilic material and an interior aqueous portion.
  • the aqueous portion is used in the present invention to contain the polynucleotide material to be delivered to the target cell.
  • the liposome forming materials used herein have a cationic group, such as a quaternary ammonium group, and one or more lipophilic groups, such as saturated or unsaturated alkyl groups having from about 6 to about 30 carbon atoms.
  • a cationic group such as a quaternary ammonium group
  • lipophilic groups such as saturated or unsaturated alkyl groups having from about 6 to about 30 carbon atoms.
  • One group of suitable materials is described in European Patent Publication No. 0187702. These materials have the formula:
  • R 1 and R 2 are the same or different and are alkyl or alkenyl of 6 to 22 carbon atoms
  • R 3 , R 4 , and R 5 are the same or different and are hydrogen, alkyl of 1 to 8 carbons, aryl, aralkyl of 7 to 11 carbons, or when two or three of R 3 , R 4 , and R 5 are taken together they form quinuclidino, piperidino, pyrrolidino, or morpholino
  • n is 1 to 8
  • X is a pharmaceutically acceptable anion, such as a halogen.
  • DOTMA N-(2,3-di-(9-(Z)-octadecenyloxy))-prop-1-yl-N,N,N-trimethylammonium chloride
  • compositions for use in the present invention has the formula:
  • R 1 and R 2 are the same or different and are alkyl or alkenyl of 5 to 21 carbon atoms
  • R 3 , R 4 , and R 5 are the same or different and are hydrogen, alkyl of 1 to 8 carbons, aryl, aralkyl of 7 to 11 carbons, or when two or three of R 3 , R 4 , and R 5 are taken together they form quinuclidino, piperidino, pyrrolidino, or morpholino
  • n is 1 to 8
  • X is a pharmaceutically acceptable anion, such as a halogen.
  • These compounds may be prepared using conventional techniques, such as nucleophilic substitution involving a carboxylic acid and an alkyl halide, by transesterification, or by condensation of an alcohol with an acid or an acid halide.
  • liposome-forming cationic lipid compounds are described in the literature. See, e.g., L. Stamatatos, et al., Biochemistry 27:3917-3925 (1988); H. Eibl, et al., Biophysical Chemistry 10:261-271 (1979).
  • Suitable liposomes for use in the present invention are commercially available.
  • DOTMA liposomes for example, are available under the trademark Lipofectin from Bethesda Research Labs, Gaithersburg, Md.
  • liposomes can be prepared from readily-available or freshly synthesized starting materials of the type previously described.
  • the preparation of DOTAP liposomes is detailed in Example 6.
  • Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Felgner, et al., Proc. Nat'l Acad. Sci. USA 84:7413-7417. Similar methods can be used to prepare liposomes from other cationic lipid materials.
  • conventional liposome forming materials can be used to prepare liposomes having negative charge or neutral charge. Such materials include phosphatidyl choline, cholesterol, phosphatidyl-ethanolamine, and the like. These materials can also advantageously be mixed with the DOTAP or DOTMA starting materials in ratios from 0% to about 75%.
  • DOPC dioleoyl-phosphatidyl choline
  • DOPG dioleoylphosphatidyl glycerol
  • DOPE dioleoylphosphatidyl ethanolamine
  • DOPG/DOPC vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water. The sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15° C.
  • negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size. Other methods are known and available to those of skill in the art.
  • DOTAP 1,2-bis(oleoyloxy)-3-(trimethylammonio)propane
  • the hexane solution was washed 3 times with an equal volume of 1:1 methanol/0.1 N aqueous NCOONa, pH 3.0, 3 times with 1:1 methanol/0.1 N aqueous NaOH, and 1 time with 1% aqueous NaCl.
  • the crude 3-bromo-1,2-bis-(oleolyloxy)propane was then stirred for 72 hours in a sealed tube with a solution of 15% trimethylamine in dry dimethyl sulfoxide (30 ml) at 25° C.
  • the purified product was a colorless, viscous oil that migrates with an R f of 0.4 on thin layer chromatography plates (silica gel G) that were developed with 50:15:5:5:2 CHCl 3 /acetone/CH 3 OH/CH 3 COOH/H 2 O.
  • Suitable template DNA for production of mRNA coding for a desired polypeptide may be prepared in accordance with standard recombinant DNA methodology. As has been previously reported (P. Kreig, et al., Nucleic Acids Res. 12:7057-7070 (1984)), a 5′ cap facilitates translation of the mRNA. Moreover, the 3′ flanking regions and the poly A tail are believed to increase the half life of the mRNA in vivo.
  • SP6 cloning vector pSP64T provides 5′ and 3′ flanking regions from ⁇ -globin, an efficiently translated mRNA.
  • the construction of this plasmid is detailed by Kreig, et al. (supra), and is hereby incorporated by this reference. Any cDNA containing an initiation codon can be introduced into this plasmid, and mRNA can be prepared from the resulting template DNA. This particular plasmid can be cut with BglII to insert any desired cDNA coding for a polypeptide of interest.
  • flanking sequences of pSP64T are purified from pSP64T as the small (approx. 150 bp) HindIII to EcoRI fragment. These sequences are then inserted into a purified linear HindIII/EcoRI fragment (approx. 2.9 k bp) from pIBI 31 (commercially available from International Biotechnologies, Inc., Newhaven, Conn. 06535) with T4 DNA ligase.
  • Resulting plasmids are screened for orientation and transformed into E. coli . These plasmids are adapted to receive any gene of interest at a unique BglII restriction site, which is situated between the two xenopus ⁇ -globin sequences.
  • a convenient marker gene for demonstrating in vivo expression of exogenous polynucleotides is chloramphenicol acetyltransferase, CAT.
  • a plasmid pSP-CAT containing the CAT gene flanked by the xenopus ⁇ -globin 5′ and 3′ sequences was produced by adding the CAT gene into the BgIII site of pSP64T.
  • the CAT gene is commonly used in molecular biology and is available from numerous sources. Both the CAT BamHI/HindIII fragment and the BgIII-cleaved pSP64T were incubated with the Klenow fragment to generate blunt ends, and were then ligated with T4 DNA ligase to form pSP-CAT.
  • the small PstI/HindIII fragment was then generated and purified, which comprises the CAT gene between the 5′ and 3′ ⁇ -globin flanking sequences of pSP64T.
  • pIBI31 International Biotechnologies, Inc.
  • PstI and HindIII the long linear sequence was purified.
  • This fragment was then combined with the CAT-gene containing sequence and the fragments were ligated with T4 DNA ligase to form a plasmid designated pT7CAT An.
  • Clones are selected on the basis of ⁇ -galactosidase activity with Xgal and ampicillin resistance.
  • the plasmid DNA from Example 3 is grown up and prepared as per Maniatis (supra), except without RNAse, using 2 CsCl spins to remove bacterial RNA.
  • E. coli containing pT7CAT An from Example 3 was grown up in ampicillin-containing LB medium. The cells were then pelleted by spinning at 5000 rpm for 10 min. in a Sorvall RC-5 centrifuge (E. I. DuPont, Burbank, Calif. 91510), resuspended in cold TE, pH 8.0, centrifuged again for 10 min.
  • the material was then centrifuged again at 10,000 rpm for 20 min., this time in an HB4 swinging bucket rotor apparatus (DuPont, supra) after which the supernatant was removed and the pellet was washed in 70% EtOH and dried at room temperature.
  • the pellet was resuspended in 3.5 ml TE, followed by addition of 3.4 g CsCl and 350 ⁇ l of 5 mg/ml EtBr.
  • the resulting material was placed in a quick seal tube, filled to the top with mineral oil. The tube was spun for 3.5 hours at 80,000 rpm in a VTi80 centrifuge (Beckman Instruments, Pasadena, Calif., 91051).
  • the band was removed, and the material was centrifuged again, making up the volume with 0.95 g CsCl/ml and 0.1 ml or 5 mg/ml EtBr/ml in TE.
  • the EtBr was then extracted with an equal volume of TE saturated N-Butanol after adding 3 volumes of TE to the band, discarding the upper phase until the upper phase is clear.
  • 2.5 vol. EtOH was added, and the material was precipitated at ⁇ 20 C for 2 hours.
  • the resultant DNA precipitate is used as a DNA template for preparation of mRNA in vitro.
  • DNA from Example 4 was linearized downstream of the poly A tail with a 5-fold excess of PstI.
  • the linearized DNA was then purified with two phenol/chloroform extractions, followed by two chloroform extractions. DNA was then precipitated with NaOAc (0.3 M) and 2 volumes of EtOH. The pellet was resuspended at about 1 mg/ml in DEP-treated deionized water.
  • a transcription buffer comprising 400 mM Tris HCl (pH 8.0), 80 mM MgCl 2 , 50 mM DTT, and 40 mM spermidine. Then, the following materials were added in order to one volume of DEP-treated water at room temperature: 1 volume T7 transcription buffer, prepared above; rATP, rCTP, and rUTP to 1 mM concentration; rGTP to 0.5 mM concentration; 7 meG (5′) ppp (5′) G cap analog (New England Biolabs, Beverly, Mass., 01951) to 0.5 mM concentration; the linearized DNA template prepared above to 0.5 mg/ml concentration; RNAsin (Promega, Madison, Wis.) to 2000 U/ml concentration; and T7 RNA polymerase (N.E. Biolabs) to 4000 U/ml concentration.
  • liposome preparation methods can be used to advantage in the practice of the present invention.
  • One particularly preferred liposome is made from DOTAP as follows:
  • a solution of 10 mg dioleoyl phosphatidylethanolamine (PE) and 10 mg DOTAP (from Example 1) in 1 ml chloroform is evaporated to dryness under a stream of nitrogen, and residual solvent is removed under vacuum overnight.
  • Liposomes are prepared by resuspending the lipids in deionized water (2 ml) and sonicating to clarity in a closed vial. These preparations are stable for at least 6 months.
  • Polynucleotide complexes were prepared by mixing 0.5 ml polynucleotide solution (e.g., from Example 5) at 0.4 mg/ml by slow addition through a syringe with constant gentle vortexing to a 0.5 ml solution of sonicated DOTMA/PE or DOTAP/PE liposomes at 20 mg/ml, at room temperature. This procedure results in positively charged complexes which will spontaneously deliver the polynucleotide into cells in vivo. Different ratios of positively charged liposome to polynucleotide can be used to suit the particular need in any particular situation. Alternatively, as reported by Felgner, et al.
  • the segment of the abdominal muscle into which the injection was made was excised, minced, and placed in a 1.5 ml disposable mortar (Kontes, Morton Grove, Ill.) together with 200 ⁇ l of an aqueous formulation having the following components: 20 mM Tris, pH 7.6; 2 mM MgCl 2 ; and 0.1% Triton X-100 surfactant.
  • the contents of the mortar were then ground for 1 minute with a disposable pestle.
  • the mortar was then covered (with Parafilm) and placed in a 1 liter Parr cell disrupter bomb (Parr Instrument Company, Moline, Ill.) and pressurized to 6 atmospheres with nitrogen at 4° C.
  • the lysates were then assayed for the presence of the CAT protein by thin-layer chromatography.
  • 75 pl of each sample (the supernatant prepared above) was incubated for two hours at 37° C. with 5 ⁇ l C14 chloramphenicol (Amersham); 20 ⁇ l 4 mM Acetyl CoA; and 50 ⁇ l 1 M Tris, pH 7.8. Thereafter, 20 pl of 4 mM Acetyl CoA was added, and the mixture was again incubated for 2 hours at 37° C.
  • the resulting solution was extracted with 1 ml EtOAc, and the organic phase was removed and lyophilized in a vacuum centrifuge (SpeedVac, Savant Co.).
  • the pellet was resuspended in 20 ⁇ l EtOAc, and was spotted onto a silica gel thin layer chromatography plate.
  • the plate was developed for 45 minutes in 95% chloroform/5% methanol, was dried, and was sprayed with a radioluminescent indicator (Enhance Spray for Surface Radiography, New England Nuclear Corp.).
  • the plate was then sandwiched with Kodak XAR5 film with overnight exposure at ⁇ 70° C., and the film was developed per manufacturer's instructions.
  • the following results were obtained: mRNA Expression FORMULATION (No. positive/total) 1. 1 ml Optimem; 37.5 ⁇ g DOTMA 0/6 2. 1 ml Optimem; 15 ⁇ g CAT RNA 3/6 3.
  • Formulation 1 plus 15 ⁇ g CAT RNA 4/6 4.
  • Optimem Serum-free media (Gibco Laboratories, Life Technologies, Inc, Grand Island, N.Y. 14072)
  • DOTMA (Lipofectin brand; Bethesda Research Labs, Gaithersburg, Md.)
  • a liposomal formulation containing mRNA coding for the gp120 protein of the HIV virus is prepared according to Examples 1 through 5, except that the gene for gp120 (pIIIenv3-1 from the Aids Research and Reagent Program, National Institute of Allergy and Infectious Disease, Rockville, Md. 20852) is inserted into the plasmid pXBG in the procedure of Example 4.
  • a volume of 200 ⁇ l of a formulation, prepared according to Example 6, and containing 200 ⁇ g/ml of gp120 mRNA and 500 ⁇ g/ml 1:1 DOTAP/PE in 10% sucrose is injected into the tail vein of mice 3 times in one day. At about 12 to 14 h after the last injection, a segment of muscle is removed from the injection site, and prepared as a cell lysate according to Example 7.
  • the HIV specific protein gp120 is identified in the lysate also according to the procedures of Example 7.
  • gp120 antibody present in serum of the mRNA vaccinated mice to protect against HIV infection is determined by a HT4-6C plaque reduction assay, as follows:
  • HT4-6C cells (CD4+HeLa cells) are obtained from Dr. Bruce Chesebro, (Rocky Mountain National Lab, Montana) and grown in culture in RPMI media (BRL, Gaithersburg, Md.). The group of cells is then divided into batches. Some of the batches are infected with HIV by adding approximately 105 to 106 infectious units of HIV to approximately 107 HT4-6C cells. Other batches are tested for the protective effect of gp120 immune serum against HIV infection by adding both the HIV and approximately 50 ⁇ l of serum from a mouse vaccinated with gp120 mRNA. After 3 days of incubation, the cells of all batches are washed, fixed and stained with crystal violet, and the number of plaques counted. The protective effect of gp120 immune serum is determined as the reduction in the number of plaques in the batches of cells treated with both gp120 mRNA-vaccinated mouse serum and HIV compared to the number in batches treated with HIV alone.
  • mice Severe combined immunodeficient mice (SCID mice (Molecular Biology Institute, (MBI), La Jolla, Calif. 92037)) were reconstituted with adult human peripheral blood lymphocytes by injection into the peritoneal cavity according to the method of Mosier (Mosier et al., Nature 335:256 (1988)). Intraperitoneal injection of 400 to 4000 infectious units of HIV-1 was then performed. The mice were maintained in a P3 level animal containment facility in sealed glove boxes.
  • SCID mice Molecular Biology Institute, (MBI), La Jolla, Calif. 92037
  • the nef mRNA was then incorporated into a formulation according to Example 6.
  • RNA/liposome complex form 200 microliter tail vein injections of a 10% sucrose solution containing 200 ⁇ g/ml NEF RNA and 500 ⁇ g/ml 1:1 DOTAP:DOPE (in RNA/liposome complex form) were performed daily on experimental animals, while control animals were likewise injected with RNA/liposome complexes containing 200 ⁇ g/ml yeast tRNA and 500 ⁇ g/ml 1:1 DOTAP/DOPE liposomes. At 2, 4 and 8 weeks post injection, biopsy specimens were obtained from injected lymphoid organs and prepared for immunohistochemistry.
  • a volume of 200 ⁇ l of the formulation, containing 200 ⁇ g/ml of nef mRNA, and 500 ⁇ g/ml 1:1 DOTAP:DOPE in 10% sucrose is injected into the tail vein of the human stem cell-containing SCID mice 3 times in one day. Following immunization, the mice are challenged by infection with an effective dose of HIV virus. Samples of blood are periodically withdrawn from the tail vein and monitored for production of the characteristic HIV protein p24 by an ELISA kit assay (Abbott Labs, Chicago, Ill.).
  • ADA human adenosine deaminase
  • the full-length sequence for the cDNA of the human adenosine deaminase (ADA) gene is obtained from the 1,300 bp EcoR1-AccI fragment of clone ADA 211 (Adrian, G. et al. Mol. Cell Biol. 4:1712 (1984). It is blunt-ended, ligated to BgIII linkers and then digested with BgIII. The modified fragment is inserted into the BgIII site of pXBG.
  • ADA mRNA is transcribed and purified according to Examples 2 through 5, and purified ADA mRNA is incorporated into a formulation according to Example 6.
  • Balb 3T3 mice are injected directly in the tail vein with 200 pl of this formulation, containing 200 ⁇ g/ml of ADA mRNA, and 500 ⁇ g/ml DOTAP in 10% sucrose.
  • a preliminary separation of human and non-human ADA is carried out by fast protein liquid chromatography (FPLC).
  • FPLC fast protein liquid chromatography
  • the proteins are fractionated on a Pharmacia (Piscataway, N.J.) MonoQ column (HR5/5) with a linear gradient from 0.05 to 0.5 M KCl, 20 mM Tris (pH 7.5).
  • Activity for ADA within the fractions is measured by reacting the fractions with 14 C-adenosine (Amersham, Chicago, Ill.) which is converted to inosine.
  • Thin layer chromatography 0.1 M NaPi pH 6.8 saturated ammonium sulfate:n-propylalcohol/100:60:2) is used to separate the radioactive inosine from the substrate adenosine.
  • mice were injected with either 100 ⁇ g of pRSVCAT DNA plasmid or 100 ⁇ g of ⁇ gCAT ⁇ gA n RNA and the muscle tissue at the injection site later tested for CAT activity.
  • mice Five to six week old female and male Balb/C mice were anesthetized by intraperitoneal injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision was made on the anterior thigh, and the quadriceps muscle was directly visualized. The DNA and RNA were injected in 0.1 ml of solution in a 1 cc syringe through a 27 gauge needle over one minute, approximately 0.5 cm from the distal insertion site of the muscle into the knee and about 0.2 cm deep. A suture was placed over the injection site for future localization, and the skin was then closed with stainless steel clips.
  • 3T3 mouse fibroblasts were also transfected in vitro with 20 ⁇ g of DNA or RNA complexed with 60 ⁇ g of LipofectinTM (BRL) in 3 ml of Opti-MemTM (Gibco), under optimal conditions described for these cells (Malone, R. et al. Proc. Nat'l. Acad. Sci. USA 86:6077-6081(1989).
  • the same fibroblasts were also transfected using calcium phosphate according to the procedure described in Ausubel et al.(Eds) Current Protocols in Molecular Biology, John Wiley and Sons, New York (1989).
  • RNA consisted of the chloramphenicol acetyl transferase (CAT) coding sequences flanked by 5′ and 3′ ⁇ -globin untranslated sequences and a 3′ poly-A tract.
  • CAT chloramphenicol acetyl transferase
  • Muscle extracts were prepared by excising the entire quadriceps, mincing the muscle into a 1.5 ml microtube containing 200 pl of a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl 2 and 0.1% Triton X), and grinding the muscle with a plastic pestle (Kontes) for one minute. In order to ensure complete disruption of the muscle cells, the muscle tissue was then placed under 600 psi of N 2 in a bomb (Parr) at 4° C. for 15 min before releasing the pressure.
  • a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl 2 and 0.1% Triton X)
  • Kontes plastic pestle
  • the muscle tissue was then placed under 600 psi of N 2 in a bomb (Parr) at 4° C. for 15 min before releasing the pressure.
  • Fibroblasts were processed similarly after they were trypsinized off the plates, taken up into media with serum, washed 2 ⁇ with PBS, and the final cell pellet suspended into 200 ⁇ l of lysis solution. 75 ⁇ l of the muscle and fibroblast extracts were assayed for CAT activity by incubating the reaction mixtures for 2 hours with 14 C-chloramphenicol, followed by extraction and thin-layer chromatography, all as described in Example 7.
  • FIG. 1 comprises autoradiograms from two separate experiments showing CAT activity within extracts of the injected quadriceps muscles.
  • Lanes 1 and 13 Control fibroblasts
  • Lanes 2 and 14 Muscle injected only with 5% sucrose
  • Lanes 3 and 15 0.005 units of non-injected, purified CAT standard
  • Lanes 4 and 16 0.05 units of purified CAT (Sigma)
  • Lanes 5 to 8 Muscle injected with 100 ⁇ g of ⁇ gCAT ⁇ gA n RNA in 5% sucrose
  • Lanes 11, 12, and 17 to 20 Muscle injected with 100 ⁇ grams pRSVCAT DNA in 5% sucrose
  • Lanes 9 and 10 20 ⁇ grams of ⁇ gCAT ⁇ gA n RNA, lipofected, with 60 ⁇ grams of DOTMA, into a 70% confluent 60 mm plate of 3T3 cells (10 6 )
  • Lanes 21, 22 20 ⁇ grams of pRSVCAT lipofected, with 60 ⁇ g of DOTMA, into a 50% confluent 60 mm plate of 3T3 cells
  • Lanes 23, 24 20 ⁇ g of pRSVCAT calcium phosphate lipofected into a 50% confluent 60 mm plate of 3T3 cells.
  • CAT activity was readily detected in all four RNA injection sites 18 hours after injection and in all six DNA injection sites 48 hours after injection. Extracts from two of the four RNA injection sites (FIG. 1, lanes 6 and 8) and from two of the six DNA injection sites (FIG. 1, lanes 11 and 20) contained levels of CAT activity comparable to the levels of CAT activity obtained from fibroblasts transiently transfected in vitro under optimal conditions (FIG. 1, lanes 9, 10, 21-24). The average total amount of CAT activity expressed in muscle was 960 pg for the RNA injections and 116 pg for the DNA injections.
  • CAT activity recovered from different muscle sites probably represents variability inherent in the injection and extraction technique, since significant variability was observed when pure CAT protein or pRSVCAT-transfected fibroblasts were injected into the muscle sites and immediately excised for measurement of CAT activity.
  • CAT activity was also recovered from abdominal muscle injected with the RNA or DNA CAT vectors, indicating that other muscle groups can take up and express polynucleotides.
  • the site of gene expression in injected muscle was determined by utilizing the pRSVLac-Z DNA vector (P. Norton and J. Coffin Molec. Cell Biol. 5:281-290 (1985)) expressing the E. coli -galactosidase gene for injection and observing the in situ cytochemical staining of muscle cells for E. coli ⁇ -galactosidase activity.
  • the quadriceps muscle of mice was exposed as described in the previous example. Quadriceps muscles were injected once with 100 ⁇ g of pRSVLAC-Z DNA in 20% sucrose. Seven days later the individual quadriceps muscles were removed in their entirety and every fifth 15 ⁇ m cross-section was histochemically stained for ⁇ -galactosidase activity.
  • the muscle biopsy was frozen in liquid N 2 -cooled isopentane. 15 ⁇ m serial sections were sliced using a cryostat and placed immediately on gelatinized slides. The slide were fixed in 1.5% glutaraldehyde in PBS for 10 minutes and stained 4 hours for ⁇ -galactosidase activity (J. Price et al. Proc. Nat'l Acad. Sci. USA 84:156-160 (1987). The muscle was counterstained with eosin.
  • RNA and DNA vectors were prepared, and the quadriceps muscles of mice injected as previously described. Muscle extracts of the entire quadriceps were prepared as described in Example 11, except that the lysis buffer was 100 mM KPi pH 7.8, 1 mM DTT, and 0.1% Triton X. 87.5 ⁇ l of the 200 ⁇ l extract was analyzed for luciferase activity (J. de Wet et al. Molec. Cell Biol. 7:725-737(1987)) using an LKB 125 I luminometer.
  • Light units were converted to picograms (pg) of luciferase using a standard curve established by measuring the light units produced by purified firefly luciferase (Analytical Luminescence Laboratory) within control muscle extract.
  • the RNA and DNA preparations prior to injection did not contain any contaminating luciferase activity.
  • Control muscle injected with 20% sucrose had no detectable luciferase activity. All the above experiments were done two to three times and specifically, the DNA time points greater than 40 days were done three times.
  • FIGS. 3A to 3 C illustrate the results of the following:
  • 3(B) Luciferase activity assayed at varying times after 20 ⁇ g of ⁇ gLUC ⁇ gA n RNA were lipofected into a million 3T3 fibroblasts (Malone, R. et al. Proc. Nat'l. Acad. Sci. USA 86:6077-6081 (1989), and after 100 ⁇ g of ⁇ gLUC ⁇ gA n RNA in 20% sucrose were injected into quadriceps.
  • 3(C) Luciferase activity assayed at varying times after pRSVL DNA was injected intramuscularly.
  • a dose-response effect was observed when quadriceps muscles were injected with various amounts of ⁇ gLuc ⁇ gA n RNA or DNA pRSVL constructs (FIG. 3A).
  • the injection of ten times more DNA resulted in luciferase activity increasing approximately ten-fold from 33 pg luciferase following the injection of 10 ⁇ g of DNA to 320 pg luciferase following the injection of 100 ⁇ g of DNA.
  • the injection of ten times more RNA also yielded approximately ten times more luciferase.
  • a million 3T3 mouse fibroblasts in a 60 mm dish were lipofected with 20 ⁇ g of DNA or RNA complexed with 60 ⁇ g of LipofectinTM (Bethesda Research Labs) in 3 ml of Opti-MEMTM (Gibco). Two days later, the cells were assayed for luciferase activity and the results from four separate plates were averaged.
  • RNA vectors Twenty ⁇ g of pRSVL DNA transfected into fibroblasts yielded a total of 120 pg of luciferase (6 pg luciferase/ ⁇ g DNA), while 25 ⁇ g injected into muscle yielded an average of 116 pg of luciferase (4.6 pg luciferase/ ⁇ g DNA; FIG. 3A). The expression from the RNA vectors was approximately seven-fold more efficient in transfected fibroblasts than in injected muscles.
  • RNA transfected into fibroblasts yielded a total of 450 pg of luciferase, while 25 ⁇ g injected into muscle yielded 74 pg of luciferase (FIGS. 3A and 3B).
  • Luciferase activity was assayed at varying times after 25 pg of ⁇ gLuc ⁇ gA n RNA or 100 ⁇ g of pRSVL DNA were injected. Following RNA injection, the average luciferase activity reached a maximum of 74 pg at 18 hours, and then quickly decreased to 2 pg at 60 hours. In transfected fibroblasts, the luciferase activity was maximal at 8 hours. Following DNA injection into muscle, substantial amounts of luciferase were present for at least 60 days.
  • FIG. 4 is an autoradiogram of a Southern blot having a sample pattern as follows:
  • Lane 1 0.05 ng of undigested pRSVL plasmid
  • Lane 2 0.05 ng of BamH1 digested pRSVL
  • Lane 4 BamH1 digest of HIRT supernatant from control muscle
  • Lane 5 BamH1 digest of cellular DNA from control, uninjected muscle
  • Lane 10 Cellular DNA (same as Lane 9) digested with BamH1 and Dpn1
  • Lane 11 Cellular DNA (Same as in Lane 9) digested with BamH1 and Mbo1
  • Lane 12 Cellular DNA digested with BgIII
  • Lane 13 HIRT supernatant digested with BgIII (Size markers (X/HindIII) are shown at the left).
  • pRSVL DNA was precipitated in ethanol and dried. The pellet was picked up with fine forceps and deposited into various muscle groups as described in the preceding examples. Five days later the muscle was analyzed for luciferase activity as described in Example 13. The DNA was efficiently expressed in different muscle groups as follows: Implant: Luciferase Activity (Light Units, LU) 25 ⁇ g pRSVL DNA Control Biceps Calf Quadriceps 428 46420 27577 159080 453 53585 34291 35512 1171 106865 53397 105176 499 40481
  • the rat lung differs from that of the human in having one large left lung off the left main bronchus. The left lung for this study was cut in half into a left upper part (LUL) and left lower part (LLL).
  • the right lung contains 4 lobes: right cranial lobe (RUL), right middle lobe (RML), right lower lobe (RLL), and an accessory lobe (AL). Extracts were prepared by mincing these lung parts into separate 1.5 ml microtubes containing 200 ⁇ l of a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl 2 and 0.1% Triton X), and grinding the lung with a plastic pestle. (Kontes) for one minute. In order to ensure complete disruption of the lung cells, the lung tissue was then placed under 600 psi of N 2 in a Parr bomb at 4° C. for 15 minutes before releasing the pressure.
  • a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl 2 and 0.1% Triton X)
  • Luciferase assays were done on 87.5 ⁇ l of lung extract out of a total volume of about 350 ⁇ l.
  • Injection RUL RLL LUL LML LLL AL Trachea Mock 22.6 22.4 21.9 21.3 20.1 19.8 — 25 ⁇ g DNA alone 21.2 21.5 21.8 21.6 21.9 21.2 — 25 ⁇ g DNA alone 21.7 21.4 21.3 — 22.2 21.5 — 250 ⁇ g DNA alone 21.7 23.2 21.9 28.5 22.6 22.0 21.3 250 ⁇ g DNA alone 22.9 22.5 33.3 23.0 25.4 24.3 21.5 250 ⁇ g DNA alone 21.8 21.5 21.8 20.4 20.7 20.8 20.7 25 ⁇ g DNA/CL 20.8 22.2 19.6 22.3 22.3 22.0 — 25 ⁇ g DNA/CL 22.9 22.0 22.7 21.7 22.8 — 22.18 25 ⁇ g DNA/CL 22.2 23.8 22.1 23.9 22.8 — 21.6 25 ⁇ g DNA/CL 20.9 20.9 20.6 20.3 — 19.3 25 ⁇ g
  • Mock Values are those for an animal that received 25 ⁇ g of DNA in 0.3 ml
  • 25 ⁇ g DNA alone represent separate animals that received intratracheal injections of 25 ⁇ g of pPGKLuc in 0.3 ml 20% sucrose.
  • 25 ⁇ g DNA/CL represent separate animals that received intratracheal injections of 25 Mg of pPGKLuc complexed with LipofectinTM in 0.3 ml 5% sucrose.
  • Luc Protein 10 4 l.u. represents an animal that received the equivalent of 30,000 light units (l.u.) of purified firefly luciferase (Sigma), and then was immediately sacrificed.
  • the DNA luciferase expression vector pPGKLuc was injected intrahepatically (1H) into the lower part of the left liver lobe in mice.
  • the pPGKLuc DNA was either injected by itself (450 Mg DNA in 1.0 ml 20% sucrose) or complexed with LipofectinTM (50 ⁇ g DNA+150 ⁇ g LipofectinTM in 1.0 ml 5% sucrose).
  • LipofectinTM 50 ⁇ g DNA+150 ⁇ g LipofectinTM in 1.0 ml 5% sucrose.
  • the left liver lobe was divided into two sections (a lower part where the lobe was injected and an upper part of the lobe distant from the injection site) and assayed for luciferase activity as described in the preceding examples.
  • mice were injected with the pXGH5 (metallothionein promoter-growth hormone fusion gene)(Selden Richard et al., Molec. Cell Biol. 6:3173-3179 (1986)) in both liver and muscle. The mice were placed on 76 mM zinc sulfate water. Later the animals were bled and the serum analyzed for growth hormone using the Nichols GH Kit.
  • pXGH5 metalothionein promoter-growth hormone fusion gene
  • mice Three mice were injected with 0.1 ml of 1 mg/ml of pXGH5 in 5% sucrose, 2 ⁇ in the quadriceps, 1 ⁇ in the hamstring muscle, 1 ⁇ in pectoralis muscle, and 1 ⁇ in trapezoid muscles on two separate days. The results were as follows: Animal No. Growth Hormone(ng/ml): Day 1 Day 2 1 0.6 0.6 2 0.8 1.0 3 0.95 0.8
  • mice were injected with a quantity of 20 ⁇ g of a plasmid construct consisting of the gp-120 gene, driven by a cytomegalovirus (CMV) promotor.
  • the DNA was injected into the quadriceps muscle of mice according to the methods described in Example 11.
  • Mouse 5 (FIG. 5A) was injected in the quadriceps muscle with 20 ⁇ g of plasmid DNA in isotonic sucrose.
  • Mouse 2 (FIG. 5B) was injected with sucrose solution alone. Blood samples were obtained prior to the injection (Day 0) at the times indicated on FIG. 5, up to more than 40 days post injection.
  • the serum from each sample was serially diluted and assayed in a standard ELISA technique assay for the detection of antibody, using recombinant gp-120 protein made in yeast as the antigen. Both IgG and IgM antibodies were detected. The study indicates that the gene reyains its signal sequence, and the protein is efficiently excreted from cells.
  • the cell line BALB/C C1.7 (TIB 80) was obtained from the American Type Tissue Culture Collection. These cells were transfected with the gp-120 gene construct described in Example 19. To 0.75 ml OptiMEMTM (Gibco. Inc.) were added 6.1 ⁇ g of DNA. The quantity of 30 ⁇ g of cationic liposomes (containing DOTMA and cholesterol in a 70:30 molar ratio) were added to another 0.75 ml OptiMEMTM. The mixtures were combined and 1.5 ml of OptiMEMTM containing 20% (v/v) fetal bovine calf serum was added.
  • OptiMEMTM containing 20% (v/v) fetal bovine calf serum
  • Two different DNA templates were constructed, both of which code for the synthesis of RNA that express the E. coli . ⁇ -galactosidase reporter gene.
  • a Lac-Z gene that contains the Kozak consensus sequence was inserted in place of the luciferase coding sequences of the p ⁇ GLuc ⁇ GA n template to generate the p ⁇ GLacZ ⁇ GA n template.
  • the pEMCLacZ ⁇ GA n template was made by replacing the 5′ ⁇ -globin untranslated sequences of p ⁇ GLacZ ⁇ GA n with the 588 bp EcoRl/Ncol fragment from the encephalomyocarditis virus (EMCV) (pE5LVPO in Parks, G.
  • EMCV encephalomyocarditis virus
  • EMC 5′ untranslated sequences had previously been shown to be Able to initiate efficient translation in vitro in reticulocytes lysates. We demonstrated that these sequences can also direct efficient translation when transfected into fibroblasts in culture. The percentage of blue cells was slightly greater in cells transfected with the uncapped EMCLacZ ⁇ GA n RNA than in cells transfected with the capped pEMCLacZ ⁇ GA n RNA.
  • pSV-G1-A pAR3126-SV40 promotor driving expression of T7 polymerase protein which is directed to the cytoplasm.
  • pSVNU-G1-A pAR3132-SV40 promotor driving expression of T7 polymerase protein which is directed to the cytoplasm.
  • mice and one newborn mouse were injected with the ⁇ gLuc ⁇ gA n mRNA containing the 5′ cap and prepared according to Example 13.
  • injections were from a stock solution of mRNA at 3.6 ⁇ g/ ⁇ l in 20% sucrose; injection volumes were 5 ⁇ 1,2 injections into each of the bilateral parietal cortex, 4 injections per mouse.
  • Tissue was assayed at 18 hours post injection, according to Example 13 using 200 ⁇ l of brain homogenate, disrupted in a Parr bomb, and 87.5 ⁇ l was taken for assay.
  • a plasmid containing the dystrophin gene under control of the Rous Sarcoma virus promoter was prepared from the Xp21 plasmid containing the complete dystrophin coding region and the SV40 poly.
  • a segment, which was cloned by Kunkel and colleagues. (Bruffle M., Monaco A P, Gillard E F, van Ommen G J, Affara N A, Ferguson-Smith M A, Kunkel L M, Lehrach H. A 10-megabase physical map of human Xp21, including the Duchenne muscular dystrophy gene. Genomics 1988 Apr 2 (3):189-202; Hoffman, E P and Kunkel, L M Dystrophin abnormalities of Duchenne's/Becher Muscular Dystrophy. Neuron Vol.
  • dystrophin gene product Functional expression of the dystrophin gene product in the dystrophic mice was detected by comparing the pattern of fluorescence observed in cross-sections of quadriceps muscle from injected animals, with the fluorescence pattern observed in normal animals.
  • Normal dystrophin expression is localized underneath the plasma membrane of the muscle fiber, so that a cross section of the quadriceps muscle give a fluorescence pattern encircling the cell.
  • dystrophin expression was quantitated by Western blot analysis using the affinity purified anti-60 kd antibody.
  • Respiratory muscles including the intercostal muscles which move the rib cage and the diaphragm, are particularly important impaired muscle groups in patients with muscular dystrophy.
  • the intercostals can be reached by injection through the skin as can the other skeletal muscle groups.
  • the diaphragm can be accessed by a surgical procedure to expose the muscle to direct injection of plasmid DNA.

Abstract

A method for delivering a naked or isolated polynucleotide to the interior of a cell in a vertebrate, comprising the interstitial introduction of a naked polynucleotide into a tissue of the vertebrate where the polynucleotide is taken up by the cells of the tissue and exerts a therapeutic effect on the vertebrate. The method can be used to deliver a therapeutic polypeptide to the cells of the vertebrate, to provide an immune response upon in vivo translation of the polynucleotide, to deliver antisense polynucleotides, to deliver receptors to the cells of the vertebrate, or to provide transitory gene therapy.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 09/452,872, filed Dec. 2, 1999, now allowed; which is a continuation of U.S. application Ser. No. 08/481,919, filed Jun. 7, 1995, now U.S. Pat. No. 6,214,804, issued Apr. 10, 2001; which is a continuation of U.S. application Ser. No. 07/496,991, filed Mar. 21, 1990, now abandoned; which is a continuation-in-part of U.S. application Ser. No. 07/467,881, filed Jan. 19, 1990, now abandoned; which is a continuation-in-part of U.S. application Ser. No. 07/326,305, filed Mar. 21, 1989, now abandoned; each of the foregoing disclosures is fully incorporated herein by reference.[0001]
  • BACKGROUND OF THE INVENTION
  • 1. Field of the Invention [0002]
  • The present invention relates to introduction of naked DNA and RNA sequences into a vertebrate to achieve controlled expression of a polypeptide. It is useful in gene therapy, vaccination, and any therapeutic situation in which a polypeptide should be administered to cells in vivo. [0003]
  • 2. Related Art [0004]
  • Current research in gene therapy has focused on “permanent” cures, in which DNA is integrated into the genome of the patient. Viral vectors are presently the most frequently used means for transforming the patient's cells and introducing DNA into the genome. In an indirect method, viral vectors, carrying new genetic information, are used to infect target cells removed from the body, and these cells are then re-implanted. Direct in vivo gene transfer into postnatal animals has been reported for formulations of DNA encapsulated in liposomes and DNA entrapped in proteoliposomes containing viral envelope receptor proteins (Nicolau et al., [0005] Proc. Natl. Acad Sci USA 80:1068-1072 (1983); Kaneda et al., Science 243:375-378 (1989); Mannino et al., Biotechniques 6:682-690 (1988). Positive results have also been described with calcium phosphate co-precipitated DNA (Benvenisty and Reshef Proc. Natl. Acad Sci USA 83:9551-9555 (1986)).
  • The clinical application of gene therapy, as well as the utilization of recombinant retrovirus vectors, has been delayed because of safety considerations. Integration of exogenous DNA into the genome of a cell can cause DNA damage and possible genetic changes in the recipient cell that could predispose to malignancy. A method which avoids these potential problems would be of significant benefit in making gene therapy safe and effective. [0006]
  • Vaccination with immunogenic proteins has eliminated or reduced the incidence of many diseases; however there are major difficulties in using proteins associated with other pathogens and disease states as immunogens. Many protein antigens are not intrinsically immunogenic. More often, they are not effective as vaccines because of the manner in which the immune system operates. [0007]
  • The immune system of vertebrates consists of several interacting components. The best characterized and most important parts are the humoral and cellular (cytolytic) branches. Humoral immunity involves antibodies, proteins which are secreted into the body fluids and which directly recognize an antigen. The cellular system, in contrast, relies on special cells which recognize and kill other cells which are producing foreign antigens. This basic functional division reflects two different strategies of immune defense. Humoral immunity is mainly directed at antigens which are exogenous to the animal whereas the cellular system responds to antigens which are actively synthesized within the animal. [0008]
  • Antibody molecules, the effectors of humoral immunity, are secreted by special B lymphoid cells, B cells, in response to antigen. Antibodies can bind to and inactivate antigen directly (neutralizing antibodies) or activate other cells of the immune system to destroy the antigen. [0009]
  • Cellular immune recognition is mediated by a special class of lymphoid cells, the cytotoxic T cells. These cells do not recognize whole antigens but instead they respond to degraded peptide fragments thereof which appear on the surface of the target cell bound to proteins called class I major histocompatibility complex (MHC) molecules. Essentially all nucleated cells have class I molecules. It is believed that proteins produced within the cell are continually degraded to peptides as part of normal cellular metabolism. These fragments are bound to the MHC molecules and are transported to the cell surface. Thus the cellular immune system is constantly monitoring the spectra of proteins produced in all cells in the body and is poised to eliminate any cells producing foreign antigens. [0010]
  • Vaccination is the process of preparing an animal to respond to an antigen. Vaccination is more complex than immune recognition and involves not only B cells and cytotoxic T cells but other types of lymphoid cells as well. During vaccination, cells which recognize the antigen (B cells or cytotoxic T cells) are clonally expanded. In addition, the population of ancillary cells (helper T cells) specific for the antigen also increase. Vaccination also involves specialized antigen presenting cells which can process the antigen and display it in a form which can stimulate one of the two pathways. [0011]
  • Vaccination has changed little since the time of Louis Pasteur. A foreign antigen is introduced into an animal where it activates specific B cells by binding to surface immunoglobulins. It is also taken up by antigen processing cells, wherein it is degraded, and appears in fragments on the surface of these cells bound to Class II MHC molecules. Peptides bound to class II molecules are capable of stimulating the helper class of T cells. Both helper T cells and activated B cells are required to produce active humoral immunization. Cellular immunity is thought to be stimulated by a similar but poorly understood mechanism. [0012]
  • Thus two different and distinct pathways of antigen processing produce exogenous antigens bound to class II MHC molecules where they can stimulate T helper cells, as well as endogenous proteins degraded and bound to class I MHC molecules and recognized by the cytotoxic class of T cells. There is little or no difference in the distribution of MHC molecules. Essentially all nucleated cells express class I molecules whereas class II MHC proteins are restricted to some few types of lymphoid cells. [0013]
  • Normal vaccination schemes will always produce a humoral immune response. They may also provide cytotoxic immunity. The humoral system protects a vaccinated individual from subsequent challenge from a pathogen and can prevent the spread of an intracellular infection if the pathogen goes through an extracellular phase during its life cycle; however, it can do relatively little to eliminate intracellular pathogens. Cytotoxic immunity complements the humoral system by eliminating the infected cells. Thus effective vaccination should activate both types of immunity. [0014]
  • A cytotoxic T cell response is necessary to remove intracellular pathogens such as viruses as well as malignant cells. It has proven difficult to present an exogenously administered antigen in adequate concentrations in conjunction with Class I molecules to assure an adequate response. This has severely hindered the development of vaccines against tumor-specific antigens (e.g., on breast or colon cancer cells), and against weakly immunogenic viral proteins (e.g., HIV, Herpes, non-A, non-B hepatitis, CMV and EBV). [0015]
  • It would be desirable to provide a cellular immune response alone in immunizing against agents such as viruses for which antibodies have been shown to enhance infectivity. It would also be useful to provide such a response against both chronic and latent viral infections and against malignant cells. [0016]
  • The use of synthetic peptide vaccines does not solve these problems because either the peptides do not readily associate with histocompatibility molecules, have a short serum half-life, are rapidly proteolyzed, or do not specifically localize to antigen-presenting monocytes and macrophages. At best, all exogenously administered antigens must compete with the universe of self-proteins for binding to antigen-presenting macrophages. [0017]
  • Major efforts have been mounted to elicit immune responses to poorly immunogenic viral proteins from the herpes viruses, non-A, non-B hepatitis, HIV, and the like. These pathogens are difficult and hazardous to propagate in vitro. As mentioned above, synthetic peptide vaccines corresponding to viral-encoded proteins have been made, but have severe pitfalls. Attempts have also been made to use vaccinia virus vectors to express proteins from other viruses. However, the results have been disappointing, since (a) recombinant vaccinia viruses may be rapidly eliminated from the circulation in already immune individuals, and (b) the administration of complex viral antigens may induce a phenomenon known as “antigenic competition,” in which weakly immunogenic portions of the virus fail to elicit an immune response because they are out-competed by other more potent regions of the administered antigen. [0018]
  • Another major problem with protein or peptide vaccines is anaphylactic reaction which can occur when injections of antigen are repeated in efforts to produce a potent immune response. In this phenomenon, IgE antibodies formed in response to the antigen cause severe and sometimes fatal allergic reactions. [0019]
  • Accordingly, there is a need for a method for invoking a safe and effective immune response to this type of protein or polypeptide. Moreover, there is a great need for a method that will associate these antigens with Class I histocompatibility antigens on the cell surface to elicit a cytotoxic T cell response, avoid anaphylaxis and proteolysis of the material in the serum, and facilitate localization of the material to monocytes and macrophages. [0020]
  • A large number of disease states can benefit from the administration of therapeutic peptides. Such peptides include lymphokines, such as interleukin-2, tumor necrosis factor, and the interferons; growth factors, such as nerve growth factor, epidermal growth factor, and human growth hormone; tissue plasminogen activator; factor VIII:C; granulocyte-macrophage colony-stimulating factor; erythropoietin; insulin; calcitonin; thymidine kinase; and the like. Moreover, selective delivery of toxic peptides (such as ricin, diphtheria toxin, or cobra venom factor) to diseased or neoplastic cells can have major therapeutic benefits. Current peptide delivery systems suffer from significant problems, including the inability to effectively incorporate functional cell surface receptors onto cell membranes, and the necessity of systemically administering large quantities of the peptide (with resultant undesirable systemic side effects) in order to deliver a therapeutic amount of the peptide into or onto the target cell. [0021]
  • These above-described problems associated with gene therapy, immunization, and delivery of therapeutic peptides to cells are addressed by the present invention. [0022]
  • SUMMARY OF THE INVENTION
  • The present invention provides a method for delivering a pharmaceutical or immunogenic polypeptide to the interior of a cell of a vertebrate in vivo, comprising the step of introducing a preparation comprising a pharmaceutically acceptable injectable carrier and a naked polynucleotide operatively coding for the polypeptide into the interstitial space of a tissue comprising the cell, whereby the naked polynucleotide is taken up into the interior of the cell and has an immunogenic or pharmacological effect on the vertebrate. Also provided is a method for introducing a polynucleotide into muscle cells in vivo, comprising the steps of providing a composition comprising a naked polynucleotide in a pharmaceutically acceptable carrier, and contacting the composition with muscle tissue of a vertebrate in vivo, whereby the polynucleotide is introduced into muscle cells of the tissue. The polynucleotide may be an antisense polynucleotide. Alternatively, the polynucleotide may code for a therapeutic peptide that is expressed by the muscle cells after the contacting step to provide therapy to the vertebrate. Similarly, it may code for an immunogenic peptide that is expressed by the muscle cells after the contacting step and which generates an immune response, thereby immunizing the vertebrate. [0023]
  • One particularly attractive aspect of the invention is a method for obtaining long term administration of a polypeptide to a vertebrate, comprising the step of introducing a naked DNA sequence operatively coding for the polypeptide interstitially into tissue of the vertebrate, whereby cells of the tissue produce the polypeptide for at least one month or at least 3 months, more preferably at least 6 months. In this embodiment of the invention, the cells producing the polypeptide are nonproliferating cells, such as muscle cells. [0024]
  • Another method according to the invention is a method for obtaining transitory expression of a polypeptide in a vertebrate, comprising the step of introducing a naked mRNA sequence operatively coding for the polypeptide interstitially into tissue of the vertebrate, whereby cells of the tissue produce the polypeptide for less than about 20 days, usually less than about 10 days, and often less than 3 or 5 days. For many of the methods of the invention, administration into solid tissue is preferred. [0025]
  • One important aspect of the invention is a method for treatment of muscular dystrophy, comprising the steps of introducing a therapeutic amount of a composition comprising a polynucleotide operatively coding for dystrophin in a pharmaceutically acceptable injectable carrier in vivo into muscle tissue of an animal suffering from muscular dystrophy, whereby the polynucleotide is taken up into the cells and dystrophin is produced in vivo. Preferably, the polynucleotide is a naked polynucleotide and the composition is introduced interstitially into the muscle tissue. [0026]
  • The present invention also includes pharmaceutical products for all of the uses contemplated in the methods described herein. For example, there is a pharmaceutical product, comprising naked polynucleotide, operatively coding for a biologically active polypeptide, in physiologically acceptable administrable form, in a container, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the polynucleotide for human or veterinary administration. Such notice, for example, may be the labeling approved by the U.S. Food and Drug Administration for prescription drugs, or the approved product insert. [0027]
  • In another embodiment, the invention provides a pharmaceutical product, comprising naked polynucleotide, operatively coding for a biologically active peptide, in solution in a physiologically acceptable injectable carrier and suitable for introduction interstitially into a tissue to cause cells of the tissue to express the polypeptide, a container enclosing the solution, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of manufacture, use, or sale of the solution of polynucleotide for human or veterinary administration. The peptide may be immunogenic and administration of the solution to a human may serve to vaccinate the human, or an animal. Similarly, the peptide may be therapeutic and administration of the solution to a vertebrate in need of therapy relating to the polypeptide will have a therapeutic effect. [0028]
  • Also provided by the present invention is a pharmaceutical product, comprising naked antisense polynucleotide, in solution in a physiologically acceptable injectable carrier and suitable for introduction interstitially into a tissue to cause cells of the tissue to take up the polynucleotide and provide a therapeutic effect, a container enclosing the solution, and a notice associated with the container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of manufacture, use, or sale of the solution of polynucleotide for human or veterinary administration. [0029]
  • One particularly important aspect of the invention relates to a pharmaceutical product for treatment of muscular dystrophy, comprising a sterile, pharmaceutically acceptable carrier, a pharmaceutically effective amount of a naked polynucleotide operatively coding for dystrophin in the carrier, and a container enclosing the carrier and the polynucleotide in sterile fashion. Preferably, the polynucleotide is DNA. [0030]
  • From yet another perspective, the invention includes a pharmaceutical product for use in supplying a biologically active polypeptide to a vertebrate, comprising a pharmaceutically effective amount of a naked polynucleotide operatively coding for the polypeptide, a container enclosing the carrier and the polynucleotide in a sterile fashion, and means associated with the container for permitting transfer of the polynucleotide from the container to the interstitial space of a tissue, whereby cells of the tissue can take up and express the polynucleotide. The means for permitting such transfer can include a conventional septum that can be penetrated, e.g., by a needle. Alternatively, when the container is a syringe, the means may be considered to comprise the plunger of the syringe or a needle attached to the syringe. [0031]
  • Containers used in the present invention will usually have at least 1, preferably at least 5 or 10, and more preferably at least 50 or 100 micrograms of polynucleotide, to provide one or more unit dosages. For many applications, the container will have at least 500 micrograms or 1 milligram, and often will contain at least 50 or 100 milligrams of polynucleotide. [0032]
  • Another aspect of the invention provides a pharmaceutical product for use in immunizing a vertebrate, comprising a pharmaceutically effective amount of a naked polynucleotide operatively coding for an immunogenic polypeptide, a sealed container enclosing the polynucleotide in a sterile fashion, and means associated with the container for permitting transfer of the polynucleotide from the container to the interstitial space of a tissue, whereby cells of the tissue can take up and express the polynucleotide. [0033]
  • Still another aspect of the present invention is the use of naked polynucleotide operatively coding for a physiologically active polypeptide in the preparation of a pharmaceutical for introduction interstitially into tissue to cause cells comprising the tissue to produce the polypeptide. The pharmaceutical, for example, may be for introduction into muscle tissue whereby muscle cells produce the polypeptide. Also contemplated is such use, wherein the peptide is dystrophin and the pharmaceutical is for treatment of muscular dystrophy. [0034]
  • Another use according to the invention is use of naked antisense polynucleotide in the preparation of a pharmaceutical for introduction interstitially into tissue of a vertebrate to inhibit translation of polynucleotide in cells of the vertebrate. [0035]
  • The tissue into which the polynucleotide is introduced can be a persistent, non-dividing cell. The polynucleotide may be either a DNA or RNA sequence. When the polynucleotide is DNA, it can also be a DNA sequence which is itself non-replicating, but is inserted into a plasmid, and the plasmid further comprises a replicator. The DNA may be a sequence engineered so as not to integrate into the host cell genome. The polynucleotide sequences may code for a polypeptide which is either contained within the cells or secreted therefrom, or may comprise a sequence which directs the secretion of the peptide. [0036]
  • The DNA sequence may also include a promoter sequence. In one preferred embodiment, the DNA sequence includes a cell-specific promoter that permits substantial transcription of the DNA only in predetermined cells. The DNA may also code for a polymerase for transcribing the DNA, and may comprise recognition sites for the polymerase and the injectable preparation may include an initial quantity of the polymerase. [0037]
  • In many instances, it is preferred that the polynucleotide is translated for a limited period of time so that the polypeptide delivery is transitory. The polypeptide may advantageously be a therapeutic polypeptide, and may comprise an enzyme, a hormone, a lymphokine, a receptor, particularly a cell surface receptor, a regulatory protein, such as a growth factor or other regulatory agent, or any other protein or peptide that one desires to deliver to a cell in a living vertebrate and for which corresponding DNA or mRNA can be obtained. [0038]
  • In preferred embodiments, the polynucleotide is introduced into muscle tissue; in other embodiments the polynucleotide is incorporated into tissues of skin, brain, lung, liver, spleen or blood. The preparation is injected into the vertebrate by a variety of routes, which may be intradermally, subdermally, intrathecally, or intravenously, or it may be placed within cavities of the body. In a preferred embodiment, the polynucleotide is injected intramuscularly. In still other embodiments, the preparation comprising the polynucleotide is impressed into the skin. Transdermal administration is also contemplated, as is inhalation. [0039]
  • In one preferred embodiment, the polynucleotide is DNA coding for both a polypeptide and a polymerase for transcribing the DNA, and the DNA includes recognition sites for the polymerase and the injectable preparation further includes a means for providing an initial quantity of the polymerase in the cell. The initial quantity of polymerase may be physically present together with the DNA. [0040]
  • Alternatively, it may be provided by including mRNA coding therefor, which mRNA is translated by the cell. In this embodiment of the invention, the DNA is preferably a plasmid. Preferably, the polymerase is phage T7 polymerase and the recognition site is a T7 origin of replication sequence. [0041]
  • In accordance with another aspect of the invention, there is provided a method for treating a disease associated with the deficiency or absence of a specific polypeptide in a vertebrate, comprising the steps of obtaining an injectable preparation comprising a pharmaceutically acceptable injectable carrier containing a naked polynucleotide coding for the specific polypeptide; introducing the injectable preparation into a vertebrate and permitting the polynucleotide to be incorporated into a cell, wherein the polypeptide is formed as the translation product of the polynucleotide, and whereby the deficiency or absence of the polypeptide is compensated for. In preferred embodiments, the preparation is introduced into muscle tissue and the method is applied repetitively. The method is advantageously applied where the deficiency or absence is due to a genetic defect. The polynucleotide is preferably a non-replicating DNA sequence; the DNA sequence may also be incorporated into a plasmid vector which comprises an origin of replication. [0042]
  • In one of the preferred embodiments, the polynucleotide codes for a non-secreted polypeptide, and the polypeptide remains in situ. According to this embodiment, when the polynucleotide codes for the polypeptide dystrophin, the method provides a therapy for Duchenne's syndrome; alternatively, when the polynucleotide codes for the polypeptide phenylalanine hydroxylase, the method comprises a therapy for phenylketonuria. In another preferred embodiment of the method, the polynucleotide codes for a polypeptide which is secreted by the cell and released into the circulation of the vertebrate; in a particularly preferred embodiment the polynucleotide codes for human growth hormone. [0043]
  • In yet another embodiment of the method, there is provided a therapy for hypercholesterolemia wherein a polynucleotide coding for a receptor associated with cholesterol homeostasis is introduced into a liver cell, and the receptor is expressed by the cell. [0044]
  • In accordance with another aspect of the present invention, there is provided a method for immunizing a vertebrate, comprising the steps of obtaining a preparation comprising an expressible polynucleotide coding for an immunogenic translation product, and introducing the preparation into a vertebrate wherein the translation product of the polynucleotide is formed by a cell of the vertebrate, which elicits an immune response against the immunogen. In one embodiment of the method, the injectable preparation comprises a pharmaceutically acceptable carrier containing an expressible polynucleotide coding for an immunogenic peptide, and on the introduction of the preparation into the vertebrate, the polynucleotide is incorporated into a cell of the vertebrate wherein an immunogenic translation product of the polynucleotide is formed, which elicits an immune response against the immunogen. [0045]
  • In an alternative embodiment, the preparation comprises one or more cells obtained from the vertebrate and transfected in vitro with the polynucleotide, whereby the polynucleotide is incorporated into said cells, where an immunogenic translation product of the polynucleotide is formed, and whereby on the introduction of the preparation into the vertebrate, an immune response against the immunogen is elicited. In any of the embodiments of the invention, the immunogenic product may be secreted by the cells, or it may be presented by a cell of the vertebrate in the context of the major histocompatibility antigens, thereby eliciting an immune response against the immunogen. The method may be practiced using non-dividing, differentiated cells from the vertebrates, which cells may be lymphocytes, obtained from a blood sample; alternatively, it may be practiced using partially differentiated skin fibroblasts which are capable of dividing. In a preferred embodiment, the method is practiced by incorporating the polynucleotide coding for an immunogenic translation product into muscle tissue. [0046]
  • The polynucleotide used for immunization is preferably an mRNA sequence, although a non-replicating DNA sequence may be used. The polynucleotide may be introduced into tissues of the body using the injectable carrier alone; liposomal preparations are preferred for methods in which in vitro transfections of cells obtained from the vertebrate are carried out. [0047]
  • The carrier preferably is isotonic, hypotonic, or weakly hypertonic, and has a relatively low ionic strength, such as provided by a sucrose solution. The preparation may further advantageously comprise a source of a cytokine which is incorporated into liposomes in the form of a polypeptide or as a polynucleotide. [0048]
  • The method may be used to selectively elicit a humoral immune response, a cellular immune response, or a mixture of these. In embodiments wherein the cell expresses major histocompatibility complex of Class I, and the immunogenic peptide is presented in the context of the Class I complex, the immune response is cellular and comprises the production of cytotoxic T-cells. [0049]
  • In one such embodiment, the immunogenic peptide is associated with a virus, is presented in the context of Class I antigens, and stimulates cytotoxic T-cells which are capable of destroying cells infected with the virus. A cytotoxic T-cell response may also be produced according the method where the polynucleotide codes for a truncated viral antigen lacking humoral epitopes. [0050]
  • In another of these embodiments, the immunogenic peptide is associated with a tumor, is presented in the context of Class I antigens, and stimulates cytotoxic T cells which are capable of destroying tumor cells. In yet another embodiment wherein the injectable preparation comprises cells taken from the animal and transfected in vitro, the cells expressing major histocompatibility antigen of class I and class II, and the immune response is both humoral and cellular and comprises the production of both antibody and cytotoxic T-cells. [0051]
  • In another embodiment, there is provided a method of immunizing a vertebrate, comprising the steps of obtaining a positively charged liposome containing an expressible polynucleotide coding for an immunogenic peptide, and introducing the liposome into a vertebrate, whereby the liposome is incorporated into a monocyte, a macrophage, or another cell, where an immunogenic translation product of the polynucleotide is formed, and the product is processed and presented by the cell in the context of the major histocompatibility complex, thereby eliciting an immune response against the immunogen. Again, the polynucleotide is preferably mRNA, although DNA may also be used. And as before, the method may be practiced without the liposome, utilizing just the polynucleotide in an injectable carrier. The present invention also encompasses the use of DNA coding for a polypeptide and for a polymerase for transcribing the DNA, and wherein the DNA includes recognition sites for the polymerase. The initial quantity of polymerase is provided by including mRNA coding therefor in the preparation, which mRNA is translated by the cell. The mRNA preferably is provided with means for retarding its degradation in the cell. This can include capping the mRNA, circularizing the mRNA, or chemically blocking the 5′ end of the mRNA. The DNA used in the invention may be in the form of linear DNA or may be a plasmid. Episomal DNA is also contemplated. One preferred polymerase is phage T7 RNA polymerase and a preferred recognition site is a T7 RNA polymerase promoter.[0052]
  • BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES
  • FIG. 1 comprises autoradiograms of chromatographic studies showing the expression of the CAT gene in mouse muscle. [0053]
  • FIGS. 2A to [0054] 2F comprise photomicrographs of muscle tissue stained for β-galactosidase activity following injection with the pRSVLac-Z DNA vector.
  • FIGS. 3A, 3B, and [0055] 3C present data for luciferase activity in muscle following the injection of βgLucβgAn into muscle.
  • FIG. 4 presents an autoradiogram of a Southern blot after analysis of extracts from pRSVL-injected muscle. [0056]
  • FIGS. 5A and 5B comprise graphs showing antibody production in mice following the injection of a gene for an immunogenic peptide. [0057]
  • FIGS. 6A and 6B comprise graphs showing antibody production in mice following the injection of mouse cells transfected with a gene for an immunogenic peptide.[0058]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The practice of the present invention requires obtaining naked polynucleotide operatively coding for a polypeptide for incorporation into vertebrate cells. A polynucleotide operatively codes for a polypeptide when it has all the genetic information necessary for expression by a target cell, such as promoters and the like. These polynucleotides can be administered to the vertebrate by any method that delivers injectable materials to cells of the vertebrate, such as by injection into the interstitial space of tissues such as muscles or skin, introduction into the circulation or into body cavities or by inhalation or insufflation. A naked polynucleotide is injected or otherwise delivered to the animal with a pharmaceutically acceptable liquid carrier. In preferred applications, the liquid carrier is aqueous or partly aqueous, comprising sterile, pyrogen-free water. The pH of the preparation is suitably adjusted and buffered. The polynucleotide can comprise a complete gene, a fragment of a gene, or several genes, together with recognition and other sequences necessary for expression. [0059]
  • In the embodiments of the invention that require use of liposomes, for example, when the polynucleotide is to be associated with a liposome, it requires a material for forming liposomes, preferably cationic or positively charged liposomes, and requires that liposomal preparations be made from these materials. With the liposomal material in hand, the polynucleotide may advantageously be used to transfect cells in vitro for use as immunizing agents, or to administer polynucleotides into bodily sites where liposomes may be taken up by phagocytic cells. [0060]
  • Polynucleotide Materials [0061]
  • The naked polynucleotide materials used according to the methods of the invention comprise DNA and RNA sequences or DNA and RNA sequences coding for polypeptides that have useful therapeutic applications. These polynucleotide sequences are naked in the sense that they are free from any delivery vehicle that can act to facilitate entry into the cell, for example, the polynucleotide sequences are free of viral sequences, particularly any viral particles which may carry genetic information. They are similarly free from, or naked with respect to, any material which promotes transfection, such as liposomal formulations, charged lipids such as Lipofectin™ or precipitating agents such as CaPO[0062] 4.
  • The DNA sequences used in these methods can be those sequences which do not integrate into the genome of the host cell. These may be non-replicating DNA sequences, or specific replicating sequences genetically engineered to lack the genome-integration ability. [0063]
  • The polynucleotide sequences of the invention are DNA or RNA sequences having a therapeutic effect after being taken up by a cell. Examples of polynucleotides that are themselves therapeutic are anti-sense DNA and RNA; DNA coding for an anti-sense RNA; or DNA coding for tRNA or rRNA to replace defective or deficient endogenous molecules. The polynucleotides of the invention can also code for therapeutic polypeptides. A polypeptide is understood to be any translation product of a polynucleotide regardless of size, and whether glycosylated or not. Therapeutic polypeptides include as a primary example, those polypeptides that can compensate for defective or deficient species in an animal, or those that act through toxic effects to limit or remove harmful cells from the body. [0064]
  • Therapeutic polynucleotides provided by the invention can also code for immunity-conferring polypeptides, which can act as endogenous immunogens to provoke a humoral or cellular response, or both. The polynucleotides employed according to the present invention can also code for an antibody. In this regard, the term “antibody” encompasses whole immunoglobulin of any class, chimeric antibodies and hybrid antibodies with dual or multiple antigen or epitope specificities, and fragments, such as F(ab)[0065] 2, Fab′, Fab and the like, including hybrid fragments. Also included within the meaning of “antibody” are conjugates of such fragments, and so-called antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference.
  • Thus, an isolated polynucleotide coding for variable regions of an antibody can be introduced, in accordance with the present invention, to enable the treated subject to produce antibody in situ. For illustrative methodology relating to obtaining antibody-encoding polynucleotides, see Ward et al. [0066] Nature, 341:544-546 (1989); Gillies et al., Biotechnol. 7:799-804 (1989); and Nakatani et al., loc. cit., 805-810 (1989). The antibody in turn would exert a therapeutic effect, for example, by binding a surface antigen associated with a pathogen. Alternatively, the encoded antibodies can be anti-idiotypic antibodies (antibodies that bind other antibodies) as described, for example, in U.S. Pat. No. 4,699,880. Such anti-idiotypic antibodies could bind endogenous or foreign antibodies in a treated individual, thereby to ameliorate or prevent pathological conditions associated with an immune response, e.g., in the context of an autoimmune disease.
  • Polynucleotide sequences of the invention preferably code for therapeutic or immunogenic polypeptides, and these sequences may be used in association with other polynucleotide sequences coding for regulatory proteins that control the expression of these polypeptides. The regulatory protein can act by binding to genomic DNA so as to regulate its transcription; [0067]
  • alternatively, it can act by binding to messenger RNA to increase or decrease its stability or translation efficiency. [0068]
  • The polynucleotide material delivered to the cells in vivo can take any number of forms, and the present invention is not limited to any particular polynucleotide coding for any particular polypeptide. Plasmids containing genes coding for a large number of physiologically active peptides and antigens or immunogens have been reported in the literature and can be readily obtained by those of skill in the art. [0069]
  • Where the polynucleotide is to be DNA, promoters suitable for use in various vertebrate systems are well known. For example, for use in murine systems, suitable strong promoters include RSV LTR, MPSV LTR, SV40 IEP, and metallothionein promoter. In humans, on the other hand, promoters such as CMV IEP may advantageously be used. All forms of DNA, whether replicating or non-replicating, which do not become integrated into the genome, and which are expressible, are within the methods contemplated by the invention. [0070]
  • With the availability of automated nucleic acid synthesis equipment, both DNA and RNA can be synthesized directly when the nucleotide sequence is known or by a combination of PCR cloning and fermentation. Moreover, when the sequence of the desired polypeptide is known, a suitable coding sequence for the polynucleotide can be inferred. [0071]
  • When the polynucleotide is mRNA, it can be readily prepared from the corresponding DNA in vitro. For example, conventional techniques utilize phage RNA polymerases SP6, T3, or T7 to prepare mRNA from DNA templates in the presence of the individual ribonucleoside triphosphates. An appropriate phage promoter, such as a T7 origin of replication site is placed in the template DNA immediately upstream of the gene to be transcribed. Systems utilizing T7 in this manner are well known, and are described in the literature, e.g., in Current Protocols in Molecular Biology, §3.8 (Vol. 1 1988). [0072]
  • One particularly preferred method for obtaining the mRNA used in the present invention is set forth in Examples 2-5. In general, however, it should be apparent that the pXGB plasmid or any similar plasmid that can be readily constructed by those of ordinary skill in the art can be used with a virtually unlimited number of cDNAs in practicing the present invention. Such plasmids may advantageously comprise a promoter for a desired RNA polymerase, followed by a 5′ untranslated region, a 3′ untranslated region, and a template for a poly A tract. There should be a unique restriction site between these 5′ and 3′ regions to facilitate the insertion of any desired cDNA into the plasmid. Then, after cloning the plasmid containing the desired gene, the plasmid is linearized by cutting in the polyadenylation region and is transcribed in vitro to form mRNA transcripts. These transcripts are preferably provided with a 5′ cap, as demonstrated in Example 5. [0073]
  • Alternatively, a 5′ untranslated sequence such as EMC can be used which does not require a 5′ cap. [0074]
  • While the foregoing represents a preferred method for preparing the mRNA, it will be apparent to those of skill in the art that many alternative methods also exist. For example, the mRNA can be prepared in commercially-available nucleotide synthesis apparatus. Alternatively, mRNA in circular form can be prepared. Exonuclease-resistant RNAs such as circular mRNA, chemically blocked mRNA, and mRNA with a 5′ cap are preferred, because of their greater half-life in vivo. [0075]
  • In particular, one preferred mRNA is a self-circularizing mRNA having the gene of interest preceded by the 5′ untranslated region of polio virus. It has been demonstrated that circular mRNA has an extremely long half-life (Harland & Misher, [0076] Development 102: 837-852 (1988)) and that the polio virus 5′ untranslated region can promote translation of mRNA without the usual 5′ cap (Pelletier & Sonnenberg, Nature 334:320-325 (1988), hereby incorporated by reference).
  • This material may be prepared from a DNA template that is self-splicing and generates circular “lariat” mRNAs, using the method of Been & Cech, Cell 47:206-216 (1986)(hereby incorporated by reference). We modify that template by including the 5′ untranslated region of the polio virus immediately upstream of the gene of interest, following the procedure of Maniatis, T. et al. [0077] Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y. (1982).
  • In addition, the present invention includes the use of mRNA that is chemically blocked at the 5′ and/or 3′ end to prevent access by RNAse. (This enzyme is an exonuclease and therefore does not cleave RNA in the middle of the chain.) Such chemical blockage can substantially lengthen the half life of the RNA in vivo. Two agents which may be used to modify RNA are available from Clonetech Laboratories, Inc., Palo Alto, Calif.: C2 AminoModifier (Catalog # 5204-1) and Amino-7-dUTP (Catalog # K1022-1). These materials add reactive groups to the RNA. After introduction of either of these agents onto an RNA molecule of interest, an appropriate reactive substituent can be linked to the RNA according to the manufacturer's instructions. By adding a group with sufficient bulk, access to the chemically modified RNA by RNAse can be prevented. [0078]
  • Transient Gene Therapy [0079]
  • Unlike gene therapies proposed in the past, one major advantage of the present invention is the transitory nature of the polynucleotide synthesis in the cells. (We refer to this as reversible gene therapy, transient gene therapy or TGT.) With mRNA introduced according to the present invention, the effect will generally last about one day. Also, in marked contrast to gene therapies proposed in the past, mRNA does not have to penetrate the nucleus to direct protein synthesis; therefore, it should have no genetic liability. [0080]
  • In some situations, however, a more prolonged effect may be desired without incorporation of the exogenous polynucleic acid into the genome of the host organism. [0081]
  • In order to provide such an effect, a preferred embodiment of the invention provides introducing a DNA sequence coding for a specific polypeptide into the cell. We have found, according to the methods of the invention, that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of about up to six months, and we have observed no evidence of integration of the DNA sequences into the genome of the cells. Alternatively, an even more prolonged effect can be achieved by introducing the DNA sequence into the cell by means of a vector plasmid having the DNA sequence inserted therein. [0082]
  • Preferably, the plasmid further comprises a replicator. Such plasmids are well known to those skilled in the art, for example, plasmid pBR322, with replicator pMB1, or plasmid pMK16, with replicator ColE1 (Ausubel, [0083] Current Protocols in Molecular Biology, John Wiley and Sons, New York (1988) §II:1.5.2.
  • Results of studies of the time course of expression of DNA and mRNA introduced into muscle cells as described in Examples 1 and 13 indicate that mRNA expression is more rapid, although shorter in duration than DNA expression. An immediate and long lived gene expression can be achieved by administering to the cell a liposomal preparation comprising both DNA and an RNA polymerase, such as the phage polymerases T7, T3, and SP6. The liposome also includes an initial source of the appropriate RNA polymerase, by either including the actual enzyme itself, or alternatively, an mRNA coding for that enzyme. When the liposome is introduced into the organism, it delivers the DNA and the initial source of RNA polymerase to the cell. The RNA polymerase, recognizing the promoters on the introduced DNA, transcribes both genes, resulting in translation products comprising more RNA polymerase and the desired polypeptide. Production of these materials continues until the introduced DNA (which is usually in the form of a plasmid) is degraded. In this manner, production of the desired polypeptide in vivo can be achieved in a few hours and be extended for one month or more. [0084]
  • Although not limited to the treatment of genetic disease, the methods of the invention can accordingly be appropriately applied to treatment strategies requiring delivery and functional expression of missing or defective genes. [0085]
  • The polynucleotides may be delivered to the interstitial space of tissues of the animal body, including those of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue. Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. [0086]
  • Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. We have discovered that in vivo muscle cells are particularly competent in their ability to take up and express polynucleotides. This ability may be due to the singular tissue architecture of muscle, comprising multinucleated cells, sarcoplasmic reticulum, and transverse tubular system. Polynucleotides may enter the muscle through the transverse tubular system, which contains extracellular fluid and extends deep into the muscle cell. It is also possible that the polynucleotides enter damaged muscle cells which then recover. [0087]
  • Muscle is also advantageously used as a site for the delivery and expression of polynucleotides in a number of therapeutic applications because animals have a proportionately large muscle mass which is conveniently accessed by direct injection through the skin; for this reason, a comparatively large dose of polynucleotides can be deposited in muscle by multiple injections, and repetitive injections, to extend therapy over long periods of time, are easily performed and can be carried out safely and without special skill or devices. [0088]
  • Muscle tissue can be used as a site for injection and expression of polynucleotides in a set of general strategies, which are exemplary and not exhaustive. First, muscle disorders related to defective or absent gene products can be treated by introducing polynucleotides coding for a non-secreted gene product into the diseased muscle tissue. In a second strategy, disorders of other organs or tissues due to the absence of a gene product, and which results in the build-up of a circulating toxic metabolite can be treated by introducing the specific therapeutic polypeptide into muscle tissue where the non-secreted gene product is expressed and clears the circulating metabolite. In a third strategy, a polynucleotide coding for an secretable therapeutic polypeptide can be injected into muscle tissue from where the polypeptide is released into the circulation to seek a metabolic target. This use is demonstrated in the expression of growth hormone gene injected into muscle, Example 18. Certain DNA segments, are known to serve as “signals” to direct secretion (Wickner, W. T. and H. F. Lodish, [0089] Science 230:400-407 (1985), and these may be advantageously employed. Finally, in immunization strategies, muscle cells may be injected with polynucleotides coding for immunogenic peptides, and these peptides will be presented by muscle cells in the context of antigens of the major histocompatibility complex to provoke a selected immune response against the immunogen.
  • Tissues other than those of muscle, and having a less efficient uptake and expression of injected polynucleotides, may nonetheless be advantageously used as injection sites to produce therapeutic polypeptides or polynucleotides under certain conditions. One such condition is the use of a polynucleotide to provide a polypeptide which to be effective must be present in association with cells of a specific type; for example, the cell surface receptors of liver cells associated with cholesterol homeostasis. (Brown and Goldstein, [0090] Science 232:34-47 (1986)). In this application, and in many others, such as those in which an enzyme or hormone is the gene product, it is not necessary to achieve high levels of expression in order to effect a valuable therapeutic result.
  • One application of TGT is in the treatment of muscular dystrophy. The genetic basis of the muscular dystrophies is just beginning to be unraveled. The gene related to Duchenne/Becker muscular dystrophy has recently been cloned and encodes a rather large protein, termed dystrophin. Retroviral vectors are unlikely to be useful, because they could not accommodate the rather large size of the cDNA (about 13 kb) for dystrophin. Very recently reported work is centered on transplanting myoblasts, but the utility of this approach remains to be determined. Clearly, an attractive approach would be to directly express the dystrophin gene within the muscle of patients with Duchennes. Since most patients die from respiratory failure, the muscles involved with respiration would be a primary target. [0091]
  • Another application is in the treatment of cystic fibrosis. The gene for cystic fibrosis was recently identified (Goodfellow, P. [0092] Nature, 341(6238):102-3 (Sep. 14, 1989); Rommens, J. et al. Science, 245(4922):1059-1065 (Sep. 8, 1989); Beardsley, T. et al., Scientific American, 261(5):28-30 (1989). Significant amelioration of the symptoms should be attainable by the expression of the dysfunctional protein within the appropriate lung cells. The bronchial epithelial cells are postulated to be appropriate target lung cells and they could be accessible to gene transfer following instillation of genes into the lung. Since cystic fibrosis is an autosomal recessive disorder one would need to achieve only about 5% of normal levels of the cystic fibrosis gene product in order to significantly ameliorate the pulmonary symptoms.
  • Biochemical genetic defects of intermediary metabolism can also be treated by TGT. These diseases include phenylketonuria, galactosemia, maple-syrup urine disease, homocystinuria, propionic acidemia, methylmalonic acidemia, and adenosine deaminase deficiency. The pathogenesis of disease in most of these disorders fits the phenylketonuria (PKU) model of a circulating toxic metabolite. That is, because of an enzyme block, a biochemical, toxic to the body, accumulates in body fluids. These disorders are ideal for gene therapy for a number of reasons. First, only 5% of normal levels of enzyme activity would have to be attained in order to significantly clear enough of the circulating toxic metabolite so that the patient is significantly improved. Second, the transferred gene could most often be expressed in a variety of tissues and still be able to clear the toxic biochemical. [0093]
  • Reversible gene therapy can also be used in treatment strategies requiring intracytoplasmic or intranuclear protein expression. Some proteins are known that are capable of regulating transcription by binding to specific promoter regions on nuclear DNA. Other proteins bind to RNA, regulating its degradation, transport from the nucleus, or translation efficiency. Proteins of this class must be delivered intracellularly for activity. Extracellular delivery of recombinant transcriptional or translational regulatory proteins would not be expected to have biological activity, but functional delivery of the DNA or RNA by TGT would be active. Representative proteins of this type that would benefit from TGT would include NEF, TAT, steroid receptor and the retinoid receptor. [0094]
  • Gene therapy can be used in a strategy to increase the resistance of an AIDS patient to HIV infection. Introducing an AIDS resistance gene, such as, for example, the NEF gene or the soluble CD4 gene to prevent budding, into an AIDS patient's T cells will render his T cells less capable of producing active AIDS virus, thus sparing the cells of the immune system and improving his ability to mount a T cell dependent immune response. Thus, in accordance with the invention, a population of the AIDS patient's own T cells is isolated from the patient's blood. These cells are then transfected in vitro and then reintroduced back into the patient's blood. The virus-resistant cells will have a selective advantage over the normal cells, and eventually repopulate the patient's lymphatic system. DNA systemic delivery to macrophages or other target cells can be used in addition to the extracorporeal treatment strategy. Although this strategy would not be expected to eradicate virus in the macrophage reservoir, it will increase the level of T cells and improve the patient's immune response. [0095]
  • In all of the systemic strategies presented herein, an effective DNA or mRNA dosage will generally be in the range of from about 0.05 μg/kg to about 50 mg/kg, usually about 0.005-5 mg/kg. However, as will be appreciated, this dosage will vary in a manner apparent to those of skill in the art according to the activity of the peptide coded for by the DNA or mRNA and the particular peptide used. For delivery of adenosine deaminase to mice or humans, for example, adequate levels of translation are achieved with a DNA or mRNA dosage of about 0.5 to 5 mg/kg. See Example 10. From this information, dosages for other peptides of known activity can be readily determined. [0096]
  • Diseases which result from deficiencies of critical proteins may be appropriately treated by introducing into specialized cells, DNA or mRNA coding for these proteins. A variety of growth factors such as nerve growth factor and fibroblast growth factor have been shown to affect neuronal cell survival in animal models of Alzheimer's disease. In the aged rat model, NGF infusions have reversed the loss of cholinergic neurons. In the fimbria-fomix lesion rat, NGF infusions or secretion from genetically-modified fibroblasts have also avoided the loss of cholinergic function. Cholinergic activity is diminished in patients with Alzheimer's. The expression within the brain of transduced genes expressing growth factors could reverse the loss of function of specific neuronal groups. [0097]
  • Introduction of DNA or mRNA by transfection of the gene for neuronal growth factor into cells lining the cranial cavity can be used in accordance with the present invention in the treatment of Alzheimer's disease. [0098]
  • In particular, the present invention treats this disease by intracranial injection of from about 10 μg to about 100 μg of DNA or mRNA into the parenchyma through use of a stereotaxic apparatus. Specifically, the injection is targeted to the cholinergic neurons in the medial septum. The DNA or mRNA injection is repeated every 1-3 days for 5′ capped, 3′ polyadenylated mRNA, and every week to 21 days for circular mRNA, and every 30 to 60 days for DNA. [0099]
  • Injection of DNA in accordance with the present invention is also contemplated. DNA would be injected in corresponding amounts; however, frequency of injection would be greatly reduced. Episomal DNA, for example, could be active for a number of months, and reinjection would only be necessary upon notable regression by the patient. [0100]
  • In addition, the enzymes responsible for neurotransmitter synthesis could be expressed from transduced genes. For example, the gene for choline acetyl transferase could be expressed within the brain cells (neurons or glial) of specific areas to increase acetylcholine levels and improve brain function. [0101]
  • The critical enzymes involved in the synthesis of other neurotransmitters such as dopamine, norepinephrine, and GABA have been cloned and available. The critical enzymes could be locally increased by gene transfer into a localized area of the brain. The increased productions of these and other neurotransmitters would have broad relevance to manipulation of localized neurotransmitter function and thus to a broad range of brain disease in which disturbed neurotransmitter function plays a crucial role. Specifically, these diseases could include schizophrenia and manic-depressive illnesses and Parkinson's Disease. It is well established that patients with Parkinson's suffer from progressively disabled motor control due to the lack of dopamine synthesis within the basal ganglia. The rate limiting step for dopamine synthesis is the conversion of tyrosine to L-DOPA by the enzyme, tyrosine hydroxylase. L-DOPA is then converted to dopamine by the ubiquitous enzyme, DOPA decarboxylase. That is why the well-established therapy with L-DOPA is effective (at least for the first few years of treatment). Gene therapy could accomplish the similar pharmacologic objective by expressing the genes for tyrosine hydroxylase and possibly DOPA decarboxylase as well. Tyrosine is readily available within the CNS. [0102]
  • The genetic form of alpha-1-antitrypsin deficiency can result in both liver and lung disease. The liver disease, which is less common, is caused by the accumulation of an abnormal protein and would be less amenable to gene therapy. The pulmonary complications, however, would be amenable to the increased expression of alpha-1-antitrypsin within the lung. This should prevent the disabling and eventually lethal emphysema from developing. [0103]
  • Alpha-1-antitrypsin deficiency also occurs in tobacco smokers since tobacco smoke decreases alpha-1-antitrypsin activity and thus serine protease activity that leads to emphysema. In addition, some recent data links tobacco smoke's anti-trypsin effect to aneurysms of the aorta. Aneurysms would also be preventable by raising blood levels of anti-1-antitrypsin since this would decrease protease activity that leads to aneurysms. [0104]
  • Patients with degenerative disease of the lung could also benefit from the expression of enzymes capable of removing other toxic metabolites which tend to accumulate in diseased lung tissue. Superoxide dismutase and catalase could be delivered by TGT to ameliorate these problems. [0105]
  • TGT can be used in treatment strategies requiring the delivery of cell surface receptors. It could be argued that there is no need to decipher methodology for functional in vivo delivery of genes. There is, after all, an established technology for the synthesis and large scale production of proteins, and proteins are the end product of gene expression. This logic applies for many protein molecules which act extracellularly or interact with cell surface receptors, such as tissue plasminogen activator (TPA), growth hormone, insulin, interferon, granulocyte-macrophage colony stimulating factor (GMCSF), erythropoietin (EPO), etc. However, the drug delivery problems associated with properly delivering a recombinant cell surface receptor to be inserted in the plasma membrane of its target cell in the proper orientation for a functional receptor have hithertofore appeared intractable. When DNA or RNA coding for a cell surface receptor is delivered intracellularly in accordance with the present invention, the resulting protein can be efficiently and functionally expressed on the target cell surface. If the problem of functional delivery of recombinant cell surface receptors remains intractable, then the only way of approaching this therapeutic modality will be through gene delivery. Similar logic for nuclear or cytoplasmic regulation of gene expression applies to nuclear regulatory factor bound to DNA to regulate (up or down) RNA transcription and to cytoplasmic regulatory factors which bind to RNA to increase or decrease translational efficiency and degradation. TGT could in this way provide therapeutic strategies for the treatment of cystic fibrosis, muscular dystrophy and hypercholesterolemia. [0106]
  • Elevated levels of cholesterol in the blood may be reduced in accordance with the present invention by supplying mRNA coding for the LDL surface receptor to hepatocytes. A slight elevation in the production of this receptor in the liver of patients with elevated LDL will have significant therapeutic benefits. Therapies based on systemic administration of recombinant proteins are not able to compete with the present invention, because simply administering the recombinant protein could not get the receptor into the plasma membrane of the target cells. The receptor must be properly inserted into the membrane in order to exert its biological effect. It is not usually necessary to regulate the level of receptor expression; the more expression the better. This simplifies the molecular biology involved in preparation of the mRNA for use in the present invention. For example, lipid/DNA or RNA complexes containing the LDL receptor gene may be prepared and supplied to the patient by repetitive I.V. injections. The lipid complexes will be taken up largely by the liver. Some of the complexes will be taken up by hepatocytes. The level of LDL receptor in the liver will increase gradually as the number of injections increases. Higher liver LDL receptor levels will lead to therapeutic lowering of LDL and cholesterol. An effective mRNA dose will generally be from about 0.1 to about 5 mg/kg. [0107]
  • Other examples of beneficial applications of TGT include the introduction of the thymidine kinase gene into macrophages of patients infected with the HIV virus. Introduction of the thymidine kinase gene into the macrophage reservoir will render those cells more capable of phosphorylating AZT. This tends to overcome their resistance to AZT therapy, making AZT capable of eradicating the HIV reservoir in macrophages. Lipid/DNA complexes containing the thymidine kinase gene can be prepared and administered to the patient through repetitive intravenous injections. The lipid complexes will be taken up largely by the macrophage reservoir leading to elevated levels of thymidine kinase in the macrophages. This will render the AZT resistant cells subject to treatment with AZT. The thymidine kinase therapy can also be focused by putting the thymidine kinase gene under the control of the HTLV III promoter. According to this strategy, the thymidine kinase would only be synthesized on infection of the cell by HIV virus, and the production of the Tat protein which activates the promoter. An analogous therapy would supply cells with the gene for diphtheria toxin under the control of the same HTLV III promoter, with the lethal result occurring in cells only after HIV infection. [0108]
  • These AIDS patients could also be treated by supplying the interferon gene to the macrophages according to the TGT method. Increased levels of localized interferon production in macrophages could render them more resistant to the consequences of HIV infection. While local levels of interferon would be high, the overall systemic levels would remain low, thereby avoiding the systemic toxic effects like those observed after recombinant interferon administration. Lipid/DNA or RNA complexes containing the interferon gene can be prepared and administered to the patient by repetitive intravenous injections. The lipid complexes will be taken up largely by the macrophage reservoir leading to elevated localized levels of interferon in the macrophages. This will render them less susceptible to HIV infection. [0109]
  • Various cancers may be treated using TGT by supplying a diphtheria toxin gene on a DNA template with a tissue specific enhancer to focus expression of the gene in the cancer cells. Intracellular expression of diphtheria toxin kills cells. These promoters could be tissue-specific such as using a pancreas-specific promoter for the pancreatic cancer. A functional diphtheria toxin gene delivered to pancreatic cells could eradicate the entire pancreas. This strategy could be used as a treatment for pancreatic cancer. The patients would have no insurmountable difficulty surviving without a pancreas. The tissue specific enhancer would ensure that expression of diphtheria toxin would only occur in pancreatic cells. DNA/lipid complexes containing the diphtheria toxin gene under the control of a tissue specific enhancer would be introduced directly into a cannulated artery feeding the pancreas. The infusion would occur on some dosing schedule for as long as necessary to eradicate the pancreatic tissue. Other lethal genes besides diphtheria toxin could be used with similar effect, such as genes for ricin or cobra venom factor or enterotoxin. [0110]
  • Also, one could treat cancer by using a cell-cycle specific promoter that would only kill cells that are rapidly cycling (dividing) such as cancer cells. Cell-cycle specific killing could also be accomplished by designing mRNA encoding killer proteins that are stable only in cycling cells (i.e. histone mRNA that is only stable during S phase). Also, one could use developmental-specific promoters such as the use of alpha-fetoprotein that is only expressed in fetal liver cells and in hepatoblastoma cells that have dedifferentiated into a more fetal state. [0111]
  • One could also treat specialized cancers by the transfer of genes such as the retinoblastoma gene (and others of that family) that suppress the cancer properties of certain cancers. [0112]
  • The TGT strategy can be used to provide a controlled, sustained delivery of peptides. Conventional drugs, as well as recombinant protein drugs, can benefit from controlled release devices. The purpose of the controlled release device is to deliver drugs over a longer time period, so that the number of doses required is reduced. This results in improvements in patient convenience and compliance. There are a wide variety of emerging technologies that are intended to achieve controlled release. [0113]
  • TGT can be used to obtain controlled delivery of therapeutic peptides. Regulated expression can be obtained by using suitable promoters, including cell-specific promoters. Suitable peptides delivered by the present invention include, for example, growth hormone, insulin, interleukins, interferons, GMCSF, EPO, and the like. Depending on the specific application, the DNA or an RNA construct selected can be designed to result in a gene product that is secreted from the injected cells and into the systemic circulation. [0114]
  • TGT can also comprise the controlled delivery of therapeutic polypeptides or peptides which is achieved by including with the polynucleotide to be expressed in the cell, an additional polynucleotide which codes for a regulatory protein which controls processes of transcription and translation. These polynucleotides comprise those which operate either to up regulate or down regulate polypeptide expression, and exert their effects either within the nucleus or by controlling protein translation events in the cytoplasm. [0115]
  • The T7 polymerase gene can be used in conjunction with a gene of interest to obtain longer duration of effect of TGT. Episomal DNA such as that obtained from the origin of replication region for the Epstein Barr virus can be used, as well as that from other origins of replication which are functionally active in mammalian cells, and preferably those that are active in human cells. This is a way to obtain expression from cells after many cell divisions, without risking unfavorable integration events that are common to retrovirus vectors. Controlled release of calcitonin could be obtained if a calcitonin gene under the control of its own promoter could be functionally introduced into some site, such as liver or skin. Cancer patients with hypercalcemia would be a group to whom this therapy could be applied. [0116]
  • Other gene therapies using TGT can include the use of a polynucleotide that has a therapeutic effect without being translated into a polypeptide. For example, TGT can be used in the delivery of anti-sense polynucleotides for turning off the expression of specific genes. Conventional anti-sense methodology suffers from poor efficacy, in part, because the oligonucleotide sequences delivered are too short. With TGT, however, full length anti-sense sequences can be delivered as easily as short oligomers. Anti-sense polynucleotides can be DNA or RNA molecules that themselves hybridize to (and, thereby, prevent transcription or translation of) an endogenous nucleotide sequence. [0117]
  • Alternatively, an anti-sense DNA may encode an RNA that hybridizes to an endogenous sequence, interfering with translation. Other uses of TGT in this vein include delivering a polynucleotide that encodes a tRNA or rRNA to replace a defective or deficient endogenous tRNA or rRNA, the presence of which causes the pathological condition. [0118]
  • Cell-specific promoters can also be used to permit expression of the gene only in the target cell. For example, certain genes are highly promoted in adults only in particular types of tumors. Similarly, tissue-specific promoters for specialized tissue, e.g., lens tissue of the eye, have also been identified and used in heterologous expression systems. [0119]
  • Beyond the therapies described, the method of the invention can be used to deliver polynucleotides to animal stock to increase production of milk in dairy cattle or muscle mass in animals that are raised for meat. [0120]
  • DNA and mRNA Vaccines [0121]
  • According to the methods of the invention, both expressible DNA and mRNA can be delivered to cells to form therein a polypeptide translation product. If the nucleic acids contain the proper control sequences, they will direct the synthesis of relatively large amounts of the encoded protein. When the DNA and mRNA delivered to the cells codes for an immunizing peptide, the methods can be applied to achieve improved and more effective immunity against infectious agents, including intracellular viruses, and also against tumor cells. [0122]
  • Since the immune systems of all vertebrates operate similarly, the applications described can be implemented in all vertebrate systems, comprising mammalian and avian species, as well as fish. [0123]
  • The methods of the invention may be applied by direct injection of the polynucleotide into cells of the animal in vivo, or by in vitro transfection of some of the animal cells which are then re-introduced into the animal body. [0124]
  • The polynucleotides may be delivered to various cells of the animal body, including muscle, skin, brain, lung, liver, spleen, or to the cells of the blood. Delivery of the polynucleotides directly in vivo is preferably to the cells of muscle or skin. The polynucleotides may be injected into muscle or skin using an injection syringe. They may also be delivered into muscle or skin using a vaccine gun. [0125]
  • It has recently been shown that cationic lipids can be used to facilitate the transfection of cells in certain applications, particularly in vitro transfection. Cationic lipid based transfection technology is preferred over other methods; it is more efficient and convenient than calcium phosphate, DEAE dextran or electroporation methods, and retrovirus mediated transfection, as discussed previously, can lead to integration events in the host cell genome that result in oncogene activation or other undesirable consequences. The knowledge that cationic lipid technology works with messenger RNA is a further advantage to this approach because RNA is turned over rapidly by intracellular nucleases and is not integrated into the host genome. A transfection system that results in high levels of reversible expression is preferred to alternative methodology requiring selection and expansion of stably transformed clones because many of the desired primary target cells do not rapidly divide in culture. [0126]
  • The ability to transfect cells at high efficiency with cationic liposomes provides an alternative method for immunization. The gene for an antigen is introduced in to cells which have been removed from an animal. The transfected cells, now expressing the antigen, are reinjected into the animal where the immune system can respond to the (now) endogenous antigen. The process can possibly be enhanced by coinjection of either an adjuvant or lymphokines to further stimulate the lymphoid cells. [0127]
  • Vaccination with nucleic acids containing a gene for an antigen may also provide a way to specifically target the cellular immune response. Cells expressing proteins which are secreted will enter the normal antigen processing pathways and produce both a humoral and cytotoxic response. The response to proteins which are not secreted is more selective. Non-secreted proteins synthesized in cells expressing only class I MHC molecules are expected to produce only a cytotoxic vaccination. Expression of the same antigen in cells bearing both class I and class II molecules may produce a more vigorous response by stimulating both cytotoxic and helper T cells. Enhancement of the immune response may also be possible by injecting the gene for the antigen along with a peptide fragment of the antigen. The antigen is presented via class I MHC molecules to the cellular immune system while the peptide is presented via class II MHC molecules to stimulate helper T cells. In any case, this method provides a way to stimulate and modulate the immune response in a way which has not previously been possible. [0128]
  • A major disadvantage of subunit vaccines is that glycoprotein antigens are seldom modified correctly in the recombinant expression systems used to make the antigens. Introducing the gene for a glycoprotein antigen will insure that the protein product is synthesized, modified and processed in the same species and cells that the pathogen protein would be. Thus, the expression of a gene for a human viral glycoprotein will contain the correct complement of sugar residues. This is important because it has been demonstrated that a substantial component of the neutralizing antibodies in some viral systems are directed at carbohydrate epitopes. [0129]
  • Any appropriate antigen which is a candidate for an immune response, whether humoral or cellular, can be used in its nucleic acid form. The source of the cells could be fibroblasts taken from an individual which provide a convenient source of cells expressing only class I MHC molecules. Alternatively, peripheral blood cells can be rapidly isolated from whole blood to provide a source of cells containing both class I and class II MHC proteins. They could be further fractionated into B cells, helper T cells, cytotoxic T cells or macrophage/monocyte cells if desired. Bone marrow cells can provide a source of less differentiated lymphoid cells. In all cases the cell will be transfected either with DNA containing a gene for the antigen or by the appropriate capped and polyadenylated mRNA transcribed from that gene or a circular RNA, chemically modified RNA, or an RNA which does not require 5′ capping. The choice of the transfecting nucleotide may depend on the duration of expression desired. For vaccination purposes, a reversible expression of the immunogenic peptide, as occurs on mRNA transfection, is preferred. Transfected cells are injected into the animal and the expressed proteins will be processed and presented to the immune system by the normal cellular pathways. [0130]
  • Such an approach has been used to produce cytotoxic immunity in model systems in mice. Cell lines, malignant continuously growing cells, can be stably transformed with DNA. When cells are injected into animals, they induce cellular immunity to the expressed antigen. The cationic lipid delivery system will allow this approach to be extended to normal, non-malignant cells taken from a patient. [0131]
  • There are several applications to this approach of targeting cellular immunity. The first is vaccination against viruses in which antibodies are known to be required or to enhanced viral infection. There are two strategies that can be applied here. One can specifically target the cellular pathway during immunization thus eliminating the enhancing antibodies. Alternatively one can vaccinate with the gene for a truncated antigen which eliminates the humoral epitopes which enhance infectivity. [0132]
  • The use of DNA or mRNA vaccine therapy could similarly provide a means to provoke an effective cytotoxic T-cell response to weakly antigenic tumors. We propose, for example, that if a tumor-specific antigen were expressed by mRNA inside a cell in an already processed form, and incorporated directly into the Class I molecules on the cell surface, a cytotoxic T cell response would be elicited. [0133]
  • A second application is that this approach provides a method to treat latent viral infections. Several viruses (for example, Hepatitis B, HIV and members of the Herpes virus group) can establish latent infections in which the virus is maintained intracellularly in an inactive or partially active form. There are few ways of treating such infections. However, by inducing a cytolytic immunity against a latent viral protein, the latently infected cells will be targeted and eliminated. [0134]
  • A related application of this approach is to the treatment of chronic pathogen infections. There are numerous examples of pathogens which replicate slowly and spread directly from cell to cell. These infections are chronic, in some cases lasting years or decades. Examples of these are the slow viruses (e.g. Visna), the Scrapie agent and HIV. One can eliminate the infected cells by inducing a cellular response to proteins of the pathogen. [0135]
  • Finally, this approach may also be applicable to the treatment of malignant disease. Vaccination to mount a cellular immune response to a protein specific to the malignant state, be it an activated oncogene, a fetal antigen or an activation marker, will result in the elimination of these cells. [0136]
  • The use of DNA/mRNA vaccines could in this way greatly enhance the immunogenicity of certain viral proteins, and cancer-specific antigens, that normally elicit a poor immune response. The mRNA vaccine technique should be applicable to the induction of cytotoxic T cell immunity against poorly immunogenic viral proteins from the Herpes viruses, non-A, non-B hepatitis, and HIV, and it would avoid the hazards and difficulties associated with in vitro propagation of these viruses. For cell surface antigens, such as viral coat proteins (e.g., HIV gp120), the antigen would be expressed on the surface of the target cell in the context of the major histocompatibility complex (MHC), which would be expected to result in a more appropriate, vigorous and realistic immune response. It is this factor that results in the more efficacious immune responses frequently observed with attenuated virus vaccines. Delivery of a single antigen gene by TGT would be much safer than attenuated viruses, which can result in a low frequency of disease due to inadequate attenuation. [0137]
  • There is an additional advantage of TGT which can be exploited during the vaccine development phase. One of the difficulties with vaccine development is the requirement to screen different structural variants of the antigen, for the optimal immune response. If the variant is derived from a recombinant source, the protein usually must be expressed and purified before it can be tested for antigenicity. This is a laborious and time consuming process. With in vitro mutagenesis, it is possible to obtain and sequence numerous clones of a given antigen. If these antigens can be screened for antigenicity at the DNA or RNA level by TGT, the vaccine development program could be made to proceed much faster. [0138]
  • Finally, in the case of the DNA/mRNA vaccines, the protein antigen is never exposed directly to serum antibody, but is always produced by the transfected cells themselves following translation of the mRNA. Hence, anaphylaxis should not be a problem. Thus, the present invention permits the patient to be immunized repeatedly without the fear of allergic reactions. The use of the DNA/mRNA vaccines of the present invention makes such immunization possible. [0139]
  • One can easily conceive of ways in which this technology can be modified to enhance still further the immunogenicity of antigens. T cell immunization can be augmented by increasing the density of Class I and Class II histocompatibility antigens on the macrophage or other cell surface and/or by inducing the transfected cell to release cytokines that promote lymphocyte proliferation. To this end, one may incorporate in the same liposomes that contain mRNA for the antigen, other mRNA species that encode interferons or interleukin-1. These cytokines are known to enhance macrophage activation. Their systemic use has been hampered because of side effects. However, when encapsulated in mRNA, along with mRNA for antigen, they should be expressed only by those cells that co-express antigen. In this situation, the induction of T cell immunity can be enhanced greatly. [0140]
  • Therapeutic Formulations [0141]
  • Polynucleotide salts: Administration of pharmaceutically acceptable salts of the polynucleotides described herein is included within the scope of the invention. Such salts may be prepared from pharmaceutically acceptable non-toxic bases including organic bases and inorganic bases. Salts derived from inorganic bases include sodium, potassium, lithium, ammonium, calcium, magnesium, and the like. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, basic amino acids, and the like. For a helpful discussion of pharmaceutical salts, see S. M. Berge et al., [0142] Journal of Pharmaceutical Sciences 66:1-19 (1977) the disclosure of which is hereby incorporated by reference.
  • Polynucleotides for injection, a preferred route of delivery, may be prepared in unit dosage form in ampules, or in multidose containers. The polynucleotides may be present in such forms as suspensions, solutions, or emulsions in oily or preferably aqueous vehicles. Alternatively, the polynucleotide salt may be in lyophilized form for reconstitution, at the time of delivery, with a suitable vehicle, such as sterile pyrogen-free water. Both liquid as well as lyophilized forms that are to be reconstituted will comprise agents, preferably buffers, in amounts necessary to suitably adjust the pH of the injected solution. For any parenteral use, particularly if the formulation is to be administered intravenously, the total concentration of solutes should be controlled to make the preparation isotonic, hypotonic, or weakly hypertonic. Nonionic materials, such as sugars, are preferred for adjusting tonicity, and sucrose is particularly preferred. Any of these forms may further comprise suitable formulatory agents, such as starch or sugar, glycerol or saline. The compositions per unit dosage, whether liquid or solid, may contain from 0.1% to 99% of polynucleotide material. [0143]
  • The units dosage ampules or multidose containers, in which the polynucleotides are packaged prior to use, may comprise an hermetically sealed container enclosing an amount of polynucleotide or solution containing a polynucleotide suitable for a pharmaceutically effective dose thereof, or multiples of an effective dose. The polynucleotide is packaged as a sterile formulation, and the hermetically sealed container is designed to preserve sterility of the formulation until use. [0144]
  • The container in which the polynucleotide is packaged is labeled, and the label bears a notice in the form prescribed by a governmental agency, for example the Food and Drug Administration, which notice is reflective of approval by the agency under Federal law, of the manufacture, use, or sale of the polynucleotide material therein for human administration. [0145]
  • Federal law requires that the use of pharmaceutical agents in the therapy of humans be approved by an agency of the Federal government. Responsibility for enforcement is the responsibility of the Food and Drug Administration, which issues appropriate regulations for securing such approval, detailed in 21 U.S.C. 301-392. Regulation for biologic material, comprising products made from the tissues of animals is provided under 42 U.S.C 262. Similar approval is required by most foreign countries. Regulations vary from country to country, but the individual procedures are well known to those in the art. [0146]
  • Dosage and Route of Administration [0147]
  • The dosage to be administered depends to a large extent on the condition and size of the subject being treated as well as the frequency of treatment and the route of administration. Regimens for continuing therapy, including dose and frequency may be guided by the initial response and clinical judgment. The parenteral route of injection into the interstitial space of tissues is preferred, although other parenteral routes, such as inhalation of an aerosol formulation, may be required in specific administration, as for example to the mucous membranes of the nose, throat, bronchial tissues or lungs. [0148]
  • In preferred protocols, a formulation comprising the naked polynucleotide in an aqueous carrier is injected into tissue in amounts of from 10 μl per site to about 1 ml per site. The concentration of polynucleotide in the formulation is from about 0.1 μg/ml to about 20 mg/ml. [0149]
  • Regulation of TGT [0150]
  • Just as DNA based gene transfer protocols require appropriate signals for transcribing (promoters, enhancers) and processing (splicing signals, polyadenylation signals) the mRNA transcript, mRNA based TGT requires the appropriate structural and sequence elements for efficient and correct translation, together with those elements which will enhance the stability of the transfected mRNA. [0151]
  • In general, translational efficiency has been found to be regulated by specific sequence elements in the 5′ non-coding or untranslated region (5′UTR) of the RNA. Positive sequence motifs include the translational initiation consensus sequence (GCC) ACCATGG (Kozak, [0152] Nucleic Acids Res. 15:8125 (1987)) and the 5 G 7 methyl GpppG cap structure (Drummond et al., Nucleic Acids Res. 13:7375 (1985)). Negative elements include stable intramolecular 5′ UTR stem-loop structures (Muesing et al., Cell 48:691(1987)) and AUG sequences or short open reading frames preceded by an appropriate AUG in the 5′ UTR (Kozak, Supra, Rao et al., Mol. and Cell. Biol. 8:284(1988)). In addition, certain sequence motifs such as the beta globin 5′ UTR may act to enhance translation (when placed adjacent to a heterologous 5′ UTR) by an unknown mechanism. There are also examples of specific 5′ UTR sequences which regulate eukaryotic translational efficiency in response to environmental signals. These include the human ferritin 5′ UTR (Hentze et al., Proc. Natl. Acad. Sci. USA 84:6730 (1987)) and the drosophila hsp 70 5′ UTR (Klemenz et al., EMBO Journal 4:2053 (1985)). Finally, there are viral 5′ UTR sequences which are able to bypass normal cap dependent translation and translational controls and mediate an efficient translation of viral or chimeric mRNAs (Dolph et al., J. of Virol. 62:2059 (1988)), Pelletier and Sonnenberg, Nature 334, 320 (1988)). mRNA based TGT protocols must therefore include appropriate 5′ UTR translational elements flanking the coding sequence for the protein of interest.
  • In addition to translational concerns, mRNA stability must be considered during the development of mRNA based TGT protocols. As a general statement, capping and 3′ polyadenylation are the major positive determinants of eukaryotic mRNA stability (Drummond, supra; Ross, [0153] Mol. Biol. Med. 5:1(1988)) and function to protect the 5′ and 3′ ends of the mRNA from degradation. However, regulatory elements which affect the stability of eukaryotic mRNAs have also been defined, and therefore must be considered in the development of mRNA TGT protocols. The most notable and clearly defined of these are the uridine rich 3′ untranslated region (3′ UTR) destabilizer sequences found in many short half-life mRNAs (Shaw and Kamen Cell 46:659 (1986)), although there is evidence that these are not the only sequence motifs which result in mRNA destabilization (Kabnick and Housman, Mol. and Cell. Biol. 8:3244 (1988)). In addition, specific regulatory sequences which modulate cellular mRNA half-life in response to environmental stimuli have also been demonstrated. These include the estrogen mediated modulation of Vitellogenin mRNA stability (Brock and Shapiro, Cell 34:207 (1983)), the iron dependent regulation of transferrin receptor mRNA stability (Mullner and Kuhn, Cell 53:815 (1988)) which is due to a specific 3′ UTR motif, the prolactin mediated control of Casein mRNA stability (Guyette et al., Cell 17:1013 (1989)), the regulation of Fibronectin mRNA stability in response to a number of stimuli (Dean et al., J. Cell. Biol. 106:2159 (1988)), and the control of Histone mRNA stability (Graves et al., Cell 48:615 (1987)). Finally, just as viral RNA sequences have evolved which bypass normal eukaryotic mRNA translational controls, likewise some viral RNA sequences seem to be able to confer stability in the absence of 3′ polyadenylation (McGrae and Woodland, Eur. J. of Biochem. 116: 467 (1981)). Some 5′, such as EMC, according to Example 21, are known to function without a cap. This cacophony of stability modulating elements must also be carefully considered in developing mRNA based TGT protocols, and can be used to modulate the effect of an mRNA treatment.
  • Liposome-Forming Materials [0154]
  • The science of forming liposomes is now well developed. Liposomes are unilamellar or multilamellar vesicles, having a membrane portion formed of lipophilic material and an interior aqueous portion. The aqueous portion is used in the present invention to contain the polynucleotide material to be delivered to the target cell. It is preferred that the liposome forming materials used herein have a cationic group, such as a quaternary ammonium group, and one or more lipophilic groups, such as saturated or unsaturated alkyl groups having from about 6 to about 30 carbon atoms. One group of suitable materials is described in European Patent Publication No. 0187702. These materials have the formula: [0155]
    Figure US20040023911A1-20040205-C00001
  • wherein R[0156] 1 and R2 are the same or different and are alkyl or alkenyl of 6 to 22 carbon atoms, R3, R4, and R5 are the same or different and are hydrogen, alkyl of 1 to 8 carbons, aryl, aralkyl of 7 to 11 carbons, or when two or three of R3, R4, and R5 are taken together they form quinuclidino, piperidino, pyrrolidino, or morpholino; n is 1 to 8, and X is a pharmaceutically acceptable anion, such as a halogen. These compounds may be prepared as detailed in the above-identified patent application; alternatively, at least one of these compounds, N-(2,3-di-(9-(Z)-octadecenyloxy))-prop-1-yl-N,N,N-trimethylammonium chloride (DOTMA), is commercially available from Bethesda Research Laboratories (BRL), Gaithersburg, Md. 20877, USA.
  • These quaternary ammonium diether compounds, however, do have some drawbacks. Because of the ether linkages, they are not readily metabolized in vivo. When long-term therapy is contemplated, there is some possibility that these materials could accumulate in tissue, ultimately resulting in lipid storage disease and toxic side effects. Accordingly, a preferred class of compositions for use in the present invention has the formula: [0157]
    Figure US20040023911A1-20040205-C00002
  • wherein R[0158] 1 and R2 are the same or different and are alkyl or alkenyl of 5 to 21 carbon atoms, R3, R4, and R5 are the same or different and are hydrogen, alkyl of 1 to 8 carbons, aryl, aralkyl of 7 to 11 carbons, or when two or three of R3, R4, and R5 are taken together they form quinuclidino, piperidino, pyrrolidino, or morpholino; n is 1 to 8, and X is a pharmaceutically acceptable anion, such as a halogen. These compounds may be prepared using conventional techniques, such as nucleophilic substitution involving a carboxylic acid and an alkyl halide, by transesterification, or by condensation of an alcohol with an acid or an acid halide.
  • Moreover, many suitable liposome-forming cationic lipid compounds are described in the literature. See, e.g., L. Stamatatos, et al., [0159] Biochemistry 27:3917-3925 (1988); H. Eibl, et al., Biophysical Chemistry 10:261-271 (1979).
  • Liposome Preparation [0160]
  • Suitable liposomes for use in the present invention are commercially available. DOTMA liposomes, for example, are available under the trademark Lipofectin from Bethesda Research Labs, Gaithersburg, Md. [0161]
  • Alternatively, liposomes can be prepared from readily-available or freshly synthesized starting materials of the type previously described. The preparation of DOTAP liposomes is detailed in Example 6. Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Felgner, et al., [0162] Proc. Nat'l Acad. Sci. USA 84:7413-7417. Similar methods can be used to prepare liposomes from other cationic lipid materials. Moreover, conventional liposome forming materials can be used to prepare liposomes having negative charge or neutral charge. Such materials include phosphatidyl choline, cholesterol, phosphatidyl-ethanolamine, and the like. These materials can also advantageously be mixed with the DOTAP or DOTMA starting materials in ratios from 0% to about 75%.
  • Conventional methods can be used to prepare other, noncationic liposomes. These liposomes do not fuse with cell walls as readily as cationic liposomes. However, they are taken up by macrophages in vivo, and are thus particularly effective for delivery of polynucleotide to these cells. For example, commercially-available dioleoyl-phosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water. The sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15° C. Alternatively, negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size. Other methods are known and available to those of skill in the art. [0163]
  • The present invention is described below in detail using the 23 examples given below; however, the methods described are broadly applicable as described herein and are not intended to be limited by the Examples. [0164]
  • EXAMPLE 1 Preparation of Liposome-Forming DOTAP
  • The cationic liposome-forming [0165] material 1,2-bis(oleoyloxy)-3-(trimethylammonio)propane (DOTAP) is prepared as reported by L. Stamatatos, et al. (supra) or H. Eibl, et al. (supra).
  • Briefly, Stamatatos, et al. report that 1 mmol of 3-bromo-1,2-propanediol (Aldrich) was acylated for 48 hours at 20° C. with 3 mmol of oleyl chloride (freshly prepared from oleic acid and oxaloyl chloride) in dry, alcohol-free diethyl ether (20 ml) containing 5 mmol of dry pyridine. The precipitate of pyridinium hydrochloride was filtered off, and the filtrate was concentrated under nitrogen and redissolved in 10 ml of hexane. The hexane solution was washed 3 times with an equal volume of 1:1 methanol/0.1 N aqueous NCOONa, pH 3.0, 3 times with 1:1 methanol/0.1 N aqueous NaOH, and 1 time with 1% aqueous NaCl. The crude 3-bromo-1,2-bis-(oleolyloxy)propane was then stirred for 72 hours in a sealed tube with a solution of 15% trimethylamine in dry dimethyl sulfoxide (30 ml) at 25° C. The products of this reaction were dissolved in chloroform (200 ml), which was repeatedly washed with 1:1 methanol/100 mM aqueous HCOONa, pH 3.0, and then evaporated in vacuo to yield a light yellow oil. This material was purified on a column of silicic acid (Bio-Sil A, Bio-Rad Laboratories), eluting with a 0-15% gradient of methanol in chloroform to give the desired product in pure form at 9-10% methanol. The purified product was a colorless, viscous oil that migrates with an R[0166] f of 0.4 on thin layer chromatography plates (silica gel G) that were developed with 50:15:5:5:2 CHCl3/acetone/CH3OH/CH3COOH/H2O.
  • EXAMPLE 2 Preparation of Plasmids for Making DNA Templates for any Gene of Interest
  • Suitable template DNA for production of mRNA coding for a desired polypeptide may be prepared in accordance with standard recombinant DNA methodology. As has been previously reported (P. Kreig, et al., [0167] Nucleic Acids Res. 12:7057-7070 (1984)), a 5′ cap facilitates translation of the mRNA. Moreover, the 3′ flanking regions and the poly A tail are believed to increase the half life of the mRNA in vivo.
  • The readily-available SP6 cloning vector pSP64T provides 5′ and 3′ flanking regions from β-globin, an efficiently translated mRNA. The construction of this plasmid is detailed by Kreig, et al. (supra), and is hereby incorporated by this reference. Any cDNA containing an initiation codon can be introduced into this plasmid, and mRNA can be prepared from the resulting template DNA. This particular plasmid can be cut with BglII to insert any desired cDNA coding for a polypeptide of interest. [0168]
  • Although good results can be obtained with pSP64T when linearized and then transcribed in vivo with SP6 RNA polymerase, we prefer to use the Xenopus β-globin flanking sequences of pSP64T with phage T7 RNA polymerase. These flanking sequences are purified from pSP64T as the small (approx. 150 bp) HindIII to EcoRI fragment. These sequences are then inserted into a purified linear HindIII/EcoRI fragment (approx. 2.9 k bp) from pIBI 31 (commercially available from International Biotechnologies, Inc., Newhaven, Conn. 06535) with T4 DNA ligase. Resulting plasmids, designated pXBG, are screened for orientation and transformed into [0169] E. coli. These plasmids are adapted to receive any gene of interest at a unique BglII restriction site, which is situated between the two xenopus β-globin sequences.
  • EXAMPLE 3 Preparation of Plasmid Coding for Chloramphenicol Acetyltransferase
  • A convenient marker gene for demonstrating in vivo expression of exogenous polynucleotides is chloramphenicol acetyltransferase, CAT. A plasmid pSP-CAT containing the CAT gene flanked by the xenopus β-[0170] globin 5′ and 3′ sequences was produced by adding the CAT gene into the BgIII site of pSP64T. We used CAT gene in the form of the small BamHI/HindIII fragment from pSV2-CAT (available from the American Type Culture Collection, Rockville, Md., Accession No. 37155). However, the CAT gene is commonly used in molecular biology and is available from numerous sources. Both the CAT BamHI/HindIII fragment and the BgIII-cleaved pSP64T were incubated with the Klenow fragment to generate blunt ends, and were then ligated with T4 DNA ligase to form pSP-CAT.
  • The small PstI/HindIII fragment was then generated and purified, which comprises the CAT gene between the 5′ and 3′ β-globin flanking sequences of pSP64T. pIBI31 (International Biotechnologies, Inc.) was cleaved with PstI and HindIII, and the long linear sequence was purified. This fragment was then combined with the CAT-gene containing sequence and the fragments were ligated with T4 DNA ligase to form a plasmid designated pT7CAT An. Clones are selected on the basis of β-galactosidase activity with Xgal and ampicillin resistance. [0171]
  • EXAMPLE 4 Preparation of Purified DNA Template
  • The plasmid DNA from Example 3 is grown up and prepared as per Maniatis (supra), except without RNAse, using 2 CsCl spins to remove bacterial RNA. Specifically, [0172] E. coli containing pT7CAT An from Example 3 was grown up in ampicillin-containing LB medium. The cells were then pelleted by spinning at 5000 rpm for 10 min. in a Sorvall RC-5 centrifuge (E. I. DuPont, Burbank, Calif. 91510), resuspended in cold TE, pH 8.0, centrifuged again for 10 min. at 5000 rpm., resuspended in a solution of 50 mM glucose, 25 mM Tris-Cl pH 8.0, 10 mM EDTA, and 40 mg/ml lysozyme. After incubation for 5 to 10 minutes with occasional inversion, 0.2 N NaOH containing 1% SDS was added, followed after 10 minutes at 0° C. with 3 M potassium acetate and 2 M acetic acid. After 10 more minutes, the material was again centrifuged at 6000 rpm, and the supernatant was removed with a pipet. The pellet was then mixed into 0.6 vol. isopropanol (−20° C.), mixed, and stored at −20° C. for 15 minutes. The material was then centrifuged again at 10,000 rpm for 20 min., this time in an HB4 swinging bucket rotor apparatus (DuPont, supra) after which the supernatant was removed and the pellet was washed in 70% EtOH and dried at room temperature. Next, the pellet was resuspended in 3.5 ml TE, followed by addition of 3.4 g CsCl and 350 μl of 5 mg/ml EtBr. The resulting material was placed in a quick seal tube, filled to the top with mineral oil. The tube was spun for 3.5 hours at 80,000 rpm in a VTi80 centrifuge (Beckman Instruments, Pasadena, Calif., 91051). The band was removed, and the material was centrifuged again, making up the volume with 0.95 g CsCl/ml and 0.1 ml or 5 mg/ml EtBr/ml in TE. The EtBr was then extracted with an equal volume of TE saturated N-Butanol after adding 3 volumes of TE to the band, discarding the upper phase until the upper phase is clear. Next, 2.5 vol. EtOH was added, and the material was precipitated at −20 C for 2 hours. The resultant DNA precipitate is used as a DNA template for preparation of mRNA in vitro.
  • EXAMPLE 5 Preparation of mRNA for Transfection
  • The DNA from Example 4 was linearized downstream of the poly A tail with a 5-fold excess of PstI. The linearized DNA was then purified with two phenol/chloroform extractions, followed by two chloroform extractions. DNA was then precipitated with NaOAc (0.3 M) and 2 volumes of EtOH. The pellet was resuspended at about 1 mg/ml in DEP-treated deionized water. [0173]
  • Next, a transcription buffer was prepared, comprising 400 mM Tris HCl (pH 8.0), 80 mM MgCl[0174] 2, 50 mM DTT, and 40 mM spermidine. Then, the following materials were added in order to one volume of DEP-treated water at room temperature: 1 volume T7 transcription buffer, prepared above; rATP, rCTP, and rUTP to 1 mM concentration; rGTP to 0.5 mM concentration; 7 meG (5′) ppp (5′) G cap analog (New England Biolabs, Beverly, Mass., 01951) to 0.5 mM concentration; the linearized DNA template prepared above to 0.5 mg/ml concentration; RNAsin (Promega, Madison, Wis.) to 2000 U/ml concentration; and T7 RNA polymerase (N.E. Biolabs) to 4000 U/ml concentration.
  • This mixture was incubated for 1 hour at 37 C. The successful transcription reaction was indicated by increasing cloudiness of the reaction mixture. [0175]
  • Following generation of the mRNA, 2 U RQ1 DNAse (Promega) per microgram of DNA template used was added and was permitted to digest the template for 15 minutes. Then, the RNA was extracted twice with chloroform/phenol and twice with chloroform. The supernatant was precipitated with 0.3 M NaOAc in 2 volumes of EtOH, and the pellet was resuspended in 100 μl DEP-treated deionized water per 500 μl transcription product. This solution was passed over an RNAse-free Sephadex G50 column ([0176] Boehringer Mannheim #100 411). The resultant mRNA was sufficiently pure to be used in transfection of vertebrates in vivo.
  • EXAMPLE 6 Preparation of Liposomes
  • A number of liposome preparation methods can be used to advantage in the practice of the present invention. One particularly preferred liposome is made from DOTAP as follows: [0177]
  • A solution of 10 mg dioleoyl phosphatidylethanolamine (PE) and 10 mg DOTAP (from Example 1) in 1 ml chloroform is evaporated to dryness under a stream of nitrogen, and residual solvent is removed under vacuum overnight. Liposomes are prepared by resuspending the lipids in deionized water (2 ml) and sonicating to clarity in a closed vial. These preparations are stable for at least 6 months. [0178]
  • Polynucleotide complexes were prepared by mixing 0.5 ml polynucleotide solution (e.g., from Example 5) at 0.4 mg/ml by slow addition through a syringe with constant gentle vortexing to a 0.5 ml solution of sonicated DOTMA/PE or DOTAP/PE liposomes at 20 mg/ml, at room temperature. This procedure results in positively charged complexes which will spontaneously deliver the polynucleotide into cells in vivo. Different ratios of positively charged liposome to polynucleotide can be used to suit the particular need in any particular situation. Alternatively, as reported by Felgner, et al. (supra), it may be advantageous to dilute the polynucleotide (DNA or RNA) with Hepes buffered saline (150 mM NaCl; 20 mM Hepes, pH 7.4) prior to combining the materials to spontaneously form liposome/polynucleotide complexes. In many instances, however, the use of solutions having low ionic strength (such as sucrose) instead of saline solution is believed to be preferable; in particular, it is believed that such solutions facilitate delivery of polynucleotide to the cell by minimizing precipitation of polynucleotide/lipid complex. [0179]
  • EXAMPLE 7 In Vivo Expression of Liposomally and Non-Liposomally Introduced mRNA in the Rat
  • The ability of mRNA coding for chloramphenicol acetyl transferase (CAT) to transfect cells in vivo and the subsequent expression of the CAT protein was demonstrated by directly injecting 0.200 ml of each of the formulations below, prepared as indicated, into the abdominal muscle of rats, forming a bleb. Six replicates of each formulation were tested. After 12 to 14 h, the segment of the abdominal muscle into which the injection was made, weighing approximately 0.1 to 0.2 grams, was excised, minced, and placed in a 1.5 ml disposable mortar (Kontes, Morton Grove, Ill.) together with 200 μl of an aqueous formulation having the following components: 20 mM Tris, pH 7.6; 2 mM MgCl[0180] 2; and 0.1% Triton X-100 surfactant. The contents of the mortar were then ground for 1 minute with a disposable pestle. The mortar was then covered (with Parafilm) and placed in a 1 liter Parr cell disrupter bomb (Parr Instrument Company, Moline, Ill.) and pressurized to 6 atmospheres with nitrogen at 4° C. After 30 minutes, the pressure was quickly released to disrupt the tissue and produce a crude lysate. The lysate was then centrifuged in a microcentrifuge at 13,000 rpm, 4° C., for 10 minutes. The supernatant was then decanted and stored at −20° C. until analyzed.
  • The lysates were then assayed for the presence of the CAT protein by thin-layer chromatography. First, 75 pl of each sample (the supernatant prepared above) was incubated for two hours at 37° C. with 5 μl C14 chloramphenicol (Amersham); 20 [0181] μl 4 mM Acetyl CoA; and 50 μl 1 M Tris, pH 7.8. Thereafter, 20 pl of 4 mM Acetyl CoA was added, and the mixture was again incubated for 2 hours at 37° C. The resulting solution was extracted with 1 ml EtOAc, and the organic phase was removed and lyophilized in a vacuum centrifuge (SpeedVac, Savant Co.). The pellet was resuspended in 20 μl EtOAc, and was spotted onto a silica gel thin layer chromatography plate. The plate was developed for 45 minutes in 95% chloroform/5% methanol, was dried, and was sprayed with a radioluminescent indicator (Enhance Spray for Surface Radiography, New England Nuclear Corp.). The plate was then sandwiched with Kodak XAR5 film with overnight exposure at −70° C., and the film was developed per manufacturer's instructions. The following results were obtained:
    mRNA Expression
    FORMULATION (No. positive/total)
    1. 1 ml Optimem; 37.5 μg DOTMA 0/6
    2. 1 ml Optimem; 15 μg CAT RNA 3/6
    3. Formulation 1 plus 15 μg CAT RNA 4/6
    4. 10% Sucrose; 37.5 μg DOTMA; 15 μg CAT RNA 3/6
    5. 10% Sucrose; 187 μg DOTMA; 75 μg CAT RNA 0/6
  • Optimem: Serum-free media (Gibco Laboratories, Life Technologies, Inc, Grand Island, N.Y. 14072) [0182]
  • DOTMA: (Lipofectin brand; Bethesda Research Labs, Gaithersburg, Md.) [0183]
  • CAT RNA: From Example 5 [0184]
  • All formulations made up in DEPC-treated RNAse-free water (International Biotechnologies, Inc., New Haven, Conn. 06535). [0185]
  • EXAMPLE 8 mRNA Vaccination of Mice to Produce the gp120 Protein of HIV Virus
  • A liposomal formulation containing mRNA coding for the gp120 protein of the HIV virus is prepared according to Examples 1 through 5, except that the gene for gp120 (pIIIenv3-1 from the Aids Research and Reagent Program, National Institute of Allergy and Infectious Disease, Rockville, Md. 20852) is inserted into the plasmid pXBG in the procedure of Example 4. A volume of 200 μl of a formulation, prepared according to Example 6, and containing 200 μg/ml of gp120 mRNA and 500 μg/ml 1:1 DOTAP/PE in 10% sucrose is injected into the tail vein of [0186] mice 3 times in one day. At about 12 to 14 h after the last injection, a segment of muscle is removed from the injection site, and prepared as a cell lysate according to Example 7. The HIV specific protein gp120 is identified in the lysate also according to the procedures of Example 7.
  • The ability of gp120 antibody present in serum of the mRNA vaccinated mice to protect against HIV infection is determined by a HT4-6C plaque reduction assay, as follows: [0187]
  • HT4-6C cells (CD4+HeLa cells) are obtained from Dr. Bruce Chesebro, (Rocky Mountain National Lab, Montana) and grown in culture in RPMI media (BRL, Gaithersburg, Md.). The group of cells is then divided into batches. Some of the batches are infected with HIV by adding approximately 105 to 106 infectious units of HIV to approximately 107 HT4-6C cells. Other batches are tested for the protective effect of gp120 immune serum against HIV infection by adding both the HIV and approximately 50 μl of serum from a mouse vaccinated with gp120 mRNA. After 3 days of incubation, the cells of all batches are washed, fixed and stained with crystal violet, and the number of plaques counted. The protective effect of gp120 immune serum is determined as the reduction in the number of plaques in the batches of cells treated with both gp120 mRNA-vaccinated mouse serum and HIV compared to the number in batches treated with HIV alone. [0188]
  • EXAMPLE 9 mRNA Vaccination of Human Stem Cell-Bearing SCID Mice with NEF mRNA Followed by HIV Challenge
  • Severe combined immunodeficient mice (SCID mice (Molecular Biology Institute, (MBI), La Jolla, Calif. 92037)) were reconstituted with adult human peripheral blood lymphocytes by injection into the peritoneal cavity according to the method of Mosier (Mosier et al., [0189] Nature 335:256 (1988)). Intraperitoneal injection of 400 to 4000 infectious units of HIV-1 was then performed. The mice were maintained in a P3 level animal containment facility in sealed glove boxes.
  • mRNA coding for the Nef protein if HIV was prepared by obtaining the nef gene in the form of a plasmid (pGM92, from the NIAID, Rockville, Md. 20852); removing the nef gene from the plasmid; inserting the nef gene in the pXBG plasmid for transcription; and purifying the transcription product nef mRNA as described in Examples 2 through 5. The nef mRNA was then incorporated into a formulation according to Example 6. 200 microliter tail vein injections of a 10% sucrose solution containing 200 μg/ml NEF RNA and 500 μg/ml 1:1 DOTAP:DOPE (in RNA/liposome complex form) were performed daily on experimental animals, while control animals were likewise injected with RNA/liposome complexes containing 200 μg/ml yeast tRNA and 500 μg/ml 1:1 DOTAP/DOPE liposomes. At 2, 4 and 8 weeks post injection, biopsy specimens were obtained from injected lymphoid organs and prepared for immunohistochemistry. At the same time points, blood samples were obtained and assayed for p24 levels by means of an ELISA kit (Abbott Labs, Chicago, Ill.) and virus titer by the plaque assay of Example 8. Immunostaining for HIV-1 was performed as described (Namikawa et al., [0190] Science 242:1684 (1988)) using polyclonal serum from a HIV infected patient. Positive cells were counted and the number of infected cells per high power field (400×) were determined. Using these assays, at least a 2 fold reduction in the number of positive staining cells was observed at 8 weeks, and titer and p24 expression was reduced by at least 50%. Together, these results indicate a moderate anti-viral effect of the (in vivo) treatment.
  • A volume of 200 μl of the formulation, containing 200 μg/ml of nef mRNA, and 500 μg/ml 1:1 DOTAP:DOPE in 10% sucrose is injected into the tail vein of the human stem cell-containing [0191] SCID mice 3 times in one day. Following immunization, the mice are challenged by infection with an effective dose of HIV virus. Samples of blood are periodically withdrawn from the tail vein and monitored for production of the characteristic HIV protein p24 by an ELISA kit assay (Abbott Labs, Chicago, Ill.).
  • EXAMPLE 10 A Method of Providing Adenosine Deaminase to Mice by In Vivo mRNA Transfection
  • The full-length sequence for the cDNA of the human adenosine deaminase (ADA) gene is obtained from the 1,300 bp EcoR1-AccI fragment of clone ADA 211 (Adrian, G. et al. [0192] Mol. Cell Biol. 4:1712 (1984). It is blunt-ended, ligated to BgIII linkers and then digested with BgIII. The modified fragment is inserted into the BgIII site of pXBG. ADA mRNA is transcribed and purified according to Examples 2 through 5, and purified ADA mRNA is incorporated into a formulation according to Example 6. Balb 3T3 mice are injected directly in the tail vein with 200 pl of this formulation, containing 200 μg/ml of ADA mRNA, and 500 μg/ml DOTAP in 10% sucrose.
  • The presence of human ADA in the tissues of the liver, skin, and muscle of the mice is confirmed by an isoelectric focusing (IEF) procedure. Tissue extracts were electrofocused between [0193] pH 4 and 5 on a non-denaturing gel. The gel was then stained for in situ ADA activity as reported by Valerio, D. et al. Gene 31:137-143 (1984).
  • A preliminary separation of human and non-human ADA is carried out by fast protein liquid chromatography (FPLC). The proteins are fractionated on a Pharmacia (Piscataway, N.J.) MonoQ column (HR5/5) with a linear gradient from 0.05 to 0.5 M KCl, 20 mM Tris (pH 7.5). Activity for ADA within the fractions is measured by reacting the fractions with [0194] 14C-adenosine (Amersham, Chicago, Ill.) which is converted to inosine. Thin layer chromatography (0.1 M NaPi pH 6.8 saturated ammonium sulfate:n-propylalcohol/100:60:2) is used to separate the radioactive inosine from the substrate adenosine.
  • EXAMPLE 11 In Vivo Expression of Pure RNA and DNA Injected Directly into the Muscles of Mice
  • The quadriceps muscles of mice were injected with either 100 μg of pRSVCAT DNA plasmid or 100 μg of βgCATβgA[0195] n RNA and the muscle tissue at the injection site later tested for CAT activity.
  • Five to six week old female and male Balb/C mice were anesthetized by intraperitoneal injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision was made on the anterior thigh, and the quadriceps muscle was directly visualized. The DNA and RNA were injected in 0.1 ml of solution in a 1 cc syringe through a 27 gauge needle over one minute, approximately 0.5 cm from the distal insertion site of the muscle into the knee and about 0.2 cm deep. A suture was placed over the injection site for future localization, and the skin was then closed with stainless steel clips. [0196]
  • 3T3 mouse fibroblasts were also transfected in vitro with 20 μg of DNA or RNA complexed with 60 μg of Lipofectin™ (BRL) in 3 ml of Opti-Mem™ (Gibco), under optimal conditions described for these cells (Malone, R. et al. [0197] Proc. Nat'l. Acad. Sci. USA 86:6077-6081(1989). The same fibroblasts were also transfected using calcium phosphate according to the procedure described in Ausubel et al.(Eds) Current Protocols in Molecular Biology, John Wiley and Sons, New York (1989).
  • The pRSVCAT DNA plasmid and βgCATβgA[0198] n RNA were prepared as described in the preceding examples. The RNA consisted of the chloramphenicol acetyl transferase (CAT) coding sequences flanked by 5′ and 3′ β-globin untranslated sequences and a 3′ poly-A tract.
  • Muscle extracts were prepared by excising the entire quadriceps, mincing the muscle into a 1.5 ml microtube containing 200 pl of a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl[0199] 2 and 0.1% Triton X), and grinding the muscle with a plastic pestle (Kontes) for one minute. In order to ensure complete disruption of the muscle cells, the muscle tissue was then placed under 600 psi of N2 in a bomb (Parr) at 4° C. for 15 min before releasing the pressure.
  • Fibroblasts were processed similarly after they were trypsinized off the plates, taken up into media with serum, washed 2×with PBS, and the final cell pellet suspended into 200 μl of lysis solution. 75 μl of the muscle and fibroblast extracts were assayed for CAT activity by incubating the reaction mixtures for 2 hours with [0200] 14C-chloramphenicol, followed by extraction and thin-layer chromatography, all as described in Example 7.
  • FIG. 1 comprises autoradiograms from two separate experiments showing CAT activity within extracts of the injected quadriceps muscles. [0201]
  • Lane numbers appear at the top of the autoradiograms and the % chloramphenicol conversions are at the bottom. Sample locations are as follows: [0202]
  • [0203] Lanes 1 and 13: Control fibroblasts
  • [0204] Lanes 2 and 14: Muscle injected only with 5% sucrose
  • [0205] Lanes 3 and 15: 0.005 units of non-injected, purified CAT standard
  • [0206] Lanes 4 and 16: 0.05 units of purified CAT (Sigma)
  • [0207] Lanes 5 to 8: Muscle injected with 100 μg of βgCATβgAn RNA in 5% sucrose
  • [0208] Lanes 11, 12, and 17 to 20: Muscle injected with 100 μgrams pRSVCAT DNA in 5% sucrose
  • [0209] Lanes 9 and 10: 20 μgrams of βgCATβgAn RNA, lipofected, with 60 βgrams of DOTMA, into a 70% confluent 60 mm plate of 3T3 cells (106)
  • [0210] Lanes 21, 22: 20 μgrams of pRSVCAT lipofected, with 60 μg of DOTMA, into a 50% confluent 60 mm plate of 3T3 cells
  • [0211] Lanes 23, 24: 20 μg of pRSVCAT calcium phosphate lipofected into a 50% confluent 60 mm plate of 3T3 cells.
  • CAT activity was readily detected in all four [0212] RNA injection sites 18 hours after injection and in all six DNA injection sites 48 hours after injection. Extracts from two of the four RNA injection sites (FIG. 1, lanes 6 and 8) and from two of the six DNA injection sites (FIG. 1, lanes 11 and 20) contained levels of CAT activity comparable to the levels of CAT activity obtained from fibroblasts transiently transfected in vitro under optimal conditions (FIG. 1, lanes 9, 10, 21-24). The average total amount of CAT activity expressed in muscle was 960 pg for the RNA injections and 116 pg for the DNA injections. The variability in CAT activity recovered from different muscle sites probably represents variability inherent in the injection and extraction technique, since significant variability was observed when pure CAT protein or pRSVCAT-transfected fibroblasts were injected into the muscle sites and immediately excised for measurement of CAT activity. CAT activity was also recovered from abdominal muscle injected with the RNA or DNA CAT vectors, indicating that other muscle groups can take up and express polynucleotides.
  • EXAMPLE 12 Site of In Vivo Expression of Pure DNA Injected Directly into the Muscles of Mice
  • The site of gene expression in injected muscle was determined by utilizing the pRSVLac-Z DNA vector (P. Norton and J. Coffin [0213] Molec. Cell Biol. 5:281-290 (1985)) expressing the E. coli-galactosidase gene for injection and observing the in situ cytochemical staining of muscle cells for E. coli β-galactosidase activity. The quadriceps muscle of mice was exposed as described in the previous example. Quadriceps muscles were injected once with 100 μg of pRSVLAC-Z DNA in 20% sucrose. Seven days later the individual quadriceps muscles were removed in their entirety and every fifth 15 μm cross-section was histochemically stained for β-galactosidase activity.
  • The muscle biopsy was frozen in liquid N[0214] 2-cooled isopentane. 15 μm serial sections were sliced using a cryostat and placed immediately on gelatinized slides. The slide were fixed in 1.5% glutaraldehyde in PBS for 10 minutes and stained 4 hours for β-galactosidase activity (J. Price et al. Proc. Nat'l Acad. Sci. USA 84:156-160 (1987). The muscle was counterstained with eosin.
  • The photographed sections (FIG. 2) are as follows: [0215]
  • (A) and (B): Cross-sections of a muscle injected with pRSVLacZ at 25×and 160× magnification, respectively. [0216]
  • (C): A longitudinal section of another muscle injected with pRSVLacZ, 160×. [0217]
  • (D), (E), and (F): Serial cross-sections of the same muscle that are 0.6 mm apart. [0218]
  • Approximately 60 muscle cells of the approximately 4000 cells (1.5%) that comprise the entire quadriceps and approximately 10-30% of the cells within the injection area were stained blue (FIGS. 2A and 2B). Control muscle injected with only a 20% sucrose solution did not show any background staining. Positive β-galactosidase staining within some individual muscle cells was at least 1.2 mm deep on serial cross-sections (FIGS. 2D, 2E, and [0219] 2F), which may be the result of either transfection into multiple nuclei or the ability of cytoplasmic proteins expressed from one nucleus to be distributed widely within the muscle cell. Longitudinal sectioning also revealed β-galactosidase staining within muscle cells for at least 400 mm (FIG. 2C). In cells adjacent to intensely blue cells, fainter blue staining often appeared in their bordering areas. This most likely represents an artifact of the histochemical β-galactosidase stain in which the reacted X-gal product diffuses before precipitating.
  • Similar results are obtained with linear DNA. [0220]
  • EXAMPLE 13 Dose-Response Effects of RNA and DNA Injected Into Muscles of Mice
  • Experiments with the firefly luciferase reporter gene (LUC) explored the effect of parameters of dose level and time on the total luciferase extracted from injected muscle. [0221]
  • The RNA and DNA vectors were prepared, and the quadriceps muscles of mice injected as previously described. Muscle extracts of the entire quadriceps were prepared as described in Example 11, except that the lysis buffer was 100 mM KPi pH 7.8, 1 mM DTT, and 0.1% Triton X. 87.5 μl of the 200 μl extract was analyzed for luciferase activity (J. de Wet et al. [0222] Molec. Cell Biol. 7:725-737(1987)) using an LKB 125I luminometer. Light units were converted to picograms (pg) of luciferase using a standard curve established by measuring the light units produced by purified firefly luciferase (Analytical Luminescence Laboratory) within control muscle extract. The RNA and DNA preparations prior to injection did not contain any contaminating luciferase activity. Control muscle injected with 20% sucrose had no detectable luciferase activity. All the above experiments were done two to three times and specifically, the DNA time points greater than 40 days were done three times.
  • The FIGS. 3A to [0223] 3C illustrate the results of the following:
  • 3(A) Luciferase activity measured 18 hours following the injection of varying amounts of ′RNA in 20% sucrose and 4 days following the injection of varying amounts of pRSVL in 20% sucrose [0224]
  • 3(B) Luciferase activity assayed at varying times after 20 μg of βgLUCβgA[0225] n RNA were lipofected into a million 3T3 fibroblasts (Malone, R. et al. Proc. Nat'l. Acad. Sci. USA 86:6077-6081 (1989), and after 100 μg of βgLUCβgAn RNA in 20% sucrose were injected into quadriceps.
  • 3(C) Luciferase activity assayed at varying times after pRSVL DNA was injected intramuscularly. [0226]
  • A. Level of Gene Expression [0227]
  • A dose-response effect was observed when quadriceps muscles were injected with various amounts of βgLucβgA[0228] n RNA or DNA pRSVL constructs (FIG. 3A). The injection of ten times more DNA resulted in luciferase activity increasing approximately ten-fold from 33 pg luciferase following the injection of 10 μg of DNA to 320 pg luciferase following the injection of 100 μg of DNA. The injection of ten times more RNA also yielded approximately ten times more luciferase. A million 3T3 mouse fibroblasts in a 60 mm dish were lipofected with 20 μg of DNA or RNA complexed with 60 μg of Lipofectin™ (Bethesda Research Labs) in 3 ml of Opti-MEM™ (Gibco). Two days later, the cells were assayed for luciferase activity and the results from four separate plates were averaged. Twenty μg of pRSVL DNA transfected into fibroblasts yielded a total of 120 pg of luciferase (6 pg luciferase/μg DNA), while 25 μg injected into muscle yielded an average of 116 pg of luciferase (4.6 pg luciferase/μg DNA; FIG. 3A). The expression from the RNA vectors was approximately seven-fold more efficient in transfected fibroblasts than in injected muscles. Twenty μg of βgLucβgAn RNA transfected into fibroblasts yielded a total of 450 pg of luciferase, while 25 μg injected into muscle yielded 74 pg of luciferase (FIGS. 3A and 3B).
  • Based on the amount of DNA delivered, the efficiency of expression from the DNA vectors was similar in both transfected fibroblasts and injected muscles. [0229]
  • B. Time Course of Expression [0230]
  • The time course was also investigated (FIGS. 3B and 3C). Luciferase activity was assayed at varying times after 25 pg of βgLucβgA[0231] n RNA or 100 μg of pRSVL DNA were injected. Following RNA injection, the average luciferase activity reached a maximum of 74 pg at 18 hours, and then quickly decreased to 2 pg at 60 hours. In transfected fibroblasts, the luciferase activity was maximal at 8 hours. Following DNA injection into muscle, substantial amounts of luciferase were present for at least 60 days.
  • The data in FIG. 3B suggest that luciferase protein and the in vitro RNA transcript have a half-life of less than 24 hours in muscle. Therefore, the persistence of luciferase activity for 60 days is not likely to be due to the stability of luciferase protein or the stability of the in vivo RNA transcript. [0232]
  • EXAMPLE 14 Persistence of DNA in Muscle Following Injection as Determined by Southern Blot Analysis
  • Preparations of muscle DNA were obtained from control, uninjected quadriceps or from quadriceps, 30 days after injection with 100 μg of pRSVL in 20% sucrose. Two entire quadriceps muscles from the same animal were pooled, minced into liquid N[0233] 2 and ground with a mortar and pestle. Total cellular DNA and HIRT supernatants were prepared (F. M. Ausubel et al.(Eds) Current Protocols in Molecular Biology, John Wiley, New York (1987). Fifteen μg of the total cellular DNA or 10 μl out of the 100 μl of HIRT supernatant were digested, run on a 1.0% agarose gel, transferred to Nytran™ (Schleicher and Schuell, N.Y.), using a vacublot apparatus (LKB) and hybridized with multiprimed 32P-luciferase probe (the HindIII-BamHI fragment of pRSVL). Following hybridization overnight, the final wash of the membrane was with 0.2×SSC containing 0.5% SDS at 68° C. Kodak XAR5 film was exposed to the membrane for 45 hours at −70° C.
  • FIG. 4 is an autoradiogram of a Southern blot having a sample pattern as follows: [0234]
  • Lane 1: 0.05 ng of undigested pRSVL plasmid [0235]
  • Lane 2: 0.05 ng of BamH1 digested pRSVL [0236]
  • Lane 3: Blank [0237]
  • Lane 4: BamH1 digest of HIRT supernatant from control muscle [0238]
  • Lane 5: BamH1 digest of cellular DNA from control, uninjected muscle [0239]
  • [0240] Lanes 6,7:BamH1 digest of HIRT supernatant from two different pools of pRSVL injected muscles
  • [0241] Lanes 8,9:BamH1 digest of cellular DNA from two different pools of pRSVL injected muscle
  • Lane 10: Cellular DNA (same as Lane 9) digested with BamH1 and Dpn1 [0242]
  • Lane 11: Cellular DNA (Same as in Lane 9) digested with BamH1 and Mbo1 [0243]
  • Lane 12: Cellular DNA digested with BgIII [0244]
  • Lane 13: HIRT supernatant digested with BgIII (Size markers (X/HindIII) are shown at the left). [0245]
  • Southern blot analysis of muscle DNA indicates that the foreign pRSVL DNA is present within the muscle tissue for at least 30 days (FIG. 4, lanes 6-9) and is similar to the levels of DNA present in muscle two and 15 days following injection. In muscle DNA digested with BamH1 (which cuts pRSVL once; FIG. 4, lanes 6-9), the presence of a 5.6 kb band that corresponds to linearized pRSVL (FIG. 4, lane 2) suggest that the DNA is present either in a circular, extrachromosomal form or in large tandem repeats of the plasmid integrated into chromosome. In muscle DNA digested with BgIII (which does not cut pRSVL), the presence of a band smaller than 10 kb (FIG. 4, [0246] lanes 12 and 13) and at the same size as the open, circular form of the plasmid pRSVL (FIG. 4, lane 1) implies that the DNA is present extrachromosomally in an open, circular form. The appearance of the pRSVL DNA in HIRT supernatants (FIG. 4, lanes 6, 7, and 13) and in bacteria rendered ampicillin-resistant following transformation with HIRT supernatants also suggest that the DNA is present unintegrated. Although the majority of the exogenous DNA appears to be extrachromosomal, low levels of chromosomal integration cannot be definitively excluded. Overexposure of the blobs did not reveal smears of hybridizing DNA larger than the 10 kb that would represent plasmid DNA integrated at random sites. The sensitivity of the pRSVL DNA is muscle to DPNI digestion (FIG. 4, lane 10) and its resistance to MboI digestion (FIG. 4, lane 11), suggests that the DNA has not replicated within the muscle cells.
  • EXAMPLE 15 In Vivo Expression of Pure DNA Implanted Directly into the Muscle of Mice
  • pRSVL DNA was precipitated in ethanol and dried. The pellet was picked up with fine forceps and deposited into various muscle groups as described in the preceding examples. Five days later the muscle was analyzed for luciferase activity as described in Example 13. The DNA was efficiently expressed in different muscle groups as follows: [0247]
    Implant: Luciferase Activity (Light Units, LU)
    25 μg pRSVL DNA Control Biceps Calf Quadriceps
    428 46420 27577 159080
    453 53585 34291 35512
    1171 106865
    53397 105176
    499 40481
  • EXAMPLE 16 Direct Gene Delivery into Lung: Intratracheal Injection of DNA, DNA/CL Complexes or Pure Protein
  • The DNA luciferase vector (pRSVL), complexed with Lipofectin™, was injected intratracheally into rats either in 20% sucrose (2 rats) or in 5% sucrose (6 rats). Two days following the injection, the rat lungs were divided into 7 sections: LUL, LLL, RUL, RML, RLL, AL, (defined as follows) and Trachea. The rat lung differs from that of the human in having one large left lung off the left main bronchus. The left lung for this study was cut in half into a left upper part (LUL) and left lower part (LLL). The right lung contains 4 lobes: right cranial lobe (RUL), right middle lobe (RML), right lower lobe (RLL), and an accessory lobe (AL). Extracts were prepared by mincing these lung parts into separate 1.5 ml microtubes containing 200 μl of a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl[0248] 2 and 0.1% Triton X), and grinding the lung with a plastic pestle. (Kontes) for one minute. In order to ensure complete disruption of the lung cells, the lung tissue was then placed under 600 psi of N2 in a Parr bomb at 4° C. for 15 minutes before releasing the pressure. Luciferase assays were done on 87.5 μl of lung extract out of a total volume of about 350 μl.
    Injection RUL RLL LUL LML LLL AL Trachea
    Mock 22.6 22.4 21.9 21.3 20.1 19.8
    25 μg DNA alone 21.2 21.5 21.8 21.6 21.9 21.2
    25 μg DNA alone 21.7 21.4 21.3 22.2 21.5
    250 μg DNA alone 21.7 23.2 21.9 28.5 22.6 22.0 21.3
    250 μg DNA alone 22.9 22.5 33.3 23.0 25.4 24.3 21.5
    250 μg DNA alone 21.8 21.5 21.8 20.4 20.7 20.8 20.7
    25 μg DNA/CL 20.8 22.2 19.6 22.3 22.3 22.0
    25 μg DNA/CL 22.9 22.0 22.7 21.7 22.8 22.18
    25 μg DNA/CL 22.2 23.8 22.1 23.9 22.8 21.6
    25 μg DNA/CL 20.9 20.9 20.9 20.6 20.3 19.3
    25 μg DNA/CL 19.8 20.0 20.3 20.2 20.1 20.3 20.1
    25 μg DNA/CL 20.5 20.5 19.8 19.5 19.9 19.9 19.8
    Luc Protein 105.3 77.1 98.7 80.0 86.3 89.6 178.9
    3 × 104 l.u.
    Blank 22.5
  • Mock: Values are those for an animal that received 25 μg of DNA in 0.3 ml [0249]
  • 20% sucrose into the esophagus. (A sample containing only water yields 22.5 l.u.) [0250]
  • 25 μg DNA alone: represent separate animals that received intratracheal injections of 25 μg of pPGKLuc in 0.3 [0251] ml 20% sucrose.
  • 25 μg DNA/CL: represent separate animals that received intratracheal injections of 25 Mg of pPGKLuc complexed with Lipofectin™ in 0.3 [0252] ml 5% sucrose.
  • The above animals were sacrificed and lung extracts prepared 2 days after injection. [0253]
  • [0254] Luc Protein 104 l.u.: represents an animal that received the equivalent of 30,000 light units (l.u.) of purified firefly luciferase (Sigma), and then was immediately sacrificed.
  • The luciferase activity in the 25 pg DNA alone and the 25 μg DNA/CL groups of animals were not greater than that in the mock animal; however, in the 250 μg DNA alone animals, three lung sections showed small but reliably elevated l.u. activity above control lung or blanks (Bold, underlined). Duplicate assays on the same extract confirmed the result. Experience with the LKB [0255] 125I luminometer indicates that these values, although just above background, indicate real luciferase activity.
  • EXAMPLE 17 Luciferase Activity in Mouse Liver Directly injected with DNA Formulations
  • The DNA luciferase expression vector pPGKLuc was injected intrahepatically (1H) into the lower part of the left liver lobe in mice. The pPGKLuc DNA was either injected by itself (450 Mg DNA in 1.0 [0256] ml 20% sucrose) or complexed with Lipofectin™ (50 μg DNA+150 μg Lipofectin™ in 1.0 ml 5% sucrose). Three days following injection, the left liver lobe was divided into two sections (a lower part where the lobe was injected and an upper part of the lobe distant from the injection site) and assayed for luciferase activity as described in the preceding examples.
    Mice Intrahepatic Luciferase Activity (Light Units, LU)
    Liver Injection Lower Upper
    Blank (20.2 LU)
    Control: 20% Sucrose Only 20.8 23.8
     50 μg pPGRLuc + Lipofectin 35.4 23.1
     50 μg pPGKLuc + Lipofectin 38.1 21.4
     50 μg pPGKLuc + Lipofectin 22.1 22.7
    450 μg pPGKLuc 43.7 29.2
    450 μg pPGKLuc 78.8 21.7
    450 μg pPGKLuc 21.7 20.8
  • Two of the three animals-that received the pure pPGKLuc injections and two of the three animals that received pPGKLuc +Lipofectin™ injections had luciferase activity significantly above background (bold, underlined). The lower part of the liver lobe, which was directly injected, had larger amounts of luciferase activity than the upper part, which was distant from the injection site. Similar results have been obtained using pRSVCAT DNA expression vector and CAT assays. Luciferase activity was not detected three days after similar preparations of pPGKLuc (+ and − Lipofectin™) were injected into the portal circulation of rats. [0257]
  • EXAMPLE 18 Expression of Growth Hormone Gene Injected into Liver and Muscle
  • Mice were injected with the pXGH5 (metallothionein promoter-growth hormone fusion gene)(Selden Richard et al., [0258] Molec. Cell Biol. 6:3173-3179 (1986)) in both liver and muscle. The mice were placed on 76 mM zinc sulfate water. Later the animals were bled and the serum analyzed for growth hormone using the Nichols GH Kit.
  • A. Two mice were injected with 20 μg of pXGH5 gene complexed with 60 μg/ml of Lipofectin in 5% sucrose. One ml of this solution was injected into the liver and the ventral and dorsal abdominal muscles were injected with 0.1 ml in 7 sites two times. Two days later, the animals were bled. The serum of one animal remained at background level, while that of the other contained 0.75 ng/ml growth hormone. [0259]
  • B. Three mice were injected with 0.1 ml of 1 mg/ml of pXGH5 in 5% sucrose, 2×in the quadriceps, 1×in the hamstring muscle, 1×in pectoralis muscle, and 1×in trapezoid muscles on two separate days. The results were as follows: [0260]
    Animal No. Growth Hormone(ng/ml): Day 1 Day 2
    1 0.6 0.6
    2 0.8 1.0
    3 0.95 0.8
  • EXAMPLE 19 Antibody Production in Mice Directly Injected with a Gene for an Immunizing Peptide
  • Mice were injected with a quantity of 20 μg of a plasmid construct consisting of the gp-120 gene, driven by a cytomegalovirus (CMV) promotor. The DNA was injected into the quadriceps muscle of mice according to the methods described in Example 11. Mouse 5 (FIG. 5A) was injected in the quadriceps muscle with 20 μg of plasmid DNA in isotonic sucrose. Mouse 2 (FIG. 5B) was injected with sucrose solution alone. Blood samples were obtained prior to the injection (Day 0) at the times indicated on FIG. 5, up to more than 40 days post injection. The serum from each sample was serially diluted and assayed in a standard ELISA technique assay for the detection of antibody, using recombinant gp-120 protein made in yeast as the antigen. Both IgG and IgM antibodies were detected. The study indicates that the gene reyains its signal sequence, and the protein is efficiently excreted from cells. [0261]
  • EXAMPLE 20 Antibody Production in Mice Injected with Cells Transfected with a Gene for an Immunizing Peptide
  • The cell line BALB/C C1.7 (TIB 80) was obtained from the American Type Tissue Culture Collection. These cells were transfected with the gp-120 gene construct described in Example 19. To 0.75 ml OptiMEM™ (Gibco. Inc.) were added 6.1 μg of DNA. The quantity of 30 μg of cationic liposomes (containing DOTMA and cholesterol in a 70:30 molar ratio) were added to another 0.75 ml OptiMEM™. The mixtures were combined and 1.5 ml of OptiMEM™ containing 20% (v/v) fetal bovine calf serum was added. This solution was poured into a 60 mm plastic petri dish containing 80% confluent cells (approximately one million total cells per plate). At 3.2 hours after lipofection, the cells were detached from the plate with trypsin and EDTA treatment, washed with OptiMEM™ and resuspended in 0.1 ml OptiMEM™ with 10% fetal calf serum. These cells were injected (IP) into mice. Mouse 12 (FIG. 6A) was injected with the transfected cells. Mouse 11 (FIG. 6B) received an identical number of untransfected cells. Blood samples were obtained prior to the injection (Day 0) and at the times indicated in FIGS. 6A and 6B. The serum samples were processed as in the preceding example. Both IgG and IgM antibodies were detected as indicated in FIGS. 6A and B. [0262]
  • EXAMPLE 21 Use of Uncapped 5′ Sequences to Direct Translation of DNA Transfected into Cells In Vitro
  • Two different DNA templates were constructed, both of which code for the synthesis of RNA that express the [0263] E. coli. β-galactosidase reporter gene. A Lac-Z gene that contains the Kozak consensus sequence was inserted in place of the luciferase coding sequences of the pβGLucβGAn template to generate the pβGLacZβGAn template. The pEMCLacZβGAn template was made by replacing the 5′ β-globin untranslated sequences of pβGLacZβGAn with the 588 bp EcoRl/Ncol fragment from the encephalomyocarditis virus (EMCV) (pE5LVPO in Parks, G. et al., J. Virology 60:376-384 (1986). These EMC 5′ untranslated sequences had previously been shown to be Able to initiate efficient translation in vitro in reticulocytes lysates. We demonstrated that these sequences can also direct efficient translation when transfected into fibroblasts in culture. The percentage of blue cells was slightly greater in cells transfected with the uncapped EMCLacZβGAn RNA than in cells transfected with the capped pEMCLacZβGAn RNA. Transfection with either uncapped or capped pEMCLacZβGAn RNA yielded a greater number of positive β-galactosidase cells than transfection with capped βGLacZβDGAn RNA. It has recently been shown that this EMC 5′ untranslated sequence, as a component of vaccinia-T7 polymerase vectors, can increase translation of an uncapped mRNA 4 to 7-fold (Elroy-Stein, 0. et al., Proc. Natl. Acad. Sci. USA 86:6126-6130 (1989). These EMC sequences thus have the ability to direct efficient translation from uncapped messengers.
  • EXAMPLE 22 T7 Polymerase Transcription in Transfected Cell Cultures
  • An SV40-T7 polymerase plasmid containing T7 polymerase protein expressed off the SV40 promotor (Dunn, J. et al., [0264] Gene 68: 259 (1988)) was co-lipofected with the pEMCLacZβGAn template DNA into 3T3 fibroblasts in culture to demonstrate that T7 polymerase transcription can occur via plasmids. Two different SV40-T7 polymerase expression vectors were used:
  • (a) pSV-G1-A: pAR3126-SV40 promotor driving expression of T7 polymerase protein which is directed to the cytoplasm. [0265]
  • (b) pSVNU-G1-A: pAR3132-SV40 promotor driving expression of T7 polymerase protein which is directed to the cytoplasm. [0266]
  • Each of these two plasmids were co-lipofected with pEMCLacZβGAn at 1:3 and 3:1 ratios into 60 mm plates of 3T3 cells. The number of blue β-galactosidase cells were counted and scored as indicated below. [0267]
    β-gal Ratio:template/ Co-Lipofectant:
    template polymerase vector pSV-G1-A pSVNU-G1-A
    βGLacZβ3GAn 3:1 0 1
    1:3 0 1
    EMCLacZβ3GAn 3:1 74 70
    1:3 45 15
  • EXAMPLE 23 Expression of Luciferase in Brain Following Directed Injection of Messenger RNA
  • Two adult mice and one newborn mouse were injected with the βgLucβgA[0268] n mRNA containing the 5′ cap and prepared according to Example 13. In the adult mice, injections were from a stock solution of mRNA at 3.6 μg/μl in 20% sucrose; injection volumes were 5 μ1,2 injections into each of the bilateral parietal cortex, 4 injections per mouse. Tissue was assayed at 18 hours post injection, according to Example 13 using 200 μl of brain homogenate, disrupted in a Parr bomb, and 87.5 μl was taken for assay.
  • The results are as follows: [0269]
    Hemisphere:
    Treatment Animal I.D. Left Right
    Sham Injection AMra 649 629
    βgLucβgAn AMrb 1,734 1,911
  • The newborn mouse was injected with 1 μl βgLucβgAn [0270]
  • (3.6 μg/μl; 20% sucrose) into the bilateral forebrain and tissues were similarly processed and analyzed. [0271]
    Hemisphere:
    Treatment Animal I.D. Left Right
    βgLucβgAn NRr 1,569 963
  • EXAMPLE 24 Functional Expression of Dystrophin in Dystrophic Mouse Muscle In Vivo
  • A plasmid containing the dystrophin gene under control of the Rous Sarcoma virus promoter was prepared from the Xp21 plasmid containing the complete dystrophin coding region and the SV40 poly. A segment, which was cloned by Kunkel and colleagues. (Brumeister M., Monaco A P, Gillard E F, van Ommen G J, Affara N A, Ferguson-Smith M A, Kunkel L M, Lehrach H. A 10-megabase physical map of human Xp21, including the Duchenne muscular dystrophy gene. [0272] Genomics 1988 Apr 2 (3):189-202; Hoffman, E P and Kunkel, L M Dystrophin abnormalities of Duchenne's/Becher Muscular Dystrophy. Neuron Vol. 2, 1019-1029 (1989); Koenig M., Monaco A P, Kunkel L M. The complete sequence of dystrophin predicts a rod-shaped cyto-skeletal protein. Cell 1988 Apr 22, 53 (2):219-26) 200 μg of the plasmid in 100 ul of phosphate buffered saline was injected into the quadriceps the mutant mouse strain lacking the dystrophin gene product (MDX mouse; Jackson labs). Expression of functional dystrophin was monitored 7 days post injection by immuno-histochemistry according to the procedures described by Watkins et al. and using the same anti-dystrophin antibody (anti-60 kd antibody with a fluorescent secondary antibody) obtained from Kunkel. Functional expression of the dystrophin gene product in the dystrophic mice was detected by comparing the pattern of fluorescence observed in cross-sections of quadriceps muscle from injected animals, with the fluorescence pattern observed in normal animals. (Watkins S. C., Hoffman E. P., Slayter H. S., Kinkel L. M., Immunoelectron microscopic localization of dystrophin in myofibres. Nature 1988, June 30; 333 (6176:863-6). Normal dystrophin expression is localized underneath the plasma membrane of the muscle fiber, so that a cross section of the quadriceps muscle give a fluorescence pattern encircling the cell. In addition dystrophin expression was quantitated by Western blot analysis using the affinity purified anti-60 kd antibody.
  • EXAMPLE 25 Administration of the Correcting Dystrophin Gene Directly into the Muscle of Patients with Duchenne's Muscular Dystrophy
  • Patients with muscular dystrophy are given multiple 200 μg injections of plasmid containing the functional dystrophin gene (see previous example) in 100 μl of phosphate buffered saline. While under light anesthesia the patients are injected at 5 cm intervals into the entire skeletal muscle mass directly through the skin without surgery. Patient recovery evaluated by monitoring twitch tension and maximum voluntary contraction. In addition, biopsies of 300-500 muscle cells from an injected area are taken for histological examination, observing muscle structure and biochemical analysis of the presence of dystrophin, which is absent in patients with Duchenne's muscular dystrophy. Respiratory muscles, including the intercostal muscles which move the rib cage and the diaphragm, are particularly important impaired muscle groups in patients with muscular dystrophy. The intercostals can be reached by injection through the skin as can the other skeletal muscle groups. The diaphragm can be accessed by a surgical procedure to expose the muscle to direct injection of plasmid DNA. [0273]
  • There will be various modifications, improvements, and applications of the disclosed invention that will be apparent to those of skill in the art, and the present application is intended to cover such embodiments. Although the present invention has been described in the context of certain preferred embodiments, it is intended that the full scope of these be measured by reference to the scope of the following claims. [0274]

Claims (65)

What is claimed is:
1. A pharmaceutical product, comprising:
naked polynucleotide, operatively coding for a biologically active polypeptide, in physiologically acceptable administrable form, in a container; and
a notice associated with said container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by said agency of said form of said polynucleotide for human or veterinary administration.
2. A pharmaceutical product, comprising:
naked polynucleotide, operatively coding for a biologically active peptide, in solution in a physiologically acceptable injectable carrier and suitable for introduction interstitially into a tissue to cause cells of said tissue to express said polypeptide;
a container enclosing said solution; and
a notice associated with said container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by said agency of manufacture, use, or sale of said solution of polynucleotide for human or veterinary administration.
3. The product of claim 2, wherein said peptide is immunogenic and administration of said solution to a human serves to vaccinate said human.
4. The product of claim 2, wherein said peptide is therapeutic and administration of said solution to a human in need of therapy relating to said polypeptide has a therapeutic effect.
5. A pharmaceutical product for treatment of muscular dystrophy, comprising:
a sterile, pharmaceutically acceptable carrier;
a pharmaceutically effective amount of a naked polynucleotide operatively coding for dystrophin solubilized in said carrier; and
a container enclosing said carrier and said polynucleotide in sterile fashion.
6. The product of claim 5, wherein said polynucleotide is DNA.
7. A pharmaceutical product for use in supplying a biologically active polypeptide to a vertebrate, comprising:
a pharmaceutically effective amount of a naked polynucleotide operatively coding for said polypeptide;
a container enclosing said polynucleotide in a sterile fashion; and
means associated with said container for permitting transfer of said polynucleotide from said container to the interstitial space of a tissue, whereby cells of said tissue can take up and express said polynucleotide.
8. The product of claim 7, wherein said container is a syringe.
9. The product of claim 7, wherein the amount of said polynucleotide in said container is at least 5 micrograms.
10. A pharmaceutical product for use in immunizing a vertebrate, comprising:
a pharmaceutically effective amount of a naked polynucleotide operatively coding for an immunogenic polypeptide;
a sealed container enclosing said polynucleotide in a sterile fashion; and
means associated with said container for permitting transfer of said polynucleotide from said container to the interstitial space of a tissue, whereby cells of said tissue can take up and express said polynucleotide.
11. The product of claim 10, wherein said container is a syringe.
12. The method of claim 10, wherein the amount of said polynucleotide in said container is at least 5 micrograms.
13. A method for delivering a pharmaceutical or immunogenic polypeptide to the interior of a cell of a vertebrate in vivo, comprising the step of:
introducing a preparation comprising a pharmaceutically acceptable injectable carrier and a naked polynucleotide operatively coding for said polypeptide into the interstitial space of a tissue comprising said cell, whereby said naked polynucleotide is taken up into the interior of said cell and has an immunogenic or pharmacological effect on said vertebrate.
14. The method of claim 13, wherein said polypeptide is immunogenic and said vertebrate develops an immune response to said polypeptide.
15. The method of claim 13, wherein said polypeptide is therapeutic.
16. The method of claim 13, wherein said polynucleotide is mRNA.
17. The method of claim 13, wherein said polynucleotide is DNA.
18. The method of claim 13, wherein said polynucleotide is a DNA sequence incorporated into a plasmid vector and said plasmid vector further comprises a replicator.
19. The method of claim 13, wherein said DNA sequence contains a promoter sequence.
20. The method of claim 19, wherein said promoter is a cell-specific promoter that permits substantial transcription of DNA only in predetermined cells.
21. The method of claim 13, wherein said polynucleotide sequence contains a sequence operatively coding for the secretion of said polypeptide.
22. The method of claim 13, wherein said polypeptide expression is transitory.
23. The method of claim 13, wherein said polypeptide is an enzyme.
24. The method of claim 13, wherein said polypeptide is an hormone.
25. The method of claim 13, wherein said polypeptide is a lymphokine.
26. The method of claim 13, wherein said polypeptide is a cell surface receptor.
27. The method of claim 13, wherein said polypeptide is a growth factor.
28. The method of claim 13, wherein said polypeptide is a regulatory protein.
29. The method of claim 13, wherein said polynucleotide is incorporated into muscle cells.
30. The method of claim 13, wherein said preparation is injected intramuscularly.
31. The method of claim 13, wherein said polynucleotide is incorporated into cells of skin, brain, lung, liver, spleen or blood.
32. The method of claim 13, wherein said preparation is injected intradermally, subdermally, intrathecally, or intravenously.
33. The method of claim 13, wherein said preparation is impressed into the skin.
34. The method of claim 13, wherein said preparation is delivered transdermally.
35. The method of claim 13, wherein said polynucleotide is a non-replicating DNA sequence operatively coding for said polypeptide and for a polymerase for transcribing said DNA, and wherein said DNA includes recognition sites for said polymerase, and said injectable preparation further includes a means for providing an initial quantity of said polymerase in said cell.
36. The method of claim 35, wherein said polymerase is phage T7 polymerase and said recognition site is a T7 origin of replication sequence.
37. A method for treating a disease associated with the deficiency or absence of a specific polypeptide in a vertebrate, comprising the step of:
introducing an injectable preparation comprising a pharmaceutically acceptable carrier and containing a naked polynucleotide operatively coding for said polypeptide into a vertebrate and permitting said polynucleotide to be incorporated into a cell, wherein said polypeptide is formed as the translation product of said polynucleotide and said deficiency or absence of said polypeptide is effectively treated.
38. The method of claim 37, wherein said preparation is injected into muscle.
39. The method of claim 37, wherein said cell is a persistent non-dividing cell.
40. A therapy for phenylketonuria according to the method set forth in claim 37, wherein said polynucleotide codes for the polypeptide phenylalanine hydroxylase.
41. The method of claim 37, wherein said polynucleotide codes for human growth hormone.
42. A therapy for hypercholesterolemia according to the method set forth in claim 37, wherein a polynucleotide operatively coding for a receptor involved in cholesterol homeostasis is incorporated into a hepatocyte, whereby said receptor is expressed by said cell.
43. A method for immunizing a vertebrate, comprising the step of:
introducing an injectable preparation comprising a pharmaceutically acceptable carrier and a naked, expressible polynucleotide operatively coding for an immunogenic peptide interstitially into tissue of a vertebrate whereby an immunogenic translation product of said polynucleotide is formed by a cell of said tissue, thereby eliciting an immune response against said immunogen.
44. The method of claim 43, wherein said immunogenic translation product is presented by said cell in the context of the major histocompatibility complex.
45. The method of claim 43, wherein said cells are muscle cells.
46. The method of claim 43, wherein said injectable preparation further comprises an adjuvant or a lymphokine.
47. The method of claim 43, wherein said immunogenic translation product is presented by a cell and provokes a humoral immune response, comprising the synthesis of antibody.
48. The method of claim 43, wherein said cell expresses major histocompatibility antigens of Class I, and said immunogenic peptide is presented in the context of Class I histocompatibility antigens and wherein said immune response is cellular and comprises the production of cytotoxic T-cells.
49. A method for introducing a polynucleotide into muscle cells in vivo, comprising the steps of:
providing a composition comprising a naked polynucleotide in a pharmaceutically acceptable carrier; and
contacting said composition with muscle tissue of a vertebrate in vivo, whereby said polynucleotide is introduced into muscle cells of said tissue.
50. The method of claim 49, wherein said polynucleotide is an antisense polynucleotide.
51. The method of claim 49, wherein said polynucleotide codes for a therapeutic peptide that is expressed by said muscle cells after said contacting step to provide therapy to said vertebrate.
52. The method of claim 49, wherein said polynucleotide codes for an immunogenic peptide that is expressed by said muscle cells after said contacting step and which generates an immune response, thereby immunizing said vertebrate.
53. A method for obtaining long term administration of a polypeptide to a vertebrate, comprising the step of introducing a naked DNA sequence operatively coding for said polypeptide interstitially into tissue of said vertebrate, whereby cells of said tissue produce said polypeptide for at least 3 months.
54. The method of claim 53, wherein said cells producing said polypeptide are nonproliferating cells.
55. The method of claim 54, wherein said cells are muscle cells.
56. A method for obtaining transitory expression of a polypeptide in a vertebrate, comprising the step of introducing a naked mRNA sequence operatively coding for said polypeptide interstitially into tissue of said vertebrate, whereby cells of said tissue produce said polypeptide for less than about 10 days.
57. The method of claim 56, wherein said tissue is solid tissue.
58. A method for treatment of muscular dystrophy, comprising the steps of:
introducing a therapeutic amount of a composition comprising a polynucleotide operatively coding for dystrophin in a pharmaceutically acceptable carrier in vivo into muscle tissue of an animal suffering from muscular dystrophy, whereby said polynucleotide is taken up into cells of said tissue and dystrophin is produced in vivo.
59. The method of claim 58, wherein said composition is introduced by means of injection.
60. The method of claim 58, wherein said polynucleotide is a naked polynucleotide and said composition is introduced interstitially into said muscle tissue.
61. A pharmaceutical product, comprising:
naked antisense polynucleotide in physiologically acceptable administrable form, in a container; and
a notice associated with said container in form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by said agency of said form of said polynucleotide for human or veterinary administration.
62. Use of naked polynucleotide operatively coding for a physiologically active polypeptide in the preparation of a pharmaceutical for introduction interstitially into tissue to cause cells comprising said tissue to produce said polypeptide.
63. Use according to claim 62, wherein said pharmaceutical is for introduction into muscle tissue whereby muscle cells produce said polypeptide.
64. Use according to claim 62, wherein said peptide is dystrophin and said pharmaceutical is for treatment of muscular dystrophy.
65. Use of naked antisense polynucleotide in the preparation of a pharmaceutical for introduction interstitially into tissue of a vertebrate to inhibit translation of polynucleotide in cells of said vertebrate.
US10/387,525 1989-03-21 2003-03-14 Expression of exogenous polynucleotide sequences in a vertebrate Abandoned US20040023911A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/387,525 US20040023911A1 (en) 1989-03-21 2003-03-14 Expression of exogenous polynucleotide sequences in a vertebrate

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US32630589A 1989-03-21 1989-03-21
US46788190A 1990-01-19 1990-01-19
US49699190A 1990-03-21 1990-03-21
US08/481,919 US6214804B1 (en) 1989-03-21 1995-06-07 Induction of a protective immune response in a mammal by injecting a DNA sequence
US09/452,872 US6710035B2 (en) 1989-03-21 1999-12-02 Generation of an immune response to a pathogen
US10/387,525 US20040023911A1 (en) 1989-03-21 2003-03-14 Expression of exogenous polynucleotide sequences in a vertebrate

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/452,872 Continuation US6710035B2 (en) 1989-03-21 1999-12-02 Generation of an immune response to a pathogen

Publications (1)

Publication Number Publication Date
US20040023911A1 true US20040023911A1 (en) 2004-02-05

Family

ID=27406452

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/187,630 Expired - Lifetime US5703055A (en) 1989-03-21 1994-01-26 Generation of antibodies through lipid mediated DNA delivery
US08/215,405 Expired - Lifetime US5580859A (en) 1989-03-21 1994-03-18 Delivery of exogenous DNA sequences in a mammal
US08/380,131 Expired - Lifetime US5589466A (en) 1989-03-21 1995-01-26 Induction of a protective immune response in a mammal by injecting a DNA sequence
US09/452,872 Expired - Fee Related US6710035B2 (en) 1989-03-21 1999-12-02 Generation of an immune response to a pathogen
US10/387,525 Abandoned US20040023911A1 (en) 1989-03-21 2003-03-14 Expression of exogenous polynucleotide sequences in a vertebrate
US10/732,416 Abandoned US20040132683A1 (en) 1989-03-21 2003-12-11 Expression of exogenous polynucleotide sequences in a vertebrate

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US08/187,630 Expired - Lifetime US5703055A (en) 1989-03-21 1994-01-26 Generation of antibodies through lipid mediated DNA delivery
US08/215,405 Expired - Lifetime US5580859A (en) 1989-03-21 1994-03-18 Delivery of exogenous DNA sequences in a mammal
US08/380,131 Expired - Lifetime US5589466A (en) 1989-03-21 1995-01-26 Induction of a protective immune response in a mammal by injecting a DNA sequence
US09/452,872 Expired - Fee Related US6710035B2 (en) 1989-03-21 1999-12-02 Generation of an immune response to a pathogen

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/732,416 Abandoned US20040132683A1 (en) 1989-03-21 2003-12-11 Expression of exogenous polynucleotide sequences in a vertebrate

Country Status (1)

Country Link
US (6) US5703055A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060024670A1 (en) * 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
US20070105193A1 (en) * 2003-05-16 2007-05-10 Vical Incorporated Severe acute respiratory syndrome DNA vaccine compositions and methods of use
US20070105799A1 (en) * 2002-09-10 2007-05-10 Vical Incorporated Codon-optimized polynucleotide-based vaccines against Bacillus anthracis infection
US7268120B1 (en) 1997-11-20 2007-09-11 Vical Incorporated Methods for treating cancer using cytokine-expressing polynucleotides

Families Citing this family (2038)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5833975A (en) * 1989-03-08 1998-11-10 Virogenetics Corporation Canarypox virus expressing cytokine and/or tumor-associated antigen DNA sequence
US7767449B1 (en) 1981-12-24 2010-08-03 Health Research Incorporated Methods using modified vaccinia virus
US6673776B1 (en) * 1989-03-21 2004-01-06 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US6867195B1 (en) * 1989-03-21 2005-03-15 Vical Incorporated Lipid-mediated polynucleotide administration to reduce likelihood of subject's becoming infected
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5698531A (en) 1989-03-31 1997-12-16 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US7238673B2 (en) * 1989-03-31 2007-07-03 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
DE69031951T2 (en) * 1989-11-16 1998-08-13 Du Pont Transformation of animal skin cells with the help of particles
US6228844B1 (en) 1991-11-12 2001-05-08 Vical Incorporated Stimulating vascular growth by administration of DNA sequences encoding VEGF
US6706694B1 (en) 1990-03-21 2004-03-16 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate
US20030186913A1 (en) * 1990-03-21 2003-10-02 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate
US6605712B1 (en) * 1990-12-20 2003-08-12 Arch Development Corporation Gene transcription and ionizing radiation: methods and compositions
US6156304A (en) * 1990-12-20 2000-12-05 University Of Pittsburgh Of The Commonwealth System Of Higher Education Gene transfer for studying and treating a connective tissue of a mammalian host
EP0659209A1 (en) * 1991-07-26 1995-06-28 The University Of Rochester Cancer therapy utilizing malignant cells
US5661133B1 (en) * 1991-11-12 1999-06-01 Univ Michigan Collateral blood vessel formation in cardiac muscle by injecting a dna sequence encoding an angiogenic protein
US5643578A (en) * 1992-03-23 1997-07-01 University Of Massachusetts Medical Center Immunization by inoculation of DNA transcription unit
US7323297B1 (en) * 1992-04-03 2008-01-29 The Regents Of The University Of California Stabilized polynucleotide complexes and methods
US5792751A (en) * 1992-04-13 1998-08-11 Baylor College Of Medicine Tranformation of cells associated with fluid spaces
US7105574B1 (en) 1999-03-26 2006-09-12 Vical Incorporated Adjuvant compositions and methods for enhancing immune responses to polynucleotide-based vaccines
US20090191227A1 (en) 2007-05-23 2009-07-30 Vical Incorporated Compositions and Methods for Enhancing Immune Responses to Vaccines
CA2142007C (en) * 1992-08-11 2007-10-30 Robert Glen Urban Immunomodulatory peptides
US6936464B1 (en) 1992-10-02 2005-08-30 Cancer Research Technology Limited Immune responses to fusion proteins
GB9223816D0 (en) 1992-11-13 1993-01-06 Medical Res Council Heat shock proteins and the treatment of tumours
GB9224584D0 (en) 1992-11-23 1993-01-13 Connaught Lab Use of outer membrane protein d15 and its peptides as vaccine against haempohilus influenzae diseases
US20040087521A1 (en) * 1993-03-18 2004-05-06 Merck & Co., Inc. Nucleic acid pharmaceuticals-influenza matrix
US5591601A (en) * 1993-05-14 1997-01-07 Ohio University Edison Animal Biotechnology Institute DNA polymerase gene expression system utilizing an RNA polymerase co-delivered with the gene expression vector system
EP1624068A1 (en) 1993-06-01 2006-02-08 Life Technologies Inc. Genetic immunization with cationic lipids
EP0702722B1 (en) * 1993-06-07 2005-08-03 Vical Incorporated Plasmids suitable for gene therapy
US5849719A (en) 1993-08-26 1998-12-15 The Regents Of The University Of California Method for treating allergic lung disease
US5985847A (en) * 1993-08-26 1999-11-16 The Regents Of The University Of California Devices for administration of naked polynucleotides which encode biologically active peptides
US5679647A (en) * 1993-08-26 1997-10-21 The Regents Of The University Of California Methods and devices for immunizing a host against tumor-associated antigens through administration of naked polynucleotides which encode tumor-associated antigenic peptides
US5804566A (en) * 1993-08-26 1998-09-08 The Regents Of The University Of California Methods and devices for immunizing a host through administration of naked polynucleotides with encode allergenic peptides
US5830877A (en) * 1993-08-26 1998-11-03 The Regents Of The University Of California Method, compositions and devices for administration of naked polynucleotides which encode antigens and immunostimulatory
US6348449B1 (en) * 1993-09-21 2002-02-19 The Trustees Of The University Of Pennsylvania Methods of inducing mucosal immunity
US5994318A (en) * 1993-10-04 1999-11-30 Albany Medical College Cochleate delivery vehicles
US5674908A (en) 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US6989434B1 (en) * 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
US5942496A (en) 1994-02-18 1999-08-24 The Regent Of The University Of Michigan Methods and compositions for multiple gene transfer into bone cells
US6551618B2 (en) 1994-03-15 2003-04-22 University Of Birmingham Compositions and methods for delivery of agents for neuronal regeneration and survival
US6727230B1 (en) 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US5739118A (en) 1994-04-01 1998-04-14 Apollon, Inc. Compositions and methods for delivery of genetic material
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
NZ290089A (en) * 1994-07-27 1999-05-28 Queensland Inst Med Res Recombinant polyepitope cytotoxic t lymphocyte (ctl) vaccines
US6290991B1 (en) 1994-12-02 2001-09-18 Quandrant Holdings Cambridge Limited Solid dose delivery vehicle and methods of making same
US6235888B1 (en) 1994-10-05 2001-05-22 The General Hospital Corporation Hepatitis C virus vaccine
US7597886B2 (en) * 1994-11-07 2009-10-06 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US7820798B2 (en) * 1994-11-07 2010-10-26 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US6359054B1 (en) 1994-11-18 2002-03-19 Supratek Pharma Inc. Polynucleotide compositions for intramuscular administration
US5641665A (en) 1994-11-28 1997-06-24 Vical Incorporated Plasmids suitable for IL-2 expression
US6383814B1 (en) 1994-12-09 2002-05-07 Genzyme Corporation Cationic amphiphiles for intracellular delivery of therapeutic molecules
FR2727867B1 (en) 1994-12-13 1997-01-31 Rhone Poulenc Rorer Sa GENE TRANSFER IN MEDULLAR MOTONURONES USING ADENOVIRAL VECTORS
US5795587A (en) 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5885971A (en) * 1995-03-24 1999-03-23 The Regents Of The University Of California Gene therapy by secretory gland expression
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression
US6531455B1 (en) * 1995-03-24 2003-03-11 The Regents Of The University Of California Delivery of polynucleotides by secretory gland expression
US5703057A (en) * 1995-04-07 1997-12-30 Board Of Regents The University Of Texas System Expression library immunization
US5935568A (en) * 1995-05-18 1999-08-10 National Jewish Medical & Research Center Gene therapy for effector cell regulation
RU2189254C2 (en) * 1995-06-06 2002-09-20 Америкэн Хоум Продактс Корпорэйшн Vaccines against hepatitis viruses
US7223739B1 (en) 1995-06-07 2007-05-29 Powderject Vaccines, Inc. Adjuvanted genetic vaccines
US20030069173A1 (en) 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US6019980A (en) * 1995-06-07 2000-02-01 Connaught Laboratories Limited Nucleic acid respiratory syncytial virus vaccines
US6017897A (en) * 1995-06-07 2000-01-25 Pasteur Merieux Connaught Canada Nucleic acid respiratory syncytial virus vaccines
US5981501A (en) * 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US7422902B1 (en) 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
AU6268696A (en) * 1995-06-07 1996-12-30 University Of North Carolina At Chapel Hill, The Aav transduction of myoblasts
US6083925A (en) * 1995-06-07 2000-07-04 Connaught Laboratories Limited Nucleic acid respiratory syncytial virus vaccines
AU723163B2 (en) * 1995-06-07 2000-08-17 Tekmira Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5766903A (en) * 1995-08-23 1998-06-16 University Technology Corporation Circular RNA and uses thereof
US7803782B2 (en) * 2003-05-28 2010-09-28 Roche Madison Inc. Intravenous delivery of polynucleotides to cells in mammalian limb
US6121246A (en) * 1995-10-20 2000-09-19 St. Elizabeth's Medical Center Of Boston, Inc. Method for treating ischemic tissue
CA2329474C (en) 1995-11-02 2002-02-26 Schering Corporation Continuous low-dose cytokine infusion therapy
JP2000500744A (en) 1995-11-09 2000-01-25 マイクロバイオロジカル リサーチ オーソリティー Microencapsulated DNA for vaccination and gene therapy
US6270795B1 (en) 1995-11-09 2001-08-07 Microbiological Research Authority Method of making microencapsulated DNA for vaccination and gene therapy
US6310046B1 (en) * 1995-11-17 2001-10-30 The United States Of America As Represented By The Secretary Of The Army Sequestrin of Plasmodium falciparum
US6537776B1 (en) 1999-06-14 2003-03-25 Diversa Corporation Synthetic ligation reassembly in directed evolution
US6747133B1 (en) 1995-12-08 2004-06-08 University Technologies International Inc. Antibodies against the tumor suppressor gene ING1
US6117633A (en) 1995-12-08 2000-09-12 University Technologies International Inc. DNA sequence encoding the tumor suppressor gene ING1
US6143522A (en) * 1995-12-08 2000-11-07 University Technologies International, Inc. Methods of modulating apoptosis
US20040259828A1 (en) * 1995-12-13 2004-12-23 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
US7507722B1 (en) 1999-11-05 2009-03-24 Roche Madison Inc. Intravascular delivery of nucleic acid
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
US5789244A (en) * 1996-01-08 1998-08-04 Canji, Inc. Compositions and methods for the treatment of cancer using recombinant viral vector delivery systems
US20040014709A1 (en) * 1996-01-08 2004-01-22 Canji, Inc. Methods and compositions for interferon therapy
US6077834A (en) * 1996-02-09 2000-06-20 Cheng; Pi-Wan Receptor ligand-facilitated delivery of biologically active molecules
EP0888086B1 (en) 1996-02-15 2005-07-27 Biosense Webster, Inc. Excavation probe
US6245747B1 (en) 1996-03-12 2001-06-12 The Board Of Regents Of The University Of Nebraska Targeted site specific antisense oligodeoxynucleotide delivery method
US6096548A (en) 1996-03-25 2000-08-01 Maxygen, Inc. Method for directing evolution of a virus
TW480282B (en) * 1996-04-24 2002-03-21 Ren Wen Corp Recombinant eucaryotic vector containing allergen gene and use of preventing and treating allergic disease thereof
DE19903693A1 (en) * 1998-04-24 1999-10-28 Centeon Pharma Gmbh Protease for activation of coagulation factor VII
WO1997042820A1 (en) 1996-05-16 1997-11-20 Duke University Tristetraprolin
AU3210997A (en) * 1996-05-24 1997-12-09 University Of Maryland At Baltimore Dna vaccines for eliciting a mucosal immune response
US8323963B2 (en) * 1996-05-29 2012-12-04 University Of Southern California Construction and use of genes encoding pathogenic epitopes for treatment of autoimmune disease
US6274136B1 (en) 1996-05-29 2001-08-14 University Of Southern California Construction and use of genes encoding pathogenic epitopes for treatment of autoimmune disease
US5849727A (en) 1996-06-28 1998-12-15 Board Of Regents Of The University Of Nebraska Compositions and methods for altering the biodistribution of biological agents
FR2750704B1 (en) 1996-07-04 1998-09-25 Rhone Poulenc Rorer Sa PROCESS FOR PRODUCING THERAPEUTIC DNA
US6444444B1 (en) * 1996-07-10 2002-09-03 Aventis Pasteur Limited Genes encoding mycobacterial proteins associated with cell binding and cell entry and uses thereof
US6696421B2 (en) 1996-07-12 2004-02-24 University Of Manitoba DNA immunization against chlamydia infection
US6235290B1 (en) * 1997-07-11 2001-05-22 University Of Manitoba DNA immunization against chlaymdia infection
US6344202B1 (en) 1996-07-12 2002-02-05 University Of Manitoba DNA immunization against chlaymdia infection
US6443974B1 (en) 1996-07-28 2002-09-03 Biosense, Inc. Electromagnetic cardiac biostimulation
US6593305B1 (en) 1996-08-02 2003-07-15 Genesense Technologies Inc. Antitumor antisense sequences directed against R1 and R2 components of ribonucleotide reductase
AU731367B2 (en) 1996-08-09 2001-03-29 Viral Technologies, Inc. HIV p-17 peptide fragment, compositions containing and methods for producing and using same
US6093400A (en) * 1996-08-09 2000-07-25 Cel Sci Corporation Modified HGP-30 peptides, conjugates, compositions and methods of use
US7001614B2 (en) * 1996-08-19 2006-02-21 The United States Of America As Represented By The Department Of Health And Human Services Liposome complexes for increased systemic delivery
US7288266B2 (en) * 1996-08-19 2007-10-30 United States Of America As Represented By The Secretary, Department Of Health And Human Services Liposome complexes for increased systemic delivery
US6770291B2 (en) * 1996-08-30 2004-08-03 The United States Of America As Represented By The Department Of Health And Human Services Liposome complexes for increased systemic delivery
AU730771B2 (en) * 1996-08-19 2001-03-15 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Novel liposome complexes for increased systemic delivery
WO1998007878A2 (en) 1996-08-23 1998-02-26 Arch Development Corporation Long-term expression of gene products by transforming muscle cells
JP2001500738A (en) * 1996-09-17 2001-01-23 カイロン コーポレイション Compositions and methods for treating intracellular diseases
US6610661B1 (en) 1996-10-11 2003-08-26 The Regents Of The University Of California Immunostimulatory polynucleotide/immunomodulatory molecule conjugates
ATE429929T1 (en) 1996-10-23 2009-05-15 Univ Pennsylvania IMPROVED VACCINES
US6387700B1 (en) * 1996-11-04 2002-05-14 The Reagents Of The University Of Michigan Cationic peptides, Cys-Trp-(LYS)n, for gene delivery
US6544523B1 (en) 1996-11-13 2003-04-08 Chiron Corporation Mutant forms of Fas ligand and uses thereof
DE19648625A1 (en) 1996-11-13 1998-05-14 Soft Gene Gmbh Microprojectile for the introduction of substances into cells by ballistic transfer
US20060002949A1 (en) 1996-11-14 2006-01-05 Army Govt. Of The Usa, As Rep. By Secretary Of The Office Of The Command Judge Advocate, Hq Usamrmc. Transcutaneous immunization without heterologous adjuvant
US6797276B1 (en) 1996-11-14 2004-09-28 The United States Of America As Represented By The Secretary Of The Army Use of penetration enhancers and barrier disruption agents to enhance the transcutaneous immune response
US6204250B1 (en) 1996-11-22 2001-03-20 The Mount Sinai Medical Center Of The City Of New York Immunization of infants
US20020103145A1 (en) * 1999-05-19 2002-08-01 Adrian Bot Immunization of infants
US6200959B1 (en) 1996-12-04 2001-03-13 Powerject Vaccines Inc. Genetic induction of anti-viral immune response and genetic vaccine for filovirus
EP0948609B1 (en) 1996-12-06 2005-03-16 Aventis Pharmaceuticals Inc. Llg polypeptides of the triacylglycerol lipase family, and compositions and methods for their use in enzymatic hydrolysis, and protein and gene therapies
US7008776B1 (en) * 1996-12-06 2006-03-07 Aventis Pharmaceuticals Inc. Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
AU6614398A (en) * 1997-01-17 1998-08-07 Institut National De La Sante Et De La Recherche Medicale Adenoviral-vector-mediated gene transfer into medullary motor neurons
US5783567A (en) * 1997-01-22 1998-07-21 Pangaea Pharmaceuticals, Inc. Microparticles for delivery of nucleic acid
US20020182258A1 (en) * 1997-01-22 2002-12-05 Zycos Inc., A Delaware Corporation Microparticles for delivery of nucleic acid
US6034072A (en) * 1997-02-10 2000-03-07 Genemedicine, Inc. IL-2 gene expression and delivery systems and uses
ES2232934T3 (en) * 1997-02-14 2005-06-01 MERCK & CO., INC. FORMULATIONS OF VACCINES BASED ON POLINUCLEOTIDES.
US20040242522A1 (en) * 1997-02-14 2004-12-02 Volkin David B. Polynucleotide vaccine formulations
AU738513B2 (en) 1997-02-28 2001-09-20 University Of Iowa Research Foundation, The Use of nucleic acids containing unmethylated CpG dinucleotide in the treatment of LPS-associated disorders
WO1998040499A1 (en) * 1997-03-10 1998-09-17 Heather Lynn Davis Gene delivery to mucosal epithelium for immunization or therapeutic purposes
US5990091A (en) 1997-03-12 1999-11-23 Virogenetics Corporation Vectors having enhanced expression, and methods of making and uses thereof
EP0969775A4 (en) * 1997-03-25 2001-06-13 Morris Laster Bone marrow as a site for transplantation
TR199902562T2 (en) 1997-04-15 2000-02-21 Farmaceutisk Laboratorium Ferring A/S De�i�tirilmi� TNF alfa molek�lleri
WO1998049321A2 (en) * 1997-04-28 1998-11-05 Rhone-Poulenc Rorer S.A. Adenovirus-mediated intratumoral delivery of an angiogenesis antagonist for the treatment of tumors
US5948925A (en) * 1997-05-06 1999-09-07 Genzyme Corporation Cationic amphiphiles containing linkers derived from neutral or positively charged amino acids
US5952516A (en) * 1997-05-08 1999-09-14 Genzyme Corporation Cationic amphiphiles containing multiplesteroid lipophilic groups
US5942634A (en) * 1997-05-09 1999-08-24 Genzyme Corporation Cationic amphiphiles for cell transfections
GB9709421D0 (en) 1997-05-10 1997-07-02 Zeneca Ltd Chemical compounds
US20030104044A1 (en) * 1997-05-14 2003-06-05 Semple Sean C. Compositions for stimulating cytokine secretion and inducing an immune response
US6287591B1 (en) 1997-05-14 2001-09-11 Inex Pharmaceuticals Corp. Charged therapeutic agents encapsulated in lipid particles containing four lipid components
EP1003531B1 (en) 1997-05-20 2007-08-22 Ottawa Health Research Institute Processes for preparing nucleic acid constructs
ATE432348T1 (en) 1997-06-06 2009-06-15 Univ California INHIBITORS OF IMMUNO-STIMULATIVE DNA SEQUENCE ACTIVITY
US6635623B1 (en) 1997-06-13 2003-10-21 Baylor College Of Medicine Lipoproteins as nucleic acid vectors
US20050096288A1 (en) * 1997-06-13 2005-05-05 Aragene, Inc. Lipoproteins as nucleic acid vectors
KR100220645B1 (en) * 1997-07-04 1999-09-15 구광시 Process for producing benzene derivatives
US6977074B2 (en) * 1997-07-10 2005-12-20 Mannkind Corporation Method of inducing a CTL response
US6994851B1 (en) 1997-07-10 2006-02-07 Mannkind Corporation Method of inducing a CTL response
US7923250B2 (en) 1997-07-30 2011-04-12 Warsaw Orthopedic, Inc. Methods of expressing LIM mineralization protein in non-osseous cells
EP1007673B1 (en) 1997-07-30 2008-12-17 Emory University Novel bone mineralization proteins, dna, vectors, expression systems
PL199014B1 (en) 1997-08-08 2008-08-29 Univ California Monoclonal antibody specifically fixing to αvß6 integrine, a hybridome producing it and application of antibody
US6716823B1 (en) 1997-08-13 2004-04-06 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom, and uses thereof
US20030125278A1 (en) * 1997-08-13 2003-07-03 Tang De-Chu C. Immunization of animals by topical applications of a salmonella-based vector
US6706693B1 (en) 1997-08-13 2004-03-16 The Uab Research Foundation Vaccination by topical application of genetic vectors
US20030045492A1 (en) * 1997-08-13 2003-03-06 Tang De-Chu C. Vaccination by topical application of recombinant vectors
US6348450B1 (en) 1997-08-13 2002-02-19 The Uab Research Foundation Noninvasive genetic immunization, expression products therefrom and uses thereof
EP1049712B1 (en) * 1997-08-14 2007-01-24 Institut Pasteur Hybrid tetanus toxoid proteins that migrate retrogradely and transynaptically into the cns
US6818627B1 (en) * 1997-08-14 2004-11-16 The Trustees Of The University Of Pennsylvania Functional fragments of HIV-1 Vpr protein and methods of using the same
US7923216B2 (en) 1997-08-14 2011-04-12 Institut Pasteur In vivo modulation of neuronal transport
US20100081197A1 (en) * 1997-08-14 2010-04-01 Sylvie Roux In vivo modulation of neuronal transport
US7923015B2 (en) * 1997-08-14 2011-04-12 Institut Pasteur Methods for direct visualization of active synapses
JP4663113B2 (en) 1997-09-05 2011-03-30 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Use of immunostimulatory oligonucleotides to prevent or reduce antigen-stimulated granulocyte-mediated inflammation
CN100391542C (en) * 1997-09-15 2008-06-04 遗传免疫有限公司 Method of delivering genes to antigen presenting cells of the skin
WO1999013896A1 (en) 1997-09-18 1999-03-25 The Trustees Of The University Of Pennsylvania Attenuated vif dna immunization cassettes for genetic vaccines
EP2292771A3 (en) * 1997-09-19 2011-07-27 Life Technologies Corporation Sense mRNA therapy
US6565885B1 (en) 1997-09-29 2003-05-20 Inhale Therapeutic Systems, Inc. Methods of spray drying pharmaceutical compositions
US20060165606A1 (en) 1997-09-29 2006-07-27 Nektar Therapeutics Pulmonary delivery particles comprising water insoluble or crystalline active agents
US6517843B1 (en) 1999-08-31 2003-02-11 Merial Reduction of porcine circovirus-2 viral load with inactivated PCV-2
US6183746B1 (en) * 1997-10-09 2001-02-06 Zycos Inc. Immunogenic peptides from the HPV E7 protein
US6013258A (en) * 1997-10-09 2000-01-11 Zycos Inc. Immunogenic peptides from the HPV E7 protein
US6875606B1 (en) 1997-10-23 2005-04-05 The United States Of America As Represented By The Department Of Veterans Affairs Human α-7 nicotinic receptor promoter
AU745805B2 (en) 1997-10-28 2002-04-11 Wyeth Compositions and methods for delivery of genetic material
ATE476508T1 (en) 1997-11-06 2010-08-15 Novartis Vaccines & Diagnostic NEISSERIAL ANTIGENS
US6914131B1 (en) * 1998-10-09 2005-07-05 Chiron S.R.L. Neisserial antigens
US6524805B1 (en) 1997-11-10 2003-02-25 George B. Stefano Methods for identifying estrogen surface receptor agonists
US7435723B2 (en) * 1997-11-21 2008-10-14 Mirus Bio Corporation Process for delivery of polynucleotides to the prostate
FR2772047B1 (en) 1997-12-05 2004-04-09 Ct Nat D Etudes Veterinaires E GENOMIC SEQUENCE AND POLYPEPTIDES OF CIRCOVIRUS ASSOCIATED WITH PIGLET LOSS DISEASE (MAP), APPLICATIONS TO DIAGNOSIS AND TO PREVENTION AND / OR TREATMENT OF INFECTION
EP1816200B1 (en) 1997-12-11 2016-03-09 Merial Postweaning multisystemic wasting syndrome virus for pigs
KR100879650B1 (en) 1997-12-11 2009-01-20 유니버시티 오브 사스카췌완 Postweaning multisystemic wasting syndrome virus from pigs
EP1042347A4 (en) * 1997-12-23 2005-02-16 Merck & Co Inc Dna pharmaceutical formulations comprising citrate or triethanolamine and combinations thereof
ATE302267T1 (en) 1998-01-08 2005-09-15 Aventis Pharma Inc TRANSGENIC RABBIT THAT EXPRESSES A FUNCTIONAL HUMAN LIPOPROTEIN(A).
WO1999036544A2 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
JP2002501743A (en) 1998-01-30 2002-01-22 ジェネセンス テクノロジーズ インコーポレイテッド Oligonucleotide sequences complementary to thioredoxin gene or thioredoxin reductase gene and methods of using same to regulate cell growth
WO1999039741A2 (en) 1998-02-03 1999-08-12 Inex Pharmaceuticals Corporation Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US6410328B1 (en) * 1998-02-03 2002-06-25 Protiva Biotherapeutics Inc. Sensitizing cells to compounds using lipid-mediated gene and compound delivery
DE69838526T2 (en) 1998-02-05 2008-07-03 Biosense Webster, Inc., Diamond Bar Device for releasing a drug in the heart
US20030129750A1 (en) * 1998-02-05 2003-07-10 Yitzhack Schwartz Homing of donor cells to a target zone in tissue using active therapeutics or substances
US20030113303A1 (en) * 1998-02-05 2003-06-19 Yitzhack Schwartz Homing of embryonic stem cells to a target zone in tissue using active therapeutics or substances
US7390619B1 (en) * 1998-02-11 2008-06-24 Maxygen, Inc. Optimization of immunomodulatory properties of genetic vaccines
US6541011B2 (en) 1998-02-11 2003-04-01 Maxygen, Inc. Antigen library immunization
US6087341A (en) * 1998-02-12 2000-07-11 The Board Of Trustees Of The Leland Standford Junior University Introduction of nucleic acid into skin cells by topical application
US6087128A (en) 1998-02-12 2000-07-11 Ndsu Research Foundation DNA encoding an avian E. coli iss
US6369201B1 (en) 1998-02-19 2002-04-09 Metamorphix International, Inc. Myostatin multimers
US6709844B1 (en) * 2000-11-16 2004-03-23 Mannkind Corporation Avoidance of undesirable replication intermediates in plasmid propagation
US20030138808A1 (en) * 1998-02-19 2003-07-24 Simard John J.L. Expression vectors encoding epitopes of target-associated antigens
DK2295065T3 (en) 1998-02-20 2014-01-06 Univ Miami Modified heat shock protein-antigen peptide complex
US20030105294A1 (en) * 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
WO1999044644A1 (en) * 1998-03-02 1999-09-10 Vanderbilt University Improved eukaryotic expression vector
US6417168B1 (en) * 1998-03-04 2002-07-09 The Trustees Of The University Of Pennsylvania Compositions and methods of treating tumors
CA2310252A1 (en) * 1998-03-06 1999-09-10 Imclone Systems Incorporated Active immunization against angiogenesis-associated antigens
CA2349562A1 (en) * 1998-03-06 1999-09-10 Crosscart, Inc. Soft tissue xenografts
AU3065999A (en) * 1998-03-10 1999-09-27 Children's Hospital Of Philadelphia, The Compositions and methods for treatment of asthma
US6579682B1 (en) 1998-03-10 2003-06-17 The Regents Of University Of California Methods and tools for identifying compounds which modulate atherosclerosis by impacting LDL-proteoglycan binding
CA2323776C (en) 1998-03-19 2010-04-27 Human Genome Sciences, Inc. Cytokine receptor common gamma chain like
US6455497B1 (en) 1998-03-25 2002-09-24 Mayo Foundation For Medical Education And Research Methods and materials for treating inflammatory diseases
US7005275B2 (en) * 1998-03-27 2006-02-28 Cytos Biotechnology Ag Inducible alphaviral gene expression system
JP4227302B2 (en) 1998-04-07 2009-02-18 コリクサ コーポレイション MYCOBACTERIUMUMBERBERCULOSIS antigen fusion protein and use thereof
US7157435B2 (en) * 1998-04-15 2007-01-02 The Regents Of The University Of California Methods for modulation of the effects of aging on the primate brain
WO1999054463A2 (en) * 1998-04-21 1999-10-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Human k+ ion channel and therapeutic applications thereof
US6841537B1 (en) 1998-04-22 2005-01-11 Protiva Biotherapeutics Inc. Combination therapy using nucleic acids and conventional drugs
US6841538B1 (en) 1998-04-22 2005-01-11 Inex Pharmaceuticals Corporation Combination therapy using nucleic acids and radio therapy
US6417169B1 (en) * 1998-04-23 2002-07-09 Genesense Technologies Inc. Insulin-like growth factor II antisense oligonucleotide sequences and methods of using same to inhibit cell growth
US6395253B2 (en) 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
US7279462B1 (en) 1998-04-27 2007-10-09 Nevagen Llc Somatic transgene immunization and related methods
US6602505B2 (en) 1998-04-30 2003-08-05 University Of Southern California Viral chimeras comprised of CAEV and HIV-1 genetic elements
EP2261349A3 (en) 1998-05-01 2012-01-11 Novartis Vaccines and Diagnostics, Inc. Neisseria meningitidis antigens and compositions
PL345241A1 (en) * 1998-05-06 2001-12-03 Upjohn Co Introduction of naked dna or rna encoding non-human vertebrate peptide hormones or cytokines into a non-human vertebrate
US6225456B1 (en) 1998-05-07 2001-05-01 University Technololy Corporation Ras suppressor SUR-5
EP1078092B1 (en) 1998-05-13 2011-08-03 Epimmune Inc. Expression vectors for stimulating an immune response and methods of using the same
US6406719B1 (en) 1998-05-13 2002-06-18 Microbiological Research Authority Encapsulation of bioactive agents
GB9810236D0 (en) 1998-05-13 1998-07-08 Microbiological Res Authority Improvements relating to encapsulation of bioactive agents
US6506889B1 (en) 1998-05-19 2003-01-14 University Technology Corporation Ras suppressor SUR-8 and related compositions and methods
US6322976B1 (en) 1998-05-28 2001-11-27 Medical Research Council Compositions and methods of disease diagnosis and therapy
US7244714B1 (en) 1998-06-12 2007-07-17 Aradigm Corporation Methods of delivering aerosolized polynucleotides to the respiratory tract
EP2036573A1 (en) 1998-06-17 2009-03-18 IDM Pharma, Inc. HLA binding peptides and their uses
CA2335460A1 (en) 1998-06-18 1999-12-23 Johns Hopkins University School Of Medicine Polymers for delivery of nucleic acids
US20030022854A1 (en) * 1998-06-25 2003-01-30 Dow Steven W. Vaccines using nucleic acid-lipid complexes
US20040247662A1 (en) * 1998-06-25 2004-12-09 Dow Steven W. Systemic immune activation method using nucleic acid-lipid complexes
US6693086B1 (en) 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
JP2002519393A (en) 1998-07-02 2002-07-02 ジエンザイム コーポレイション Transgene expression in polarized cells.
US7922709B2 (en) * 1998-07-13 2011-04-12 Genetronics, Inc. Enhanced delivery of naked DNA to skin by non-invasive in vivo electroporation
CA2337652C (en) 1998-07-13 2013-03-26 Genetronics, Inc. Skin and muscle-targeted gene therapy by pulsed electrical field
US6916490B1 (en) 1998-07-23 2005-07-12 UAB Research Center Controlled release of bioactive substances
EP1100545A4 (en) * 1998-07-29 2002-03-20 Invitrogen Corp Regulated expression of recombinant proteins using rna viruses
GB9817662D0 (en) 1998-08-13 1998-10-07 Crocker Peter J Substance delivery
US6333051B1 (en) * 1998-09-03 2001-12-25 Supratek Pharma, Inc. Nanogel networks and biological agent compositions thereof
US6696089B2 (en) 1998-09-03 2004-02-24 Board Of Regents Of The University Of Nebraska Nanogel networks including polyion polymer fragments and biological agent compositions thereof
AU5911899A (en) * 1998-09-09 2000-03-27 Genzyme Corporation Methylation of plasmid vectors
EP0998945A1 (en) * 1998-09-30 2000-05-10 Transgene S.A. Use of magnesium (Mg2+) for the enhancement of gene delivery in gene therapy
US6387888B1 (en) 1998-09-30 2002-05-14 American Foundation For Biological Research, Inc. Immunotherapy of cancer through expression of truncated tumor or tumor-associated antigen
EP1121437B1 (en) 1998-10-15 2008-02-20 Novartis Vaccines and Diagnostics, Inc. Metastatic breast and colon cancer regulated genes
DK1123396T3 (en) * 1998-10-19 2006-07-10 Powderject Vaccines Inc Minimal promoters and uses thereof
US6770282B1 (en) * 1998-10-23 2004-08-03 Heska Corporation Cationic lipid-mediated enhancement of nucleic acid immunization of cats
US6913749B2 (en) 1998-11-02 2005-07-05 Resistentia Pharmaceuticals Ab Immunogenic polypeptides for inducing anti-self IgE responses
US6881723B1 (en) 1998-11-05 2005-04-19 Powderject Vaccines, Inc. Nucleic acid constructs
EP1129064B1 (en) 1998-11-12 2008-01-09 Invitrogen Corporation Transfection reagents
KR20020007281A (en) * 1998-11-16 2002-01-26 링순 두안 Generation of antibodies using polynucleotide vaccination in avian species
US6656498B1 (en) * 1998-11-25 2003-12-02 Vanderbilt University Cationic liposomes for gene transfer
US6716810B1 (en) * 1998-12-09 2004-04-06 Eleanor Roosevelt Institute Composition and method for regulation of body weight and associated conditions
US6331405B1 (en) * 1998-12-10 2001-12-18 The Rockefeller University Receptor for Mycobacterium leprae and methods of use thereof
ES2310055T3 (en) 1998-12-16 2008-12-16 Novartis Vaccines & Diagnostic CYCLIN DEPENDENT HUMAN KINASE (HPNQALRE).
US6441156B1 (en) * 1998-12-30 2002-08-27 The United States Of America As Represented By The Department Of Health And Human Services Calcium channel compositions and methods of use thereof
US7935805B1 (en) 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
US6602705B1 (en) 1998-12-31 2003-08-05 Chiron Corporation Expression of HIV polypeptides and production of virus-like particles
CA2363067C (en) 1999-01-12 2012-03-20 David J. Grainger Compounds and methods to inhibit or augment an inflammatory response
US7625859B1 (en) 2000-02-16 2009-12-01 Oregon Health & Science University HER-2 binding antagonists
US7393823B1 (en) 1999-01-20 2008-07-01 Oregon Health And Science University HER-2 binding antagonists
AU776150B2 (en) * 1999-01-28 2004-08-26 Medical College Of Georgia Research Institute, Inc. Composition and method for (in vivo) and (in vitro) attenuation of gene expression using double stranded RNA
WO2000044764A1 (en) * 1999-01-28 2000-08-03 Smithkline Beecham Corporation Mvd
DE60007290T2 (en) 1999-01-28 2004-09-23 Cyto Pulse Sciences, Inc. INTRODUCTION OF MACROMOLECULES IN CELLS
PT1150918E (en) 1999-02-03 2005-01-31 Biosante Pharmaceuticals Inc METHOD OF MANUFACTURE OF CALCIUM PHOSPHATE THERAPEUTIC PARTICLES
US20040258763A1 (en) * 1999-02-03 2004-12-23 Bell Steve J.D. Methods of manufacture and use of calcium phosphate particles containing allergens
US20020054914A1 (en) * 1999-02-03 2002-05-09 Tulin Morcol Compositions and methods for therapuetic agents complexed with calcium phosphate and encased by casein
WO2000047227A2 (en) 1999-02-09 2000-08-17 Powderject Vaccines, Inc. Mycobacterium tuberculosis, immunization
US6121000A (en) * 1999-02-11 2000-09-19 Genesense Technologies, Inc. Antitumor antisense sequences directed against R1 and R2 components of ribonucleotide reductase
US6592623B1 (en) 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US7615373B2 (en) * 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US20040018226A1 (en) * 1999-02-25 2004-01-29 Wnek Gary E. Electroprocessing of materials useful in drug delivery and cell encapsulation
US20020081732A1 (en) * 2000-10-18 2002-06-27 Bowlin Gary L. Electroprocessing in drug delivery and cell encapsulation
US20040116032A1 (en) * 1999-02-25 2004-06-17 Bowlin Gary L. Electroprocessed collagen
CA2363779A1 (en) 1999-02-26 2000-08-31 Human Genome Sciences, Inc. Human endokine alpha and methods of use
US6656696B2 (en) 1999-02-26 2003-12-02 Cyclacel Compositions and methods for monitoring the phosphorylation of natural binding partners
US6670144B1 (en) 1999-02-26 2003-12-30 Cyclacel, Ltd. Compositions and methods for monitoring the phosphorylation of natural binding partners
US6465199B1 (en) 1999-02-26 2002-10-15 Cyclacel, Ltd. Compositions and methods for monitoring the modification of natural binding partners
JP2002538090A (en) * 1999-03-03 2002-11-12 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Vaccine and gene therapy compositions and methods of making and using the same
EP1171189B1 (en) 1999-03-25 2016-05-11 Inovio Pharmaceuticals, Inc. Apparatus for reducing electroporation-mediated muscle reaction and pain response
US6678558B1 (en) * 1999-03-25 2004-01-13 Genetronics, Inc. Method and apparatus for reducing electroporation-mediated muscle reaction and pain response
ATE549032T1 (en) * 1999-03-26 2012-03-15 Vical Inc ADJUVANCE COMPOUNDS FOR IMPROVEMENT OF IMMUNE RESPONSES TO POLYNUCLEOTIDE-BASED VACCINES
US6864235B1 (en) * 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US6911429B2 (en) * 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
JP4932086B2 (en) 1999-04-08 2012-05-16 インターセル ユーエスエイ、インコーポレイテッド Dry formulation for transcutaneous immunization
DK1175497T3 (en) 1999-04-14 2010-05-31 Novartis Vaccines & Diagnostic Compositions and Methods for Generating an Immune Response by Utilizing Alpha Virus-Based Vector Systems
AU778737B2 (en) 1999-04-14 2004-12-16 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
EP1171586B1 (en) 1999-04-21 2013-06-12 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibiting the function of polynucleotide sequences
JP5000807B2 (en) * 1999-04-27 2012-08-15 ネバジェン エルエルシー Somatic cell transgene immunization and related methods
WO2000066741A2 (en) 1999-04-30 2000-11-09 Chiron S.P.A. Conserved neisserial antigens
CN1196495C (en) 1999-04-30 2005-04-13 宾夕法尼亚大学理事会 Mutant human CD80 and copositions for and methods of making and using the same
US20040009936A1 (en) * 1999-05-03 2004-01-15 Tang De-Chu C. Vaccine and drug delivery by topical application of vectors and vector extracts
WO2000067825A1 (en) 1999-05-07 2000-11-16 Microheart, Inc. Apparatus and method for delivering therapeutic and diagnostic agents
US7171264B1 (en) * 1999-05-10 2007-01-30 Genetronics, Inc. Intradermal delivery of active agents by needle-free injection and electroporation
US6520950B1 (en) 1999-05-10 2003-02-18 Genetronics, Inc. Method of electroporation-enhanced delivery of active agents
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
AU5043900A (en) * 1999-05-24 2000-12-12 Mayo Foundation For Medical Education And Research Adenovirus vectors encoding brain natriuretic peptide
US6696424B1 (en) 1999-05-28 2004-02-24 Vical Incorporated Cytofectin dimers and methods of use thereof
TR199901199A2 (en) * 1999-05-31 2001-09-21 Kocagöz Tanil Bacterial transformation kit
US6774120B1 (en) * 1999-06-01 2004-08-10 Sarah Ferber Methods of inducing regulated pancreatic hormone production in non-pancreatic islet tissues
US8778899B2 (en) 1999-06-01 2014-07-15 Sarah Ferber Methods of inducing regulated pancreatic hormone production in non-pancreatic islet tissues
ATE342089T1 (en) 1999-06-02 2006-11-15 Boston Scient Ltd DRUG DELIVERY DEVICES
US7147633B2 (en) 1999-06-02 2006-12-12 Boston Scientific Scimed, Inc. Method and apparatus for treatment of atrial fibrillation
EP1246532A2 (en) * 1999-06-07 2002-10-09 Mirus Corporation Delivery of single stranded dna for expression
US6468984B1 (en) 1999-06-08 2002-10-22 Innovo Biotechnologies Ltd. DNA vaccine for protecting an avian against infectious bursal disease virus
US6943152B1 (en) 1999-06-10 2005-09-13 Merial DNA vaccine-PCV
US6355246B1 (en) 1999-06-10 2002-03-12 Board Of Trustees Of Michigan State University Feline calicivirus isolated from cat urine and vaccines thereof
US20010048940A1 (en) * 1999-06-18 2001-12-06 Jennifer D. Tousignant Cationic amphiphile micellar complexes
WO2001000663A2 (en) * 1999-06-28 2001-01-04 Oklahoma Medical Research Foundation Catalytically active recombinant memapsin and methods of use thereof
US6770740B1 (en) 1999-07-13 2004-08-03 The Regents Of The University Of Michigan Crosslinked DNA condensate compositions and gene delivery methods
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
US20030078499A1 (en) 1999-08-12 2003-04-24 Eppstein Jonathan A. Microporation of tissue for delivery of bioactive agents
US20050249794A1 (en) * 1999-08-27 2005-11-10 Semple Sean C Compositions for stimulating cytokine secretion and inducing an immune response
AUPQ259399A0 (en) * 1999-09-01 1999-09-23 Lustre Investments Pte Ltd Therapeutic agents
CA2382026A1 (en) 1999-09-02 2001-03-08 Ruth A. Vrable Vaccine to control equine protozoal myeloencephalitis in horses
US7419668B1 (en) 1999-09-02 2008-09-02 Board Of Trustees Of Michigan State University Vaccine to control equine protozoal myeloencephalitis in horses
US20050100928A1 (en) * 1999-09-16 2005-05-12 Zycos Inc., A Delaware Corporation Nucleic acids encoding polyepitope polypeptides
US6660260B1 (en) * 1999-09-21 2003-12-09 Mayo Foundation For Medical Education And Research Bioprosthetic heart valves
US6632663B1 (en) 1999-09-22 2003-10-14 Aventis Pasteur Limited DNA immunization against chlamydia infection
EP1808180A3 (en) 1999-10-22 2010-12-22 Sanofi Pasteur Limited Modified GP 100 and uses thereof
ES2541830T3 (en) 1999-10-29 2015-07-27 Novartis Vaccines And Diagnositics S.R.L. Neisseria antigenic peptides
US7196066B1 (en) * 1999-11-03 2007-03-27 Powderject Vaccines, Inc. DNA-vaccines based on constructs derived from the genomes of human and animal pathogens
US20040234539A1 (en) * 1999-11-03 2004-11-25 Powderject Research Limited Nucleic acid vaccine compositions having a mammalian cd80/cd86 gene promoter driving antigen expression
US7214369B2 (en) * 2003-05-05 2007-05-08 Mirus Bio Corporation Devices and processes for distribution of genetic material to mammalian limb
US7642248B2 (en) * 1999-11-05 2010-01-05 Roche Madison Inc Devices and processes for distribution of genetic material to mammalian limb
EP1246649B1 (en) 1999-11-05 2006-10-18 Mirus Bio Corporation Intravascular delivery of nucleic acid
US20040109874A1 (en) * 1999-11-10 2004-06-10 Powderject Vaccines, Inc. Induction of mucosal immunity by vaccination via the skin route
WO2001036489A2 (en) 1999-11-12 2001-05-25 Merck Patent Gmbh Erythropoietin forms with improved properties
EP1232264B1 (en) 1999-11-18 2009-10-21 Novartis Vaccines and Diagnostics, Inc. Human fgf-21 gene and gene expression products
CA2386341A1 (en) 1999-11-18 2001-05-25 Epimmune Inc. Heteroclitic analogs and related methods
US20050037086A1 (en) * 1999-11-19 2005-02-17 Zycos Inc., A Delaware Corporation Continuous-flow method for preparing microparticles
US20070077256A1 (en) 1999-11-19 2007-04-05 Los Angeles Biomedical Research Institute Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US20040072785A1 (en) * 1999-11-23 2004-04-15 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
ES2170622B1 (en) * 1999-12-03 2004-05-16 Consejo Superior De Investigaciones Cientificas CLONES AND INFECTIVE VECTORS DERIVED FROM CORONAVIRUS AND ITS APPLICATIONS.
US6407211B1 (en) * 1999-12-17 2002-06-18 Mayo Foundation For Medical Education And Research Chimeric natriuretic peptides
DK1248646T3 (en) * 1999-12-21 2008-05-19 Merial Sas Preparations and vaccines containing antigen (s) from cryptosporidium parvum and from other enteric pathogen
IL150358A0 (en) * 1999-12-22 2002-12-01 Univ Pennsylvania Cosmid dna constructs and methods of making the same
CA2398207A1 (en) * 1999-12-22 2001-06-28 The Ohio State University Research Foundation Methods for protecting against lethal infection with bacillus anthracis
EP1842549A3 (en) 1999-12-28 2007-10-17 Pharmexa Inc. Optimized minigenes and peptides encoded thereby
EP2275129A3 (en) 2000-01-17 2013-11-06 Novartis Vaccines and Diagnostics S.r.l. Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
US6372722B1 (en) 2000-01-19 2002-04-16 Genteric, Inc. Method for nucleic acid transfection of cells
US6852705B2 (en) 2000-01-21 2005-02-08 Merial DNA vaccines for farm animals, in particular bovines and porcines
US20040002472A1 (en) * 2000-01-21 2004-01-01 Audonnet Jean-Christophe Francis Vaccination or immunization using a prime-boost regimen
US7078388B2 (en) * 2000-01-21 2006-07-18 Merial DNA vaccines for farm animals, in particular bovines and porcines
US7041443B2 (en) * 2000-02-08 2006-05-09 Regents Of The University Of Minnesota Porcine reproductive and respiratory syndrome virus and methods of use
CA2399832C (en) 2000-02-11 2011-09-20 Stephen D. Gillies Enhancing the circulating half-life of antibody-based fusion proteins
AU780270B2 (en) * 2000-02-23 2005-03-10 Association Francaise Contre Les Myopathies Treatment of immune diseases
ES2281409T3 (en) * 2000-02-28 2007-10-01 Novartis Vaccines And Diagnostics S.R.L. NEISSERIA PROTEIN HETEROLOGY EXPRESSION.
US8623379B2 (en) * 2000-03-02 2014-01-07 Emory University Compositions and methods for generating an immune response
US20020106798A1 (en) 2000-03-02 2002-08-08 Robinson Harriet L. DNA expression vectors and methods of use
US20040105871A1 (en) * 2000-03-02 2004-06-03 Robinson Harriet L. Compositions and methods for generating an immune response
WO2002072754A2 (en) * 2001-03-08 2002-09-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mva expressing modified hiv envelope, gag, and pol genes
EP2075255A1 (en) 2000-03-08 2009-07-01 Novartis Vaccines and Diagnostics, Inc. Human FGF-23 gene and gene expression products
US6953658B2 (en) 2000-03-09 2005-10-11 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating gastrointestinal cancer
CA2402530C (en) 2000-03-13 2014-01-14 Cornell Research Foundation, Inc. Blocking leukocyte emigration and inflammation by interfering with cd99
ATE450621T2 (en) 2000-03-30 2009-12-15 Whitehead Biomedical Inst MEDIATORS OF RNA INTERFERENCE THAT ARE RNA SEQUENCE SPECIFIC
GB0018307D0 (en) 2000-07-26 2000-09-13 Aventis Pharm Prod Inc Polypeptides
EP1274720A4 (en) 2000-04-12 2004-08-18 Human Genome Sciences Inc Albumin fusion proteins
WO2001079299A1 (en) 2000-04-13 2001-10-25 The Rockefeller University Enhancement of antibody-mediated immune responses
EP1278551A2 (en) * 2000-04-21 2003-01-29 Vical Incorporated Compositions and methods for (in vivo) delivery of polynucleotide-based therapeutics
US6861234B1 (en) 2000-04-28 2005-03-01 Mannkind Corporation Method of epitope discovery
JP2003531865A (en) * 2000-04-28 2003-10-28 アメリカ合衆国 Improving immunogenicity using a combination of DNA and vaccinia virus vector vaccines
JP2003535824A (en) * 2000-04-28 2003-12-02 シーティーエル イムノセラピーズ コーポレーション Epitope tuning in antigen presenting cells
US20030215425A1 (en) * 2001-12-07 2003-11-20 Simard John J. L. Epitope synchronization in antigen presenting cells
EP1278542A2 (en) * 2000-05-05 2003-01-29 Cytos Biotechnology AG Molecular antigen arrays and vaccines
CA2382133C (en) 2000-05-10 2010-11-23 Alliance Pharmaceutical Corporation Phospholipid-based powders for drug delivery
US7871598B1 (en) 2000-05-10 2011-01-18 Novartis Ag Stable metal ion-lipid powdered pharmaceutical compositions for drug delivery and methods of use
US8404217B2 (en) 2000-05-10 2013-03-26 Novartis Ag Formulation for pulmonary administration of antifungal agents, and associated methods of manufacture and use
AU2001258102B2 (en) 2000-05-10 2007-03-01 Aventis Pasteur Limited Immunogenic polypeptides encoded by mage minigenes and uses thereof
AU2001256609A1 (en) * 2000-05-12 2001-11-20 Cyril John Higgins Method of organized retrieval of world-wide web pages
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
EP1950297A2 (en) 2000-05-31 2008-07-30 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating neoplastic disease using chemotherapy and radiation sensitizers
AU2001266694C1 (en) 2000-06-02 2005-09-01 University Of Connecticut Health Center Complexes of alpha (2) macroglobulin and antigenic molecules for immunotherapy
US7700359B2 (en) * 2000-06-02 2010-04-20 Novartis Vaccines And Diagnostics, Inc. Gene products differentially expressed in cancerous cells
EP1292285A4 (en) * 2000-06-02 2009-07-22 Eisai Corp North America Delivery systems for bioactive agents
EP1294949A4 (en) 2000-06-15 2004-08-25 Human Genome Sciences Inc Human tumor necrosis factor delta and epsilon
ES2305087T3 (en) 2000-06-15 2008-11-01 Novartis Vaccines And Diagnostics, Inc. POLINUCLEOTIDOS RELATED TO CANCER DE COLON.
US20040204379A1 (en) * 2000-06-19 2004-10-14 Cheng Seng H. Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
US20020068691A1 (en) * 2000-06-21 2002-06-06 Susana Salceda Method of diagnosing, monitoring, staging, imaging and treating breast cancer
AU2001271621A1 (en) 2000-06-28 2002-01-08 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating colon cancer
EP2075582A3 (en) 2000-07-12 2010-01-06 Agensys, Inc. Novel tumor antigen useful in diagnosis and therapy of bladder, ovary, lung and kidney cancers
AU2001273432A1 (en) * 2000-07-13 2002-01-30 Invitrogen Corporation Methods and compositions for rapid protein and peptide extraction and isolation using a lysis matrix
EP1305435B1 (en) * 2000-07-27 2007-12-19 The Trustees Of The University Of Pennsylvania Use of herpes simplex virus glycoprotein d to suppress immune responses
US6680059B2 (en) * 2000-08-29 2004-01-20 Tripep Ab Vaccines containing ribavirin and methods of use thereof
AU2001292151B2 (en) 2000-08-17 2006-05-04 Tripep Ab Vaccines containing ribavirin and methods of use thereof
US7022830B2 (en) * 2000-08-17 2006-04-04 Tripep Ab Hepatitis C virus codon optimized non-structural NS3/4A fusion gene
RU2286172C2 (en) * 2000-08-17 2006-10-27 Трипеп Аб Ribavirin-containing vaccines and methods for their application
IL154596A0 (en) 2000-08-25 2003-09-17 Basf Plant Science Gmbh Plant polynucleotides encoding novel prenyl proteases
WO2002018578A2 (en) 2000-08-28 2002-03-07 Agensys, Inc. Nucleic acid and corresponding protein entitled 85p1b3 useful in treatment and detection of cancer
AU2001287023A1 (en) 2000-09-01 2002-03-13 Virginia Commonwealth University Intellectual Property Foundation Plasma-derived-fibrin-based matrices and tissue
EP1315756A2 (en) * 2000-09-01 2003-06-04 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed fibrin-based matrices and tissues
EP1320377B1 (en) 2000-09-01 2011-03-23 Epimmune Inc. Hla binding peptides and their uses
EP1315816B1 (en) 2000-09-08 2010-12-29 University Of Maryland Biotechnology Institute Genetically engineered co-expression dna vaccines, construction methods and uses thereof
UA83458C2 (en) 2000-09-18 2008-07-25 Байоджен Айдек Ма Інк. The isolated polypeptide baff-r (the receptor of the factor of activation of b-cells of the family tnf)
ATE384119T1 (en) * 2000-09-29 2008-02-15 Cargill Inc ISOPRENOID PRODUCTION
US8105825B2 (en) 2000-10-03 2012-01-31 Intrexon Corporation Multiple inducible gene regulation system
CA2425152A1 (en) 2000-10-04 2002-04-11 The Trustees Of The University Of Pennsylvania Highly expressible genes
CA2425648A1 (en) 2000-10-19 2002-04-19 Epimmune Inc. Hla class i and ii binding peptides and their uses
EP2027867A1 (en) 2000-10-20 2009-02-25 Genetics Institute, LLC Method and composition for inhibition of tumor growth and enhancing an immune response
CA2425303A1 (en) 2000-10-27 2002-05-02 John Telford Nucleic acids and proteins from streptococcus groups a & b
US7060442B2 (en) * 2000-10-30 2006-06-13 Regents Of The University Of Michigan Modulators on Nod2 signaling
CA2363493C (en) * 2000-11-22 2006-06-06 Xuemei Cao Attenuated forms of bovine viral diarrhea virus
WO2002041833A2 (en) * 2000-11-22 2002-05-30 Cargill Incorporated Carotenoid biosynthesis
KR20030067696A (en) 2000-11-30 2003-08-14 모렉큐랄 스킨케어 리미티드 Diagnosis and treatment of disease
US20040259247A1 (en) 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
US7393921B2 (en) * 2000-12-04 2008-07-01 Institute For Systems Biology Prostate-specific polypeptide pamp and encoding nucleic acid molecules
CA2430379A1 (en) 2000-12-07 2002-06-13 Chiron Corporation Endogenous retroviruses up-regulated in prostate cancer
US6803200B2 (en) 2000-12-12 2004-10-12 Invitrogen Corporation Compositions and methods for the release of nucleic acid molecules from solid matrices
US6472176B2 (en) 2000-12-14 2002-10-29 Genvec, Inc. Polynucleotide encoding chimeric protein and related vector, cell, and method of expression thereof
AU2002231736A1 (en) 2000-12-22 2002-07-08 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Use of repulsive guidance molecule (rgm) and its modulators
IL155726A0 (en) 2000-12-28 2003-11-23 Wyeth Corp Recombinant protective protein from streptococcus pneumoniae
EP1363663B1 (en) 2001-01-12 2011-03-02 Novartis Vaccines and Diagnostics, Inc. Nucleic acid mucosal immunization
US7128911B2 (en) 2001-01-19 2006-10-31 Cytos Biotechnology Ag Antigen arrays for treatment of bone disease
US7094409B2 (en) * 2001-01-19 2006-08-22 Cytos Biotechnology Ag Antigen arrays for treatment of allergic eosinophilic diseases
US7264810B2 (en) 2001-01-19 2007-09-04 Cytos Biotechnology Ag Molecular antigen array
US20050003474A1 (en) * 2001-01-26 2005-01-06 Desouza Mervyn L. Carotenoid biosynthesis
US20040078846A1 (en) * 2002-01-25 2004-04-22 Desouza Mervyn L. Carotenoid biosynthesis
WO2002059337A1 (en) * 2001-01-26 2002-08-01 Georgetown University School Of Medicine Anti-apoptopic gene scc-s2 and diagnostic and therapeutic uses thereof
US20020108132A1 (en) * 2001-02-02 2002-08-08 Avigenics Inc. Production of a monoclonal antibody by a transgenic chicken
WO2002064612A2 (en) 2001-02-09 2002-08-22 Human Genome Sciences, Inc. Human g-protein chemokine receptor (ccr5) hdgnr10
JP4955905B2 (en) 2001-02-20 2012-06-20 イントレキソン コーポレーション Chimeric retinoid X receptors and their use in novel ecdysone receptor-based inducible gene expression systems
CA2445796C (en) 2001-02-20 2014-09-16 Rheogene, Inc. Novel substitution mutant receptors and their use in a nuclear receptor-based inducible gene expression system
US7094892B2 (en) * 2001-02-22 2006-08-22 The Research Foundation Of State University Of New York Nucleic acids encoding opiate receptors
WO2002070665A2 (en) 2001-03-02 2002-09-12 The Rockefeller University Recombinant hybrid allergen constructs with reduced allergenicity that retain immunogenicity of the natural allergen
US6924358B2 (en) 2001-03-05 2005-08-02 Agensys, Inc. 121P1F1: a tissue specific protein highly expressed in various cancers
CA2440221C (en) 2001-03-07 2013-02-05 Merck Patent Gesellschaft Mit Beschraenkter Haftung Expression technology for proteins containing a hybrid isotype antibody moiety
AU2002254174A1 (en) * 2001-03-13 2002-09-24 Vical Incorporated Interferon-beta polynucleotide therapy for autoimmune and inflammatory diseases
EP1367947B1 (en) 2001-03-14 2008-01-16 Boston Scientific Scimed, Inc. Apparatus for treatment of atrial fibrillation
US7271240B2 (en) 2001-03-14 2007-09-18 Agensys, Inc. 125P5C8: a tissue specific protein highly expressed in various cancers
WO2002074345A2 (en) * 2001-03-16 2002-09-26 Johns Hopkins University School Of Medicine Immune modulation by transduced hematopoietic stem cells expressing antigens and antigen-presenting cell regulatory molecules
WO2002077012A2 (en) * 2001-03-23 2002-10-03 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Human papilloma virus immunoreative peptides
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
US20030096414A1 (en) * 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
US20040028651A1 (en) * 2001-03-29 2004-02-12 Karrupiah Muthumani Composition and methods of using hiv vpr
US7541512B2 (en) * 2001-03-30 2009-06-02 Synageva Biopharma Corp. Avians containing a lysozyme promoter transgene
EP1478751A4 (en) * 2001-03-30 2005-10-19 Avigenics Inc Avian lysozyme promoter
US7176300B2 (en) * 2001-03-30 2007-02-13 Avigenics, Inc. Avian lysozyme promoter
WO2002079415A2 (en) 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
WO2002081642A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
AU2002305151A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
US8034791B2 (en) 2001-04-06 2011-10-11 The University Of Chicago Activation of Egr-1 promoter by DNA damaging chemotherapeutics
AU2002303261A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
US20040242523A1 (en) * 2003-03-06 2004-12-02 Ana-Farber Cancer Institue And The Univiersity Of Chicago Chemo-inducible cancer gene therapy
AU2002303262A1 (en) * 2001-04-06 2002-10-21 Georgetown University Gene shinc-1 and diagnostic and therapeutic uses thereof
ATE541937T1 (en) * 2001-04-06 2012-02-15 Univ Chicago CHEMOTHERAPEUTIC INTRODUCTION OF EGR-1 PROMOTER ACTIVITY IN GENE THERAPY
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20030191073A1 (en) 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
US20070128229A1 (en) * 2002-04-12 2007-06-07 Wyeth Surface proteins of Streptococcus pyogenes
CA2443493A1 (en) * 2001-04-13 2002-10-24 Wyeth Surface proteins of streptococcus pyogenes
CA2444632A1 (en) 2001-04-13 2002-10-24 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
EP1572868A4 (en) 2001-04-16 2007-04-04 Wyeth Corp Novel streptococcus pneumoniae open reading frames encoding polypeptide antigens and uses thereof
US20030077829A1 (en) * 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
BR0209177A (en) 2001-05-03 2004-10-05 Merck Patent Gmbh Recombinant tumor specific antibody and use
WO2002095002A2 (en) 2001-05-22 2002-11-28 University Of Chicago N4 virion single-stranded dna dependent rna polymerase
KR20030022094A (en) * 2001-05-24 2003-03-15 (주)휴먼디엔에이테크놀로지 Novel keratinocyte growth factor-2 analogue in hair follicle
AU2002329176A1 (en) * 2001-05-25 2002-12-23 The Trustees Of The University Of Pennsylvania Targeted particles and methods of using the same
EP2305699B1 (en) * 2001-06-05 2014-08-13 CureVac GmbH Stabilised mRNA with increased G/C content which is optimised for translation in its coded areas for the vaccination against sleeping sickness, leishmaniosis and toxoplasmosis
CA2452015C (en) 2001-07-05 2012-07-03 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
US20030198621A1 (en) 2001-07-05 2003-10-23 Megede Jan Zur Polynucleotides encoding antigenic HIV type B and/or type C polypeptides, polypeptides and uses thereof
CA2452593C (en) * 2001-07-12 2015-02-03 Arexis Ab Triple polypeptide complexes
WO2003015716A2 (en) * 2001-08-13 2003-02-27 Ige Therapeutics, Inc. Immunoglobulin e vaccines and methods of use thereof
US20030185892A1 (en) * 2001-08-17 2003-10-02 Bell Steve J. D. Intraocular delivery compositions and methods
RU2335295C2 (en) * 2001-08-20 2008-10-10 Юниверсити Оф Коннектикут Хелт Сентер Method of obtaining compositions including heat shock proteins or alpha-2-macroglobulin, suitable for treatment of malignant tumour and infectious disease
EP2287186B1 (en) 2001-09-06 2014-12-31 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
US6800462B2 (en) * 2001-09-10 2004-10-05 Abgenomics Corporation Production of recombinant proteins in vivo and use for generating antibodies
US20030049694A1 (en) * 2001-09-10 2003-03-13 Chung-Hsiun Wu Production of fusion proteins and use for identifying binding molecules
US7550650B2 (en) * 2001-09-18 2009-06-23 Synageva Biopharma Corp. Production of a transgenic avian by cytoplasmic injection
US7108975B2 (en) * 2001-09-21 2006-09-19 Regents Of The University Of Michigan Atlastin
US7582425B2 (en) * 2001-09-21 2009-09-01 The Regents Of The University Of Michigan Atlastin
US20040023910A1 (en) * 2001-09-28 2004-02-05 Zhiming Zhang Use of cyr61 in the treatment and diagnosis of human uterine leiomyomas
AU2002362753B9 (en) * 2001-10-05 2008-05-15 The Trustees Of The University Of Pennsylvania Compositions for and methods of treating and preventing sirs/sepsis
US7115266B2 (en) 2001-10-05 2006-10-03 Cytos Biotechnology Ag Angiotensin peptide-carrier conjugates and uses thereof
US20030134811A1 (en) * 2001-10-09 2003-07-17 John Jackson Methods and compositions comprising hydroxyapatite useful for the administration of therapeutic agents
MX339524B (en) 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
US20040166490A1 (en) * 2002-12-17 2004-08-26 Morris David W. Novel therapeutic targets in cancer
US20040126762A1 (en) * 2002-12-17 2004-07-01 Morris David W. Novel compositions and methods in cancer
AU2002348417B9 (en) * 2001-10-26 2010-02-04 Baylor College Of Medicine A composition and method to alter lean body mass and bone properties in a subject
US20030125292A1 (en) * 2001-11-07 2003-07-03 Sean Semple Mucoscal vaccine and methods for using the same
DE60238864D1 (en) * 2001-11-07 2011-02-17 Mankind Corp FOR EPITOPES OF ANTIGENIC ENCODING EXPRESSION VECTORS AND METHOD FOR THEIR DESIGN
EP1487854B1 (en) 2001-11-09 2010-04-28 Georgetown University Novel isoform of vascular endothelial cell growth inhibitor (vegi)
EP2301552A1 (en) 2001-11-21 2011-03-30 The Board Of Trustees Of The Leland Stanford Junior University Polynucleotide therapy
US20030113784A1 (en) * 2001-11-29 2003-06-19 Jaime Flores-Riveros Regulated expression of recombinant DNA
US20040197778A1 (en) * 2002-12-26 2004-10-07 Sagres Discovery, Inc. Novel compositions and methods in cancer
US7335761B2 (en) * 2001-11-30 2008-02-26 Avigenics, Inc. Avian gene expression controlling regions
US20100333219A1 (en) * 2001-11-30 2010-12-30 Synageva Biopharma Corp. Methods of protein production using ovomucoid regulatory regions
US6875588B2 (en) 2001-11-30 2005-04-05 Avigenics, Inc. Ovomucoid promoter and methods of use
US20060040262A1 (en) * 2002-12-27 2006-02-23 Morris David W Novel compositions and methods in cancer
US7294507B2 (en) * 2001-11-30 2007-11-13 Avigenics, Inc. Ovomucoid promoters and methods of use
US20040180344A1 (en) * 2003-03-14 2004-09-16 Morris David W. Novel therapeutic targets in cancer
AU2002351332A1 (en) * 2001-12-07 2003-06-23 Chiron Corporation Endogenous retrovirus polypeptides linked to oncogenic transformation
BR0214869A (en) 2001-12-11 2005-03-08 Advisys Inc Plasmid-mediated supplementation for treatment of chronically ill individuals
NZ546711A (en) 2001-12-12 2008-06-30 Chiron Srl Immunisation against chlamydia trachomatis
US20030181409A1 (en) * 2001-12-14 2003-09-25 The Regents Of The University Of California Methods of inhibiting fertility
ES2364636T3 (en) 2001-12-19 2011-09-08 Novartis Ag PULMONARY ADMINISTRATION OF AMINOGLUCOSIDS.
DE10162480A1 (en) 2001-12-19 2003-08-07 Ingmar Hoerr The application of mRNA for use as a therapeutic agent against tumor diseases
US7049121B2 (en) * 2001-12-20 2006-05-23 Applied Molecular Evolution Butyrylcholinesterase variant polypeptides with increased catalytic efficiency and methods of use
US6989261B2 (en) * 2001-12-20 2006-01-24 Eli Lilly And Company Butyrylcholinesterase variant polypeptides with increased catalytic efficiency and methods of use
EP2277888A3 (en) 2001-12-21 2011-04-27 Human Genome Sciences, Inc. Fusion proteins of albumin and erythropoietin
US6867004B2 (en) * 2001-12-21 2005-03-15 Focus Technologies, Inc. Methods of using oxidized fungal antigens in antibody testing
US20030134423A1 (en) * 2002-01-04 2003-07-17 Chu Yong Liang Compounds for delivering substances into cells
CA2472282A1 (en) * 2002-01-08 2003-07-17 Chiron Corporation Gene products differentially expressed in cancerous breast cells and their methods of use
US20030219459A1 (en) * 2002-01-18 2003-11-27 Cytos Biotechnology Ag Prion protein carrier-conjugates
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
CA2475003A1 (en) 2002-02-01 2003-08-07 Sequitur, Inc. Double-stranded oligonucleotides
WO2003068190A1 (en) * 2002-02-13 2003-08-21 Northeastern University Intracellular delivery of therapeutic agents
WO2003070905A2 (en) * 2002-02-15 2003-08-28 Zycos, Inc. Electroporation methods for introducing bioactive agents into cells
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
US7245963B2 (en) * 2002-03-07 2007-07-17 Advisys, Inc. Electrode assembly for constant-current electroporation and use
US8209006B2 (en) * 2002-03-07 2012-06-26 Vgx Pharmaceuticals, Inc. Constant current electroporation device and methods of use
CA2478822C (en) 2002-03-11 2016-07-12 Altea Therapeutics Corporation Transdermal drug delivery patch system, method of making same and method of using same
US9918665B2 (en) 2002-03-11 2018-03-20 Nitto Denko Corporation Transdermal porator and patch system and method for using same
US8116860B2 (en) 2002-03-11 2012-02-14 Altea Therapeutics Corporation Transdermal porator and patch system and method for using same
CN1646160A (en) 2002-03-13 2005-07-27 拜奥根Idec马萨诸塞公司 Anti-alpha V beta 6 antibodies
US7135562B2 (en) 2002-03-14 2006-11-14 University Of Cincinnati Avian iFABP gene expression controlling region
KR100981471B1 (en) 2002-03-15 2010-09-10 더 큐레이터스 오브 더 유니버시티 오브 미주리 Mutants of the p4 protein of nontypable haemophilus influenzae with reduced enzymatic activity
US20040241723A1 (en) * 2002-03-18 2004-12-02 Marquess Foley Leigh Shaw Systems and methods for improving protein and milk production of dairy herds
KR100562824B1 (en) * 2002-03-20 2006-03-23 주식회사 바이로메드 Hybrid hepatocyte growth factor gene which has a high expression efficiency and expresses two heterotypes of hepatocyte growth factor
JP2005520543A (en) 2002-03-21 2005-07-14 サイグレス ディスカバリー, インコーポレイテッド Novel compositions and methods in cancer
US20040009182A1 (en) * 2002-04-01 2004-01-15 Myers Robert C. Method and compositions using anthrax immune globulin to provide passive immunity against lethal infections from bacillus anthracis
US20040038303A1 (en) * 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
DE60315628T2 (en) 2002-04-09 2008-06-05 Sanofi Pasteur Ltd., Toronto MODIFIED CEA NUCLEIC ACID AND EXPRESSION VECTORS
US7138512B2 (en) * 2002-04-10 2006-11-21 Georgetown University Gene SHINC-2 and diagnostic and therapeutic uses thereof
US7244565B2 (en) * 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
US6984389B2 (en) 2002-04-25 2006-01-10 University Of Connecticut Health Center Using heat shock proteins to improve the therapeutic benefit of a non-vaccine treatment modality
IL164799A0 (en) 2002-04-25 2005-12-18 Univ Connecticut Using heat shock proteins to improve the therapeutic benefit of a non-vaccine treatment modality
US20040180438A1 (en) * 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
US7354908B2 (en) 2002-04-30 2008-04-08 University Of South Florida Materials and methods for prevention and treatment of RNA viral diseases
AU2003301841A1 (en) * 2002-05-01 2004-06-07 National Institutes Of Health Immunotherapy regimens in hiv-infected patients
US20040126400A1 (en) * 2002-05-03 2004-07-01 Iversen Patrick L. Delivery of therapeutic compounds via microparticles or microbubbles
EP1549352A4 (en) 2002-05-06 2005-07-27 Nucleonics Inc Methods for delivery of nucleic acids
US20040013649A1 (en) * 2002-05-10 2004-01-22 Inex Pharmaceuticals Corporation Cancer vaccines and methods of using the same
US20040009944A1 (en) * 2002-05-10 2004-01-15 Inex Pharmaceuticals Corporation Methylated immunostimulatory oligonucleotides and methods of using the same
US20030228691A1 (en) * 2002-05-17 2003-12-11 Lewis David L. Processes for inhibiting gene expression using polynucleotides
CA2486464A1 (en) 2002-05-21 2003-11-27 Resistentia Pharmaceuticals Ab Chimeric ige polypeptides and host cells
US20030228317A1 (en) * 2002-05-22 2003-12-11 Prafulla Gokhale Gene BRCC-1 and diagnostic and therapeutic uses thereof
JP4563171B2 (en) 2002-05-24 2010-10-13 シェーリング コーポレイション Neutralizing human anti-IGFR antibody
US20040033601A1 (en) * 2002-05-30 2004-02-19 Davidson Eric H. Gene regulatory networks and methods of interdiction for controlling the differentiation state of a cell
CN100418981C (en) 2002-06-10 2008-09-17 瓦西尼斯公司 Gene differentially expressed in breast and bladder cancer and encoded polypeptides
CA2489292A1 (en) * 2002-06-13 2003-12-24 Chiron Corporation Vectors for expression of hml-2 polypeptides
DE60335186D1 (en) * 2002-06-20 2011-01-13 Cytos Biotechnology Ag PACKAGED VIRUS PARTICLES IN COMBINATION WITH CPG FOR USE AS ADJUVANTIES WITH ALLERGENS. MANUFACTURING METHOD AND USE
US8029803B2 (en) * 2002-06-20 2011-10-04 Paladin Labs, Inc. Chimeric antigens for eliciting an immune response
AU2003243663A1 (en) 2002-06-20 2004-01-06 The Trustees Of The University Of Pennsylvania Vaccines for suppressing ige-mediated allergic disease and methods for using the same
US8025873B2 (en) 2002-06-20 2011-09-27 Paladin Labs, Inc. Chimeric antigens for eliciting an immune response
US7527966B2 (en) 2002-06-26 2009-05-05 Transgenrx, Inc. Gene regulation in transgenic animals using a transposon-based vector
EP1539245A2 (en) * 2002-06-26 2005-06-15 The Penn State Research Foundation Methods and materials for treating human papillomavirus infections
US20040235011A1 (en) * 2002-06-26 2004-11-25 Cooper Richard K. Production of multimeric proteins
US20040172667A1 (en) 2002-06-26 2004-09-02 Cooper Richard K. Administration of transposon-based vectors to reproductive organs
ES2354607T3 (en) 2002-06-28 2011-03-16 Protiva Biotherapeutics Inc. PROCEDURE AND APPLIANCE TO PRODUCE LIPOSOMES.
SI1546357T1 (en) * 2002-06-28 2013-10-30 Iowa State University Research Foundation, Inc. Immunogenic mycoplasma hyopneumoniae polypeptides
US7375093B2 (en) 2002-07-05 2008-05-20 Intrexon Corporation Ketone ligands for modulating the expression of exogenous genes via an ecdysone receptor complex
US6916474B2 (en) 2002-07-15 2005-07-12 Board Of Regents, The University Of Texas System Antibodies with increased affinities for anthrax antigens
JP4726483B2 (en) 2002-07-17 2011-07-20 サイトス バイオテクノロジー アーゲー Molecular antigen array
KR20120041739A (en) * 2002-07-18 2012-05-02 사이토스 바이오테크놀로지 아게 Hapten-carrier conjugates and uses thereof
DE60336902D1 (en) 2002-07-19 2011-06-09 Cytos Biotechnology Ag VACCINE COMPOSITIONS CONTAINING AMYLOID BETA 1-6 ANTIGENARRAYS
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
WO2004011060A2 (en) * 2002-07-26 2004-02-05 Mirus Corporation Delivery of molecules and complexes to mammalian cells in vivo
WO2004011624A2 (en) * 2002-07-31 2004-02-05 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
US20080176812A1 (en) * 2002-08-05 2008-07-24 Davidson Beverly L Allele-specific silencing of disease genes
US20040241854A1 (en) 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
US20050106731A1 (en) * 2002-08-05 2005-05-19 Davidson Beverly L. siRNA-mediated gene silencing with viral vectors
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
US20040023390A1 (en) * 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
US20050255086A1 (en) * 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US20050042646A1 (en) * 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
KR101170653B1 (en) 2002-08-12 2012-08-03 제네렉스, 인코포레이티드 Methods and compositions concerning poxviruses and cancer
US20040081653A1 (en) 2002-08-16 2004-04-29 Raitano Arthur B. Nucleic acids and corresponding proteins entitled 251P5G2 useful in treatment and detection of cancer
US20060251620A1 (en) * 2002-08-22 2006-11-09 Lidia Ivanova Inducible alphaviral/orip based gene expression system
US7785608B2 (en) * 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
US7595303B1 (en) * 2002-09-05 2009-09-29 University Of South Florida Genetic adjuvants for immunotherapy
WO2004022094A1 (en) * 2002-09-05 2004-03-18 Resistentia Pharmaceuticals Ab Allergy vaccines
US20050196382A1 (en) * 2002-09-13 2005-09-08 Replicor, Inc. Antiviral oligonucleotides targeting viral families
CN1694959B (en) * 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
CA2503905A1 (en) * 2002-09-16 2004-03-25 Plexxikon, Inc. Crystal structure of pim-1 kinase
US7741437B2 (en) 2002-09-19 2010-06-22 The United States Of America As Represented By The Department Of Health And Human Services P. ariasi polypeptides, p. perniciosus polypeptides and methods of use
US7074598B2 (en) * 2002-09-25 2006-07-11 Mayo Foundation For Medical Education And Research Detection of vancomycin-resistant enterococcus spp.
SI1546088T1 (en) 2002-10-03 2015-04-30 Novaremed Ltd. Compounds for use in the treatment of autoimmune diseases, immuno-allergical diseases and organ or tissue transplantation rejection
US20060079453A1 (en) 2002-10-03 2006-04-13 John Sidney Hla binding peptides and their uses
WO2004032713A2 (en) * 2002-10-04 2004-04-22 Nanomatrix, Inc. Sealants for skin and other tissues
US20060147450A1 (en) * 2002-10-04 2006-07-06 Shelton David L Methods for treating cardiac arrhythmia and preventing death due to cardiac arrhythmia using ngf antagonists
UA80447C2 (en) 2002-10-08 2007-09-25 Methods for treating pain by administering nerve growth factor antagonist and opioid analgesic
RU2338555C2 (en) * 2002-10-08 2008-11-20 Ринат Ньюросайенс Корп. Method of treatment of postoperative pain by administering of antagonist of factor of growth of nerves and compositions containing factor of growth of nerves
AU2003304238A1 (en) 2002-10-08 2005-01-13 Rinat Neuroscience Corp. Methods for treating post-surgical pain by administering an anti-nerve growth factor antagonist antibody and compositions containing the same
US6863731B2 (en) * 2002-10-18 2005-03-08 Controls Corporation Of America System for deposition of inert barrier coating to increase corrosion resistance
EP1552025A4 (en) * 2002-10-18 2006-12-13 Cylene Pharmaceuticals Inc Processes for identifying quadruplex-targeted antiviral molecules
WO2004037175A2 (en) * 2002-10-21 2004-05-06 Mgi Pharma Biologics, Inc. Compositions and methods for treating human papillomavirus-mediated disease
AU2003287267A1 (en) 2002-10-29 2004-05-25 Centro De Pesquisas Goncalo Moniz Lutzomyia longipalpis polypeptides and methods of use
EP2279746B1 (en) 2002-11-15 2013-10-02 Novartis Vaccines and Diagnostics S.r.l. Surface proteins in neisseria meningitidis
US7811813B2 (en) 2002-11-21 2010-10-12 The Board Of Trustees Of The Leland Stanford Junior University Methods and immune modulatory nucleic acid compositions for preventing and treating disease
JP2006508163A (en) 2002-11-27 2006-03-09 アジェンシス, インコーポレイテッド Nucleic acids and corresponding proteins referred to as 24P4C12 useful in the treatment and detection of cancer
MXPA05005909A (en) * 2002-12-02 2005-08-26 Resistentia Pharmaceuticals Ab Methods and materials for treating inflammatory conditions using a polypeptide comprising a self-c5 amino acid segment and a non-self amino acid segment.
CA2507626A1 (en) * 2002-12-04 2004-06-17 Applied Molecular Evolution Inc. Butyrylcholinesterase variants that alter the activity of chemotherapeutic agents
US7354593B2 (en) 2002-12-09 2008-04-08 Merial Limited Coccidial vaccine and methods of making and using same
EP1903056A3 (en) 2002-12-10 2008-05-07 Idm Pharma, Inc. HLA-A1, -A2 -A3, -A24, -B7, and -B44 binding peptides comprising tumor associated antigen epitopes, and compositions thereof
ES2346205T3 (en) 2002-12-17 2010-10-13 Merck Patent Gmbh HUMANIZED ANTIBODY (H14.18) OF ANTIBODY 14.18 OF MOUSE THAT LINKS TO GD2 AND ITS FUSION WITH IL-2.
PT2311848E (en) 2002-12-23 2013-10-03 Vical Inc Codon-optimized polynucleotide-based vaccines against human cytomegalovirus infection
JP4917263B2 (en) * 2002-12-23 2012-04-18 バイカル インコーポレイテッド Method for lyophilizing nucleic acid / block copolymer / cationic surfactant complex
US20050089521A1 (en) * 2002-12-23 2005-04-28 Shelton David L. Methods for treating taxol-induced sensory neuropathy
EP1581201A4 (en) * 2002-12-23 2011-03-30 Vical Inc Method for producing sterile polynucleotide based medicaments
US7569364B2 (en) 2002-12-24 2009-08-04 Pfizer Inc. Anti-NGF antibodies and methods using same
US9498530B2 (en) 2002-12-24 2016-11-22 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
SI1575517T1 (en) * 2002-12-24 2012-06-29 Rinat Neuroscience Corp Anti-ngf antibodies and methods using same
ATE457716T1 (en) 2002-12-30 2010-03-15 Angiotech Int Ag RELEASE OF ACTIVE INGREDIENTS FROM QUICK-GELLING POLYMER COMPOSITION
AU2004206250B8 (en) 2003-01-21 2009-09-17 Bristol-Myers Squibb Company Polynucleotide encoding a novel acyl coenzyme a, monoacylglycerol acyltransferase-3 (MGAT3), and uses thereof
CN1980571A (en) * 2003-01-24 2007-06-13 阿维季尼克斯股份有限公司 Exogenous proteins expressed in avians and their eggs
US20050048573A1 (en) * 2003-02-03 2005-03-03 Plexxikon, Inc. PDE5A crystal structure and uses
US7304161B2 (en) 2003-02-10 2007-12-04 Intrexon Corporation Diaclhydrazine ligands for modulating the expression of exogenous genes in mammalian systems via an ecdysone receptor complex
EP2343315A3 (en) 2003-02-10 2011-11-23 Agensys, Inc. Nucleic acid and corresponding protein named 158P1D7 useful in the treatment and detection of bladder and other cancers
US20070218071A1 (en) * 2003-09-15 2007-09-20 Morris David W Novel therapeutic targets in cancer
US20040170982A1 (en) * 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
US20070149449A1 (en) 2003-02-14 2007-06-28 Morris David W Therapeutic targets in cancer
US7767387B2 (en) * 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
US20040248798A1 (en) 2003-02-14 2004-12-09 Peter Sutovsky Contraceptive methods and compositions related to proteasomal interference
PL379983A1 (en) 2003-02-19 2006-11-27 Rinat Neuroscience Corp. Methods for treating pain by administering a nerve growth factor antagonist and an nsaid and compositions containing the same
RU2324493C2 (en) * 2003-02-20 2008-05-20 Юниверсити Оф Коннектикут Хелт Сентер Method of application of compositions containing heat-shock proteins or alpha-2-macroglobulin for treatment of cancer and infectious diseases
CA2516425A1 (en) * 2003-02-21 2005-02-24 The Penn State Research Foundation Rna interference compositions and methods
ES2531204T3 (en) * 2003-02-25 2015-03-11 Vaccibody As Modified antibody
US7456315B2 (en) 2003-02-28 2008-11-25 Intrexon Corporation Bioavailable diacylhydrazine ligands for modulating the expression of exogenous genes via an ecdysone receptor complex
US20050170431A1 (en) * 2003-02-28 2005-08-04 Plexxikon, Inc. PYK2 crystal structure and uses
US7262027B2 (en) * 2003-03-14 2007-08-28 Medical College Of Ohio Polypeptide and DNA immunization against Coccidioides spp. infections
EP2390352A1 (en) 2003-03-18 2011-11-30 Quantum Genetics Ireland Limited Systems and methods for improving protein and milk production of dairy herds
EP1620127A4 (en) * 2003-03-20 2007-04-04 Rinat Neuroscience Corp Methods for treating taxol-induced gut disorder
MXPA05008526A (en) * 2003-03-26 2005-10-20 Wyeth Corp Immunogenic composition and methods.
US7507727B2 (en) * 2003-04-07 2009-03-24 Cylene Pharmaceuticals, Inc. Substituted quinobenzoxazine analogs and methods of using thereof
US7354916B2 (en) * 2003-04-07 2008-04-08 Cylene Pharmaceuticals Substituted quinobenzoxazine analogs
JP2006522827A (en) * 2003-04-07 2006-10-05 サイリーン ファーマシューティカルズ インコーポレーティッド Substituted quinobenzoxazine analogs
EP1610795A4 (en) * 2003-04-07 2007-10-24 Cylene Pharmaceuticals Inc Heterocyclic substituted 1,4-dihydro-4-oxo-1,8-naphthpyridine analogs
JP4789208B2 (en) 2003-04-09 2011-10-12 バイオデリバリー サイエンシーズ インターナショナル インコーポレイティッド Swirl composition for protein expression
US20050013854A1 (en) 2003-04-09 2005-01-20 Mannino Raphael J. Novel encochleation methods, cochleates and methods of use
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
CA2522751A1 (en) * 2003-04-16 2004-11-04 Wyeth Holdings Corporation Novel immunogenic compositions for the prevention and treatment of meningococcal disease
ES2456666T3 (en) 2003-04-18 2014-04-23 Biotech Synergy, Inc. HLA-A2 antigen peptides associated with a tumor and compositions
TW200424214A (en) * 2003-04-21 2004-11-16 Advisys Inc Plasmid mediated GHRH supplementation for renal failures
EP2236619A3 (en) 2003-04-25 2011-12-14 Dana-Farber Cancer Institute, Inc. BCL2L12 polypeptide activators and inhibitors
US20070037151A1 (en) * 2003-04-28 2007-02-15 Babe Lilia M Cd4+ human papillomavirus (hpv) epitopes
WO2004098634A2 (en) * 2003-04-30 2004-11-18 Government Of The United States Of America As Represented By The Sercretary Of The Department Of Health And Human Services National Institutes Of Health Protein arginine n-methyltransferase 2 (prmt-2)
US7468273B2 (en) 2003-05-01 2008-12-23 Meial Limited Canine GHRH gene, polypeptides and methods of use
US20050256042A1 (en) * 2003-05-09 2005-11-17 Jeffers Michael E Methods of preventing and treating alimentary mucositis
WO2004102198A2 (en) * 2003-05-15 2004-11-25 Cytos Biotechnology Ag Selection of b cells with specificity of interest: method of preparation and use
EP2319524B1 (en) 2003-05-30 2013-08-21 Agensys, Inc. Prostate stem cell antigen (PSCA) variants and subsequences thereof
AU2004245074A1 (en) * 2003-06-04 2004-12-16 Canji, Inc Methods and compositions for interferon therapy
JP4980716B2 (en) 2003-06-12 2012-07-18 アルナイラム ファーマシューティカルズ, インコーポレイテッド Conserved HBV and HCV sequences useful for gene silencing
NZ570709A (en) * 2003-06-13 2010-04-30 Univ Pennsylvania Nucleic acid sequences encoding and compositions comprising IgE signal peptide and/or IL-15 and methods for using the same
US8008265B2 (en) 2003-06-13 2011-08-30 The Trustees Of The University Of Pennsylvania Vaccines, immunotherapeutics and methods for using the same
US8183011B2 (en) 2003-06-13 2012-05-22 University Of Medicine And Dentistry Of New Jersey RNA interferases and methods of use thereof
WO2004112825A2 (en) * 2003-06-17 2004-12-29 Mannkind Corporation Combinations of tumor-associated antigens for the treatment of various types of cancers
ATE485303T1 (en) * 2003-06-20 2010-11-15 Mayo Foundation ISOFORMS OF BRAIN NATRIURETIC PEPTIDE
US7261882B2 (en) 2003-06-23 2007-08-28 Reagents Of The University Of Colorado Methods for treating neuropathic pain by administering IL-10 polypeptides
US20050079548A1 (en) * 2003-07-07 2005-04-14 Plexxikon, Inc. Ligand development using PDE4B crystal structures
US20070224615A1 (en) * 2003-07-09 2007-09-27 Invitrogen Corporation Methods for assaying protein-protein interactions
CN1894581B (en) 2003-07-09 2012-02-01 生命技术公司 Method for assaying protein-protein interaction
EP1644508A1 (en) * 2003-07-11 2006-04-12 Cytos Biotechnology AG Gene expression system
CN101291653B (en) * 2003-07-16 2012-06-27 普洛体维生物治疗公司 Lipid encapsulated interfering rna
US20060269531A1 (en) * 2003-07-18 2006-11-30 Eastern Virginia Medical School Apparatus for generating electrical pulses and methods of using the same
US7361357B2 (en) 2003-07-29 2008-04-22 Pfizer Inc. Safe mutant viral vaccines
US7663017B2 (en) 2003-07-30 2010-02-16 Institut Pasteur Transgenic mice having a human major histocompatability complex (MHC) phenotype, experimental uses and applications
WO2005012498A2 (en) * 2003-08-01 2005-02-10 Regents Of The University Of Minnesota Production of porphyrins
US8007805B2 (en) * 2003-08-08 2011-08-30 Paladin Labs, Inc. Chimeric antigens for breaking host tolerance to foreign antigens
US20060035242A1 (en) 2004-08-13 2006-02-16 Michelitsch Melissa D Prion-specific peptide reagents
WO2005040388A2 (en) * 2003-08-22 2005-05-06 Nucleonics Inc. Eukariotic expression systems for expression of inhibitory rna in multiple intracellular compartments
AU2004271951B2 (en) * 2003-09-05 2008-08-21 Genencor International, Inc. HPV CD8+ T-cell epitopes
CA2551022C (en) * 2003-09-15 2013-06-04 Protiva Biotherapeutics, Inc. Polyethyleneglycol-modified lipid compounds and uses thereof
JP4773352B2 (en) 2003-09-17 2011-09-14 デューク ユニバーシティ Consensus / ancestral immunogen
US20070281896A1 (en) * 2003-09-30 2007-12-06 Morris David W Novel compositions and methods in cancer
US8562970B2 (en) * 2003-10-08 2013-10-22 Sanofi Pasteur Limited Modified CEA/B7 vector
WO2005034979A2 (en) * 2003-10-11 2005-04-21 Inex Pharmaceuticals Corporation Methods and compositions for enhancing innate immunity and antibody dependent cellular cytotoxicity
DE10347710B4 (en) 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Recombinant vaccines and their use
RU2401843C2 (en) 2003-10-16 2010-10-20 Микромет Аг Multispecific deimmunising cd3-binders
US8016811B2 (en) 2003-10-24 2011-09-13 Altea Therapeutics Corporation Method for transdermal delivery of permeant substances
WO2005049794A2 (en) 2003-11-13 2005-06-02 University Of Georgia Research Foundation, Inc. Methods of characterizing infectious bursal disease virus
US20050277127A1 (en) * 2003-11-26 2005-12-15 Epitomics, Inc. High-throughput method of DNA immunogen preparation and immunization
US20050287118A1 (en) * 2003-11-26 2005-12-29 Epitomics, Inc. Bacterial plasmid with immunological adjuvant function and uses thereof
JP2005173484A (en) * 2003-12-15 2005-06-30 Canon Inc Image forming apparatus and process cartridge
US20070066641A1 (en) * 2003-12-19 2007-03-22 Prabha Ibrahim Compounds and methods for development of RET modulators
PL1696920T3 (en) * 2003-12-19 2015-03-31 Plexxikon Inc Compounds and methods for development of ret modulators
KR20070001932A (en) 2003-12-23 2007-01-04 리나트 뉴로사이언스 코퍼레이션 Agonist anti-trkc antibodies and methods using same
US8071364B2 (en) 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
WO2005063279A1 (en) * 2003-12-29 2005-07-14 Universitätsklinikum Münster Means for stimulation and activation of hair growth by il-15
WO2005065719A1 (en) * 2004-01-12 2005-07-21 Genesense Technologies Inc. Antisense oligonucleotides directed to ribonucleotide reductase r2 and uses thereof in combination therapies for the treatment of cancer
US20060205070A1 (en) * 2004-01-13 2006-09-14 The Government Of The Usa, As Represented By The Secretary, Department Of Health And Human Services HIV TEV compositions and methods of use
AU2005205181A1 (en) * 2004-01-20 2005-07-28 Cytos Biotechnology Ag Particle-induced ghrelin immune response
US7432057B2 (en) * 2004-01-30 2008-10-07 Michigan State University Genetic test for PSE-susceptible turkeys
DE602005021756D1 (en) * 2004-02-13 2010-07-22 Nod Pharmaceuticals Inc PARTICLES WITH CORE OF CALCIUM PHOSPHATE NANOPARTICLES, BIOMOLECOL AND GALLENIC ACID, METHOD OF MANUFACTURE AND THERAPEUTIC USE
US20050181035A1 (en) * 2004-02-17 2005-08-18 Dow Steven W. Systemic immune activation method using non CpG nucleic acids
CA2556911C (en) 2004-02-19 2013-07-30 The Governors Of The University Of Alberta Leptin promoter polymorphisms and uses thereof
US20050209148A1 (en) * 2004-02-20 2005-09-22 Arnon Rosenthal Methods of treating obesity or diabetes using NT-4/5
DE602005021873D1 (en) 2004-03-12 2010-07-29 Univ Georgia NEW VACCINE ADJUVANT AND ITS MANUFACTURE AND USE
CA2559586A1 (en) * 2004-03-18 2005-09-29 Fred Hutchinson Cancer Research Center Methods and compositions involving s-ship promoter regions
US7504243B2 (en) 2004-03-19 2009-03-17 The United States Of America As Represented By The Department Of Health And Human Services Methods for the production of biliverdin
CN104292321A (en) 2004-03-29 2015-01-21 株式会社嘉尔药物 Novel modified galectin 9 protein and use thereof
EP1737294B1 (en) 2004-04-03 2020-12-02 Boehringer Ingelheim Animal Health USA Inc. Method and apparatus for automatic jet injection of bird eggs
JP5301152B2 (en) * 2004-04-07 2013-09-25 ライナット ニューロサイエンス コーポレイション Method for treating bone cancer pain by administering a nerve growth factor antagonist
GB0408164D0 (en) * 2004-04-13 2004-05-19 Immune Targeting Systems Ltd Antigen delivery vectors and constructs
GB0716992D0 (en) 2007-08-31 2007-10-10 Immune Targeting Systems Its L Influenza antigen delivery vectors and constructs
US20050260652A1 (en) * 2004-04-15 2005-11-24 The General Hospital Corporation Compositions and methods that modulate RNA interference
US7429459B2 (en) 2004-04-20 2008-09-30 Galapagos N.V. Methods for identifying a compound that inhibits the procession of amyloid-beta protein production in a mammalian cell expressing APP and overexpressing a G-protein coupled receptor in the cell
ES2427177T3 (en) 2004-04-27 2013-10-29 Galapagos N.V. Methods, agents and tests for the detection of compounds to induce differentiation of undifferentiated mammalian cells to give rise to osteoblasts
WO2005105136A1 (en) * 2004-04-27 2005-11-10 University Of South Florida Nanogene therapy for cell proliferation disorders
US7935510B2 (en) 2004-04-30 2011-05-03 Intrexon Corporation Mutant receptors and their use in a nuclear receptor-based inducible gene expression system
US7303881B2 (en) * 2004-04-30 2007-12-04 Pds Biotechnology Corporation Antigen delivery compositions and methods of use
EP1742627A4 (en) 2004-05-06 2009-08-26 Plexxikon Inc Pde4b inhibitors and uses therefor
US20060094651A1 (en) * 2004-11-03 2006-05-04 Cyrus Karkaria Formulations and methods of production of FGF-20
EP1598428A1 (en) 2004-05-18 2005-11-23 Georg Dewald Methods and kits to detect Hereditary angioedema type III
WO2005116657A2 (en) 2004-05-24 2005-12-08 Universität Zu Köln Identification of ergothioneine transporter and therapeutic uses thereof
JP4651663B2 (en) 2004-05-28 2011-03-16 アジェンシス,インコーポレイテッド Antibodies and related molecules that bind to PSCA proteins
EP1602926A1 (en) 2004-06-04 2005-12-07 University of Geneva Novel means and methods for the treatment of hearing loss and phantom hearing
WO2005121348A1 (en) * 2004-06-07 2005-12-22 Protiva Biotherapeutics, Inc. Lipid encapsulated interfering rna
CA2569645C (en) 2004-06-07 2014-10-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods of use
EP2256198A1 (en) 2004-06-14 2010-12-01 Galapagos N.V. Methods for identification, and compounds useful for the treatment of degenerative and inflammatory diseases
US20060159689A1 (en) * 2004-06-17 2006-07-20 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
US20060008468A1 (en) * 2004-06-17 2006-01-12 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
TW200613554A (en) 2004-06-17 2006-05-01 Wyeth Corp Plasmid having three complete transcriptional units and immunogenic compositions for inducing an immune response to HIV
KR20070028547A (en) * 2004-06-18 2007-03-12 리전츠 오브 더 유니버스티 오브 미네소타 Identifying virally infected and vaccinated organisms
WO2005124342A2 (en) 2004-06-21 2005-12-29 Galapagos N.V. Methods and means for treatment of osteoarthritis
WO2006002262A2 (en) 2004-06-21 2006-01-05 The Board Of Trustees Of The Leland Stanford Junior University Genes and pathways differentially expressed in bipolar disorder and/or major depressive disorder
CA2572439A1 (en) * 2004-07-02 2006-01-12 Protiva Biotherapeutics, Inc. Immunostimulatory sirna molecules and uses therefor
ES2673972T3 (en) 2004-07-09 2018-06-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Soluble forms of the glycoprotein G of the hendra and nipah viruses
US20090221440A1 (en) * 2004-07-12 2009-09-03 Board Of Regents, The University Of Texas System Methods and compositions related to identifying protein-protein interactions
US7604798B2 (en) * 2004-07-15 2009-10-20 Northwestern University Methods and compositions for importing nucleic acids into cell nuclei
US20090214588A1 (en) 2004-07-16 2009-08-27 Nabel Gary J Vaccines against aids comprising cmv/r-nucleic acid constructs
CA2574572A1 (en) * 2004-07-19 2006-10-26 Baylor College Of Medicine Modulation of cytokine signaling regulators and applications for immunotherapy
WO2006007712A1 (en) * 2004-07-19 2006-01-26 Protiva Biotherapeutics, Inc. Methods comprising polyethylene glycol-lipid conjugates for delivery of therapeutic agents
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
TWI355389B (en) 2004-07-30 2012-01-01 Rinat Neuroscience Corp Antibodies directed against amyloid-beta peptide a
US20060039949A1 (en) * 2004-08-20 2006-02-23 Nycz Jeffrey H Acetabular cup with controlled release of an osteoinductive formulation
US7985581B2 (en) 2004-08-23 2011-07-26 Alnylam Pharmaceuticals, Inc. Multiple RNA polymerase III promoter expression constructs
US20060045902A1 (en) * 2004-09-01 2006-03-02 Serbousek Jon C Polymeric wrap for in vivo delivery of osteoinductive formulations
DE102004042546A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immune stimulation
US7605168B2 (en) * 2004-09-03 2009-10-20 Plexxikon, Inc. PDE4B inhibitors
EP1804835B9 (en) 2004-09-13 2012-05-02 Genzyme Corporation Multimeric constructs
US20060057184A1 (en) * 2004-09-16 2006-03-16 Nycz Jeffrey H Process to treat avascular necrosis (AVN) with osteoinductive materials
ATE505541T1 (en) 2004-09-24 2011-04-15 Alnylam Pharmaceuticals Inc TARGETING INTERMEDIATE PRODUCTS FOR COUNTER STRAND REPLICATION OF SINGLE STRAND VIRUSES BY RNAI
US20110098554A1 (en) * 2004-09-29 2011-04-28 Tel Hashomer Medical Research Infrastructure And Services Ltd. Monitoring of convection enhanced drug delivery
US20060088607A1 (en) * 2004-10-01 2006-04-27 Stefano George B Nutritional supplement compositions
AU2005294347A1 (en) * 2004-10-05 2006-04-20 Oregon Health And Science University Compositions and methods for treating disease
AU2005293752A1 (en) * 2004-10-13 2006-04-20 Ablynx N.V. Single domain camelide anti-amyloid beta antibodies and polypeptides comprising the same for the treatment and diagnosis of degenarative neural diseases such as Alzheimer's disease
US7485468B2 (en) 2004-10-15 2009-02-03 Galapagos Bv Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
US20090281025A1 (en) * 2004-10-18 2009-11-12 Mount Sinai School Of Medicine Of New York University Inhibition of tumor growth and metastasis by atf2-derived peptides
CN101175508A (en) 2004-10-21 2008-05-07 惠氏公司 Immunogenic compositions of staphylococcus epidermidis polypeptide antigens
WO2006047227A1 (en) * 2004-10-21 2006-05-04 Massachusetts Institute Of Technology Compositions and methods for treatment of hypertrophic tissues
ZA200701792B (en) * 2004-10-25 2008-12-31 Cytos Biotechnology Ag Gastric inhibitory polypeptide (GIP) antigen arrays and uses thereof
US20090041812A1 (en) * 2004-11-01 2009-02-12 Bell Steve J D Therapeutic Calcium Phosphate Particles in Use for Aesthetic of Cosmetic Medicine, and Methods of Manufacture and Use
EP2266602A3 (en) 2004-11-01 2011-08-10 Novartis Vaccines and Diagnostics, Inc. Combination approaches for generating immune responses
WO2006053061A2 (en) * 2004-11-10 2006-05-18 Kansas State University Research Foundation Porcine reproductive and respiratory syndrome virus receptor components and uses thereof
EP2302054B1 (en) 2004-11-12 2014-07-16 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
US20060147449A1 (en) * 2004-11-15 2006-07-06 Brass Lawrence F Method of using CD100 (or Sema4D) to mediate platelet activation and inflammatory responses
AU2005306533B2 (en) * 2004-11-17 2012-05-31 Arbutus Biopharma Corporation siRNA silencing of apolipoprotein B
WO2007050095A2 (en) * 2004-11-19 2007-05-03 The Trustees Of The University Of Pennsylvania Improved vaccines and methods for using the same
US20060115462A1 (en) * 2004-12-01 2006-06-01 Vladimir Subbotin Direct DNA delivery to bone cells
WO2006063301A1 (en) * 2004-12-10 2006-06-15 Maxcyte, Inc. Genetically modified tumor cells as cancer vaccines
US7939490B2 (en) * 2004-12-13 2011-05-10 University Of Maryland, Baltimore TWEAK as a therapeutic target for treating central nervous system diseases associated with cerebral edema and cell death
EP2316942B1 (en) 2004-12-22 2021-04-21 Alnylam Pharmaceuticals, Inc. Conserved hbv and hcv sequences useful for gene silencing
US20060165711A1 (en) * 2004-12-29 2006-07-27 Bot Adrian I Methods to elicit, enhance and sustain immune responses against MHC class I-restricted epitopes, for prophylactic or therapeutic purposes
PL1833506T3 (en) * 2004-12-29 2016-01-29 Mannkind Corp Use of compositions comprising various tumor-associated antigens as anti-cancer vaccines
EP1835929B8 (en) 2005-01-06 2016-07-27 Novo Nordisk A/S Anti-kir combination treatments and methods
US9034650B2 (en) 2005-02-02 2015-05-19 Intrexon Corporation Site-specific serine recombinases and methods of their use
WO2006086284A2 (en) * 2005-02-11 2006-08-17 Merck & Co., Inc. Adenovirus serotype 26 vectors, nucleic acid and viruses produced thereby
EP2634252B1 (en) 2005-02-11 2018-12-19 University of Southern California Method of expressing proteins with disulfide bridges
JP2008530245A (en) 2005-02-18 2008-08-07 ノバルティス ヴァクシンズ アンド ダイアグノスティクス, インコーポレイテッド Antigens from uropathogenic strains
EP1863511A2 (en) 2005-02-23 2007-12-12 UAB Research Foundation Alkyl-glycoside enhanced vaccination
US20060194740A1 (en) * 2005-02-25 2006-08-31 Ulevitch Richard J NOD1 as an anti-tumor agent
PL2325305T3 (en) 2005-02-25 2014-07-31 Oncotherapy Science Inc Peptide vaccines for lung cancers expressing TTK, URLC10 or KOC1 polypeptides
WO2006093030A1 (en) 2005-02-28 2006-09-08 Oncotherapy Science, Inc. Epitope peptides derived from vascular endothelial growth factor receptor 1 and vaccines containing these peptides
US20090142338A1 (en) * 2005-03-04 2009-06-04 Curedm, Inc. Methods and Compositions for Treating Type 1 and Type 2 Diabetes Mellitus and Related Conditions
JP2008531730A (en) * 2005-03-04 2008-08-14 キュアーディーエム、インク. Methods and pharmaceutical compositions for treating type I diabetes mellitus and other conditions
WO2006099574A2 (en) * 2005-03-16 2006-09-21 Tai June Yoo Cockroach allergen gene expression and delivery systems and uses
US7829673B2 (en) 2005-03-23 2010-11-09 Genmab A/S Antibodies against CD38 for treatment of multiple myeloma
RU2413735C2 (en) 2005-03-31 2011-03-10 Эдженсис, Инк. Antibodies and related molecules binding with proteins 161p2f10b
US20070092449A1 (en) * 2005-04-05 2007-04-26 Rafael Vazquez-Martinez Methods for direct visualization of active synapses
WO2006110599A2 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cacna1e in cancer diagnosis, detection and treatment
JP2008535494A (en) 2005-04-07 2008-09-04 サグレシュ ディスカバリー, インコーポレイテッド Cancer-related gene (PRLR)
US7691579B2 (en) * 2005-04-15 2010-04-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods and compositions for producing an enhanced immune response to a human papillomavirus immunogen
EP1712241A1 (en) 2005-04-15 2006-10-18 Centre National De La Recherche Scientifique (Cnrs) Composition for treating cancer adapted for intra-tumoral administration and uses thereof
EP2266656B1 (en) 2005-04-18 2020-10-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Devices for delivering a biologically active agent to the ocular sphere of a subject
EP1881845B1 (en) 2005-04-25 2010-03-24 Merial Ltd. Nipah virus vaccines
AR054260A1 (en) * 2005-04-26 2007-06-13 Rinat Neuroscience Corp METHODS OF TREATMENT OF DISEASES OF THE LOWER MOTOR NEURONE AND COMPOSITIONS USED IN THE SAME
UY29504A1 (en) 2005-04-29 2006-10-31 Rinat Neuroscience Corp DIRECTED ANTIBODIES AGAINST BETA AMYLOID PEPTIDE AND METHODS USING THE SAME.
US8048446B2 (en) * 2005-05-10 2011-11-01 Drexel University Electrospun blends of natural and synthetic polymer fibers as tissue engineering scaffolds
EP1882035A2 (en) 2005-05-12 2008-01-30 Novartis AG Genes and proteins of brachyspira hyodysenteriae and use of same for diagnosis and therapy
AU2006272951A1 (en) 2005-05-17 2007-02-01 Plexxikon, Inc. Pyrrol (2,3-b) pyridine derivatives protein kinase inhibitors
DK2949668T3 (en) 2005-05-18 2019-11-11 Ablynx Nv IMPROVED NANOBODIES TM AGAINST TUMOR CANCER FACTOR-ALFA
DE102005023170A1 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized formulation for mRNA
PT3415535T (en) 2005-05-20 2021-02-02 Ablynx Nv Improved nanobodies tm for the treatment of aggregation-mediated disorders
WO2006128083A2 (en) 2005-05-25 2006-11-30 Curedm, Inc. Human proislet peptide, derivatives and analogs thereof, and methods of using same
WO2007031867A2 (en) 2005-05-25 2007-03-22 Tripep Ab A hepatitis c virus non-stru tural ns3/4a fusion gene
EP2816118B1 (en) 2005-05-31 2018-10-17 The Regents of the University of Colorado, a body corporate Methods for delivering genes
ES2413079T3 (en) 2005-06-17 2013-07-15 Mannkind Corporation Methods and compositions for triggering multivalent immune responses against dominant and subdominant epitopes expressed in cancer cells and tumor stroma
DK1893612T3 (en) 2005-06-22 2011-11-21 Plexxikon Inc Pyrrole [2,3-B] pyridine derivatives as protein kinase inhibitors
FR2887457B1 (en) * 2005-06-23 2007-10-05 Fond Bettencourt Schueller TRANSCUTANE TARGETING VACCINATION
US7868159B2 (en) * 2005-06-23 2011-01-11 Baylor College Of Medicine Modulation of negative immune regulators and applications for immunotherapy
EP1904631B1 (en) 2005-06-24 2012-05-16 Regents Of The University Of Minnesota Prrs viruses, infectious clones, mutants thereof, and methods of use
CN101437539B (en) * 2005-07-05 2013-10-02 康奈尔研究基金会(有限公司) Blocking leukocyte emigration and inflammation by interfering with CD99l2
CN104072614B (en) 2005-07-08 2017-04-26 生物基因Ma公司 Anti-alpha[v]beta[6] antibodies and uses thereof
KR20080039929A (en) 2005-07-22 2008-05-07 와이스 테라퓨틱스 가부시키가이샤 Anti-cd26 antibodies and methods of use thereof
PL1910839T3 (en) 2005-07-27 2016-11-30 Colon cancer related gene tom34
CN101563096A (en) 2005-07-28 2009-10-21 辉瑞产品公司 Methods of vaccine administration, new feline caliciviruses, and treatments for immunizing animals against feline paraovirus and feline herpes virus
EP1922083A2 (en) * 2005-08-10 2008-05-21 Oklahoma Medical Research Foundation Truncated memapsin 2 for use for treating alzheimer's disease
AU2006283101B2 (en) 2005-08-23 2013-03-07 Beth Israel Deaconess Medical Center Polyvalent vaccine
US7951377B2 (en) 2005-08-23 2011-05-31 Los Alamos National Security, Llc Mosaic clade M human immunodeficiency virus type 1 (HIV-1) envelope immunogens
US20070048254A1 (en) * 2005-08-24 2007-03-01 Mirus Bio Corporation Generation of dendritic cells
US20080241184A1 (en) 2005-08-25 2008-10-02 Jules Maarten Minke Canine influenza vaccines
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
US7666584B2 (en) * 2005-09-01 2010-02-23 Philadelphia Health & Education Coporation Identification of a pin specific gene and protein (PIN-1) useful as a diagnostic treatment for prostate cancer
EP1928537B1 (en) 2005-09-02 2015-01-21 Intercell USA, Inc. Devices for transcutaneous delivery of vaccines and transdermal delivery of drugs
EP1924279A2 (en) * 2005-09-07 2008-05-28 The Secretary of State for Defence Adjuvanted vaccine
CA2621982C (en) 2005-09-07 2017-11-28 Jennerex Biotherapeutics Ulc Systemic treatment of metastatic and/or systemically-disseminated cancers using gm-csf-expressing poxviruses
US8980246B2 (en) 2005-09-07 2015-03-17 Sillajen Biotherapeutics, Inc. Oncolytic vaccinia virus cancer therapy
US7803901B2 (en) * 2005-09-16 2010-09-28 Mayo Foundation For Medical Education And Research Polypeptides with natriuresis activity
DE102005046490A1 (en) 2005-09-28 2007-03-29 Johannes-Gutenberg-Universität Mainz New nucleic acid molecule comprising promoter, a transcriptable nucleic acid sequence, a first and second nucleic acid sequence for producing modified RNA with transcriptional stability and translational efficiency
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
GB0519871D0 (en) * 2005-09-30 2005-11-09 Secr Defence Immunogenic agents
AU2006301846A1 (en) * 2005-10-13 2007-04-19 Akshaya Bio Inc. Chimeric antigen containing hepatitis C virus polypeptide and FC fragment for eliciting an immune response
MX2008005405A (en) * 2005-10-28 2008-09-11 Florida Internat University Bo Horse: human chimeric antibodies.
CN101346393B (en) * 2005-11-02 2015-07-22 普洛体维生物治疗公司 Modified siRNA molecules and uses thereof
WO2007056113A2 (en) * 2005-11-02 2007-05-18 Cylene Pharmaceuticals, Inc. Methods for targeting quadruplex sequences
JP5366554B2 (en) 2005-11-12 2013-12-11 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー FGF2-related methods for diagnosing and treating depression
CA2629522A1 (en) 2005-11-14 2007-05-18 Merial Limited Gene therapy for renal failure
US20070253969A1 (en) * 2005-11-14 2007-11-01 Freda Stevenson Materials and methods relating to DNA vaccination
US7771995B2 (en) 2005-11-14 2010-08-10 Merial Limited Plasmid encoding human BMP-7
DK3045182T3 (en) 2005-11-14 2018-03-19 Teva Pharmaceuticals Int Gmbh ANTAGONIST ANTIBODIES AGAINST CALCITONIN GEN-RELATED PEPTIDE TO TREAT HEAT
ES2514316T3 (en) 2005-11-22 2014-10-28 Novartis Vaccines And Diagnostics, Inc. Norovirus and Sapovirus virus-like particles (VLPs)
US8026354B2 (en) 2005-11-23 2011-09-27 Institut Pasteur Recombinant plasmodium falciparum merozoite surface proteins 4 and 5 and their use
EP1795540A1 (en) 2005-11-30 2007-06-13 Imaxio Multimeric complexes of antigens and an adjuvant
KR101045165B1 (en) 2005-12-07 2011-06-30 화이자 프로덕츠 인크. Marked bovine viral diarrhea virus vaccines
US7745158B2 (en) * 2005-12-14 2010-06-29 Kimberly-Clark Worldwide, Inc. Detection of secreted aspartyl proteases from Candida species
LT1976880T (en) 2005-12-21 2016-10-10 Amgen Research (Munich) Gmbh Pharmaceutical compositions with resistance to soluble cea
US8178660B2 (en) 2006-01-13 2012-05-15 The Trustees Of The University Of Pennsylvania Vaccines and immunotherapeutics using codon optimized IL-15 and methods for using the same
EP1998800A2 (en) 2006-01-18 2008-12-10 University Of Chicago Compositions and methods related to staphylococcal bacterium proteins
NL2000439C2 (en) 2006-01-20 2009-03-16 Quark Biotech Therapeutic applications of inhibitors of RTP801.
TW200808352A (en) * 2006-02-02 2008-02-16 Rinat Neuroscience Corp Methods for treating unwanted weight loss or eating disorders by administering a TRKB agonist
JP2009525319A (en) * 2006-02-02 2009-07-09 ライナット ニューロサイエンス コーポレイション Method of treating obesity by administering a trkB antagonist
EP2253957B1 (en) 2006-03-14 2013-05-15 Oregon Health and Science University Methods for producing an immune response to tuberculosis.
US20090130212A1 (en) * 2006-05-15 2009-05-21 Physical Pharmaceutica, Llc Composition and improved method for preparation of small particles
US7862821B2 (en) 2006-06-01 2011-01-04 Merial Limited Recombinant vaccine against bluetongue virus
CN103709252B (en) 2006-06-07 2016-12-07 生物联合公司 Identify antibody and the using method thereof of the sugary epi-position of CD-43 and CEA expressed on cancerous cell
WO2007146831A2 (en) * 2006-06-08 2007-12-21 Cylene Pharmaceuticals, Inc. Quinolone analogs derivatized with sulfonic acid, sulfonate or sulfonamide
US20110112086A1 (en) * 2006-06-08 2011-05-12 Cylene Pharmaceuticals, Inc. Pyridinone analogs
US20100015168A1 (en) 2006-06-09 2010-01-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
EP2383341A1 (en) * 2006-06-12 2011-11-02 Exegenics, Inc., D/b/a Opko Health, Inc. Compositions and methods for siRNA inhibition of angiogenesis
WO2007149493A2 (en) * 2006-06-22 2007-12-27 Immugen Inc Restoration of hearing loss
US7871624B2 (en) * 2006-06-27 2011-01-18 Saint Louis University Chimeral polypeptide composition for cross-placenta delivery
US8784833B2 (en) * 2006-06-27 2014-07-22 Saint Louis University Prenatal enzyme replacement therapy for hypophosphatasia
US7572618B2 (en) 2006-06-30 2009-08-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
EP2046374A4 (en) 2006-07-10 2010-05-05 Biogen Idec Inc Compositions and methods for inhibiting growth of smad4-deficient cancers
US8323664B2 (en) * 2006-07-25 2012-12-04 The Secretary Of State For Defence Live vaccine strains of Francisella
ATE543097T1 (en) 2006-07-27 2012-02-15 Univ Maryland CELLULAR RECEPTOR FOR ANTIPROLIFERATIVE FACTOR
EP3489251B1 (en) 2006-07-28 2021-03-17 The Trustees of the University of Pennsylvania Hiv consensus proteins and vaccines made therefrom
BRPI0714981A2 (en) * 2006-07-28 2013-08-13 Sanofi Aventis A method of making the same comprising an inhibitor that inhibits the activity of a complex iii protein, a vehicle and a member of the tumor necrosis factor superfamily, a method for conducting an assay to determine whether a compound is an inhibitor. of tnf suppression activity of a complex iii protein and use of a compound
WO2008021872A1 (en) 2006-08-08 2008-02-21 Mayo Foundation For Medical Education And Research Diuretic and natriuretic polypeptides
US20100166788A1 (en) 2006-08-16 2010-07-01 Novartis Vaccines And Diagnostics Immunogens from uropathogenic escherichia coli
EP2295445A1 (en) 2006-09-08 2011-03-16 Mayo Foundation for Medical Education and Research Aquaretic and natriuretic polypeptides lacking vasodilatory activity
US7872118B2 (en) * 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
US8481023B2 (en) 2006-09-15 2013-07-09 Ottawa Hospital Research Institute Oncolytic rhabdovirus
SG10201502098YA (en) 2006-10-17 2015-05-28 Oncotherapy Science Inc Peptide Vaccines For Cancers Expressing MPHOSPH1 OR DEPDC1 Polypeptides
ES2501947T3 (en) 2006-10-19 2014-10-02 Csl Limited Interleukin alpha 1 receptor high affinity antibody antagonists
CA2666682C (en) 2006-10-19 2014-07-08 Merck & Co., Inc. Anti-il-13r.alpha.1 antibodies and their uses thereof
US20080118524A1 (en) * 2006-10-20 2008-05-22 Stefan Persson Anti-IgE Vaccines
MX2009004221A (en) * 2006-10-20 2009-09-10 Univ Arizona State Modified cyanobacteria.
JP2010507387A (en) 2006-10-25 2010-03-11 クアーク・ファーマスーティカルス、インコーポレイテッド Novel siRNA and method of using the same
US8202967B2 (en) 2006-10-27 2012-06-19 Boehringer Ingelheim Vetmedica, Inc. H5 proteins, nucleic acid molecules and vectors encoding for those, and their medicinal use
EP2799547B1 (en) 2006-11-08 2016-12-21 Veritas Bio, LLC In Vivo Delivery of RNA to a Target Cell
US8785400B2 (en) * 2006-11-22 2014-07-22 Curedm Group Holdings, Llc Methods and compositions relating to islet cell neogenesis
WO2008063888A2 (en) 2006-11-22 2008-05-29 Plexxikon, Inc. Compounds modulating c-fms and/or c-kit activity and uses therefor
ATE555128T1 (en) 2006-11-30 2012-05-15 Res Dev Foundation IMPROVED IMMUNOLOBULIN LIBRARIES
RU2486907C2 (en) 2006-12-04 2013-07-10 Джонс Хопкинс Юниверсити Imidised biopolymer adhesive and hydrogel
WO2008074678A1 (en) 2006-12-18 2008-06-26 F. Hoffmann-La Roche Ag Novel use of inhibitors of soluble epoxide hydrolase
AU2007336242B2 (en) 2006-12-19 2012-08-30 Ablynx N.V. Amino acid sequences directed against GPCRs and polypeptides comprising the same for the treatment of GPCR-related diseases and disorders
EP2102244A2 (en) 2006-12-19 2009-09-23 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders
AU2007337809A1 (en) * 2006-12-20 2008-07-03 Rinat Neuroscience Corporation TrkB agonists for treating autoimmune disorders
WO2008079909A1 (en) 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2,3-b] pyridines as kinase modulators
EP2094701A2 (en) 2006-12-21 2009-09-02 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
PE20081581A1 (en) * 2006-12-21 2008-11-12 Plexxikon Inc PIRROLO [2,3-b] PYRIDINES COMPOUNDS AS KINASE MODULATORS
AR064642A1 (en) * 2006-12-22 2009-04-15 Wyeth Corp POLINUCLEOTIDE VECTOR THAT INCLUDES IT RECOMBINATING CELL THAT UNDERSTANDS THE VECTOR POLYPEPTIDE, ANTIBODY, COMPOSITION THAT UNDERSTANDS THE POLINUCLEOTIDE, VECTOR, RECOMBINATING CELL POLYPEPTIDE OR ANTIBODY, USE OF THE COMPOSITION AND A COMPOSITION AND A METHOD
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2008092019A1 (en) * 2007-01-25 2008-07-31 Mayo Foundation For Medical Education And Research Fgf-23 polypeptides
KR101471445B1 (en) * 2007-01-26 2014-12-15 시나게바 바이오파르마, 코포레이션 Transgene expression in avians
WO2008097927A2 (en) * 2007-02-06 2008-08-14 Tai June Yoo Treatment and prevention of neurodegenerative diseases using gene therapy
TWI596109B (en) 2007-02-21 2017-08-21 腫瘤療法 科學股份有限公司 Peptide vaccines for cancers expressing tumor-associated antigens
CA2678404C (en) 2007-02-28 2019-03-19 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Brachyury polypeptides and methods for use
KR20080084528A (en) 2007-03-15 2008-09-19 제네렉스 바이오테라퓨틱스 인크. Oncolytic vaccinia virus cancer therapy
EP2139447A2 (en) * 2007-03-20 2010-01-06 Harold Brem Gm-csf cosmeceutical compositions and methods of use thereof
EP2136778A2 (en) * 2007-03-20 2009-12-30 Harold Brem Growth factor mediated cosmeceuticals and use thereof to enhance skin quality
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
JP2010524851A (en) 2007-04-03 2010-07-22 マイクロメット アーゲー Species-specific binding domains
BRPI0809594A2 (en) 2007-04-03 2019-08-27 Micromet Ag polypeptide, nucleic acid sequence, vector, host, process for producing a polypeptide, pharmaceutical composition, use of a polypeptide, method for preventing, treating or ameliorating a disease in an individual in need thereof, kit, method for the identification of a polypeptide (s)
WO2008124768A1 (en) 2007-04-09 2008-10-16 The General Hospital Corporation Hemojuvelin fusion proteins and uses thereof
EP2155777A2 (en) 2007-04-10 2010-02-24 The Administrators of the Tulane Educational Fund Soluble and membrane-anchored forms of lassa virus subunit proteins
TW201425333A (en) 2007-04-11 2014-07-01 Oncotherapy Science Inc TEM8 peptides and vaccines comprising the same
US20090010923A1 (en) * 2007-04-24 2009-01-08 University Of Maryland, Baltimore Treatment of cancer with anti-muscarinic receptor agents
AU2008247819B2 (en) 2007-05-01 2013-02-14 Research Development Foundation Immunoglobulin Fc libraries
EP2152736B1 (en) 2007-05-03 2017-07-05 Lysomab GmbH Complement factor h-derived short consensus repeat-antibody constructs
CN104761466A (en) 2007-05-29 2015-07-08 英特拉克森公司 Chiral diacylhydrazine ligands for modulating expression of exogenous genes via ecdysone receptor complex
GB0710529D0 (en) 2007-06-01 2007-07-11 Circassia Ltd Vaccine
CN101796186B (en) 2007-06-07 2013-05-22 韦克福里斯特大学健康科学院 Inkjet gene printing
MX2009013766A (en) 2007-06-20 2010-02-01 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of bone and joint degenerative diseases.
MX2010000617A (en) 2007-07-17 2010-05-17 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor.
EP2171053B1 (en) 2007-07-20 2014-04-23 Mayo Foundation for Medical Education and Research Natriuretic polypeptides
US8889622B2 (en) * 2007-07-25 2014-11-18 Washington University Methods of inhibiting seizure in a subject
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
CN101821284B (en) 2007-08-03 2014-11-19 贝林格尔·英格海姆维特梅迪卡有限公司 Novel genes and proteins of brachyspira hyodysenteriac and uses thereof
US20120150023A1 (en) * 2007-08-06 2012-06-14 Kaspar Roger L Microneedle arrays for active agent delivery
JP2010535591A (en) 2007-08-06 2010-11-25 トランスダーム, インコーポレイテッド Microneedle array formed from polymer film
US8551493B2 (en) 2007-08-15 2013-10-08 Circassia Limited Peptide with reduced dimer formation
US8071561B2 (en) 2007-08-16 2011-12-06 Chrontech Pharma Ab Immunogen platform
US20090214593A1 (en) * 2007-08-16 2009-08-27 Tripep Ab Immunogen platform
WO2009026233A2 (en) * 2007-08-17 2009-02-26 Northwestern University Self assembling peptide systems and methods
KR20100049084A (en) 2007-08-23 2010-05-11 인트렉손 코포레이션 Methods and compositions for diagnosing disease
PT2193142E (en) * 2007-08-30 2015-04-22 Curedm Group Holdings Llc Compositions and methods of using proislet peptides and analogs thereof
EP2666784B1 (en) 2007-08-31 2017-04-05 University Of Chicago Methods and compositions related to immunizing against staphylococcal lung diseases and conditions
US7855049B2 (en) * 2007-08-31 2010-12-21 Biocrine Ab Inositol pyrophosphates determine exocytotic capacity
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US8357656B2 (en) * 2007-09-15 2013-01-22 Mayo Foundation For Medical Education And Research Natriuretic peptide receptor-C agonists
JP2010538647A (en) * 2007-09-17 2010-12-16 ローム アンド ハース カンパニー Compositions and methods for modifying physiological responses in plants
EP2042193A1 (en) * 2007-09-28 2009-04-01 Biomay AG RNA Vaccines
BRPI0817233A2 (en) 2007-09-28 2012-11-06 Intrexon Corp therapeutic constructs of trca gene and bireactors for the expression of biotherapeutic molecules, and uses thereof
PT2214491T (en) * 2007-10-05 2016-10-25 Senhwa Biosciences Inc Quinolone analogs and methods related thereto
JP2010539993A (en) * 2007-10-08 2010-12-24 イントレキソン コーポレーション Use for the treatment of genetically engineered dendritic cells and cancer
GB0720250D0 (en) 2007-10-17 2007-11-28 Univ Edinburgh Immunogenic compositions containing escherichia coli h7 flagella and methods of use thereof
EP3085707B1 (en) 2007-11-01 2019-02-27 Mayo Foundation for Medical Education and Research Hla-dr binding peptides and their uses
DK2222697T3 (en) 2007-11-01 2013-03-11 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
EP2222344A4 (en) * 2007-11-30 2012-11-07 Baylor College Medicine Dendritic cell vaccine compositions and uses of same
JP5737944B2 (en) 2007-12-17 2015-06-17 ファイザー・リミテッドPfizer Limited Treatment of interstitial cystitis
SG186669A1 (en) 2007-12-18 2013-01-30 Bioalliance Cv Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
BRPI0906429B1 (en) 2008-01-10 2021-08-03 Research Development Foundation METHOD OF IDENTIFYING AN E. CHAFFEENSIS INFECTION IN AN INDIVIDUAL, USE OF ONE OR MORE SYNTHETIC POLYPEPTIDE AND KIT
JP2011509743A (en) 2008-01-17 2011-03-31 ジェネトロニクス,インコーポレイティド Variable current density single needle electroporation system and method
US20090202606A1 (en) * 2008-01-25 2009-08-13 Viromed Co., Ltd. Treatment and Prevention of Cardiac Conditions Using Two or More Isoforms of Hepatocyte Growth Factor
MX2010008168A (en) 2008-01-25 2011-02-24 P53 Inc P53 biomarkers.
EP2252702B1 (en) * 2008-02-08 2014-01-29 Mayo Foundation for Medical Education and Research Detection of clostridium difficile
EP2363407A1 (en) 2008-02-28 2011-09-07 Murdoch University Novel sequences of Brachyspira, immunogenic compositions, methods for preparation and use thereof
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
CN102014937A (en) 2008-03-03 2011-04-13 迈阿密大学 Allogeneic cancer cell-based immunotherapy
CA2718904C (en) 2008-03-17 2017-01-03 The Scripps Research Institute Combined chemical and genetic approaches for generation of induced pluripotent stem cells
CN102036677A (en) 2008-03-20 2011-04-27 迈阿密大学 Heat shock protein GP96 vaccination and methods of using same
WO2009117773A1 (en) 2008-03-27 2009-10-01 Murdoch University Novel sequences of brachyspira, immunogenic compositions, methods for preparation and use thereof
JP5744719B2 (en) * 2008-04-04 2015-07-08 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Chikungunya virus protein consensus sequence, nucleic acid molecule encoding the same, and compositions and methods using the same
AU2009231598B2 (en) 2008-04-04 2015-03-12 The Trustees Of The University Of Pennsylvania Vaccines and immunotherapeutics using IL-28 and compositions and methods of using the same
WO2009124931A2 (en) 2008-04-07 2009-10-15 Ablynx Nv Amino acid sequences directed against the notch pathways and uses thereof
EP2274437B1 (en) 2008-04-10 2015-12-23 Cell Signaling Technology, Inc. Compositions and methods for detecting egfr mutations in cancer
JP5475753B2 (en) * 2008-04-15 2014-04-16 プロチバ バイオセラピューティクス インコーポレイティッド Lipid formulations for nucleic acid delivery
CA2721203C (en) 2008-04-16 2016-11-01 Howard University Inhibitors of protein phosphatase-1 and uses thereof
CN110075113A (en) 2008-04-17 2019-08-02 Pds生物科技公司 Immune response is stimulated by the enantiomer of cation lipid
WO2009137829A2 (en) 2008-05-09 2009-11-12 Wake Forest University Health Sciences Directed stem cell recruitment
EP2853269B1 (en) 2008-05-19 2019-05-01 Advaxis, Inc. Dual delivery system for heterologous antigens comprising a recombinant Listeria strain attenuated by mutation of dal/dat and deletion of ActA comprising a nucleic acid molecule encoding an listeriolysin O - prostate specific anigen fusion protein
US9650639B2 (en) 2008-05-19 2017-05-16 Advaxis, Inc. Dual delivery system for heterologous antigens
US9017660B2 (en) 2009-11-11 2015-04-28 Advaxis, Inc. Compositions and methods for prevention of escape mutation in the treatment of Her2/neu over-expressing tumors
US8093043B2 (en) * 2008-06-04 2012-01-10 New York University β-TrCP1, β-TrCP2 and RSK1 or RSK2 inhibitors and methods for sensitizing target cells to apoptosis
EP3279314A1 (en) 2008-06-04 2018-02-07 Cellular Dynamics International, Inc. Methods for the production of ips cells using non-viral approach
CA2635187A1 (en) * 2008-06-05 2009-12-05 The Royal Institution For The Advancement Of Learning/Mcgill University Oligonucleotide duplexes and uses thereof
WO2009147684A2 (en) 2008-06-06 2009-12-10 Quark Pharmaceuticals, Inc. Compositions and methods for treatment of ear disorders
WO2009150623A1 (en) 2008-06-13 2009-12-17 Pfizer Inc Treatment of chronic prostatitis
EP2303318A2 (en) 2008-06-20 2011-04-06 Wyeth LLC Compositions and methods of use of orf1358 from beta-hemolytic streptococcal strains
EP2307447B1 (en) * 2008-07-02 2016-03-09 Mayo Foundation For Medical Education And Research Natriuretic polypeptides with unique pharmacologic profiles
CA2737146A1 (en) 2008-07-25 2010-01-28 The Regents Of The University Of Colorado Clip inhibitors and methods of modulating immune function
WO2010019569A1 (en) 2008-08-12 2010-02-18 Cellular Dynamics International. Inc. Methods for the production of ips cells
EP2433639A3 (en) 2008-08-15 2012-06-27 Circassia Limited Vaccine comprising Amb a 1 peptides for use in the treatment of ragweed allergy
US8912146B2 (en) 2008-08-18 2014-12-16 University Of Maryland, Baltimore Derivatives of APF and methods of use
EP2329044B1 (en) 2008-08-27 2016-05-18 Oncotherapy Science, Inc. Prmt1 for target genes of cancer therapy and diagnosis
AU2009290848A1 (en) 2008-09-11 2010-03-18 Galapagos Nv Method for identifying compounds useful for increasing the functional activity and cell surface expression of CF-associated mutant Cystic Fibrosis transmembrane conductance regulator
TWI516501B (en) 2008-09-12 2016-01-11 禮納特神經系統科學公司 Pcsk9 antagonists
US20110287026A1 (en) 2008-09-23 2011-11-24 President And Fellows Of Harvard College Sirt4 and uses thereof
US9157097B2 (en) 2008-09-25 2015-10-13 Proteovec Holding, L.L.C. Vectors for production of growth hormone
US9150880B2 (en) 2008-09-25 2015-10-06 Proteovec Holding, L.L.C. Vectors for production of antibodies
RS54900B1 (en) 2008-10-01 2016-10-31 Amgen Res (Munich) Gmbh Cross-species-specific psmaxcd3 bispecific single chain antibody
EP2352765B1 (en) 2008-10-01 2018-01-03 Amgen Research (Munich) GmbH Cross-species-specific single domain bispecific single chain antibody
EP2341929B1 (en) 2008-10-06 2017-01-25 University Of Chicago Compositions and methods related to bacterial emp proteins
EP2743265B1 (en) 2008-10-09 2017-03-15 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010048252A1 (en) * 2008-10-23 2010-04-29 Intervet International B.V. Lawsonia intracellularis vaccines
US8642550B2 (en) 2008-10-24 2014-02-04 Mayo Foundation For Medical Education And Research Chimeric natriuretic peptides without hypotensive inducing capability
US8921536B2 (en) 2008-10-29 2014-12-30 The Trustees Of The University Of Pennsylvania HCV vaccines and methods for using the same
JP5753090B2 (en) 2008-10-29 2015-07-22 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Improved HCV vaccine and method of use thereof
NZ592368A (en) 2008-11-05 2013-11-29 Wyeth Llc Multicomponent immunogenic composition for the prevention of beta-hemolytic streptococcal (bhs) disease
WO2010056901A2 (en) 2008-11-13 2010-05-20 University Of Southern California Method of expressing proteins with disulfide bridges with enhanced yields and activity
EP2361093A2 (en) 2008-11-26 2011-08-31 Five Prime Therapeutics, Inc. Compositions and methods for regulating collagen and smooth muscle actin expression by serpine2
MX2011005820A (en) * 2008-12-03 2011-06-21 Pfizer Bovine viral diarrhea virus with a modified erns protein.
WO2010065834A1 (en) 2008-12-04 2010-06-10 Opko Ophthalmics, Llc Compositions and methods for selective inhibition of pro-angiogenic vegf isoforms
TWI500932B (en) 2008-12-05 2015-09-21 Oncotherapy Science Inc Wdrpuh epitope peptides and vaccines containing the same
UA109633C2 (en) 2008-12-09 2015-09-25 HUMAN ANTIBODY AGAINST TISSUE FACTOR
WO2010068738A1 (en) 2008-12-10 2010-06-17 Dana-Farber Cancer Institute, Inc. Mek mutations conferring resistance to mek inhibitors
BRPI0922572A2 (en) 2008-12-17 2019-09-24 Scripps Research Inst method for culturing pluripotent cells, pluripotent mammalian cell culture, cell culture medium, isolated pluripotent animal cell, and method for increasing pluripotence of a mammalian cell.
WO2010080452A2 (en) 2008-12-18 2010-07-15 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS AND METHODS OF USE THEREOF
US8455438B2 (en) 2008-12-29 2013-06-04 Mayo Foundation For Medical Education And Research Natriuretic polypeptides for reducing or preventing restenosis
CN102307477B (en) * 2009-01-05 2015-07-29 埃皮托吉尼西斯股份有限公司 Adjunvant composition and using method
US9181315B2 (en) 2009-01-08 2015-11-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for induced brown fat differentiation
GB0900455D0 (en) 2009-01-13 2009-02-11 Secr Defence Vaccine
EP2387627B1 (en) 2009-01-15 2016-03-30 Adaptive Biotechnologies Corporation Adaptive immunity profiling and methods for generation of monoclonal antibodies
EP2379720B1 (en) 2009-01-20 2016-08-17 Alona Zilberberg Mir-21 promoter driven targeted cancer therapy
US9050287B2 (en) 2009-01-23 2015-06-09 The Trustees Of The University Of Pennsylvania Vaccines for human papilloma virus and methods for using the same
CA2653478A1 (en) 2009-01-23 2010-07-23 Gregg Martin Automated wash system for industrial vehicles
EP2391635B1 (en) 2009-01-28 2017-04-26 Epimmune Inc. Pan-dr binding polypeptides and uses thereof
GB0901411D0 (en) 2009-01-29 2009-03-11 Secr Defence Treatment
GB0901423D0 (en) 2009-01-29 2009-03-11 Secr Defence Treatment
GB0901593D0 (en) 2009-01-30 2009-03-11 Touchlight Genetics Ltd Production of closed linear DNA
WO2010086828A2 (en) 2009-02-02 2010-08-05 Rinat Neuroscience Corporation Agonist anti-trkb monoclonal antibodies
EP2891662A1 (en) 2009-02-05 2015-07-08 Circassia Limited Grass peptides for vaccine
TWI469791B (en) 2009-02-18 2015-01-21 Oncotherapy Science Inc Foxm1 peptides and vaccines containing the same
WO2010096561A1 (en) 2009-02-18 2010-08-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Synthetic hiv/siv gag proteins and uses thereof
EP2221066A1 (en) 2009-02-18 2010-08-25 Sanofi-Aventis Use of VgII3 activity modulator for the modulation of adipogenesis
WO2010094733A2 (en) 2009-02-19 2010-08-26 Biofocus Dpi B.V. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
EP2398480A1 (en) 2009-02-19 2011-12-28 Galapagos N.V. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
US20120004160A1 (en) 2009-02-19 2012-01-05 Glaxo Group Ltd. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
US8664183B2 (en) 2009-02-27 2014-03-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services SPANX-B polypeptides and their use
AU2010220103A1 (en) 2009-03-06 2011-09-22 Novartis Ag Chlamydia antigens
CN102356155B (en) 2009-03-18 2016-02-24 肿瘤疗法科学股份有限公司 NEIL3 peptide and comprise its vaccine
WO2010107778A1 (en) 2009-03-18 2010-09-23 Wake Forest University Health Sciences Flagellin fusion proteins and use thereof to induce immune responses against pseudomonas aeruginosa
CN102378766A (en) 2009-03-23 2012-03-14 夸克医药公司 Compounds compositions and methods of treating cancer and fibrotic diseases
US8993509B2 (en) 2009-03-31 2015-03-31 Robert Zimmerman Method for treatment of cachexia by administering inhibitors of adipose triglyceride lipase expression or activity
WO2010112569A1 (en) 2009-03-31 2010-10-07 Robert Zimmermann Modulation of adipose triglyceride lipase for prevention and treatment of cachexia, loss of weight and muscle atrophy and methods of screening therefor
WO2010115841A1 (en) 2009-04-01 2010-10-14 Galapagos Nv Methods and means for treatment of osteoarthritis
SI3281947T1 (en) 2009-04-03 2020-07-31 The University Of Chicago Compositions and methods related to protein a (spa) variants
WO2010114928A2 (en) 2009-04-03 2010-10-07 F.Hoffmann-La Roche Ag Compositions and uses thereof
CA2757030C (en) 2009-04-03 2019-01-15 Merial Limited Vectors comprising newcastle disease viruses and compositions thereof
WO2010118243A2 (en) 2009-04-08 2010-10-14 Genentech, Inc. Use of il-27 antagonists to treat lupus
US9150881B2 (en) 2009-04-09 2015-10-06 Proteovec Holding, L.L.C. Production of proteins using transposon-based vectors
WO2010115998A2 (en) 2009-04-10 2010-10-14 Ablynx Nv Improved amino acid sequences directed against il-6r and polypeptides comprising the same for the treatment of il-6r related diseases and disorders
SG175092A1 (en) 2009-04-14 2011-11-28 Novartis Ag Compositions for immunising against staphylococcus aerus
GB0906234D0 (en) 2009-04-14 2009-05-20 Secr Defence Vaccine
JP5822822B2 (en) 2009-04-17 2015-11-24 ニューヨーク ユニバーシティ Peptides that target TNF family receptors and antagonize TNF action, compositions, methods and uses thereof
US8748381B2 (en) 2009-04-28 2014-06-10 Vanderbilt University Compositions and methods for the treatment of disorders involving epithelial cell apoptosis
US20100284977A1 (en) * 2009-04-28 2010-11-11 University Of South Carolina Expression of Anti-Nociceptive Compounds from Endogenously Regulated Promoters
EP2432499A2 (en) 2009-05-20 2012-03-28 Schering Corporation Modulation of pilr receptors to treat microbial infections
TWI507204B (en) 2009-05-26 2015-11-11 Oncotherapy Science Inc Cdc45l peptides and vaccines including the same
WO2010141312A2 (en) 2009-06-01 2010-12-09 Wake Forest University Health Sciences Flagellin fusion proteins and conjugates comprising pneumococcus antigens and methods of using the same
MX340541B (en) 2009-06-05 2016-07-13 Alblynx Nv Monovalent, bivalent and trivalent anti human respiratory syncytial virus (hrsv) nanobody constructs for the prevention and/or treatment of respiratory tract infections.
KR101813464B1 (en) 2009-06-05 2018-01-30 셀룰러 다이내믹스 인터내셔널, 인코포레이티드 Reprogramming T cells and hematopoietic cells
US8637482B2 (en) 2009-06-08 2014-01-28 Quark Pharmaceuticals, Inc. Methods for treating chronic kidney disease
WO2010146511A1 (en) 2009-06-17 2010-12-23 Pfizer Limited Treatment of overactive bladder
US9018187B2 (en) 2009-07-01 2015-04-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
ES2613498T3 (en) 2009-07-01 2017-05-24 Protiva Biotherapeutics Inc. New lipid formulations for the delivery of therapeutic agents to solid tumors
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
EP2451475A2 (en) 2009-07-06 2012-05-16 Novartis AG Self replicating rna molecules and uses thereof
ES2662716T3 (en) 2009-07-07 2018-04-09 Glaxosmithkline Biologicals Sa Preserved immunogens of Escherichia coli
US20120108514A1 (en) 2009-07-09 2012-05-03 University Of Iowa Research Foundation Long acting atrial natriuretic peptide (la-anp) and methods for use thereof
ES2526996T3 (en) 2009-07-16 2015-01-19 Novartis Ag Detoxified immunogens from Escherichia coli
US8329724B2 (en) 2009-08-03 2012-12-11 Hoffmann-La Roche Inc. Process for the manufacture of pharmaceutically active compounds
WO2011015572A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
WO2011015573A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
WO2011022589A2 (en) * 2009-08-20 2011-02-24 Mayo Foundation For Medical Education And Research Detection of enterovirus
US20110053803A1 (en) 2009-08-26 2011-03-03 Xin Ge Methods for creating antibody libraries
US20110059111A1 (en) 2009-09-01 2011-03-10 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Mammalian receptors as targets for antibody and active vaccination therapy against mold infections
AR078253A1 (en) 2009-09-02 2011-10-26 Boehringer Ingelheim Vetmed METHODS TO REDUCE ANTIVIRICAL ACTIVITY IN PCV-2 COMPOSITIONS AND PCV-2 COMPOSITIONS WITH BETTER IMMUNOGENICITY
GB0915794D0 (en) 2009-09-09 2009-10-07 Ucl Business Plc Screening method and treatment
JP2013504600A (en) 2009-09-14 2013-02-07 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Vaccine and immunotherapeutic agent comprising IL-15 receptor α and / or nucleic acid molecule encoding the same, and method of using the same
KR101838472B1 (en) 2009-09-14 2018-03-15 신라젠(주) Oncolytic vaccinia virus combination cancer therapy
WO2011043823A2 (en) * 2009-10-09 2011-04-14 Georgetown University Polypeptides that home to atherosclerotic plaque
MX337982B (en) 2009-10-16 2016-03-30 Scripps Research Inst Induction of pluripotent cells.
WO2011066048A1 (en) 2009-10-22 2011-06-03 Thomas Jefferson University Cell-based anti-cancer compositions and methods of making and using the same
KR101851699B1 (en) 2009-11-02 2018-04-24 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Foot and mouth disease virus (fmdv) consensus proteins, coding sequences therefor and vaccines made therefrom
NZ629615A (en) 2009-11-06 2016-01-29 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor
GB0919690D0 (en) 2009-11-10 2009-12-23 Guy S And St Thomas S Nhs Foun compositions for immunising against staphylococcus aureus
US10016617B2 (en) 2009-11-11 2018-07-10 The Trustees Of The University Of Pennsylvania Combination immuno therapy and radiotherapy for the treatment of Her-2-positive cancers
EP2502076B1 (en) 2009-11-20 2018-04-18 Oregon Health and Science University Methods for detecting a mycobacterium tuberculosis infection
CA2776568A1 (en) 2009-11-26 2011-06-03 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
WO2011064382A1 (en) 2009-11-30 2011-06-03 Ablynx N.V. Improved amino acid sequences directed against human respiratory syncytial virus (hrsv) and polypeptides comprising the same for the prevention and/or treatment of respiratory tract infections
AU2010326132B9 (en) 2009-12-01 2014-10-02 Translate Bio, Inc. Delivery of mRNA for the augmentation of proteins and enzymes in human genetic diseases
CA2780069C (en) 2009-12-08 2018-07-17 Abbott Gmbh & Co. Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
EP2510098B1 (en) 2009-12-09 2015-02-11 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the cns
MX365946B (en) 2009-12-10 2019-06-19 Turnstone Lp Oncolytic rhabdovirus.
TW201136604A (en) 2009-12-14 2011-11-01 Oncotherapy Science Inc TMEM22 peptides and vaccines including the same
WO2011084357A1 (en) 2009-12-17 2011-07-14 Schering Corporation Modulation of pilr to treat immune disorders
TW201125583A (en) * 2009-12-23 2011-08-01 Bioalliance Cv Anti-EpCAM antibodies that induce apoptosis of cancer cells and methods using same
WO2011084193A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
US8298820B2 (en) 2010-01-26 2012-10-30 The Trustees Of The University Of Pennsylvania Influenza nucleic acid molecules and vaccines made therefrom
WO2011097640A1 (en) 2010-02-08 2011-08-11 The Trustees Of The University Of Pennsylvania Nucleic acid molecules encoding rantes, and compositions comprising and methods of using the same
WO2011100508A2 (en) 2010-02-12 2011-08-18 Arizona Board Of Regents For And On Behalf Of Arizona State University Methods and compositions related to glycoprotein-immunoglobulin fusions
GB201002559D0 (en) 2010-02-15 2010-03-31 Circassia Ltd Birch peptides for vaccine
US8298535B2 (en) 2010-02-24 2012-10-30 Rinat Neuroscience Corp. Anti-IL-7 receptor antibodies
EP2538943B1 (en) 2010-02-25 2016-03-30 Dana-Farber Cancer Institute, Inc. Braf mutations conferring resistance to braf inhibitors
GB201003333D0 (en) 2010-02-26 2010-04-14 Novartis Ag Immunogenic proteins and compositions
US20130052221A1 (en) 2010-02-26 2013-02-28 The Govt. of the U.S, as represented by The Sec. of The Dept. of Health and Human Services Dna-protein vaccination protocols
WO2011108930A1 (en) 2010-03-04 2011-09-09 Interna Technologies Bv A MiRNA MOLECULE DEFINED BY ITS SOURCE AND ITS DIAGNOSTIC AND THERAPEUTIC USES IN DISEASES OR CONDITIONS ASSOCIATED WITH EMT
US9068011B2 (en) 2010-03-10 2015-06-30 Genmab A+S Monoclonal antibodies against c-Met
PE20130393A1 (en) 2010-03-11 2013-04-07 Rinat Neuroscience Corp ANTIBODIES WITH PH-DEPENDENT ANTIGEN UNION
BR112012022641A2 (en) 2010-03-11 2017-02-14 Oncotherapy Science Inc hjurp peptides and vaccines that include the same
WO2011112599A2 (en) 2010-03-12 2011-09-15 The United States Of America, As Represented By The Secretary. Department Of Health & Human Services Immunogenic pote peptides and methods of use
GB201004475D0 (en) 2010-03-17 2010-05-05 Isis Innovation Gene silencing
AU2011227050B2 (en) 2010-03-19 2016-12-08 University Of South Alabama Methods and compositions for the treatment of cancer
US20130195800A1 (en) 2010-03-23 2013-08-01 Intrexon Corporation Vectors Conditionally Expressing Therapeutic Proteins, Host Cells Comprising the Vectors, and Uses Thereof
EP2550362B1 (en) 2010-03-25 2017-01-04 Oregon Health&Science University Cmv glycoproteins and recombinant vectors
EP3199623B1 (en) 2010-03-31 2021-07-28 The Scripps Research Institute Reprogramming cells
GB201005625D0 (en) 2010-04-01 2010-05-19 Novartis Ag Immunogenic proteins and compositions
TWI653333B (en) 2010-04-01 2019-03-11 安進研究(慕尼黑)有限責任公司 Cross-species specific PSMAxCD3 bispecific single chain antibody
TWI538685B (en) 2010-04-02 2016-06-21 腫瘤療法 科學股份有限公司 Ect2 peptides and vaccines including the same
JP2013523818A (en) 2010-04-05 2013-06-17 ザ・ユニバーシティー・オブ・シカゴ Compositions and methods relating to protein A (SpA) antibodies as enhancers of immune responses
WO2011126976A1 (en) 2010-04-07 2011-10-13 Vanderbilt University Reovirus vaccines and methods of use therefor
CA2796601C (en) 2010-04-19 2019-03-26 Research Development Foundation Rtef-1 variants and uses thereof
WO2011133931A1 (en) 2010-04-22 2011-10-27 Genentech, Inc. Use of il-27 antagonists for treating inflammatory bowel disease
EP3699266A1 (en) 2010-05-14 2020-08-26 The General Hospital Corporation Neoantigen specific cytotoxic t cells for use in treating cancer
GB201008267D0 (en) 2010-05-18 2010-06-30 Univ Edinburgh Cationic lipids
CA3051311A1 (en) 2010-05-27 2011-12-01 Genmab A/S Monoclonal antibodies against her2
JP6082344B2 (en) 2010-05-27 2017-02-15 ゲンマブ エー/エス Monoclonal antibody against HER2 epitope
WO2011156588A1 (en) 2010-06-09 2011-12-15 Dana-Farber Cancer Institute, Inc. A mek 1 mutation conferring resistance to raf and mek inhibitors
WO2011154453A1 (en) 2010-06-09 2011-12-15 Genmab A/S Antibodies against human cd38
EP2580320B1 (en) 2010-06-14 2018-08-01 The Scripps Research Institute Reprogramming of cells to a new fate
WO2011157741A2 (en) 2010-06-15 2011-12-22 Genmab A/S Human antibody drug conjugates against tissue factor
ES2670842T3 (en) 2010-06-15 2018-06-01 Cellular Dynamics International, Inc. Generation of induced pluripotent stem cells from small volumes of peripheral blood
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
CA2801928C (en) 2010-06-24 2018-04-10 Quark Pharmaceuticals, Inc. Double stranded rna compounds to rhoa and use thereof
JP2013532971A (en) 2010-06-25 2013-08-22 バッシボディ アクスイェ セルスカプ Homodimeric protein construct
US9006417B2 (en) 2010-06-30 2015-04-14 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
JP6002128B2 (en) 2010-07-02 2016-10-05 ザ・ユニバーシティ・オブ・シカゴThe University Of Chicago Compositions and methods related to protein A (SpA) variants
NZ719520A (en) 2010-07-06 2017-07-28 Int Tech Bv Mirna and its diagnostic and therapeutic uses in diseases or conditions associated with melanoma, or in diseases or conditions associated with activated braf pathway
CA2804595C (en) 2010-07-07 2018-11-13 Cellular Dynamics International, Inc. Endothelial cell production by programming
US8747844B2 (en) 2010-07-30 2014-06-10 Saint Louis University Methods of treating pain
CA2806858C (en) 2010-08-04 2021-06-15 Cellular Dynamics International, Inc. Reprogramming immortalized b cells
GB201013153D0 (en) 2010-08-04 2010-09-22 Touchlight Genetics Ltd Primer for production of closed linear DNA
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
GB201014026D0 (en) 2010-08-20 2010-10-06 Ucl Business Plc Treatment
ES2850973T3 (en) 2010-08-23 2021-09-01 Wyeth Llc Stable formulations of rLP2086 antigens from Neisseria meningitidis
WO2012025759A2 (en) 2010-08-26 2012-03-01 Isis Innovation Limited Method
CA2809127C (en) 2010-08-31 2019-04-02 Merial Limited Newcastle disease virus vectored herpesvirus vaccines
US20140023683A1 (en) 2010-09-08 2014-01-23 The Uab Research Foundation Identification of transmitted hepatitis c virus (hcv) genomes by single genome amplification
US9095540B2 (en) 2010-09-09 2015-08-04 The University Of Chicago Methods and compositions involving protective staphylococcal antigens
ES2585328T5 (en) 2010-09-10 2022-12-14 Wyeth Llc Non-lipidated variants of Neisseria meningitidis ORF2086 antigens
US9084746B2 (en) 2010-09-22 2015-07-21 The Regents Of The University Of Colorado, A Body Corporate Therapeutic applications of SMAD7
EP2621522A2 (en) 2010-09-27 2013-08-07 China Agricultural University Combined antigen and dna vaccine for preventing and treating autoimmune diseases
AU2011308567B2 (en) 2010-10-01 2015-09-03 Fundacion Centro Nacional De Investigaciones Oncologicas, Carlos Iii Manipulation of stem cell function by p53 isoforms
WO2012138377A2 (en) 2010-10-01 2012-10-11 Trustees Of The University Of Pennsylvania The use of listeria vaccine vectors to reverse vaccine unresponsiveness in parasitically infected individuals
EP4108671A1 (en) 2010-10-01 2022-12-28 ModernaTX, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2012052748A1 (en) 2010-10-18 2012-04-26 Isis Innovation Limited Method for immunising a subject against mycobacterium tuberculosis or mycobacterium bovis
AR083533A1 (en) 2010-10-22 2013-03-06 Boehringer Ingelheim Vetmed PROTEINS OF HEMAGLUTININ 5 (H5) FOR THE TREATMENT AND PREVENTION OF INFLECTIONS OF FLU
WO2012056008A1 (en) 2010-10-28 2012-05-03 Jonas Nilsson Diagnosis and treatment of alzheimer's disease
US9539427B2 (en) 2010-11-08 2017-01-10 The Johns Hopkins University Methods for improving heart function
CA2817709C (en) 2010-11-12 2021-06-01 The Trustees Of The University Of Pennsylvania Consensus prostate antigens, nucleic acid molecule encoding the same and vaccine and uses comprising the same
US20120141537A1 (en) 2010-11-18 2012-06-07 Pease Larry R Enhancing t cell activation using altered mhc-peptide ligands
WO2012075337A2 (en) 2010-12-01 2012-06-07 Spinal Modulation, Inc. Directed delivery of agents to neural anatomy
WO2012072769A1 (en) 2010-12-01 2012-06-07 Novartis Ag Pneumococcal rrgb epitopes and clade combinations
WO2012078536A2 (en) 2010-12-06 2012-06-14 Quark Pharmaceuticals, Inc. Double stranded oligonucleotide compounds comprising positional modifications
AP2013006918A0 (en) 2010-12-09 2013-06-30 Univ Pennsylvania Use of chimeric antigen receptormodified T-cells to treat cancer
EP2655601A4 (en) 2010-12-22 2014-09-10 Fate Therapeutics Inc Cell culture platform for single cell sorting and enhanced reprogramming of ipscs
WO2012090073A2 (en) 2010-12-30 2012-07-05 The Netherlands Cancer Institute Methods and compositions for predicting chemotherapy sensitivity
JP6121910B2 (en) 2011-01-04 2017-04-26 シラジェン バイオセラピューティクス インコーポレイテッド Generation of antibodies against tumor antigens and tumor-specific complement-dependent cytotoxicity by administration of oncolytic vaccinia virus
EP2474617A1 (en) 2011-01-11 2012-07-11 InteRNA Technologies BV Mir for treating neo-angiogenesis
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
US9428565B2 (en) 2011-01-31 2016-08-30 The General Hospital Corporation Treatment and bioluminescent visualization using multimodal TRAIL molecules
JP6099573B2 (en) 2011-01-31 2017-03-22 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Nucleic acid molecules encoding novel herpes antigens, vaccines containing them and methods of use thereof
KR20130112950A (en) 2011-02-04 2013-10-14 조에티스 엘엘씨 Compositions for canine respiratory disease complex
PL2670432T3 (en) 2011-02-04 2021-10-11 Zoetis Services Llc Immunogenic bordetella bronchiseptica compositions
JP5941069B2 (en) 2011-02-07 2016-06-29 プレキシコン インコーポレーテッドPlexxikon Inc. Compounds and methods for kinase regulation and indications therefor
EP2673299B1 (en) 2011-02-07 2017-05-10 Research Development Foundation Engineered immunoglobulin fc polypeptides
US9574179B2 (en) 2011-02-08 2017-02-21 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
EA032364B1 (en) 2011-02-11 2019-05-31 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания Nucleic acid molecule encoding hepatitis b virus core protein and vaccine comprising the same
US9238679B2 (en) 2011-02-11 2016-01-19 The Trustees Of The University Of Pennslyvania Nucleic acid molecule encoding hepatitis B virus core protein and surface antigen protein and vaccine comprising the same
AR085279A1 (en) 2011-02-21 2013-09-18 Plexxikon Inc SOLID FORMS OF {3- [5- (4-CHLORINE-PHENYL) -1H-PIRROLO [2,3-B] PIRIDINA-3-CARBONIL] -2,4-DIFLUOR-PHENIL} -AMIDE OF PROPANE ACID-1- SULFONIC
EP2492279A1 (en) 2011-02-25 2012-08-29 Laboratorios Del. Dr. Esteve, S.A. Rapid immunogen selection method using lentiviral display
US9796979B2 (en) 2011-03-03 2017-10-24 Quark Pharmaceuticals Inc. Oligonucleotide modulators of the toll-like receptor pathway
CN103562387A (en) 2011-03-03 2014-02-05 夸克医药公司 Oligonucleotide modulators of the toll-like receptor pathway
US9402919B2 (en) 2011-03-04 2016-08-02 Intrexon Corporation Vectors conditionally expressing protein
AU2012229218B2 (en) 2011-03-11 2017-03-02 Advaxis, Inc. Listeria-based adjuvants
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
AU2012234259A1 (en) 2011-03-31 2013-10-03 Libera-Korner, Jeanette Perfluorinated compounds for the non-viral transfer of nucleic acids
US9458456B2 (en) 2011-04-01 2016-10-04 University Of South Alabama Methods and compositions for the diagnosis, classification, and treatment of cancer
WO2012138789A2 (en) 2011-04-04 2012-10-11 Netherlands Cancer Institute Methods and compositions for predicting resistance to anticancer treatment
WO2012138783A2 (en) 2011-04-04 2012-10-11 Netherlands Cancer Institute Methods and compositions for predicting resistance to anticancer treatment
US9499610B2 (en) 2011-04-08 2016-11-22 H. Lundbeck A/S Antibodies specific to pyroglutamated Aβ
US9085631B2 (en) 2011-04-08 2015-07-21 Nov Vac APS Proteins and nucleic acids useful in vaccines targeting Staphylococcus aureus
AU2012245395A1 (en) 2011-04-20 2013-11-14 Merial, Inc. Adjuvanted rabies vaccine with improved viscosity profile
JP6177231B2 (en) 2011-04-20 2017-08-09 ゲンマブ エー/エス Bispecific antibody against HER2
WO2012143524A2 (en) 2011-04-20 2012-10-26 Genmab A/S Bispecific antibodies against her2 and cd3
CA2834288A1 (en) 2011-04-25 2012-11-01 Advanced Bioscience Laboratories, Inc. Truncated hiv envelope proteins (env), methods and compositions related thereto
US8945588B2 (en) 2011-05-06 2015-02-03 The University Of Chicago Methods and compositions involving protective staphylococcal antigens, such as EBH polypeptides
RU2631487C2 (en) 2011-05-17 2017-09-22 Плексксикон Инк. Kinases modulation and indications for its use
ES2931180T3 (en) 2011-05-19 2022-12-27 Fund Publica Andaluza Progreso Y Salud Highly inducible dual promoter lentiviral Tet-on type system
AU2012261237B2 (en) 2011-05-24 2017-06-01 BioNTech SE Individualized vaccines for cancer
US9034581B2 (en) 2011-05-26 2015-05-19 Roche Molecular Systems, Inc. Compositions and methods for detection of Staphylococcus aureus
WO2012166493A1 (en) 2011-06-01 2012-12-06 Merial Limited Needle-free administration of prrsv vaccines
CN103906840B (en) 2011-06-07 2018-01-30 艾比欧公司 Recombinant protein with PNGase F by co-expressing deglycosylation in vivo
KR102128248B1 (en) 2011-06-08 2020-07-01 샤이어 휴먼 지네틱 테라피즈 인크. Lipid nanoparticle compositions and methods for mrna delivery
US9364532B2 (en) 2011-06-08 2016-06-14 Children's Hospital Of Eastern Ontario Research Institute Inc. Compositions and methods for glioblastoma treatment
EP2691530B1 (en) 2011-06-10 2018-03-07 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
US20140275211A1 (en) 2011-06-21 2014-09-18 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
WO2012178118A1 (en) 2011-06-24 2012-12-27 Epitogenesis Inc. Pharmaceutical compositions, comprising a combination of select carriers, vitamins, tannins and flavonoids as antigen-specific immuno-modulators
EP3508219A1 (en) 2011-07-06 2019-07-10 GlaxoSmithKline Biologicals S.A. Self-replicating rna prime - protein boost vaccines
EP3332802A1 (en) 2011-07-06 2018-06-13 GlaxoSmithKline Biologicals SA Immunogenic combination compositions and uses thereof
EP3662935A1 (en) 2011-07-11 2020-06-10 Inovio Pharmaceuticals, Inc. Cross-protective arenavirus vaccines and their method of use
US20130040302A1 (en) 2011-07-11 2013-02-14 Thomas J. Burke Methods for cell reprogramming and genome engineering
EP2546358A1 (en) 2011-07-15 2013-01-16 Laboratorios Del. Dr. Esteve, S.A. Methods and reagents for efficient control of HIV progression
PL2734544T3 (en) 2011-07-18 2021-05-31 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for inhibiting polyomavirus-associated pathology
EP2738255B1 (en) 2011-07-29 2016-11-23 Tokushima University Erap1-derived peptide and use thereof
JP6041361B2 (en) 2011-08-12 2016-12-07 メリアル インコーポレイテッド Vacuum storage of biological products, especially vaccines
WO2013024582A1 (en) 2011-08-12 2013-02-21 Oncotherapy Science, Inc. Mphosph1 peptides and vaccines including the same
JP6317670B2 (en) 2011-08-15 2018-04-25 ザ・ユニバーシティ・オブ・シカゴThe University Of Chicago Compositions and methods related to antibodies to staphylococcal protein A
AR088028A1 (en) 2011-08-15 2014-05-07 Boehringer Ingelheim Vetmed PROTEINS H5, FROM H5N1 FOR MEDICINAL USE
WO2013026015A1 (en) 2011-08-18 2013-02-21 Dana-Farber Cancer Institute, Inc. Muc1 ligand traps for use in treating cancers
US20130071375A1 (en) 2011-08-22 2013-03-21 Saint Louis University Compositions and methods for treating inflammation
WO2013028527A1 (en) 2011-08-23 2013-02-28 Indiana University Research And Technology Corporation Compositions and methods for treating cancer
CN103906761B (en) 2011-08-30 2016-12-21 梅约医学教育与研究基金会 Profit sodium polypeptide
WO2013033092A2 (en) 2011-09-03 2013-03-07 Boehringer Ingelheim Vetmedica Gmbh Streptococcus suis pilus antigens
EP2568289A3 (en) 2011-09-12 2013-04-03 International AIDS Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2013039792A1 (en) 2011-09-12 2013-03-21 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Immunogens based on an hiv-1 gp120 v1v2 epitope
CA2848368C (en) 2011-09-13 2023-02-14 Dana-Farber Cancer Institute, Inc. Compositions and methods for brown fat induction and activity using fndc5
AU2012312520A1 (en) 2011-09-20 2014-05-08 The University Of North Carolina At Chapel Hill Regulation of sodium channels by PLUNC proteins
US9708384B2 (en) 2011-09-22 2017-07-18 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
CN103974724B (en) 2011-10-03 2019-08-30 现代泰克斯公司 Nucleosides, nucleotide and nucleic acid of modification and application thereof
EP3896759A1 (en) 2011-10-05 2021-10-20 OneD Material, Inc. Silicon nanostructure active materials for lithium ion batteries and processes, compositions, components, and devices related thereto
WO2013053765A1 (en) 2011-10-11 2013-04-18 Proyecto De Biomedicina Cima, S.L. A non-human animal model of mucosa-associated lymphoid tissue (malt) lymphoma
EA033242B1 (en) 2011-10-12 2019-09-30 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания Improved nucleic acids and human papilloma virus proteins, vaccines and compositions based thereon and methods of inducing an immune response against hpv using the same
EP2766388A1 (en) 2011-10-12 2014-08-20 Møller, Niels Iversen Peptides derived from campylobacter jejuni and their use in vaccination
WO2013055911A1 (en) 2011-10-14 2013-04-18 Dana-Farber Cancer Institute, Inc. Znf365/zfp365 biomarker predictive of anti-cancer response
JP5898324B2 (en) 2011-10-24 2016-04-06 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Improved HCV vaccine and method of use thereof
US9402894B2 (en) 2011-10-27 2016-08-02 International Aids Vaccine Initiative Viral particles derived from an enveloped virus
EP2771364B1 (en) 2011-10-27 2019-05-22 Genmab A/S Production of heterodimeric proteins
CA3122778A1 (en) 2011-10-28 2013-05-02 Oncotherapy Science, Inc. Topk peptides and vaccines including the same
WO2013070821A1 (en) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the nervous system
SG11201401699WA (en) 2011-11-11 2014-09-26 Rinat Neuroscience Corp Antibodies specific for trop-2 and their uses
ES2894724T3 (en) 2011-12-02 2022-02-15 Rhode Island Hospital Falciparum malaria vaccine
CA2859387A1 (en) 2011-12-16 2013-06-20 Moderna Therapeutics, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013093629A2 (en) 2011-12-20 2013-06-27 Netherlands Cancer Institute Modular vaccines, methods and compositions related thereto
EP2794659A1 (en) 2011-12-22 2014-10-29 Rinat Neuroscience Corp. Human growth hormone receptor antagonist antibodies and methods of use thereof
WO2013093693A1 (en) 2011-12-22 2013-06-27 Rinat Neuroscience Corp. Staphylococcus aureus specific antibodies and uses thereof
EP3369818B1 (en) 2011-12-22 2021-06-09 InteRNA Technologies B.V. Mirna for treating head and neck cancer
US9611305B2 (en) 2012-01-06 2017-04-04 Mayo Foundation For Medical Education And Research Treating cardiovascular or renal diseases
US9464291B2 (en) 2012-01-06 2016-10-11 University Of South Alabama Methods and compositions for the treatment of cancer
DK3381461T3 (en) 2012-01-09 2021-05-17 Serpin Pharma Llc PEPTIDES AND PROCEDURES FOR USING IT
EP2802657B1 (en) 2012-01-12 2018-05-02 Quark Pharmaceuticals, Inc. Combination therapy for treating hearing and balance disorders
ES2676725T3 (en) 2012-01-27 2018-07-24 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of diseases associated with the degeneration of neurites
KR102091297B1 (en) 2012-02-03 2020-03-20 에프. 호프만-라 로슈 아게 Bispecific antibody molecules with antigen-transfected t-cells and their use in medicine
CN103239734B (en) 2012-02-10 2016-02-24 北京艾棣维欣生物技术有限公司 For preventing and/or treating the vaccine of respiratory syncytial virus infection
KR20140135715A (en) 2012-02-22 2014-11-26 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Use of icos-based cars to enhance antitumor activity and car persistence
EP4275699A3 (en) 2012-02-22 2024-01-03 The Trustees of the University of Pennsylvania Use of the cd2 signaling domain in second-generation chimeric antigen receptors
BR112014020499A2 (en) 2012-02-22 2019-09-24 Univ Pennsylvania isolated nucleic acid sequence, t-cell, vector, and persistent t-cell population
MX2018011291A (en) 2012-03-09 2023-01-31 Pfizer Neisseria meningitidis compositions and methods thereof.
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
SG10201700392UA (en) 2012-03-12 2017-03-30 Advaxis Inc Suppressor cell function inhibition following listeria vaccine treatment
WO2013138776A1 (en) 2012-03-16 2013-09-19 Merial Limited Novel methods for providing long-term protective immunity against rabies in animals, based upon administration of replication-deficient flavivirus expressing rabies g
RS58794B1 (en) * 2012-03-26 2019-07-31 Biontech Rna Pharmaceuticals Gmbh Rna formulation for immunotherapy
WO2013143555A1 (en) * 2012-03-26 2013-10-03 Biontech Ag Rna formulation for immunotherapy
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
AU2013243951A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
MX363667B (en) 2012-04-10 2019-03-29 Univ Pennsylvania Human respiratory syncytial virus consensus antigens, nucleic acid constructs and vaccines made therefrom, and methods of using same.
BR112014026861A2 (en) 2012-04-26 2018-05-15 Univ Chicago coagulase staphylococcal antigens and methods of use
CN104703622B (en) 2012-04-26 2017-05-24 芝加哥大学 Compositions and methods related to antibodies that neutralize coagulase activity during staphylococcus aureus disease
PE20142406A1 (en) 2012-05-04 2015-01-23 Pfizer ANTIGENS ASSOCIATED WITH PROSTATE AND VACCINE-BASED IMMUNOTHERAPY REGIMES
US9150570B2 (en) 2012-05-31 2015-10-06 Plexxikon Inc. Synthesis of heterocyclic compounds
US10632189B2 (en) 2012-05-31 2020-04-28 Zoetis Services Llc Canine respiratory coronavirus for treatment and protection against bacterial infections
US20150267192A1 (en) 2012-06-08 2015-09-24 Shire Human Genetic Therapies, Inc. Nuclease resistant polynucleotides and uses thereof
EP2679596B1 (en) 2012-06-27 2017-04-12 International Aids Vaccine Initiative HIV-1 env glycoprotein variant
CA2877948C (en) 2012-07-02 2019-07-30 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Paramyxovirus and methods of use
US11180572B2 (en) 2012-07-06 2021-11-23 Genmab B.V. Dimeric protein with triple mutations
CN104853770A (en) 2012-07-06 2015-08-19 诺华股份有限公司 Immunogenic compositions and uses thereof
WO2014010232A1 (en) 2012-07-10 2014-01-16 Oncotherapy Science, Inc. Ly6k epitope peptides for th1 cells and vaccines containing the same
WO2014010231A1 (en) 2012-07-10 2014-01-16 Oncotherapy Science, Inc. Kif20a epitope peptides for th1 cells and vaccines containing the same
BR112015000638A2 (en) 2012-07-13 2017-08-08 Univ Pennsylvania isolated nucleic acid sequence, cell, methods for stimulating a cell-mediated immune response to a target cell or tissue population in a mammal, for providing antitumor immunity in a mammal, for treating a mammal having a disease, disorder or condition associated with high expression of a tumor antigen
IN2014DN11156A (en) 2012-07-13 2015-10-02 Univ Pennsylvania
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
EP2698377A1 (en) 2012-08-17 2014-02-19 Laboratorios Del. Dr. Esteve, S.A. Enhanced rapid immunogen selection method for HIV gp120 variants
US9982313B2 (en) 2012-08-17 2018-05-29 Roche Molecular Systems, Inc. Compositions and methods for detection of herpes simplex virus 1 and 2
WO2014035474A1 (en) 2012-08-30 2014-03-06 The General Hospital Corporation Compositions and methods for treating cancer
WO2014039513A2 (en) 2012-09-04 2014-03-13 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive t cell transfer
JP6283861B2 (en) 2012-09-11 2018-02-28 オンコセラピー・サイエンス株式会社 UBE2T peptide and vaccine containing the same
ES2704855T3 (en) 2012-09-12 2019-03-20 Quark Pharmaceuticals Inc Double chain oligonucleotide molecules for p53 and methods of using them
WO2014043289A2 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
ES2752190T3 (en) 2012-09-14 2020-04-03 Us Health Brachyury protein, adenoviral vectors encoding Brachyury protein and their use
EP2897639A4 (en) 2012-09-21 2016-05-04 Frank Bedu-Addo Improved vaccine compositions and methods of use
ES2792124T3 (en) 2012-09-26 2020-11-10 Benchmark Animal Health Ltd Subunit Immersion Vaccines for Fish
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
WO2014065945A1 (en) 2012-10-23 2014-05-01 The Board Of Regents Of The University Of Texas System Antibodies with engineered igg fc domains
EP2917348A1 (en) 2012-11-06 2015-09-16 InteRNA Technologies B.V. Combination for use in treating diseases or conditions associated with melanoma, or treating diseases or conditions associated with activated b-raf pathway
AU2013343099A1 (en) 2012-11-09 2015-05-14 Pfizer Inc. Platelet-derived growth factor B specific antibodies and compositions and uses thereof
CN104244973B (en) 2012-11-16 2017-10-20 美国联合生物医学公司 For the urgent vaccine based on synthetic peptide of aftosa (FMD)
JP6144355B2 (en) 2012-11-26 2017-06-07 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chemically modified mRNA
JP6484558B2 (en) 2012-11-28 2019-03-13 バイオエヌテック エールエヌアー ファーマシューティカルズ ゲーエムベーハーBiontech Rna Pharmaceuticals Gmbh Combination of cancer vaccine
ES2656510T3 (en) 2012-11-30 2018-02-27 Glaxosmithkline Biologicals S.A. Pseudomonas antigens and antigen combination
WO2014093702A1 (en) 2012-12-12 2014-06-19 The Usa, As Represented By The Secretary, Department Of Health And Human Services Hiv therapeutics and methods of making and using same
SG10201707569YA (en) 2012-12-12 2017-10-30 Broad Inst Inc Delivery, Engineering and Optimization of Systems, Methods and Compositions for Sequence Manipulation and Therapeutic Applications
US10220082B2 (en) 2012-12-13 2019-03-05 Inovio Pharmaceuticals, Inc. WT1 vaccine
WO2014107739A1 (en) 2013-01-07 2014-07-10 Eleven Biotherapeutics, Inc. Antibodies against pcsk9
WO2014116721A1 (en) 2013-01-22 2014-07-31 The Arizona Board Of Regents For And On Behalf Of Arizona State University Geminiviral vector for expression of rituximab
TWI682941B (en) 2013-02-01 2020-01-21 美商再生元醫藥公司 Antibodies comprising chimeric constant domains
WO2014120975A1 (en) 2013-02-01 2014-08-07 California Institute Of Technology Antibody-mediated immunocontraception
US9534041B2 (en) 2013-02-12 2017-01-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that neutralize a norovirus
US9938334B2 (en) 2013-02-15 2018-04-10 Mayo Foundation For Medical Education And Research Insulin secreting polypeptides
CN105121631B (en) 2013-02-20 2019-04-19 瑞泽恩制药公司 The genetic modification of rat
UY35340A (en) 2013-02-20 2014-09-30 Novartis Ag EFFECTIVE MARKING OF HUMAN LEUKEMIA USING CELLS DESIGNED WITH AN ANTIGEN CHEMERIC RECEIVER ANTI-CD123
SG11201505896YA (en) 2013-02-20 2015-09-29 Novartis Ag Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
RU2684211C2 (en) 2013-02-21 2019-04-04 Тёрнстоун Лимитед Партнершип Vaccine composition
WO2014127825A1 (en) 2013-02-21 2014-08-28 Boehringer Ingelheim Vetmedica Gmbh H5 proteins of h5n1 influenza virus for use as a medicament
US20140242595A1 (en) 2013-02-22 2014-08-28 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US10086093B2 (en) 2013-02-28 2018-10-02 The General Hospital Corporation miRNA profiling compositions and methods of use
US20160010094A1 (en) 2013-03-01 2016-01-14 University Of Pretoria Transgenic cell selection
US8957044B2 (en) 2013-03-01 2015-02-17 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
EP2964665B1 (en) 2013-03-08 2018-08-01 Pfizer Inc Immunogenic fusion polypeptides
EP2964774B1 (en) 2013-03-08 2020-05-06 The Regents of The University of Colorado, A Body Corporate Ptd-smad7 therapeutics
WO2014164697A1 (en) 2013-03-12 2014-10-09 Merial Limited Reverse genetics schmallenberg virus vaccine compositions, and methods of use thereof
TWI658049B (en) 2013-03-12 2019-05-01 腫瘤療法 科學股份有限公司 Kntc2 peptides and vaccines containing the same
US10227369B2 (en) 2013-03-12 2019-03-12 The Johns Hopkins University Short-chain fatty acid hexosamine analogs and their use in tissue engineering applications
EA034056B1 (en) 2013-03-12 2019-12-23 Дзе Трастиз Оф Дзе Юниверсити Оф Пенсильвания Improved vaccines for human papilloma virus and methods for using the same
PL2968470T3 (en) 2013-03-12 2021-05-31 The General Hospital Corporation Modified mullerian inhibiting substance (mis) proteins and uses thereof for the treatment of diseases
JP2016522675A (en) 2013-03-14 2016-08-04 ガラパゴス・ナムローゼ・フェンノートシャップGalapagos N.V. Molecular targets useful in the treatment of diseases associated with epithelial-mesenchymal transition and inhibitors of said targets
EP2972322B1 (en) 2013-03-14 2019-03-06 Galapagos NV Molecular targets and compounds, and methods to identify the same, useful in the treatment of fibrotic diseases
WO2014152955A1 (en) 2013-03-14 2014-09-25 Regeneron Pharmaceuticals, Inc. Apelin fusion proteins and uses thereof
KR20150128687A (en) 2013-03-14 2015-11-18 샤이어 휴먼 지네틱 테라피즈 인크. Methods for purification of messenger rna
JP2016518812A (en) 2013-03-14 2016-06-30 ガラパゴス・ナムローゼ・フェンノートシャップGalapagos N.V. Molecular targets useful in the treatment of fibrosis and inhibitors of said targets
BR112015022868B1 (en) 2013-03-14 2023-05-16 Ethris Gmbh CFTR MRNA COMPOSITIONS AND RELATED USES AND METHODS
BR112015022367B1 (en) 2013-03-15 2021-06-22 The Trustees Of The University Of Pennsylvania VACCINE, NUCLEIC ACID MOLECULE, AND AMINO ACID MOLECULE
KR102276405B1 (en) 2013-03-15 2021-07-12 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 Foot and mouth disease virus (fmdv) consensus proteins, coding sequences therefor and vaccines made therefrom
CN105051068A (en) 2013-03-15 2015-11-11 戴埃克斯有限公司 Anti-plasma kallikrein antibodies
WO2014144466A1 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
WO2014143739A2 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP2968606B1 (en) 2013-03-15 2020-10-07 Loma Linda University Treatment of autoimmune diseases
TWI654206B (en) 2013-03-16 2019-03-21 諾華公司 Treatment of cancer with a humanized anti-CD19 chimeric antigen receptor
WO2014153636A1 (en) 2013-03-27 2014-10-02 Immunovaccine Technologies Inc. Method for improving the efficacy of a survivin vaccine in the treatment of cancer
CN105377292A (en) 2013-04-07 2016-03-02 博德研究所 Compositions and methods for personalized neoplasia vaccines
CA3182572A1 (en) 2013-04-17 2014-10-23 Genzyme Corporation Use of an il17 inhibitor for treating and preventing macular degeneration
RU2015147721A (en) 2013-05-07 2017-06-15 Ринат Нейросаенз Корпорэйшн ANTIBODIES AGAINST GLUCAGON RECEPTOR AND METHODS OF USING THEM
EP2994163B1 (en) 2013-05-09 2019-08-28 The United States of America, as represented by The Secretary, Department of Health and Human Services Single-domain vhh antibodies directed to norovirus gi.1 and gii.4 and their use
WO2014180490A1 (en) 2013-05-10 2014-11-13 Biontech Ag Predicting immunogenicity of t cell epitopes
EP2996697B1 (en) 2013-05-15 2019-06-26 Robert Kruse Intracellular translation of circular rna
CA2913155A1 (en) 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
US20160130585A1 (en) 2013-05-28 2016-05-12 The Johns Hopkins University Aptamers for the treatment of sickle cell disease
US10364451B2 (en) 2013-05-30 2019-07-30 Duke University Polymer conjugates having reduced antigenicity and methods of using the same
WO2014194244A1 (en) 2013-05-30 2014-12-04 Duke University Enzyme-catalyzed synthesis of site-specific and stoichiometric biomolecule-polymer conjugates
US10392611B2 (en) 2013-05-30 2019-08-27 Duke University Polymer conjugates having reduced antigenicity and methods of using the same
US9982236B2 (en) 2013-06-13 2018-05-29 Orgenesis Ltd. Cell populations, methods of transdifferentiation and methods of use thereof
KR20160019553A (en) 2013-06-17 2016-02-19 더 브로드 인스티튜트, 인코퍼레이티드 Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
MX2015017312A (en) 2013-06-17 2017-04-10 Broad Inst Inc Delivery and use of the crispr-cas systems, vectors and compositions for hepatic targeting and therapy.
EP3597755A1 (en) 2013-06-17 2020-01-22 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
BR122023002590B1 (en) 2013-07-05 2023-12-05 Genmab A/S HUMANIZED OR CHIMERICAL ANTIBODY BINDING TO HUMAN CD3, BIESPECIFIC ANTIBODY, MICROORGANISM, COMPOSITION, PHARMACEUTICAL COMPOSITION, USE THEREOF, METHOD FOR PRODUCING AN ANTIBODY, DIAGNOSTIC COMPOSITION, IN VITRO METHOD FOR DETECTING THE PRESENCE OF CD3 ANTIGEN, OR AN EXP CELL RESASTING CD3 , IN A SAMPLE, KIT TO DETECT THE PRESENCE OF CD3 ANTIGEN, OR A CELL THAT EXPRESSES CD3, IN A SAMPLE AND ANTI-IDIOTYPIC ANTIBODY
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
WO2015009787A1 (en) 2013-07-19 2015-01-22 The Johns Hopkins University Biomaterials comprising hyaluronic acid binding peptides and extracellular matrix binding peptides for hyaluronic acid retention and tissue engineering applications
CA2919835A1 (en) 2013-07-31 2015-02-05 Board Of Trustees Of The University Of Arkansas Compositions for and methods of treating and preventing cancer targeting tumor associated carbohydrate antigens
AR097102A1 (en) 2013-08-02 2016-02-17 Pfizer ANTI-CXCR4 ANTIBODY AND ANTIBODY AND DRUG CONJUGATES
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US20150065381A1 (en) 2013-09-05 2015-03-05 International Aids Vaccine Initiative Methods of identifying novel hiv-1 immunogens
KR101905278B1 (en) 2013-09-08 2018-10-08 화이자 인코포레이티드 Neisseria meningitidis compositions and methods thereof
US11819555B2 (en) 2013-09-09 2023-11-21 Figene, Llc Gene therapy for the regeneration of chondrocytes or cartilage type cells
KR102433548B1 (en) 2013-09-16 2022-08-18 체엠엠 - 포르슝스첸트룸 퓨어 몰레쿨라레 메디친 게엠베하 Mutant calreticulin for the diagnosis of myeloid malignancies
EP3049106A1 (en) 2013-09-25 2016-08-03 Zoetis Services LLC Pcv2b divergent vaccine composition and methods of use
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
AU2014332352B9 (en) 2013-10-07 2017-05-11 Inovio Pharmaceuticals, Inc. Vaccines with Interleukin-33 as an adjuvant
KR102358807B1 (en) 2013-10-11 2022-02-08 리제너론 파마슈티칼스 인코포레이티드 Metabolically optimized cell culture
JP6502931B2 (en) 2013-10-11 2019-04-17 アメリカ合衆国 TEM 8 antibody and use thereof
EP3060258A1 (en) 2013-10-22 2016-08-31 Shire Human Genetic Therapies, Inc. Mrna therapy for phenylketonuria
SG11201602452SA (en) 2013-10-22 2016-05-30 Viromed Co Ltd Composition for preventing or treating amyotrophic lateral sclerosis using two or more isoforms of hepatocyte growth factor
EP3501605B1 (en) 2013-10-22 2023-06-28 Translate Bio, Inc. Mrna therapy for argininosuccinate synthetase deficiency
ES2806575T3 (en) 2013-11-01 2021-02-18 Curevac Ag Modified RNA with decreased immunostimulatory properties
WO2015070027A1 (en) 2013-11-07 2015-05-14 University Of Southern California Use of ikk epsilon inhibitors to activate nfat and t cell response
WO2015070009A2 (en) 2013-11-08 2015-05-14 The Board Of Regents Of The University Of Texas System Vh4 antibodies against gray matter neuron and astrocyte
US10028902B2 (en) 2013-11-08 2018-07-24 Baylor Research Institute Nuclear localization of GLP-1 stimulates myocardial regeneration and reverses heart failure
US10072288B2 (en) 2013-11-11 2018-09-11 Roche Molecular Systems, Inc. Detecting single nucleotide polymorphism using overlapped primer and melting probe
WO2015073580A1 (en) 2013-11-13 2015-05-21 Pfizer Inc. Tumor necrosis factor-like ligand 1a specific antibodies and compositions and uses thereof
WO2015073291A1 (en) 2013-11-14 2015-05-21 The Trustees Of The University Of Pennsylvania Hiv-1 env dna vaccine plus protein boost
US10801070B2 (en) 2013-11-25 2020-10-13 The Broad Institute, Inc. Compositions and methods for diagnosing, evaluating and treating cancer
AU2014352638B2 (en) 2013-11-25 2018-08-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors to control HIV infection
IL276210B2 (en) 2013-11-29 2024-01-01 Inovio Pharmaceuticals Inc Mers-cov vaccine
EP4227685A3 (en) 2013-12-03 2024-02-28 Evaxion Biotech A/S Proteins and nucleic acids useful in vaccines targeting staphylococcus aureus
US11725237B2 (en) 2013-12-05 2023-08-15 The Broad Institute Inc. Polymorphic gene typing and somatic change detection using sequencing data
WO2015087187A1 (en) 2013-12-10 2015-06-18 Rinat Neuroscience Corp. Anti-sclerostin antibodies
DK3079712T3 (en) 2013-12-11 2022-04-25 Massachusetts Gen Hospital USE OF MÜLLER INHIBITANT SUBSTANCE (MIS) PROTEINS FOR CONTRACEPTION AND OVARIAN RESERVE PRESERVATION
WO2015089419A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
CA2932436A1 (en) 2013-12-12 2015-06-18 The Broad Institute, Inc. Compositions and methods of use of crispr-cas systems in nucleotide repeat disorders
EP3080259B1 (en) 2013-12-12 2023-02-01 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions with new architectures for sequence manipulation
RU2016128077A (en) 2013-12-12 2018-12-06 Те Брод Инститьют Инк. DELIVERY, APPLICATION AND APPLICATIONS IN THE THERAPY OF CRISPR-CAS SYSTEMS AND COMPOSITIONS FOR TREATMENT OF CONDITIONED HBV AND VIRAL DISEASES AND DISORDERS
MX2016007328A (en) 2013-12-12 2017-07-19 Broad Inst Inc Delivery, use and therapeutic applications of the crispr-cas systems and compositions for genome editing.
EP3079716B1 (en) 2013-12-13 2019-05-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Multi-epitope tarp peptide vaccine and uses thereof
RU2714902C2 (en) 2013-12-19 2020-02-20 Новартис Аг Chimeric human mesotheliogen antigen receptors and use thereof
CN106456724A (en) 2013-12-20 2017-02-22 博德研究所 Combination therapy with neoantigen vaccine
WO2015095770A1 (en) 2013-12-20 2015-06-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Immunogenic jc polyomavirus compositions and methods of use
WO2015103549A1 (en) 2014-01-03 2015-07-09 The United States Of America, As Represented By The Secretary Department Of Health And Human Services Neutralizing antibodies to hiv-1 env and their use
EP4070818A3 (en) 2014-01-06 2023-01-11 The Trustees of the University of Pennsylvania Pd1 and pdl1 antibodies and vaccine combinations and use of same for immunotherapy
US10398772B2 (en) 2014-01-08 2019-09-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Ras pathways as markers of protection against HIV and methods to improve vaccine efficacy
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
MX2016009954A (en) 2014-01-29 2017-02-23 Dana Farber Cancer Inst Inc Antibodies against the muc1-c/extracellular domain (muc1-c/ecd).
WO2015120309A1 (en) 2014-02-06 2015-08-13 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
EP3105332A4 (en) 2014-02-14 2018-01-10 University of Utah Research Foundation Methods and compositions for inhibiting retinopathy of prematurity
US20170037091A1 (en) 2014-02-25 2017-02-09 Research Development Foundation Sty peptides for inhibition of angiogenesis
KR102340553B1 (en) 2014-03-04 2021-12-21 페이트 세러퓨틱스, 인코포레이티드 Improved reprogramming methods and cell culture platforms
KR101605421B1 (en) 2014-03-05 2016-03-23 국립암센터 A monoclonal antibody which specifically recognizes B cell lymphoma and use thereof
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
PE20161439A1 (en) 2014-03-21 2017-01-26 Teva Pharmaceuticals Int Gmbh ANTIBODIES ANTAGONISTS DIRECTED AGAINST THE PEPTIDE RELATED TO THE CALCITONIN GENE AND METHODS USING THE SAME
AU2015241107B2 (en) 2014-04-03 2019-10-03 Boehringer Ingelheim Animal Health USA Inc. Porcine epidemic diarrhea virus vaccine
KR102651707B1 (en) 2014-04-07 2024-03-29 노파르티스 아게 Treatment of cancer using anti-cd19 chimeric antigen receptor
US20170107486A1 (en) 2014-04-21 2017-04-20 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
CA3177878A1 (en) 2014-04-23 2015-10-29 Modernatx, Inc. Nucleic acid vaccines
US11624093B2 (en) 2014-04-24 2023-04-11 Dana-Farber Cancer Institute, Inc. Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response
AU2015249312B2 (en) 2014-04-25 2021-07-29 Translate Bio, Inc. Methods for purification of messenger RNA
EP3137098A4 (en) 2014-04-29 2017-11-01 Mayo Foundation for Medical Education and Research Butyrylcholinesterases having an enhanced ability to hydrolyze acyl ghrelin
US10308697B2 (en) 2014-04-30 2019-06-04 President And Fellows Of Harvard College Fusion proteins for treating cancer and related methods
WO2015173398A1 (en) 2014-05-15 2015-11-19 Cemm - Forschungszentrum Für Molekulare Medizin Gmbh Antagonists of slc38a9 and their use in therapy
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US10132818B2 (en) 2014-07-08 2018-11-20 New York University Tau imaging ligands and their uses in the diagnosis and treatment of tauopathy
LT3169706T (en) 2014-07-11 2020-03-25 Genmab A/S Antibodies binding axl
EP3169310A1 (en) 2014-07-15 2017-05-24 Life Technologies Corporation Compositions with lipid aggregates and methods for efficient delivery of molecules to cells
WO2016011226A1 (en) 2014-07-16 2016-01-21 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
EP3172237A2 (en) 2014-07-21 2017-05-31 Novartis AG Treatment of cancer using humanized anti-bcma chimeric antigen receptor
US20170209492A1 (en) 2014-07-31 2017-07-27 Novartis Ag Subset-optimized chimeric antigen receptor-containing t-cells
JP6811706B2 (en) 2014-07-31 2021-01-13 ザ ホンコン ユニヴァーシティ オブ サイエンス アンド テクノロジー Human monoclonal antibodies against EPHA4 and their use
EP3590954A3 (en) 2014-08-04 2020-03-25 OncoTherapy Science, Inc. Koc1-derived peptide and vaccine including same
KR20220165831A (en) 2014-08-04 2022-12-15 온코세라피 사이언스 가부시키가이샤 Urlc10-derived peptide and vaccine containing same
MX2017001650A (en) 2014-08-04 2017-04-27 Oncotherapy Science Inc Cdca1-derived peptide and vaccine containing same.
WO2016025510A1 (en) 2014-08-12 2016-02-18 Rappolee Daniel A Systems and methods to detect stem cell stress and uses thereof
JP6919118B2 (en) 2014-08-14 2021-08-18 ノバルティス アーゲー Treatment of cancer with GFRα-4 chimeric antigen receptor
ES2780904T3 (en) 2014-08-17 2020-08-27 Broad Inst Inc Genomic editing using Cas9 nickases
AR102333A1 (en) 2014-08-19 2017-02-22 Regeneron Pharma EFFICIENT SELECTIVITY OF RECOMBINED PROTEINS
BR112017003104A2 (en) 2014-08-19 2017-12-05 Novartis Ag cancer treatment using an anti-cd123 chimeric antigen receptor
CN107072963B (en) 2014-09-03 2020-07-07 吉倪塞思公司 Therapeutic nanoparticles and related compositions, methods and systems
EP3189067B1 (en) 2014-09-04 2021-05-19 The United States of America, as represented by The Secretary, Department of Health and Human Services Recombinant hiv-1 envelope proteins and their use
WO2016040488A2 (en) 2014-09-10 2016-03-17 Georgetown University Compositions and methods of using interleukin-4 induced gene 1 (il4i1)
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016049258A2 (en) 2014-09-25 2016-03-31 The Broad Institute Inc. Functional screening with optimized functional crispr-cas systems
WO2016045732A1 (en) 2014-09-25 2016-03-31 Biontech Rna Pharmaceuticals Gmbh Stable formulations of lipids and liposomes
US10166288B2 (en) 2014-10-01 2019-01-01 The Trustees Of The University Of Pennsylvania Vaccines having an antigen and interleukin-21 as an adjuvant
AU2015328411C1 (en) 2014-10-06 2022-03-03 Dana-Farber Cancer Institute, Inc. Angiopoietin-2 biomarkers predictive of anti-immune checkpoint response
SG11201702895SA (en) 2014-10-08 2017-05-30 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
AU2015328163B2 (en) 2014-10-09 2020-10-15 Dana-Farber Cancer Institute, Inc. Multiple-variable IL-2 dose regimen for treating immune disorders
JP6930913B2 (en) 2014-10-14 2021-09-01 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニアThe Regents Of The University Of California Usage of CDK9 and BRD4 inhibitors to inhibit inflammation
NZ731659A (en) 2014-11-03 2018-10-26 Merial Inc Methods of using microneedle vaccine formulations to elicit in animals protective immunity against rabies virus
US20180169211A1 (en) 2014-11-13 2018-06-21 Evaxion Biotech Aps Peptides derived from acinetobacter baumannii and their use in vaccination
MA40929A (en) 2014-11-14 2017-09-20 Regeneron Pharma HIGH AFINITY ANTIBODIES PRODUCTION PROCESS
US10160795B2 (en) 2014-11-14 2018-12-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to Ebola virus glycoprotein and their use
US9345718B1 (en) 2014-11-17 2016-05-24 Cellectar Biosciences, Inc. Phospholipid ether analogs as cancer-targeting drug vehicles
US9920381B2 (en) 2014-12-02 2018-03-20 Roche Molecular Systems, Inc. Compositions and methods for detecting MECC-containing methicillin-resistant Staphylococcus aureus
US20180334490A1 (en) 2014-12-03 2018-11-22 Qilong H. Wu Methods for b cell preconditioning in car therapy
US10072070B2 (en) 2014-12-05 2018-09-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Potent anti-influenza A neuraminidase subtype N1 antibody
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
EP3230452A1 (en) 2014-12-12 2017-10-18 The Broad Institute Inc. Dead guides for crispr transcription factors
EP3230451B1 (en) 2014-12-12 2021-04-07 The Broad Institute, Inc. Protected guide rnas (pgrnas)
WO2016094880A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Delivery, use and therapeutic applications of crispr systems and compositions for genome editing as to hematopoietic stem cells (hscs)
WO2016094874A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Escorted and functionalized guides for crispr-cas systems
WO2016100977A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Methods for profiling the t-cel- receptor repertoire
EP3234193B1 (en) 2014-12-19 2020-07-15 Massachusetts Institute of Technology Molecular biomarkers for cancer immunotherapy
EP3234192B1 (en) 2014-12-19 2021-07-14 The Broad Institute, Inc. Unbiased identification of double-strand breaks and genomic rearrangement by genome-wide insert capture sequencing
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
AU2015369725A1 (en) 2014-12-24 2017-06-29 Massachusetts Institute Of Technology CRISPR having or associated with destabilization domains
EP3236998A1 (en) 2014-12-24 2017-11-01 The U.S.A. as represented by the Secretary, Department of Health and Human Services Recombinant metapneumovirus f proteins and their use
MA41296A (en) 2014-12-30 2017-11-07 Orgenesis Ltd TRANSDIFFERENTIATION PROCESSES AND METHODS FOR USING THE SAME
CA2972635A1 (en) 2015-01-06 2016-07-14 Immunovaccine Technologies Inc. Lipid a mimics, methods of preparation, and uses thereof
US10434162B2 (en) 2015-01-12 2019-10-08 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting Klebsiella pneumoniae
CN107438665A (en) 2015-01-28 2017-12-05 沙特基础全球技术有限公司 For efficiently producing the method and composition of bio-fuel and/or biomass
US11116838B2 (en) 2015-01-29 2021-09-14 The Trustees Of The University Of Pennsylvania Checkpoint inhibitor and vaccine combinations and use of same for immunotherapy
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
EP3253786A4 (en) 2015-02-06 2018-10-17 The University of North Carolina at Chapel Hill Optimized human clotting factor viii gene expression cassettes and their use
AU2016219511B2 (en) 2015-02-09 2020-11-12 Research Development Foundation Engineered immunoglobulin Fc polypeptides displaying improved complement activation
WO2016128060A1 (en) 2015-02-12 2016-08-18 Biontech Ag Predicting t cell epitopes useful for vaccination
US10758558B2 (en) 2015-02-13 2020-09-01 Translate Bio Ma, Inc. Hybrid oligonucleotides and uses thereof
AU2016221318B2 (en) 2015-02-19 2020-06-25 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
US20170151281A1 (en) 2015-02-19 2017-06-01 Batu Biologics, Inc. Chimeric antigen receptor dendritic cell (car-dc) for treatment of cancer
US11421229B2 (en) 2015-02-20 2022-08-23 Baylor College Of Medicine p63 inactivation for the treatment of heart failure
WO2016135130A1 (en) 2015-02-23 2016-09-01 Serini Guido Non-natural semaphorins 3 and their medical use
KR20170140180A (en) 2015-02-24 2017-12-20 더 유나이티드 스테이츠 오브 어메리카, 애즈 리프리젠티드 바이 더 세크러테리, 디파트먼트 오브 헬쓰 앤드 휴먼 서비씨즈 Middle east respiratory syndrome coronavirus immunogens, antibodies, and their use
GB201503453D0 (en) 2015-03-01 2015-04-15 Jain Arjun Endothelin-1"sponge"
MA41629A (en) 2015-03-04 2018-01-09 Center For Human Reproduction COMPOSITIONS AND METHODS OF USE OF ANTI-MÜLLERIAN HORMONE FOR THE TREATMENT OF INFERTILITY
CA2977532A1 (en) 2015-03-06 2016-09-15 Dana-Farber Cancer Institute, Inc. Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers
WO2016145578A1 (en) 2015-03-13 2016-09-22 Syz Cell Therapy Co. Methods of cancer treatment using activated t cells
EP3069730A3 (en) 2015-03-20 2017-03-15 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
AU2016235541B2 (en) 2015-03-20 2021-04-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to gp120 and their use
US9931394B2 (en) 2015-03-23 2018-04-03 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US10385115B2 (en) 2015-03-26 2019-08-20 Duke University Fibronectin type III domain-based fusion proteins
WO2016160166A1 (en) 2015-03-30 2016-10-06 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Immunogenic rsv polypeptides
SG11201707490SA (en) 2015-04-03 2017-10-30 Eureka Therapeutics Inc Constructs targeting afp peptide/mhc complexes and uses thereof
US9758575B2 (en) 2015-04-06 2017-09-12 Yung Shin Pharmaceutical Industrial Co. Ltd. Antibodies which specifically bind to canine vascular endothelial growth factor and uses thereof
US10160755B2 (en) 2015-04-08 2018-12-25 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
IL254817B2 (en) 2015-04-08 2023-12-01 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
TWI787645B (en) 2015-04-13 2022-12-21 美商輝瑞股份有限公司 Cd3-specific antibodies, therapeutic bispecific antibodies and their uses
EP3283529B1 (en) 2015-04-17 2023-06-07 The General Hospital Corporation Agents, systems and methods for treating cancer
EP3286211A1 (en) 2015-04-23 2018-02-28 Novartis AG Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
US20190031759A1 (en) 2015-04-30 2019-01-31 Technion Research & Development Foundation Limited Chimeric antigen receptors and methods of their use
WO2016178996A1 (en) 2015-05-01 2016-11-10 The Regents Of The University Of California Glycan-dependent immunotherapeutic molecules
US10624964B2 (en) 2015-05-01 2020-04-21 The Trustees Of The University Of Pennsylvania Methods and compositions for stimulating immune response using potent immunostimulatory RNA motifs
US10093930B2 (en) 2015-05-06 2018-10-09 Mayo Foundation For Medical Education And Research Targeting WSB1 and pVHL to treat cancer
WO2016180430A1 (en) 2015-05-08 2016-11-17 Curevac Ag Method for producing rna
WO2016183420A1 (en) 2015-05-13 2016-11-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Methods and compositions for inducing an immune response using conserved element constructs
AU2016264323B2 (en) 2015-05-18 2021-02-18 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
CR20230191A (en) 2015-05-20 2023-07-06 Dana Farber Cancer Inst Inc SHARED NEOANTIGENS (Div. exp 2017-584)
CN107849148B (en) 2015-05-21 2023-09-19 哈普恩治疗公司 Trispecific binding proteins and methods of use
WO2016196366A1 (en) 2015-05-29 2016-12-08 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Extension of replicative lifespan in diseases of premature aging using p53 isoforms
WO2016196975A1 (en) 2015-06-03 2016-12-08 The United States Of America, As Represented By The Secretary Department Of Health & Human Services Neutralizing antibodies to hiv-1 env and their use
TW202241500A (en) 2015-06-09 2022-11-01 美商博德研究所有限公司 Formulations for neoplasia vaccines and methods of preparing thereof
SG10201912329YA (en) 2015-06-18 2020-02-27 Broad Inst Inc Crispr Enzyme Mutations Reducing Off-Target Effects
US9790490B2 (en) 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems
CA3012607A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Crispr enzymes and systems
CA3012631A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Novel crispr enzymes and systems
WO2016205745A2 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Cell sorting
AU2016282772B2 (en) 2015-06-23 2019-09-05 Boehringer Ingelheim Animal Health USA Inc. PRRSV minor protein-containing recombinant viral vectors and methods of making and use thereof
US10550379B2 (en) 2015-06-29 2020-02-04 The Board Of Trustees Of The Leland Stanford Junior University Degron fusion constructs and methods for controlling protein production
WO2017005670A1 (en) 2015-07-04 2017-01-12 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting pseudomonas aeruginosa
CA2991805A1 (en) 2015-07-10 2017-01-19 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
GB201512215D0 (en) 2015-07-13 2015-08-19 Lundbeck & Co As H Agents,uses and methods
JO3711B1 (en) 2015-07-13 2021-01-31 H Lundbeck As Antibodies specific for hyperphosphorylated tau and methods of use thereof
WO2017015334A1 (en) 2015-07-21 2017-01-26 Saint Louis University Compositions and methods for diagnosing and treating endometriosis-related infertility
CN108137705B (en) 2015-07-21 2022-11-08 武田药品工业株式会社 Monoclonal antibody inhibitor of factor XIIA
WO2017015619A1 (en) 2015-07-23 2017-01-26 The Regents Of The University Of California Antibodies to coagulation factor xia and uses thereof
US10829484B2 (en) 2015-07-28 2020-11-10 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
US10550164B2 (en) 2015-08-03 2020-02-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Brachyury deletion mutants, non-yeast vectors encoding Brachyury deletion mutants, and their use
EP3331557B1 (en) 2015-08-04 2021-04-07 Duke University Genetically encoded intrinsically disordered stealth polymers for delivery and methods of using same
US10988528B2 (en) 2015-08-13 2021-04-27 New York University Antibody-based molecules specific for the truncated ASP421 epitope of Tau and their uses in the diagnosis and treatment of tauopathy
ES2954969T3 (en) 2015-08-13 2023-11-27 Univ New York Antibody-based molecules selective for the {p}Ser404 epitope of Tau and their uses in the diagnosis and treatment of tauopathy
WO2017031370A1 (en) 2015-08-18 2017-02-23 The Broad Institute, Inc. Methods and compositions for altering function and structure of chromatin loops and/or domains
IL302353A (en) 2015-08-19 2023-06-01 Pfizer Tissue factor pathway inhibitor antibodies and uses thereof
CA2996975C (en) 2015-08-28 2022-02-15 Serpin Pharma, Llc Methods for treatment of diseases
US10526408B2 (en) 2015-08-28 2020-01-07 Research Development Foundation Engineered antibody FC variants
BR112017028224A2 (en) 2015-08-31 2018-09-11 Boehringer Ingelheim Vetmedica Gmbh composition, and method for protecting a piglet against a pestivirus-associated disease.
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
EP3350220B1 (en) 2015-09-15 2021-05-19 Scholar Rock, Inc. Anti-pro/latent-myostatin antibodies and uses thereof
AU2016323106A1 (en) 2015-09-16 2018-03-29 Boehringer Ingelheim Animal Health USA Inc. Salmonella Choleraesuis-Salmonella Typhimurium vaccines
US10392441B2 (en) 2015-10-07 2019-08-27 United States Of America, As Represented By The Secretary, Department Of Health And Human Services IL-7R-alpha specific antibodies for treating acute lymphoblastic leukemia
WO2017059902A1 (en) 2015-10-07 2017-04-13 Biontech Rna Pharmaceuticals Gmbh 3' utr sequences for stabilization of rna
EP3360886B1 (en) 2015-10-08 2023-01-25 Oncotherapy Science, Inc. Foxm1-derived peptide, and vaccine including same
WO2017062855A1 (en) 2015-10-09 2017-04-13 Monsanto Technology Llc Novel rna-guided nucleases and uses thereof
WO2017069958A2 (en) 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
AU2016334401A1 (en) 2015-10-10 2018-04-26 Intrexon Corporation Improved therapeutic control of proteolytically sensitive, destabilized forms of interleukin-12
EP3362570A4 (en) 2015-10-16 2019-03-20 Fate Therapeutics, Inc. Platform for the induction & maintenance of ground state pluripotency
US11207393B2 (en) 2015-10-16 2021-12-28 President And Fellows Of Harvard College Regulatory T cell PD-1 modulation for regulating T cell effector immune responses
US10865381B2 (en) 2015-10-20 2020-12-15 FUJIFILM Cellular Dynamics, Inc. Multi-lineage hematopoietic precursor cell production by genetic programming
US20180311336A1 (en) * 2015-10-22 2018-11-01 Moderna TX, Inc. Broad spectrum influenza virus vaccine
CA3002822A1 (en) * 2015-10-22 2017-04-27 Modernatx, Inc. Herpes simplex virus vaccine
CA3002820A1 (en) * 2015-10-22 2017-04-27 Modernatx, Inc. Respiratory syncytial virus vaccine
JP2018532777A (en) * 2015-10-22 2018-11-08 モデルナティーエックス, インコーポレイテッド Cancer vaccine
CA3024543A1 (en) 2015-10-22 2017-04-27 The Broad Institute, Inc. Type vi-b crispr enzymes and systems
US10138298B2 (en) 2015-10-23 2018-11-27 The Regents Of The University Of California Anti-IL-2 antibodies and compositions and uses thereof
SG11201802895QA (en) 2015-10-23 2018-05-30 Eureka Therapeutics Inc Antibody/t-cell receptor chimeric constructs and uses thereof
WO2017075037A1 (en) 2015-10-27 2017-05-04 Scholar Rock, Inc. Primed growth factors and uses thereof
EP3368687B1 (en) 2015-10-27 2021-09-29 The Broad Institute, Inc. Compositions and methods for targeting cancer-specific sequence variations
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
WO2017075478A2 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by use of immune cell gene signatures
US20210106661A1 (en) 2015-10-29 2021-04-15 Dana-Farber Cancer Institute, Inc. Methods for identification, assessment, prevention, and treatment of metabolic disorders using pm20d1 and n-lipidated amino acids
CA3003145A1 (en) 2015-10-30 2017-05-04 Gay M. Crooks Methods of generating t-cells from stem cells and immunotherapeutic methods using the t-cells
SI3370733T1 (en) 2015-11-02 2021-11-30 Board Of Regents The University Of Texas System Methods of cd40 activation and immune checkpoint blockade
EP4011911A1 (en) 2015-11-03 2022-06-15 The United States of America as represented by The Secretary Department of Health and Human Services Neutralizing antibodies to hiv-1 gp41 and their use
CN108884159A (en) 2015-11-07 2018-11-23 茂体外尔公司 The composition use for cancer treatment blocked comprising tumor suppressor gene treatment and immunologic test point
AU2016354440B2 (en) 2015-11-09 2023-09-28 The Children's Hospital Of Philadelphia Glypican 2 as a cancer marker and therapeutic target
AU2016354590B2 (en) 2015-11-13 2023-11-23 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
CN106699889A (en) 2015-11-18 2017-05-24 礼进生物医药科技(上海)有限公司 PD-1 resisting antibody and treatment application thereof
CN108601789A (en) 2015-11-20 2018-09-28 生华生物科技股份有限公司 Fourth Ring quinolone analogs combination treatment for treating cancer
WO2017095823A1 (en) 2015-11-30 2017-06-08 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
RU2018123825A (en) 2015-12-07 2020-01-15 Плексксикон Инк. COMPOUNDS AND METHODS FOR MODULATION OF KINASES, AND INDICATIONS FOR THIS
US10590169B2 (en) 2015-12-09 2020-03-17 Virogin Biotech Canada Ltd Compositions and methods for inhibiting CD279 interactions
US9957282B2 (en) 2015-12-14 2018-05-01 Senhwa Biosciences, Inc. Crystalline forms of quinolone analogs and their salts
EP3389630B1 (en) 2015-12-16 2023-11-08 Gritstone bio, Inc. Neoantigen identification, manufacture, and use
US20190233814A1 (en) 2015-12-18 2019-08-01 The Broad Institute, Inc. Novel crispr enzymes and systems
US20190015520A1 (en) 2015-12-21 2019-01-17 Duke University Polymer conjugates having reduced antigenicity and methods of using the same
US11752213B2 (en) 2015-12-21 2023-09-12 Duke University Surfaces having reduced non-specific binding and antigenicity
BR112018012784A2 (en) 2015-12-22 2019-02-05 Univ Muenchen Tech nucleic acids encoding proline and alanine residue-rich repetitive amino acid sequences that have low repeat nucleotide sequences
AU2016380351B2 (en) 2015-12-29 2023-04-06 Monsanto Technology Llc Novel CRISPR-associated transposases and uses thereof
US11441146B2 (en) 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
BR112018013967A2 (en) 2016-01-19 2019-02-05 Pfizer cancer vaccines
ES2904593T3 (en) 2016-01-21 2022-04-05 Pfizer Monospecific and bispecific antibodies to epidermal growth factor receptor variant III and CD3 and their uses
AU2017214656B2 (en) 2016-02-05 2022-08-25 Inovio Pharmaceuticals, Inc. Cancer vaccines and methods of treatment using the same
WO2017139392A1 (en) 2016-02-08 2017-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant hiv-1 envelope proteins and their use
EP3419994A4 (en) 2016-02-22 2019-11-06 The University of North Carolina at Chapel Hill Peptide inhibitors of calcium channels
ES2754785T3 (en) 2016-02-22 2020-04-20 Caribou Biosciences Inc DNA repair results modulation procedures
US10946084B2 (en) 2016-02-22 2021-03-16 Evaxion Biotech Aps Proteins and nucleic acids useful in vaccines targeting Staphylococcus aureus
KR20180118175A (en) 2016-03-04 2018-10-30 노파르티스 아게 Cells expressing multiple chimeric antigen receptor (CAR) molecules and their uses
EP3216458A1 (en) 2016-03-07 2017-09-13 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Modified vascular endothelial growth factor a (vegf-a) and its medical use
EP3426291A1 (en) 2016-03-09 2019-01-16 The U.S.A. as represented by the Secretary, Department of Health and Human Services Recombinant hiv-1 envelope proteins and their use
JP6962927B2 (en) 2016-03-11 2021-11-05 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Compositions and methods for detecting Zika virus
US20210309965A1 (en) 2016-03-21 2021-10-07 Dana-Farber Cancer Institute, Inc. T-cell exhaustion state-specific gene expression regulators and uses thereof
US11020160B2 (en) 2016-03-21 2021-06-01 Warsaw Orthopedic, Inc. Surgical injection system and method
CA3015570A1 (en) 2016-03-29 2017-10-05 Peter Kwong Substitutions-modified prefusion rsv f proteins and their use
US20190112380A1 (en) 2016-03-29 2019-04-18 University Of Southern California Chimeric antigen receptors targeting cancer
US20190177391A1 (en) 2016-03-31 2019-06-13 Baylor Research Institute Angiopoietin-like protein 8 (angptl8)
US11246924B2 (en) 2016-04-01 2022-02-15 Duke University Alpha-helical peptide nanofibers as a self-adjuvanting vaccine platform
HRP20230457T1 (en) 2016-04-15 2023-07-21 Novartis Ag Compositions and methods for selective expression of chimeric antigen receptors
US20190346442A1 (en) 2016-04-18 2019-11-14 The Broad Institute, Inc. Improved hla epitope prediction
US20200263190A1 (en) 2016-04-19 2020-08-20 The Broad Institute, Inc. Novel crispr enzymes and systems
CA3026112A1 (en) 2016-04-19 2017-10-26 The Broad Institute, Inc. Cpf1 complexes with reduced indel activity
WO2017184768A1 (en) 2016-04-19 2017-10-26 The Broad Institute Inc. Novel crispr enzymes and systems
US10709814B2 (en) 2016-04-22 2020-07-14 Warsaw Orthopedic, Inc. Osteoimplant comprising an insoluble fibrous polymer
KR20240011868A (en) 2016-04-29 2024-01-26 이노비오 파마수티컬즈, 인크. The in vivo use of chondroitinase and/or hyaluronidase to enhance delivery of an agent
WO2017192589A1 (en) 2016-05-02 2017-11-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to influenza ha and their use and identification
IL263079B1 (en) 2016-05-18 2024-01-01 Modernatx Inc Polynucleotides encoding relaxin
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2017201488A1 (en) 2016-05-20 2017-11-23 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US20200317738A1 (en) 2016-05-26 2020-10-08 University College Cork - National University Of Ireland, Cork Methods for increasing proliferation of mammalian cells
WO2017203051A1 (en) 2016-05-26 2017-11-30 University College Cork - National University Of Ireland, Cork An engineered gram positive bacterium
EP3464617B1 (en) 2016-05-27 2021-04-07 Roche Diagnostics GmbH Compositions and methods for detection of trichomonas vaginalis
US10612101B2 (en) 2016-05-27 2020-04-07 Roche Molecular Systems, Inc. Compositions and methods for detection of Mycoplasma genitalium
WO2017210476A1 (en) 2016-06-01 2017-12-07 Duke University Nonfouling biosensors
EP3464375A2 (en) 2016-06-02 2019-04-10 Novartis AG Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2017216384A1 (en) 2016-06-17 2017-12-21 Evaxion Biotech Aps Vaccination targeting ichthyophthirius multifiliis
CN109642231A (en) 2016-06-17 2019-04-16 博德研究所 VI type CRISPR ortholog and system
EP3471544B1 (en) 2016-06-21 2021-12-22 The Curators Of The University Of Missouri Modified dystrophin proteins
WO2017220787A1 (en) 2016-06-24 2017-12-28 Evaxion Biotech Aps Vaccines against aearomonas salmonicida infection
US20210222164A1 (en) 2016-06-29 2021-07-22 The Broad Institute, Inc. Crispr-cas systems having destabilization domain
CA3026858A1 (en) 2016-06-30 2018-01-04 F. Hoffmann-La Roche Ag Improved adoptive t-cell therapy
JP7099967B2 (en) 2016-07-01 2022-07-12 リサーチ ディベロップメント ファウンデーション Elimination of Proliferative Cells from Stem Cell-Derived Grafts
MA45655B1 (en) 2016-07-12 2021-04-30 H Lundbeck As Hyperphosphorylated tau protein specific antibodies and methods of use thereof
JP2018035137A (en) 2016-07-13 2018-03-08 マブイミューン ダイアグノスティックス エイジーMabimmune Diagnostics Ag Novel anti-fibroblast activated protein (FAP) binding agent and use thereof
MX2018015853A (en) 2016-07-14 2019-08-21 Genmab As Multispecific antibodies against cd40 and cd137.
CN110461315A (en) 2016-07-15 2019-11-15 诺华股份有限公司 Cytokines release syndrome is treated and prevented using with the Chimeric antigen receptor of kinase inhibitor combination
JP7178568B2 (en) 2016-07-18 2022-11-28 ナショナル リサーチ カウンシル オブ カナダ CAR immune cells targeting carcinoembryonic antigen-related cell adhesion molecule 6 for treating cancer
EP3889167A1 (en) 2016-07-22 2021-10-06 Evaxion Biotech ApS Chimeric proteins for inducing immunity towards infection with s. aureus
BR112019001570A2 (en) 2016-07-28 2019-07-09 Novartis Ag chimeric antigen receptor combination therapies and pd-1 inhibitors
BR112019002035A2 (en) 2016-08-01 2019-05-14 Novartis Ag cancer treatment using a chimeric antigen receptor in combination with an inhibitor of a m2 pro-macrophage molecule
AU2017306432A1 (en) 2016-08-02 2019-03-21 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018024562A1 (en) 2016-08-02 2018-02-08 Roche Diagnostics Gmbh Helper oligonucleotide for improving efficiency of amplification and detection/quantitation of nucleic acids
US20200283743A1 (en) 2016-08-17 2020-09-10 The Broad Institute, Inc. Novel crispr enzymes and systems
CN110114461A (en) 2016-08-17 2019-08-09 博德研究所 Novel C RISPR enzyme and system
WO2018035429A1 (en) 2016-08-18 2018-02-22 Wisconsin Alumni Research Foundation Peptides that inhibit syndecan-1 activation of vla-4 and igf-1r
US20190262399A1 (en) 2016-09-07 2019-08-29 The Broad Institute, Inc. Compositions and methods for evaluating and modulating immune responses
EP3512868A4 (en) 2016-09-14 2020-04-15 Duke University Triblock polypeptide-based nanoparticles for the delivery of hydrophilic drugs
US11865163B2 (en) 2016-09-15 2024-01-09 Mayo Foundation For Medical Education And Research Methods and materials for using butyrylcholinesterases to treat cancer
EA201990717A1 (en) 2016-09-20 2019-10-31 NEW VACCINE AGAINST PIG INFLUENZA
MX2019003161A (en) 2016-09-20 2019-05-27 Boehringer Ingelheim Vetmedica Gmbh New ehv insertion site orf70.
US11136409B2 (en) 2016-09-20 2021-10-05 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of AML using USP10 biomarkers and modulators
EP3515504B1 (en) 2016-09-20 2023-07-12 Boehringer Ingelheim Vetmedica GmbH Canine adenovirus vectors
TWI780070B (en) 2016-09-20 2022-10-11 德商百靈佳殷格翰維美迪加股份有限公司 New promoters
TW201815766A (en) 2016-09-22 2018-05-01 美商普雷辛肯公司 Compounds and methods for IDO and TDO modulation, and indications therefor
JP2020500150A (en) 2016-09-23 2020-01-09 デューク ユニバーシティ Non-repetitive and non-structural polypeptides with lower critical solution temperature behavior
EP3518968B1 (en) 2016-10-03 2022-01-05 The U.S.A. as represented by the Secretary, Department of Health and Human Services Hiv-1 env fusion peptide immunogens and their use
EP3523423A1 (en) 2016-10-05 2019-08-14 FUJIFILM Cellular Dynamics, Inc. Methods for directed differentiation of pluripotent stem cells to hla homozygous immune cells
CA3038690A1 (en) 2016-10-05 2018-04-12 FUJIFILM Cellular Dynamics, Inc. Generating mature lineages from induced pluripotent stem cells with mecp2 disruption
ES2875959T3 (en) 2016-10-07 2021-11-11 Tcr2 Therapeutics Inc Compositions and methods for T-cell receptor reprogramming using fusion proteins
WO2018067991A1 (en) 2016-10-07 2018-04-12 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
WO2018081289A2 (en) 2016-10-25 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion piv f immunogens and their use
WO2018081318A1 (en) 2016-10-25 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion coronavirus spike proteins and their use
WO2018081832A1 (en) 2016-10-31 2018-05-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Peptide fragments from filoviruses and their uses
US10172933B2 (en) 2016-10-31 2019-01-08 The United States Of America, As Represented By The Secretary Of Agriculture Mosaic vaccines for serotype a foot-and-mouth disease virus
WO2018083154A1 (en) 2016-11-03 2018-05-11 Boehringer Ingelheim Vetmedica Gmbh Vaccine against porcine parvovirus
ES2942749T3 (en) 2016-11-03 2023-06-06 Boehringer Ingelheim Vetmedica Gmbh Vaccine against porcine parvovirus and porcine reproductive and respiratory syndrome virus, and production methods thereof
US10793923B2 (en) 2016-11-09 2020-10-06 Roche Molecular Systems, Inc. Compositions and methods for detection of BK virus
EP3538660A1 (en) 2016-11-09 2019-09-18 Intrexon Corporation Frataxin expression constructs
US11072642B2 (en) 2016-11-09 2021-07-27 Mayo Foundation For Medical Education And Research MANP analogues
EP3541378A4 (en) 2016-11-16 2020-10-07 University of South Florida ALLOSTERIC ANTAGONISTS OF GPRC6a AND THEIR USE IN MITIGATING PROTEINOPATHIES
AU2017363311A1 (en) 2016-11-22 2019-06-13 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US20200010849A1 (en) 2016-11-23 2020-01-09 Gritstone Oncology, Inc. Viral delivery of neoantigens
JP7215997B2 (en) 2016-11-23 2023-01-31 ハープーン セラピューティクス,インク. Trispecific proteins targeting prostate specific membrane antigen (PSMA) and methods of use
WO2018098354A1 (en) 2016-11-23 2018-05-31 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
US10376495B2 (en) 2016-11-23 2019-08-13 University Of South Florida Small molecules that mimic or antagonize actions of granulocyte colony-stimulating-factor (G-CSF)
US11198723B2 (en) 2016-12-05 2021-12-14 The Administrators Of The Tulane Educational Fund Arenavirus monoclonal antibodies and uses
WO2018111902A1 (en) 2016-12-12 2018-06-21 Multivir Inc. Methods and compositions comprising viral gene therapy and an immune checkpoint inhibitor for treatment and prevention of cancer and infectious diseases
JP7144755B2 (en) 2016-12-16 2022-09-30 ハー・ルンドベック・アクチエゼルスカベット Drugs, uses and methods
CA3043790A1 (en) 2016-12-16 2018-06-21 Institute For Research In Biomedicine Novel recombinant prefusion rsv f proteins and uses thereof
EP3558348A1 (en) 2016-12-21 2019-10-30 TCR2 Therapeutics Inc. Engineered t cells for the treatment of cancer
US20190211377A1 (en) 2016-12-22 2019-07-11 Roche Molecular Systems, Inc. Cobra probes to detect a marker for epidemic ribotypes of clostridium difficile
WO2018136202A2 (en) 2016-12-23 2018-07-26 Plexxikon Inc. Compounds and methods for cdk8 modulation and indications therefor
MA47205A (en) 2017-01-04 2019-11-13 H Lundbeck As SPECIFIC ANTIBODIES OF HYPERPHOSPHORYLATED TAU PROTEIN TO TREAT EYE DISEASES
US11718648B2 (en) 2017-01-05 2023-08-08 Evaxion Biotech A/S Vaccines targeting Pseudomonas aeruginosa
EP3568470B1 (en) 2017-01-10 2022-07-06 Christiana Care Health Services, Inc. Methods for in vitro site-directed mutagenesis using gene editing technologies
WO2018132732A1 (en) 2017-01-12 2018-07-19 Duke University Genetically encoded lipid-polypeptide hybrid biomaterials that exhibit temperature triggered hierarchical self-assembly
TW201831517A (en) 2017-01-12 2018-09-01 美商優瑞科生物技術公司 Constructs targeting histone h3 peptide/mhc complexes and uses thereof
EP3574116A1 (en) 2017-01-24 2019-12-04 The Broad Institute, Inc. Compositions and methods for detecting a mutant variant of a polynucleotide
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
EA201991762A1 (en) 2017-01-30 2020-01-23 Бёрингер Ингельхайм Энимал Хелс Ю-Эс-Эй Инк. PIG CORONAVIRUS VACCINES
KR102567845B1 (en) 2017-01-31 2023-08-17 화이자 인코포레이티드 Neisseria meningitidis compositions and methods thereof
BR112019015797A2 (en) 2017-02-01 2020-03-17 Modernatx, Inc. IMMUNOMODULATORY THERAPEUTIC MRNA COMPOSITIONS THAT CODE ACTIVATING ONCOGEN MUTATION PEPTIDES
CN108395482B (en) 2017-02-08 2021-02-05 西比曼生物科技(香港)有限公司 Construction of targeting CD20 antigen chimeric antigen receptor and activity identification of engineered T cell thereof
US11021535B2 (en) 2017-02-10 2021-06-01 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
US10813935B2 (en) 2017-02-23 2020-10-27 Transgenex Nanobiotech, Inc. Methods and compositions for treating drug resistance in cancer
US10272052B2 (en) 2017-02-24 2019-04-30 University Of South Florida Compositions and methods for the treatment of tauopathies
US11318155B2 (en) 2017-02-24 2022-05-03 University Of South Florida Hsp90 activator Aha1 drives production of pathological tau aggregates
WO2018157072A1 (en) 2017-02-27 2018-08-30 Life Technologies Corporation Expansion of populations of t cells by the use of modified serum free media
EP3585417B1 (en) 2017-02-27 2023-02-22 Translate Bio, Inc. Method of making a codon-optimized cftr mrna
WO2018160754A2 (en) 2017-02-28 2018-09-07 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
US11198735B2 (en) 2017-03-03 2021-12-14 Rinat Neuroscience Corp. Anti-GITR antibodies and methods of use thereof
SG11201908088RA (en) 2017-03-09 2019-10-30 Genmab As Antibodies against pd-l1
WO2018170256A1 (en) 2017-03-15 2018-09-20 Modernatx, Inc. Herpes simplex virus vaccine
US11739308B2 (en) 2017-03-15 2023-08-29 The Broad Institute, Inc. Cas13b orthologues CRISPR enzymes and systems
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US20200056190A1 (en) 2017-03-16 2020-02-20 Pfizer Inc. Tyrosine prototrophy
WO2018176031A1 (en) 2017-03-24 2018-09-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Glycan-masked engineered outer domains of hiv-1 gp120 and their use
US10675283B2 (en) 2017-03-24 2020-06-09 University Of South Florida Compositions and methods for white to beige adipogenesis
CR20190440A (en) 2017-03-27 2019-11-12 Hoffmann La Roche Improved antigen binding receptors
AU2018241625A1 (en) 2017-03-27 2019-09-05 F. Hoffmann-La Roche Ag Improved antigen binding receptor formats
US10876101B2 (en) 2017-03-28 2020-12-29 Locanabio, Inc. CRISPR-associated (Cas) protein
US20200270359A1 (en) 2017-03-31 2020-08-27 Genmab Holding B.V. Bispecific anti-cd37 antibodies, monoclonal anti-cd37 antibodies and methods of use thereof
US20230241207A1 (en) 2017-04-03 2023-08-03 Neon Therapeutics, Inc. Protein antigens and uses thereof
AU2018251801A1 (en) 2017-04-12 2019-11-07 Massachusetts Institute Of Technology Novel type VI crispr orthologs and systems
WO2018191490A1 (en) 2017-04-13 2018-10-18 The Trustees Of The University Of Pennsylvania Use of gene editing to generate universal tcr re-directed t cells for adoptive immunotherapy
WO2018191750A2 (en) 2017-04-14 2018-10-18 The Broad Institute Inc. Novel delivery of large payloads
CA3059938A1 (en) 2017-04-14 2018-10-18 Kodiak Sciences Inc. Complement factor d antagonist antibodies and conjugates thereof
DK3612557T3 (en) 2017-04-18 2022-04-19 Fujifilm Cellular Dynamics Inc ANTIGEN-SPECIFIC IMMUNE EFFECTOR CELLS
AU2018258049A1 (en) 2017-04-26 2019-12-12 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
SG10201913656TA (en) 2017-04-26 2020-03-30 Eureka Therapeutics Inc Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
EP3615055A1 (en) 2017-04-28 2020-03-04 Novartis AG Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018204777A2 (en) 2017-05-05 2018-11-08 The Broad Institute, Inc. Methods for identification and modification of lncrna associated with target genotypes and phenotypes
SG11201910101SA (en) 2017-05-08 2019-11-28 Gritstone Oncology Inc Alphavirus neoantigen vectors
EP3635106A4 (en) 2017-05-08 2021-01-06 Orgenesis Ltd. Transdifferentiated cell populations and methods of use thereof
JP7340458B2 (en) 2017-05-09 2023-09-07 アメリカ合衆国 Combination of PDL1 and TGFbeta blockade in patients with HPV+ malignancies
KR102376863B1 (en) 2017-05-12 2022-03-21 하푼 테라퓨틱스, 인크. mesothelin binding protein
EP3621648A4 (en) 2017-05-12 2021-01-20 Harpoon Therapeutics, Inc. Msln targeting trispecific proteins and methods of use
WO2018213320A1 (en) 2017-05-15 2018-11-22 Duke University Recombinant production of hybrid lipid-biopolymer materials that self-assemble and encapsulate agents
CA3063531A1 (en) 2017-05-16 2018-11-22 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mrna encoding cftr
WO2018213803A1 (en) 2017-05-19 2018-11-22 Neon Therapeutics, Inc. Immunogenic neoantigen identification
CA3006798A1 (en) 2017-06-02 2018-12-02 Pfizer Inc. Antibodies specific for flt3 and their uses
WO2018226685A2 (en) 2017-06-06 2018-12-13 Dana-Farber Cancer Institute, Inc. Methods for sensitizing cancer cells to t cell-mediated killing by modulating molecular pathways
US20200247897A1 (en) 2017-06-07 2020-08-06 Genmab B.V. Therapeutic antibodies based on mutated igg hexamers
US10428067B2 (en) 2017-06-07 2019-10-01 Plexxikon Inc. Compounds and methods for kinase modulation
EP3638295A1 (en) 2017-06-13 2020-04-22 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
GB2576680B (en) 2017-06-13 2022-03-23 Bostongene Corp Systems and methods for generating, visualizing and classifying molecular functional profiles
US11786607B2 (en) 2017-06-15 2023-10-17 Modernatx, Inc. RNA formulations
US20210093667A1 (en) 2017-06-26 2021-04-01 The Broad Institute, Inc. Crispr/cas-adenine deaminase based compositions, systems, and methods for targeted nucleic acid editing
US11680083B2 (en) 2017-06-30 2023-06-20 Duke University Order and disorder as a design principle for stimuli-responsive biopolymer networks
US10392616B2 (en) 2017-06-30 2019-08-27 Arbor Biotechnologies, Inc. CRISPR RNA targeting enzymes and systems and uses thereof
EP3648789B9 (en) 2017-07-03 2023-09-27 Université de Strasbourg Mtmr2-s polypeptide for use in the treatment of myopathies
GB201710838D0 (en) 2017-07-05 2017-08-16 Ucl Business Plc Bispecific antibodies
BR112020000413A2 (en) 2017-07-11 2020-07-21 Pfizer Inc. immunogenic compositions comprising cea muc1 and tert
JP7083362B2 (en) 2017-07-12 2022-06-10 ベーリンガー インゲルハイム アニマル ヘルス ユーエスエイ インコーポレイテッド Senecavirus A immunogenic composition and its method
BR112020000679A2 (en) 2017-07-13 2020-07-14 Massachusetts Institute Of Technology targeting the hdac2-sp3 complex to enhance synaptic function
US10760137B2 (en) 2017-07-18 2020-09-01 Roche Molecular Systems, Inc. Compositions and methods for detection of Babesia
US10894833B2 (en) 2017-07-20 2021-01-19 H. Lundbeck A/S Agents, uses and methods for treatment
WO2019016784A1 (en) 2017-07-21 2019-01-24 Universidade De Coimbra Anti-nucleolin antibody
BR102017016440A2 (en) 2017-07-31 2019-03-19 Universidade Federal Do Rio Grande Do Sul COMPOSITION FOR CENTRAL NERVOUS SYSTEM GENE THERAPY, OBTAINING AND USE PROCESS
AU2018309339A1 (en) 2017-08-04 2020-02-20 BioNTech SE Binding agents binding to PD-L1 and CD137 and use thereof
US11833178B2 (en) 2017-08-25 2023-12-05 University College Cork—National University of Ireland, Cork Bifidobacterium longum for treating obesity and weight management
CA3073211A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. Methods of making lipid nanoparticles
CN109456943A (en) 2017-09-06 2019-03-12 亘喜生物科技(上海)有限公司 Universal Chimeric antigen receptor T cell technology of preparing
JP2020533986A (en) 2017-09-15 2020-11-26 ライフ テクノロジーズ コーポレーション Compositions and methods for culturing and growing cells
BR112020005519A2 (en) 2017-09-20 2020-10-27 The University Of British Columbia new anti-hla-a2 antibodies and their uses
EP3684397A4 (en) 2017-09-21 2021-08-18 The Broad Institute, Inc. Systems, methods, and compositions for targeted nucleic acid editing
TW201923084A (en) 2017-09-23 2019-06-16 德商百靈佳殷格翰維美迪加股份有限公司 PARAMYXOVIRIDAE expression system
CN109554348A (en) 2017-09-27 2019-04-02 亘喜生物科技(上海)有限公司 It can induce the engineering immunocyte of secretion anti-cd 47 antibody
WO2019063661A1 (en) 2017-09-29 2019-04-04 Roche Diagnostics Gmbh Compositions and methods for detection of trichomonas vaginalis
EP3692370A2 (en) 2017-10-04 2020-08-12 OPKO Pharmaceuticals, LLC Articles and methods directed to personalized therapy of cancer
JP7227237B2 (en) 2017-10-10 2023-02-21 グリットストーン バイオ インコーポレイテッド Identification of neoantigens using hotspots
EA202090739A1 (en) 2017-10-13 2020-09-07 Харпун Терапьютикс, Инк. PROTEINS BINDING ANTIGEN OF MATURING B-CELLS
CN111630070A (en) 2017-10-13 2020-09-04 哈普恩治疗公司 Trispecific proteins and methods of use
US11136356B2 (en) 2017-10-16 2021-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant HIV-1 envelope proteins and their use
EP3701014A1 (en) 2017-10-25 2020-09-02 Roche Diagnostics GmbH Improved modified/mutant bacterial luciferases
WO2019084338A1 (en) 2017-10-25 2019-05-02 University Of South Florida Drug-induced activation of the reelin signaling system
US11547614B2 (en) 2017-10-31 2023-01-10 The Broad Institute, Inc. Methods and compositions for studying cell evolution
US20210179709A1 (en) 2017-10-31 2021-06-17 Novartis Ag Anti-car compositions and methods
US20200255830A1 (en) 2017-11-02 2020-08-13 Arbor Biotechnologies, Inc. Novel crispr-associated transposon systems and components
US11497762B2 (en) 2017-11-03 2022-11-15 Interna Technologies B.V. MiRNA molecule, equivalent, antagomir, or source thereof for treating and/or diagnosing a condition and/or a disease associated with neuronal deficiency or for neuronal (re)generation
WO2019092027A1 (en) 2017-11-09 2019-05-16 Boehringer Ingelheim Vetmedica Gmbh Sapelovirus immunogenic compositions and uses thereof
CN111542333A (en) 2017-11-11 2020-08-14 斯特拉斯堡大学 Compositions and methods for treating X-linked centronuclear myopathy
US11612618B2 (en) 2017-11-14 2023-03-28 Henry Ford Health System Compositions for use in the treatment and prevention of cardiovascular disorders resulting from cerebrovascular injury
WO2019099639A1 (en) 2017-11-15 2019-05-23 Navartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
CA3083097A1 (en) 2017-11-22 2019-05-31 Gritstone Oncology, Inc. Reducing junction epitope presentation for neoantigens
EP3713587B1 (en) 2017-11-24 2024-03-27 University College Cork-National University of Ireland, Cork A composition comprising a cohort of bacteria
WO2019108541A1 (en) 2017-11-28 2019-06-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Recombinant rsv g proteins and their use
AU2018375738A1 (en) 2017-11-30 2020-06-11 Novartis Ag BCMA-targeting chimeric antigen receptor, and uses thereof
JP2021506255A (en) 2017-12-13 2021-02-22 イノビオ ファーマシューティカルズ,インコーポレイティド Cancer vaccines targeting PRAME and their use
BR112020010542A2 (en) 2017-12-13 2020-12-01 Inovio Pharmaceuticals, Inc. nucleic acid molecules, vector, composition, proteins, uses of the above, and vaccines
MX2020006226A (en) 2017-12-13 2020-11-18 Inovio Pharmaceuticals Inc Cancer vaccines targeting mesothelin and uses thereof.
US11793867B2 (en) 2017-12-18 2023-10-24 Biontech Us Inc. Neoantigens and uses thereof
WO2019136029A1 (en) 2018-01-02 2019-07-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to ebola virus glycoprotein and their use
CA3087537A1 (en) 2018-01-04 2019-07-11 Jan-willem THEUNISSEN Anti-tissue factor antibodies, antibody-drug conjugates, and related methods
US11254704B2 (en) 2018-01-05 2022-02-22 University Of South Florida Compounds and methods for reducing or inhibiting aggregation of proteins in a subject
MX2020007011A (en) 2018-01-05 2020-12-03 Ottawa Hospital Res Inst Modified vaccinia vectors.
CN112218651A (en) 2018-01-08 2021-01-12 诺华公司 Immunopotentiating RNA for combination with chimeric antigen receptor therapy
WO2019144047A1 (en) 2018-01-18 2019-07-25 University Of South Florida Dead antigen stimulated immature heterogenous dendritic cells as therapeutics for diseases
WO2019143934A1 (en) 2018-01-19 2019-07-25 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-cancer activity of scbg3a2 and lps
WO2019145399A1 (en) 2018-01-24 2019-08-01 Evaxion Biotech Aps Vaccines for prophylaxis of s. aureus infections
US20210038659A1 (en) 2018-01-31 2021-02-11 Novartis Ag Combination therapy using a chimeric antigen receptor
SG11202006883QA (en) 2018-02-01 2020-08-28 Pfizer Chimeric antigen receptors targeting cd70
AU2019215075A1 (en) 2018-02-01 2020-08-20 Pfizer Inc. Antibodies specific for CD70 and their uses
KR20200141986A (en) 2018-02-12 2020-12-21 다이어비티스-프리, 인크. Improved antagonistic anti-human CD40 monoclonal antibody
US11491206B1 (en) 2018-02-13 2022-11-08 Duke University Compositions and methods for the treatment of trail-resistant cancer
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
CN112334133A (en) 2018-02-15 2021-02-05 生华生物科技股份有限公司 Quinolone analogs and salts thereof, compositions, and methods of use thereof
WO2019165122A1 (en) 2018-02-21 2019-08-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to hiv-1 env and their use
AR115256A1 (en) 2018-02-23 2020-12-16 Boehringer Ingelheim Vetmedica Gmbh RECOMBINANT VIRAL VECTOR SYSTEMS EXPRESSING EXOGENOUS FELINE PARAMYXOVIRUS GENES AND VACCINES MADE FROM THESE
KR20200128116A (en) 2018-02-28 2020-11-11 화이자 인코포레이티드 IL-15 variants and uses thereof
WO2019175198A2 (en) 2018-03-12 2019-09-19 Genmab A/S Antibodies
ES2953541T3 (en) 2018-03-14 2023-11-14 Arbor Biotechnologies Inc Novel CRISPR DNA and RNA Targeting Systems and Enzymes
CN112041444A (en) 2018-03-14 2020-12-04 阿伯生物技术公司 Novel CRISPR DNA targeting enzymes and systems
CN112166123B (en) 2018-03-14 2022-09-30 北京轩义医药科技有限公司 Anti-claudin 18.2 antibodies
EP3539975A1 (en) 2018-03-15 2019-09-18 Fundació Privada Institut d'Investigació Oncològica de Vall-Hebron Micropeptides and uses thereof
EP3765053A2 (en) 2018-03-16 2021-01-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from urocortin 3 and uses thereof for the diagnosis and treatment of type 1 diabetes
EP3765064A1 (en) 2018-03-16 2021-01-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from pcsk2 and uses thereof for the diagnosis and treatment of type 1 diabetes
WO2019175380A2 (en) 2018-03-16 2019-09-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from secretogranin v and uses thereof for the diagnosis and treatment of type 1 diabetes
WO2020036635A2 (en) 2018-03-19 2020-02-20 Multivir Inc. Methods and compositions comprising tumor suppressor gene therapy and cd122/cd132 agonists for the treatment of cancer
CA3091960A1 (en) 2018-03-19 2019-09-26 Boehringer Ingelheim Vetmedica Gmbh New ehv insertion site ul43
AR115002A1 (en) 2018-03-19 2020-11-18 Boehringer Ingelheim Vetmedica Gmbh EHV WITH UL18 AND / OR UL8 INACTIVATED
EA202092216A1 (en) 2018-03-26 2021-06-11 Бёрингер Ингельхайм Энимал Хелс Ю-Эс-Эй Инк. METHOD FOR OBTAINING IMMUNOGENIC COMPOSITION
WO2019186274A2 (en) 2018-03-30 2019-10-03 University Of Geneva Micro rna expression constructs and uses thereof
WO2019183924A1 (en) 2018-03-30 2019-10-03 Syz Cell Therapy Co. Improved multiple antigen specific cell therapy methods
US10968257B2 (en) 2018-04-03 2021-04-06 The Broad Institute, Inc. Target recognition motifs and uses thereof
CN116514995A (en) 2018-04-12 2023-08-01 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting BCMA and preparation method and application thereof
WO2019196088A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of obtaining tumor-specific t cell receptors
WO2019196087A1 (en) 2018-04-13 2019-10-17 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific t cells
WO2019207051A1 (en) 2018-04-25 2019-10-31 Università Degli Studi Di Torino Medical use of combinations of non-natural semaphorins 3 and antimetabolites
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
US20210396739A1 (en) 2018-05-01 2021-12-23 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
US20210087530A1 (en) 2018-05-08 2021-03-25 Life Technologies Corporation Compositions and methods for culturing and expanding cells
WO2019222555A1 (en) 2018-05-16 2019-11-21 Arbor Biotechnologies, Inc. Novel crispr-associated systems and components
EP3797121A1 (en) 2018-05-23 2021-03-31 Pfizer Inc Antibodies specific for cd3 and uses thereof
CA3100829A1 (en) 2018-05-23 2019-11-28 Pfizer Inc. Antibodies specific for gucy2c and uses thereof
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
BR112020024292A2 (en) 2018-06-06 2021-03-02 Massachusetts Institute Of Technology circular rna for translation into eukaryotic cells
SG11202011830SA (en) 2018-06-13 2020-12-30 Novartis Ag Bcma chimeric antigen receptors and uses thereof
US20210371539A1 (en) 2018-06-22 2021-12-02 Genmab Holding B.V. Anti-cd37 antibodies and anti-cd20 antibodies, compositions and methods of use thereof
GB201811382D0 (en) 2018-07-11 2018-08-29 Taylor John Hermon Vaccine
US20200017600A1 (en) 2018-07-13 2020-01-16 Genmab A/S Variants of cd38 antibody and uses thereof
WO2020012038A1 (en) 2018-07-13 2020-01-16 Genmab A/S Trogocytosis-mediated therapy using cd38 antibodies
WO2020018142A1 (en) 2018-07-16 2020-01-23 Arbor Biotechnologies, Inc. Novel crispr dna targeting enzymes and systems
EP3604507A1 (en) 2018-07-30 2020-02-05 University College Cork-National University of Ireland, Cork An omega-transaminase enzyme
TW202023581A (en) 2018-08-02 2020-07-01 日商腫瘤療法 科學股份有限公司 Cdca1-derived peptide and vaccine containing same
WO2020028806A1 (en) 2018-08-02 2020-02-06 Duke University Dual agonist fusion proteins
US11890339B2 (en) 2018-08-03 2024-02-06 The United States of Americam as represented by the Secretary, Department of Health and Human Services Nipah virus immunogens and their use
CA3106035A1 (en) 2018-08-07 2020-02-13 The Broad Institute, Inc. Cas12b enzymes and systems
SG11202101169PA (en) 2018-08-10 2021-03-30 Eutilex Co Ltd Chimeric antigen receptor that binds hla-dr and car-t cell
WO2020041380A1 (en) 2018-08-20 2020-02-27 The Broad Institute, Inc. Methods and compositions for optochemical control of crispr-cas9
WO2020041793A1 (en) 2018-08-24 2020-02-27 Translate Bio, Inc. Methods for purification of messenger rna
CN113164556A (en) 2018-08-30 2021-07-23 特纳亚治疗股份有限公司 Cardiac cell reprogramming with cardiac myoprotein and ASCL1
US20210253666A1 (en) 2018-08-30 2021-08-19 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
JP7320601B2 (en) 2018-09-11 2023-08-03 上▲海▼市公共▲衛▼生▲臨▼床中心 Broad-spectrum anti-influenza vaccine immunogen and its use
WO2020053808A1 (en) 2018-09-12 2020-03-19 Georg Dewald Method of diagnosing vasoregulatory disorders
CN112867505A (en) 2018-09-20 2021-05-28 勃林格殷格翰动物保健有限公司 Modified PEDV spike protein
US10905758B2 (en) 2018-09-20 2021-02-02 Boehringer Ingelheim Vetmedica Gmbh Intranasal vector vaccine against porcine epidemic diarrhea
SG11202103022WA (en) 2018-09-25 2021-04-29 Harpoon Therapeutics Inc Dll3 binding proteins and methods of use
CN110950953B (en) 2018-09-26 2022-05-13 福州拓新天成生物科技有限公司 Monoclonal antibody against B7-H3 and application thereof in cell therapy
US20220000932A1 (en) 2018-09-28 2022-01-06 Henry Ford Health System Use of extracellular vesicles in combination with tissue plasminogen activator and/or thrombectomy to treat stroke
EP3856779A1 (en) 2018-09-28 2021-08-04 Novartis AG Cd22 chimeric antigen receptor (car) therapies
US20210347851A1 (en) 2018-09-28 2021-11-11 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020072700A1 (en) 2018-10-02 2020-04-09 Dana-Farber Cancer Institute, Inc. Hla single allele lines
WO2020070313A1 (en) 2018-10-04 2020-04-09 Genmab Holding B.V. Pharmaceutical compositions comprising bispecific anti-cd37 antibodies
MX2021004462A (en) 2018-10-19 2021-06-04 Univ Rochester Immune modulators in combination with radiation treatment for advanced pancreatic cancer.
US20220111031A1 (en) 2018-10-22 2022-04-14 Evaxion Biotech Aps Vaccines targeting M. catharrhalis
US20210340188A1 (en) 2018-10-22 2021-11-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Servic Recombinant gp120 protein with v1-loop deletion
AU2019365100A1 (en) 2018-10-22 2021-06-03 University Of Rochester Genome editing by directed non-homologous DNA insertion using a retroviral integrase-Cas9 fusion protein
WO2020102454A1 (en) 2018-11-13 2020-05-22 Regents Of The University Of Minnesota Cd40 targeted peptides and uses thereof
US20210403597A1 (en) 2018-11-16 2021-12-30 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof
EP3889264A4 (en) 2018-11-30 2022-10-12 Tokushima University Therapeutic agent for breast caner comprising big3-phb2 interaction-inhibiting peptide derived from phb2
EP3891302A1 (en) 2018-12-03 2021-10-13 F. Hoffmann-La Roche AG Compositions and methods for detection of candida auris
US20220031830A1 (en) 2018-12-04 2022-02-03 The Rockefeller University Hiv vaccine immunogens
EP3898958A1 (en) 2018-12-17 2021-10-27 The Broad Institute, Inc. Crispr-associated transposase systems and methods of use thereof
US20220062394A1 (en) 2018-12-17 2022-03-03 The Broad Institute, Inc. Methods for identifying neoantigens
US20220089694A1 (en) 2018-12-20 2022-03-24 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Ebola virus glycoprotein-specific monoclonal antibodies and uses thereof
WO2020140007A1 (en) 2018-12-28 2020-07-02 University Of Rochester Gene therapy for best1 dominant mutations
WO2020150287A1 (en) 2019-01-14 2020-07-23 University Of Rochester Targeted nuclear rna cleavage and polyadenylation with crispr-cas
GB201900858D0 (en) 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020174044A1 (en) 2019-02-27 2020-09-03 Evaxion Biotech Aps Vaccines targeting h. influenzae
SG11202109225WA (en) 2019-03-08 2021-09-29 Obsidian Therapeutics Inc Cd40l compositions and methods for tunable regulation
RU2742000C2 (en) 2019-03-13 2021-02-01 Общество С Ограниченной Ответственностью "Анабион" Isolated alternative intracellular signalling domain of chimeric antigen receptor and chimeric antigen receptor including it
WO2020186213A1 (en) 2019-03-14 2020-09-17 The Broad Institute, Inc. Novel nucleic acid modifiers
KR20230146132A (en) 2019-03-15 2023-10-18 카티전 테라퓨틱스, 인코포레이티드 Anti-bcma chimeric antigen receptors
US20220177863A1 (en) 2019-03-18 2022-06-09 The Broad Institute, Inc. Type vii crispr proteins and systems
EP3941505A1 (en) 2019-03-20 2022-01-26 Centre national de la recherche scientifique Amphiphysin / bin1 for the treatment of autosomal dominant centronuclear myopathy
CN111793627A (en) 2019-04-08 2020-10-20 中国科学院上海生命科学研究院 RNA fixed-point editing by utilizing artificially constructed RNA editing enzyme and related application
US20220220170A1 (en) 2019-04-24 2022-07-14 University Of Massachusetts Aav capsid chimeric antigen receptors and uses thereof
EP3959320A1 (en) 2019-04-24 2022-03-02 Novartis AG Compositions and methods for selective protein degradation
US20220227853A1 (en) 2019-05-03 2022-07-21 The United States Of America,As Represented By The Secretary,Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
CA3139346A1 (en) 2019-05-07 2020-11-12 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cell targeting bcma and use thereof
EP3964238A4 (en) 2019-05-07 2022-09-14 Gracell Biotechnologies (Shanghai) Co., Ltd. Bcma-targeting engineered immune cell and use thereof
US20220220539A1 (en) 2019-05-07 2022-07-14 Roche Molecular Systems, Inc. Compositions and methods for detection of neisseria gonorroheae
US20230085439A1 (en) 2019-05-21 2023-03-16 University Of Georgia Research Foundation, Inc. Antibodies that bind human metapneumovirus fusion protein and their use
IL288284B1 (en) 2019-05-22 2024-02-01 Massachusetts Inst Technology Circular rna compositions and methods
BR122024002387A2 (en) 2019-05-30 2024-03-12 Gritstone Bio, Inc. ADENOVIRUS VECTORS, PHARMACEUTICAL COMPOSITION, ISOLATED NUCLEOTIDE SEQUENCE, ISOLATED CELL, VECTOR, KIT, USES OF A VECTOR, METHOD FOR MAKING THE VECTOR, METHODS FOR PRODUCING A VIRUS AND VIRAL VECTOR
US20220380800A1 (en) 2019-07-02 2022-12-01 M6P Therapeutics (Switzerland) Llc Vector compositions and methods of using same for treatment of lysosomal storage disorders
EA202290208A1 (en) 2019-07-02 2022-03-25 Дзе Юнайтед Стейтс Оф Эмерика, Эз Репрезентед Бай Дзе Секретэри, Дипартмент Оф Хелт Энд Хьюман Сервисиз MONOCLONAL ANTIBODIES THAT BIND EGFRvIII AND THEIR APPLICATIONS
US20210005283A1 (en) 2019-07-03 2021-01-07 Bostongene Corporation Techniques for bias correction in sequence data
EP3997226A1 (en) 2019-07-11 2022-05-18 Tenaya Therapeutics, Inc. Cardiac cell reprogramming with micrornas and other factors
US11512314B2 (en) 2019-07-12 2022-11-29 Duke University Amphiphilic polynucleotides
AU2020317009A1 (en) 2019-07-19 2022-02-03 The Board Of Trustees Of The Leland Stanford Junior University Chimeric antigen receptors containing Glypican 2 binding domains
JP2022544015A (en) 2019-07-23 2022-10-17 ユニバーシティ オブ ロチェスター Targeted RNA Cleavage with CRISPR-Cas
EP4004240B1 (en) 2019-07-25 2024-03-13 F. Hoffmann-La Roche AG Compositions and methods for detection of epstein barr virus (ebv)
CN112300997A (en) 2019-08-01 2021-02-02 上海赛比曼生物科技有限公司 Universal CAR-T cell and preparation and application thereof
EP4017518A1 (en) 2019-08-20 2022-06-29 Nuritas Limited Peptides for treating muscle atrophy
EP3783012A1 (en) 2019-08-20 2021-02-24 Nuritas Limited An antimicrobial peptide
US11739382B2 (en) 2019-08-27 2023-08-29 Roche Molecular Systems, Inc. Compositions and methods for amplification and detection of hepatitis B virus RNA, including HBV RNA transcribed from cccDNA
MX2022002578A (en) 2019-09-02 2022-06-02 Inst Curie Immunotherapy targeting tumor neoantigenic peptides.
US20240050529A1 (en) 2019-10-07 2024-02-15 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
CA3157509A1 (en) 2019-10-10 2021-04-15 Kodiak Sciences Inc. Methods of treating an eye disorder
EP4041773A1 (en) 2019-10-11 2022-08-17 Beth Israel Deaconess Medical Center, Inc. Anti-tn antibodies and uses thereof
US20210147525A1 (en) 2019-10-18 2021-05-20 The Regents Of The University Of California Methods and compositions for treating pathogenic blood vessel disorders
CA3154368A1 (en) 2019-10-22 2021-04-29 Nora KHALDI Treatment of non-alcoholic fatty liver disease
EP4048304A1 (en) 2019-10-22 2022-08-31 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
KR102237349B1 (en) 2019-10-23 2021-04-07 한국과학기술연구원 Pharmaceutical composition for the treatment or prevention of nicotine addiction and withdrawal comprising miRNA
CN110760007B (en) 2019-11-21 2022-08-26 博生吉医药科技(苏州)有限公司 CD7-CAR-T cell and preparation and application thereof
CN114945382A (en) 2019-11-26 2022-08-26 诺华股份有限公司 CD19 and CD22 chimeric antigen receptors and uses thereof
ES2961245T3 (en) 2019-12-04 2024-03-11 Orna Therapeutics Inc Circular RNA compositions and methods
WO2021113644A1 (en) 2019-12-05 2021-06-10 Multivir Inc. Combinations comprising a cd8+ t cell enhancer, an immune checkpoint inhibitor and radiotherapy for targeted and abscopal effects for the treatment of cancer
WO2021119497A1 (en) 2019-12-11 2021-06-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mumps and measles virus immunogens and their use
EP4081666A2 (en) 2019-12-27 2022-11-02 F. Hoffmann-La Roche AG Compositions and methods for detecting methicillin-resistant staphylococcus aureus
WO2021140123A1 (en) 2020-01-06 2021-07-15 Evaxion Biotech Aps Vaccines targeting neisseria gonorrhoeae
WO2021142376A1 (en) 2020-01-08 2021-07-15 Obsidian Therapeutics, Inc. Compositions and methods for tunable regulation of transcription
US20230067811A1 (en) 2020-01-24 2023-03-02 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
JP2023513135A (en) 2020-02-06 2023-03-30 ベーリンガー インゲルハイム フェトメディカ ゲーエムベーハー Polypeptides useful for detecting anti-rhabdovirus antibodies
CA3168903A1 (en) 2020-02-07 2021-08-12 University Of Rochester Ribozyme-mediated rna assembly and expression
US20230078498A1 (en) 2020-02-07 2023-03-16 University Of Rochester Targeted Translation of RNA with CRISPR-Cas13 to Enhance Protein Synthesis
WO2021163365A1 (en) 2020-02-11 2021-08-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Sars-cov-2 vaccine
WO2021168292A1 (en) 2020-02-20 2021-08-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Epstein-barr virus monoclonal antibodies and uses thereof
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
JP2023511780A (en) 2020-02-25 2023-03-22 イノビオ ファーマシューティカルズ,インコーポレイティド Vaccines against coronavirus and how to use them
WO2021173674A1 (en) 2020-02-26 2021-09-02 A2 Biotherapeutics, Inc. Polypeptides targeting mage-a3 peptide-mhc complexes and methods of use thereof
JP2023516945A (en) 2020-02-28 2023-04-21 上海復宏漢霖生物技術股▲フン▼有限公司 Anti-CD137 constructs and uses thereof
JP2023516941A (en) 2020-02-28 2023-04-21 上海復宏漢霖生物技術股▲フン▼有限公司 Anti-CD137 constructs, multispecific antibodies and uses thereof
CN115552022A (en) 2020-03-02 2022-12-30 特纳亚治疗股份有限公司 MicroRNA-controlled gene vector expressed by cardiac muscle cells
US11213482B1 (en) 2020-03-05 2022-01-04 University of Pittsburgh—Of the Commonwealth System of Higher Educat SARS-CoV-2 subunit vaccine and microneedle array delivery system
CA3166337A1 (en) 2020-03-09 2021-09-16 F. Hoffmann-La Roche Ag Compositions and methods for detecting severe acute respiratory syndrome coronavirus 2 (sars-cov-2), influenza a and influenza b
IL296362A (en) 2020-03-18 2022-11-01 Genmab As Antibodies binding to b7h4
JP2023520359A (en) 2020-03-25 2023-05-17 エラスムス・ユニヴァーシティ・メディカル・センター・ロッテルダム Reporter system for radionuclide imaging
EP3885440A1 (en) 2020-03-26 2021-09-29 Splicebio, S.L. Split inteins and their uses
WO2021195525A1 (en) 2020-03-27 2021-09-30 University Of Rochester Crispr-cas13 crrna arrays
WO2021195519A1 (en) 2020-03-27 2021-09-30 University Of Rochester Targeted destruction of viral rna by crispr-cas13
WO2021205325A1 (en) 2020-04-08 2021-10-14 Pfizer Inc. Anti-gucy2c antibodies and uses thereof
CN113527507A (en) 2020-04-16 2021-10-22 上海赛比曼生物科技有限公司 Chimeric antigen receptor targeting CD22 and preparation method and application thereof
CN116096407A (en) 2020-04-29 2023-05-09 美国政府(由卫生和人类服务部的部长所代表) Recombinant human metapneumovirus F protein and application thereof
WO2021224850A1 (en) 2020-05-06 2021-11-11 Crispr Therapeutics Ag Mask peptides and masked anti-ptk7 antibodies comprising such
EP4149952A1 (en) 2020-05-12 2023-03-22 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
MX2022014248A (en) 2020-05-14 2022-12-02 Inovio Pharmaceuticals Inc Vaccines for recurrent respiratory papillomatosis and methods of using the same.
CA3184802A1 (en) 2020-05-26 2021-12-02 Inserm (Institut National De La Sante Et De La Recherche Medicale) Severe acute respiratory syndrome coronavirus 2 (sars-cov-2) polypeptides and uses thereof for vaccine purposes
US20230220057A1 (en) 2020-05-27 2023-07-13 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
IL298254A (en) 2020-05-29 2023-01-01 Fujifilm Cellular Dynamics Inc Bilayer of retinal pigmented epithelium and photoreceptors and use thereof
CN116033912A (en) 2020-05-29 2023-04-28 富士胶片细胞动力公司 Retinal pigment epithelium and photoreceptor double cell aggregates and methods of use thereof
WO2021249116A1 (en) 2020-06-10 2021-12-16 Sichuan Clover Biopharmaceuticals, Inc. Coronavirus vaccine compositions, methods, and uses thereof
AU2021288224A1 (en) 2020-06-11 2023-01-05 Novartis Ag ZBTB32 inhibitors and uses thereof
IL298874A (en) 2020-06-12 2023-02-01 Univ Rochester ENCODING AND EXPRESSION OF ACE-tRNAs
EP4182455A1 (en) 2020-07-15 2023-05-24 University of Rochester Targeted rna cleavage with dcasl3-rnase fusion proteins
WO2022011651A1 (en) 2020-07-16 2022-01-20 上海交通大学 Immunotherapy method of targeted chemokine and cytokine delivery by mesenchymal stem cell
JP2023533793A (en) 2020-07-17 2023-08-04 ファイザー・インク Therapeutic antibodies and their uses
WO2022016114A1 (en) 2020-07-17 2022-01-20 Instill Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
CA3189671A1 (en) 2020-07-17 2022-01-20 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
JP2022025558A (en) 2020-07-29 2022-02-10 学校法人帝京大学 miR-96-5p INHIBITOR AND PHARMACEUTICAL COMPOSITION CONTAINING THE SAME
CA3190227A1 (en) 2020-07-30 2022-02-03 Pfizer Inc. Cells having gene duplications and uses thereof
WO2022029051A1 (en) 2020-08-03 2022-02-10 F. Hoffmann-La Roche Ag Improved antigen binding receptors
KR20230046313A (en) 2020-08-06 2023-04-05 그릿스톤 바이오, 인코포레이티드 Multi-epitope vaccine cassette
WO2022035860A2 (en) 2020-08-10 2022-02-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Replication-competent adenovirus type 4-hiv env vaccines and their use
BR112023004321A2 (en) 2020-09-10 2023-04-04 Genmab As METHOD FOR TREATMENT OF DIFFUSED GRAND B-CELL LYMPHOMA IN A HUMAN SUBJECT
CA3190349A1 (en) 2020-09-10 2022-03-17 Brian Elliott Bispecific antibodies against cd3 and cd20 for treating chronic lymphocytic leukemia
CA3193508A1 (en) 2020-09-23 2022-03-31 Myeloid Therapeutics, Inc. Improved methods and compositions for expression of nucleic acids in cells
WO2022068912A1 (en) 2020-09-30 2022-04-07 Huigene Therapeutics Co., Ltd. Engineered crispr/cas13 system and uses thereof
WO2022188039A1 (en) 2021-03-09 2022-09-15 Huigene Therapeutics Co., Ltd. Engineered crispr/cas13 system and uses thereof
AU2021351187A1 (en) 2020-10-02 2023-04-20 Genmab A/S Antibodies capable of binding to ROR2 and bispecific antibodies binding to ROR2 and CD3
EP4225361A1 (en) 2020-10-05 2023-08-16 Boehringer Ingelheim Animal Health USA Inc. Fusion protein comprising circoviridae capsid protein, and chimeric virus-like particles composed thereof
US20220160866A1 (en) 2020-10-05 2022-05-26 Boehringer Ingelheim Vetmedica Gmbh Fusion protein useful for vaccination against rotavirus
WO2022074098A1 (en) 2020-10-08 2022-04-14 Fundació Privada Institut D'investigació Oncològica De Vall Hebron Method for the identification of cancer neoantigens
EP4343004A2 (en) 2020-10-19 2024-03-27 Dana-Farber Cancer Institute, Inc. Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
WO2022093745A1 (en) 2020-10-26 2022-05-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies targeting sars coronavirus spike protein and uses thereof
AR124562A1 (en) 2020-10-28 2023-04-12 Hoffmann La Roche ENHANCED ANTIGEN BINDING RECEPTORS
MX2023005201A (en) 2020-11-04 2023-06-28 Myeloid Therapeutics Inc Engineered chimeric fusion protein compositions and methods of use thereof.
CN116802738A (en) 2020-11-06 2023-09-22 亚马逊科技公司 Selection of neoantigens for personalized cancer vaccines
US20240033358A1 (en) 2020-11-13 2024-02-01 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022104104A2 (en) 2020-11-13 2022-05-19 Dana-Farber Cancer Institute, Inc. Personalized fusion cell vaccines
WO2022101463A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the last c-terminal residues m31/41 of zikv m ectodomain for triggering apoptotic cell death
CN114525306A (en) 2020-11-23 2022-05-24 博生吉医药科技(苏州)有限公司 Preparation method and application of CD7-CAR-T cell
EP4251170A1 (en) 2020-11-25 2023-10-04 Akagera Medicines, Inc. Lipid nanoparticles for delivery of nucleic acids, and related methods of use
US20240029829A1 (en) 2020-12-04 2024-01-25 Bostongene Corporation Hierarchical machine learning techniques for identifying molecular categories from expression data
US20220218614A1 (en) 2020-12-04 2022-07-14 Tidal Therapeutics, Inc. Ionizable cationic lipids and lipid nanoparticles, and methods of synthesis and use thereof
JP2023552546A (en) 2020-12-04 2023-12-18 エフ. ホフマン-ラ ロシュ アーゲー Compositions and methods for detecting malaria
TW202237638A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
TW202237639A (en) 2020-12-09 2022-10-01 日商武田藥品工業股份有限公司 Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
AU2021400424A1 (en) 2020-12-14 2023-07-06 Biontech Us Inc. Tissue-specific antigens for cancer immunotherapy
WO2022132904A1 (en) 2020-12-17 2022-06-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies targeting sars-cov-2
EP4267736A1 (en) 2020-12-22 2023-11-01 Helmholtz Zentrum München Forschungszentrum für Gesundheit und Umwelt GmbH Application of crispr/cas13 for therapy of rna virus and/or bacterium induced diseases
BR112023011689A2 (en) 2020-12-23 2023-10-31 Regeneron Pharma METHOD FOR OBTAINING AN ANTIBODY PRODUCING CELL, ANTIBODY, AND MAMMALIAN HOST CELL
US20220205020A1 (en) 2020-12-30 2022-06-30 Roche Molecular Systems, Inc. Compositions and methods for detection of bacteria and fungi associated with bacterial and candida vaginosis
EP4271482A2 (en) 2020-12-31 2023-11-08 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
EP4276113A1 (en) 2021-01-08 2023-11-15 Beijing Hanmi Pharmaceutical Co., Ltd. Antibody specifically binding to cd47 and antigen-binding fragment thereof
KR20230129483A (en) 2021-01-08 2023-09-08 베이징 한미 파마슈티컬 컴퍼니 리미티드 Antibodies and antigen-binding fragments that specifically bind to PD-L1
CN116710482A (en) 2021-01-08 2023-09-05 北京韩美药品有限公司 Antibodies and antigen binding fragments thereof that specifically bind 4-1BB
CN114763381A (en) 2021-01-13 2022-07-19 博生吉医药科技(苏州)有限公司 B7-H3 chimeric antigen receptor modified T cell and application thereof
WO2022159176A1 (en) 2021-01-19 2022-07-28 Amazon Technologies, Inc. A deep learning model for predicting tumor-specific neoantigen mhc class i or class ii immunogenicity
AU2021421391A1 (en) 2021-01-24 2023-07-20 Michael David FORREST Inhibitors of atp synthase - cosmetic and therapeutic uses
WO2022159793A2 (en) 2021-01-25 2022-07-28 Dana-Farber Cancer Institute, Inc. Methods and compositions for identifying neuroendocrine prostate cancer
US20230173045A1 (en) 2021-02-05 2023-06-08 Amazon Technologies, Inc. Ranking neoantigens for personalized cancer vaccine
WO2022167570A1 (en) 2021-02-05 2022-08-11 F. Hoffmann-La Roche Ag Compositions and methods for detection of human parainfluenza viruses 1-4 (hpiv 1-4)
CN114907485A (en) 2021-02-08 2022-08-16 浙江大学 Chimeric antigen receptor with endogenous protein molecule replacing single domain antibody
EP4291306A1 (en) 2021-02-09 2023-12-20 University of Georgia Research Foundation, Inc. Human monoclonal antibodies against pneumococcal antigens
CA3209136A1 (en) 2021-02-09 2022-08-18 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Antibodies targeting the spike protein of coronaviruses
CA3202849A1 (en) 2021-02-09 2022-08-18 University Of Houston System Oncolytic virus for systemic delivery and enhanced anti-tumor activities
WO2022175815A1 (en) 2021-02-19 2022-08-25 Pfizer Inc. Methods of protecting rna
WO2022178438A1 (en) 2021-02-22 2022-08-25 Duke University Non-immunogenic poegma-aptamer conjugates
CN115427561A (en) 2021-03-09 2022-12-02 辉大(上海)生物科技有限公司 Engineered CRISPR/Cas13 systems and uses thereof
JP2024510217A (en) 2021-03-11 2024-03-06 アンスティテュ・クリー Transmembrane neoantigen peptide
AU2022235060A1 (en) 2021-03-11 2023-09-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Tumor neoantigenic peptides and uses thereof
KR20230172630A (en) 2021-03-11 2023-12-22 므네모 테라퓨틱스 Tumor Neoantigenic Peptide
KR20230156921A (en) 2021-03-12 2023-11-15 젠맵 에이/에스 Non-activating antibody variants
CN117321690A (en) 2021-03-15 2023-12-29 亚马逊科技公司 Method for optimizing the coverage of a heterogeneous malignancy with tumor vaccine antigens
US20220290221A1 (en) 2021-03-15 2022-09-15 Roche Molecular Systems, Inc. Compositions and methods for detecting severe acute respiratory syndrome coronavirus 2 (sars-cov-2) variants having spike protein mutations
EP4070670A1 (en) 2021-04-08 2022-10-12 University College Cork-National University of Ireland Cork Lacticaseibacillus paracasei em025-11 and uses thereof
AU2022259536A1 (en) 2021-04-14 2023-11-09 University College Cork - National University Of Ireland, Cork Treatment of cerebrovascular events and neurological disorders
WO2022219168A1 (en) 2021-04-14 2022-10-20 University College Cork - National University Of Ireland, Cork Psg1 for use in the treatment of osteoarthritis
AU2022261125A1 (en) 2021-04-23 2023-11-23 University Of Rochester Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas fusion protein and methods of treatment
WO2022232612A1 (en) 2021-04-29 2022-11-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Lassa virus-specific nanobodies and methods of their use
WO2022232648A1 (en) 2021-04-29 2022-11-03 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Prefusion-stabilized lassa virus glycoprotein complex and its use
US20220372580A1 (en) 2021-04-29 2022-11-24 Bostongene Corporation Machine learning techniques for estimating tumor cell expression in complex tumor tissue
CN117242173A (en) 2021-05-03 2023-12-15 安斯泰来再生医药协会 Method for producing mature corneal endothelial cells
CN117858960A (en) 2021-05-06 2024-04-09 豪夫迈·罗氏有限公司 Compositions and methods for detecting hepatitis delta virus by dual target assays
TW202309268A (en) 2021-05-07 2023-03-01 安斯泰來再生醫藥協會 Methods of generating mature hepatocytes
AU2022280051A1 (en) 2021-05-26 2023-11-23 FUJIFILM Cellular Dynamics, Inc. Methods to prevent rapid silencing of genes in pluripotent stem cells
CN117813108A (en) 2021-05-27 2024-04-02 亚马逊科技公司 Multicomponent chemical composition of peptide-based neoantigen vaccine
TW202307210A (en) 2021-06-01 2023-02-16 瑞士商諾華公司 Cd19 and cd22 chimeric antigen receptors and uses thereof
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
CN117500831A (en) 2021-06-09 2024-02-02 美国政府(由卫生和人类服务部的部长所代表) Cross-species single domain antibodies targeting PD-L1 for the treatment of solid tumors
WO2022266192A1 (en) 2021-06-16 2022-12-22 Instil Bio, Inc. Receptors providing targeted costimulation for adoptive cell therapy
CN115477705B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on granzyme B
CN115477704B (en) 2021-06-16 2024-02-23 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cells constructed based on LOX1
WO2022266660A1 (en) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
AU2022303363A1 (en) 2021-06-29 2024-01-18 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
CA3224564A1 (en) 2021-07-05 2023-01-12 Andreas Holm MATTSSON Vaccines targeting neisseria gonorrhoeae
CA3226042A1 (en) 2021-07-05 2023-01-12 Regeneron Pharmaceuticals, Inc. Utilization of antibodies to shape antibody responses to an antigen
TW202317633A (en) 2021-07-08 2023-05-01 美商舒泰神(加州)生物科技有限公司 Antibodies specifically recognizing tnfr2 and uses thereof
CN115812082A (en) 2021-07-14 2023-03-17 舒泰神(北京)生物制药股份有限公司 Antibody specifically recognizing CD40 and application thereof
WO2023288278A1 (en) 2021-07-16 2023-01-19 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
CA3219606A1 (en) 2021-07-22 2023-01-26 F. Hoffmann-La Roche Ag Heterodimeric fc domain antibodies
WO2023012627A1 (en) 2021-08-02 2023-02-09 Pfizer Inc. Improved expression vectors and uses thereof
AU2022323509A1 (en) 2021-08-03 2024-02-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hiv-1 vaccination and samt-247 microbicide to prevent hiv-1 infection
AR127743A1 (en) 2021-09-06 2024-02-28 Genmab Bv ANTIBODIES CAPABLE OF BINDING CD27, VARIANTS AND USES THEREOF
AU2022345251A1 (en) 2021-09-17 2024-03-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Synthetic humanized llama nanobody library and use thereof to identify sars-cov-2 neutralizing antibodies
CA3232870A1 (en) 2021-09-22 2023-03-30 Richard B. Gaynor Coronavirus vaccines and methods of use
CA3233097A1 (en) 2021-09-30 2023-04-06 Katherine Diane GRIBBLE Compositions and methods for treating kcnq4-associated hearing loss
CA3234153A1 (en) 2021-10-08 2023-04-13 David Satijn Antibodies binding to cd30 and cd3
WO2023076881A1 (en) 2021-10-26 2023-05-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies targeting the s2 subunit of sars-cov-2 spike protein
CN114015674A (en) 2021-11-02 2022-02-08 辉二(上海)生物科技有限公司 Novel CRISPR-Cas12i system
WO2023079032A1 (en) 2021-11-05 2023-05-11 F. Hoffmann-La Roche Ag Compositions and methods for detection of malaria
WO2023086961A1 (en) 2021-11-12 2023-05-19 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Sars-cov-2 spike fused to a hepatitis b surface antigen
WO2023089556A1 (en) 2021-11-22 2023-05-25 Pfizer Inc. Reducing risk of antigen mimicry in immunogenic medicaments
WO2023089186A1 (en) 2021-11-22 2023-05-25 F. Hoffmann-La Roche Ag Compositions and methods for detecting vana and/or vanb genes associated with multidrug resistance
WO2023094413A1 (en) 2021-11-25 2023-06-01 F. Hoffmann-La Roche Ag Improved antigen binding receptors
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
WO2023144779A1 (en) 2022-01-28 2023-08-03 Pfizer Inc. Coronavirus antigen variants
US20230245479A1 (en) 2022-01-31 2023-08-03 Bostongene Corporation Machine learning techniques for cytometry
WO2023148598A1 (en) 2022-02-02 2023-08-10 Pfizer Inc. Cysteine prototrophy
WO2023150753A1 (en) 2022-02-07 2023-08-10 University Of Rochester Optimized sequences for enhanced trna expression or/and nonsense mutation suppression
WO2023154824A1 (en) 2022-02-10 2023-08-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies that broadly target coronaviruses
WO2023158732A1 (en) 2022-02-16 2023-08-24 Dana-Farber Cancer Institute, Inc. Methods for decreasing pathologic alpha-synuclein using agents that modulate fndc5 or biologically active fragments thereof
US20230349919A1 (en) 2022-03-15 2023-11-02 Regeneron Pharmaceuticals, Inc. Methods Of Mapping Antigen Specificity To Antibody-Secreting Cells
US20230295661A1 (en) 2022-03-16 2023-09-21 University Of Houston System Persistent hsv gene delivery system
WO2023178229A1 (en) 2022-03-16 2023-09-21 Amazon Technologies, Inc. Monitoring circulating tumor dna to improve subclone penetration of follow-up neoantigen cancer vaccines
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023180511A1 (en) 2022-03-25 2023-09-28 F. Hoffmann-La Roche Ag Improved chimeric receptors
WO2023192827A1 (en) 2022-03-26 2023-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Bispecific antibodies to hiv-1 env and their use
WO2023192835A1 (en) 2022-03-27 2023-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Base-covered hiv-1 envelope ectodomains and their use
WO2023192881A1 (en) 2022-03-28 2023-10-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to hiv-1 env and their use
WO2023194913A1 (en) 2022-04-05 2023-10-12 Boehringer Ingelheim Vetmedica Gmbh Immunogenic composition useful for vaccination against rotavirus
GB202205265D0 (en) 2022-04-11 2022-05-25 Mogrify Ltd Cell conversion
WO2023213393A1 (en) 2022-05-04 2023-11-09 Evaxion Biotech A/S Staphylococcal protein variants and truncates
GB202206507D0 (en) 2022-05-04 2022-06-15 Antion Biosciences Sa Expression construct
WO2023220645A1 (en) 2022-05-10 2023-11-16 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Vaccine for human t-lymphotropic virus-1
WO2023230295A1 (en) 2022-05-25 2023-11-30 BioNTech SE Rna compositions for delivery of monkeypox antigens and related methods
WO2023240156A1 (en) 2022-06-08 2023-12-14 Tidal Therapeutics, Inc. Ionizable cationic lipids and lipid nanoparticles, and methods of synthesis and use thereof
WO2023239940A1 (en) 2022-06-10 2023-12-14 Research Development Foundation Engineered fcriib selective igg1 fc variants and uses thereof
WO2024003046A1 (en) 2022-06-27 2024-01-04 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Viral load-dependent crispr/cas13-system
WO2024006911A1 (en) 2022-06-29 2024-01-04 FUJIFILM Holdings America Corporation Ipsc-derived astrocytes and methods of use thereof
WO2024003260A1 (en) 2022-06-30 2024-01-04 F. Hoffmann-La Roche Ag Compositions and methods for detecting lymphogranuloma venereum (lgv) serovars of chlamydia trachomatis
WO2024011033A1 (en) 2022-07-07 2024-01-11 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Immunogens and methods for inducing an immune response
WO2024015702A1 (en) 2022-07-15 2024-01-18 Amazon Technologies, Inc. Personalized longitudinal analysis of circulating material to monitor and adapt neoantigen cancer vaccines
WO2024015561A1 (en) 2022-07-15 2024-01-18 Bostongene Corporation Techniques for detecting homologous recombination deficiency (hrd)
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024030829A1 (en) 2022-08-01 2024-02-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind to the underside of influenza viral neuraminidase
WO2024042042A1 (en) 2022-08-24 2024-02-29 F. Hoffmann-La Roche Ag Compositions and methods for detecting monkeypox virus
WO2024054822A1 (en) 2022-09-07 2024-03-14 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Engineered sars-cov-2 antibodies with increased neutralization breadth
WO2024064826A1 (en) 2022-09-22 2024-03-28 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1617545C2 (en) 1967-06-19 1983-11-03 Diether Prof. Dr. Bern Jachertz Use of informational ribonucleic acid as a vaccine
US3931397A (en) * 1971-11-05 1976-01-06 Beecham Group Limited Biologically active material
GB1594097A (en) * 1976-12-16 1981-07-30 Int Inst Of Differentiation Production of specific immune nucleic acids cell dialysates and antibodies
US4394448A (en) * 1978-02-24 1983-07-19 Szoka Jr Francis C Method of inserting DNA into living cells
DE2942780A1 (en) 1979-10-23 1981-05-21 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V., 3400 Göttingen EUKARYOTIC CELLS, EUKARYOTIC PROTOPLASTICS AND MULTI-CELL EUKARYOTIC LIVING ORGANISMS CONTAINING DNA IN LIPID VESICLES, METHODS FOR THE PRODUCTION OF GENE PRODUCTS, FOR IMMEDIATING, AND DEFECTED BREADING
EP0091539B2 (en) 1982-03-31 1996-11-27 Ajinomoto Co., Inc. Gene coding for interleukin-2 polypeptide, recombinant DNA carrying said gene, cell lines possessing the recombinant DNA,and method for producing interleukin-2 using said cells
NZ209840A (en) * 1983-10-17 1988-11-29 Kaji Akira A method of inhibiting viral propagation by hybridising dna with the viral rna thus blocking its action
US4761375A (en) 1984-05-08 1988-08-02 Genetics Institute, Inc. Human interleukin-2 cDNA sequence
WO1986000930A1 (en) 1984-07-20 1986-02-13 Worcester Foundation For Experimental Biology Retroviral vaccines and vectors and methods for their construction
US4699880A (en) * 1984-09-25 1987-10-13 Immunomedics, Inc. Method of producing monoclonal anti-idiotype antibody
US4945050A (en) * 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
US4946787A (en) * 1985-01-07 1990-08-07 Syntex (U.S.A.) Inc. N-(ω,(ω-1)-dialkyloxy)- and N-(ω,(ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5049386A (en) * 1985-01-07 1991-09-17 Syntex (U.S.A.) Inc. N-ω,(ω-1)-dialkyloxy)- and N-(ω,(ω-1)-dialkenyloxy)Alk-1-YL-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
ZA8681B (en) * 1985-01-07 1987-08-26 Syntex Inc 1,2-dialkoxy-omega-trialkylammonium cationic surfactants
US4806463A (en) * 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
US4704692A (en) * 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
JPS63102682A (en) * 1986-10-20 1988-05-07 Medeisa Shinyaku Kk Transduction of dna into eukaryocyte
EP0273085A1 (en) * 1986-12-29 1988-07-06 IntraCel Corporation A method for internalizing nucleic acids into eukaryotic cells
JP2547768B2 (en) 1987-05-19 1996-10-23 キヤノン株式会社 Optical magnetic recording medium
JP2627899B2 (en) * 1987-08-19 1997-07-09 株式会社 ビタミン研究所 Production method of gene-encapsulated liposome
WO1989009271A1 (en) 1988-03-21 1989-10-05 Viagene, Inc. Recombinant retroviruses
WO1990001543A1 (en) * 1988-07-29 1990-02-22 Intracel Corporation Method for the genetic expression of heterologous proteins by cells transfected in vivo
US5693622A (en) * 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
EP1001032A3 (en) 1989-08-18 2005-02-23 Chiron Corporation Recombinant retroviruses delivering vector constructs to target cells
JP2805092B2 (en) * 1989-09-14 1998-09-30 ソニー株式会社 Disk drive device
DE69031951T2 (en) 1989-11-16 1998-08-13 Du Pont Transformation of animal skin cells with the help of particles
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5661133B1 (en) 1991-11-12 1999-06-01 Univ Michigan Collateral blood vessel formation in cardiac muscle by injecting a dna sequence encoding an angiogenic protein
WO1993014629A1 (en) 1992-01-27 1993-08-05 North Carolina State University Gene transfer in birds by introduction of dna into muscle in ovo
WO1997019675A2 (en) 1995-11-30 1997-06-05 Vical Incorporated Complex cationic lipids
US5994317A (en) 1996-04-09 1999-11-30 Vical Incorporated Quaternary cytofectins
DE19650157A1 (en) * 1996-12-04 1998-06-10 Basf Coatings Ag Process for coating substrates, preferably of metal

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7268120B1 (en) 1997-11-20 2007-09-11 Vical Incorporated Methods for treating cancer using cytokine-expressing polynucleotides
US20070225243A1 (en) * 1997-11-20 2007-09-27 Holly Horton Treatment of cancer using cytokine-expressing polynucleotides and compositions therefor
US7470675B2 (en) 1997-11-20 2008-12-30 Vical Incorporated Methods for treating cancer using interferon-ω-expressing polynucleotides
US20070105799A1 (en) * 2002-09-10 2007-05-10 Vical Incorporated Codon-optimized polynucleotide-based vaccines against Bacillus anthracis infection
US20070105193A1 (en) * 2003-05-16 2007-05-10 Vical Incorporated Severe acute respiratory syndrome DNA vaccine compositions and methods of use
US8080642B2 (en) 2003-05-16 2011-12-20 Vical Incorporated Severe acute respiratory syndrome DNA compositions and methods of use
US20060024670A1 (en) * 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
US20070286869A1 (en) * 2004-05-18 2007-12-13 Vical Incorporated Influenza virus vaccine composition and methods of use
US7537768B2 (en) 2004-05-18 2009-05-26 Vical Incorporated Influenza virus vaccine composition and methods of use
US7785603B2 (en) 2004-05-18 2010-08-31 Vical Incorporated Influenza virus vaccine composition and methods of use
US8128938B1 (en) 2004-05-18 2012-03-06 Vical Incorporated Influenza virus vaccine composition and methods of use
US8821890B2 (en) 2004-05-18 2014-09-02 Vical Incorporated Influenza virus vaccine composition and methods of use

Also Published As

Publication number Publication date
US20040132683A1 (en) 2004-07-08
US6710035B2 (en) 2004-03-23
US5703055A (en) 1997-12-30
US5580859A (en) 1996-12-03
US5589466A (en) 1996-12-31

Similar Documents

Publication Publication Date Title
US5589466A (en) Induction of a protective immune response in a mammal by injecting a DNA sequence
US6673776B1 (en) Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US7250404B2 (en) Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US6214804B1 (en) Induction of a protective immune response in a mammal by injecting a DNA sequence
EP0465529B1 (en) Expression of exogenous polynucleotide sequences in a vertebrate
US5693622A (en) Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
US6228844B1 (en) Stimulating vascular growth by administration of DNA sequences encoding VEGF
US5676954A (en) Method of in vivo delivery of functioning foreign genes
AU2776697A (en) Cationic virosomes as transfer system for genetic material
US6706694B1 (en) Expression of exogenous polynucleotide sequences in a vertebrate
JP2002316997A (en) Complex for transducing objective anionic substance into cell
US20030186913A1 (en) Expression of exogenous polynucleotide sequences in a vertebrate
WO1998040499A1 (en) Gene delivery to mucosal epithelium for immunization or therapeutic purposes
KR19990063814A (en) Pharmaceutical compositions useful for nucleic acid transfection and uses thereof
US7364750B2 (en) Autogene nucleic acids encoding a secretable RNA polymerase
Felgner et al. WITHDRAWN APPLICATION AS PER THE LATEST USPTO WITHDRAWN LIST
Gould-Fogerite et al. Liposomes: use as gene transfer vehicles and vaccines.
Fletcher et al. 470. Enhanced Transposition Activities of Mutant Sleeping Beauty Transposases
US20050220806A1 (en) Method of transporting physiological polymer using protein having rxp repeated sequence
JP2006517799A (en) A non-immunogenic selectable marker resistant to cardiac glycosides

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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