US6214804B1 - Induction of a protective immune response in a mammal by injecting a DNA sequence - Google Patents

Induction of a protective immune response in a mammal by injecting a DNA sequence Download PDF

Info

Publication number
US6214804B1
US6214804B1 US08/481,919 US48191995A US6214804B1 US 6214804 B1 US6214804 B1 US 6214804B1 US 48191995 A US48191995 A US 48191995A US 6214804 B1 US6214804 B1 US 6214804B1
Authority
US
United States
Prior art keywords
mammal
dna
cells
construct
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.)
Expired - Lifetime
Application number
US08/481,919
Inventor
Philip L. Felgner
Jon Asher Wolff
Gary H. Rhodes
Robert Wallace Malone
Dennis A. 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.)
Fresh Tracks Therapeutics Inc
Original Assignee
Vical Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vical Inc filed Critical Vical Inc
Priority to US08/481,919 priority Critical patent/US6214804B1/en
Priority to US09/452,872 priority patent/US6710035B2/en
Application granted granted Critical
Publication of US6214804B1 publication Critical patent/US6214804B1/en
Priority to US10/387,525 priority patent/US20040023911A1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • 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
    • 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.
  • 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.
  • MHC molecules 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.
  • 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.
  • 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 granulocyte-macrophage colony-stimulating factor
  • erythropoietin insulin
  • calcitonin thymidine kinase
  • thymidine kinase and the like.
  • toxic peptides such as ricin, diphtheria toxin,
  • FIG. 1 comprises autoradiograms of chromatographic studies showing the expression of the CAT gene in mouse muscle.
  • FIGS. 2A-2F comprise photomicrographs of muscle tissue stained for beta-galactosidase activity following injection with the pRSVLac-Z DNA vector.
  • FIGS. 3A, 3 B, and 3 C present data for luciferase activity in muscle following the injection of ⁇ gLucBgA 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 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 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.
  • 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.
  • 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 tissuess 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. Alternatively, it may be provided by including mRNA coding therefor, which mRNA is translated by the cell.
  • 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 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.
  • 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 in accordance with the present invention, 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.
  • 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; alternatively, it 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. 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).
  • 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, 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 more prolonged effect may be desired without incorporation of the exogenous polynucleic acid into the genome of the host organism.
  • 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 ColEl (Ausubel, Current Protocols in Molecular Biology , John Wiley and Sons, New York (1988) ⁇ II:1.5.2.
  • RNA 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.
  • muscle tissue 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.
  • NGF infusions have reversed the loss of cholinergic neurons.
  • 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 lost of function of specific neuronal groups.
  • 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.
  • 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.
  • 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 possible DOPA decarboxylase as well.
  • Tyrosine is readily available within the CNS.
  • 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 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
  • DNA or RNA coding for a cell surface receptor 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.
  • 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.
  • cancer cells 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).
  • 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.
  • 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 include the use of a polynucleotide that has a therapeutic effect without being translated into a polypeptide.
  • 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 the 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.
  • 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 TGT-Tretrachloro-2-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-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.
  • in vitro mutagenesis it is possible to obtain and sequence numerous clones of a given antigen. If these antigen 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 tisues 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.
  • RNA Ribonucleic Acids Res. 13: 7375 (1985)
  • GCC translational initiation consensus sequence
  • 5 G 7 methyl GpppG cap structure 5 G 7 methyl GpppG cap structure
  • 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.
  • 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).
  • DOTMA 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, an d1 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.
  • 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.
  • a transcription buffer comprising 400 mM Tris HCl (pH 8.0), 80 mM MgCl 2 , 50 m 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; 7meG(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.
  • T7 RNA polymerase N.E. Biolabs
  • This mixture was incubated for 1 hour at 37 C.
  • the successful transcription reaction was indicated by increasing cloudiness of the reaction mixture.
  • RNAse-free Sephadex G50 column Boehringer Mannheim #100 411). The resultant mRNA was sufficiently pure to be used in transfection of vertebrates in vivo.
  • 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.
  • CAT chloramphenicol acetyl transferase
  • 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 the 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 ⁇ l of each sample (the supernatant prepared above) was incubated for two hours at 37° C. with 5 ⁇ l C 14 chloramphenicol (Amersham); 20 ⁇ l 4 mM Acetyl CoA; and 50 ⁇ l 1 M Tris, pH 7.8. Thereafter, 20 ⁇ l 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. 10% Sucrose; 37.5 ⁇ g DOTMA; 15 ⁇ g CAT RNA 3/6 5. 10% Sucrose; 187 ⁇ g DOTMA; 75 ⁇ g CAT RNA 0/6
  • 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, Mont.) 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 10 5 to 10 6 infectious units of HIV to approximately 10 7 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 imunodeficient 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.
  • 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 ug/ml NEF RNA and 500 ug/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 ug/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 ⁇ l 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 The quadriceps muscles of mice were injected with either 100 ⁇ grams of pRSVCAT DNA plasmid or 100 ⁇ grams 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).
  • BRL LipofectinTM
  • Opti-MemTM Opti-MemTM
  • 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 ⁇ l 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 C 14 -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. Lane numbers appear at the top of the autoradiograms and the % chloramphenicol conversions are at the bottom. Sample locations are as follows:
  • 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. coil ⁇ -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.
  • the photographed sections are as follows:
  • 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 1251 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:
  • a dose-response effect was observed when quadriceps muscles were injected with various amounts of ⁇ gLuc ⁇ gA n RNA or DNA pRSVL constructs (FIG. 3 A).
  • 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. 3 A). 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. 3 A and 3 B). Based on the amount of DNA delivered, the efficiency of expression from the DNA vectors was similar in both transfected fibroblasts and injected muscles.
  • Luciferase activity was assayed at varying times after 25 ⁇ g 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.
  • 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.
  • Preparations of muscle DNA were obtained from control, uninjected quadriceps or from quadriceps, 30 days after injection with 100 ⁇ g of pRSVL in 20% sucrose. Two entire quadriceps muscles from the same animal were pooled, minced into liquid N 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).
  • 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
  • Lanes 6,7 BamH1 digest of HIRT supernatant from two different pools of pRSVL injected muscles
  • 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
  • Lane 12 Cellular DNA digested with BgIII
  • Lane 13 HIRT supernatant digested with BgIII
  • 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:
  • 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. Luciferase assays were done on 87.5 ⁇ l of lung extract out of a total volume of about 350 ⁇ l.
  • Blank 22.5 Mock Values are those for an animal that received 25 ⁇ g of DNA in 0.3 ml 20% sucrose into the esophagus. (A sample containing only water yields 22.5 l.u.) 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 Lipofectin TM in 0.3 ml 5% sucrose. The above animals were sacrificed and lung extracts prepared 2 days after injection. 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 luciferase activity in the 25 ⁇ g DNA alone and the 25 ⁇ g DNA/CL groups of animals were not greater than that in the mock animal; however, in the 250 ⁇ g 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 1251 luminometer indicates that these values, although just above background, indicate real luciferase activity.
  • the DNA luciferase expression vector pPGKLuc was injected intrahepatically (IH) 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 (metalothionien 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 metalothionien promoter-growth hormone fusion gene
  • mice Two mice were injected with 20 ⁇ g of pXGH5 gene complexed with 60 ⁇ g/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.
  • 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:
  • 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 retains 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 EcoR1/Nco1 fragment from the encephalomyocarditis virus (EMCV).
  • SV40-T7 polymerase plasmid containing T7 polymerase protein expressed off the SV40 promotor was co-lipofected with the pEMCLacZ ⁇ GAn 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:
  • 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.
  • Each of these two plasmids were co-lipofected with pEMCLacZ ⁇ GAn at 1:3 and 3:1 ratios into a 60 mm plates of 3T3 cells.
  • the number of blue ⁇ -galactosidase cells were counted and scored as indicated below.
  • ⁇ -gal Ratio template polymerase vector pSV-G1-A pSVNU-G1-A ⁇ GLacZ ⁇ GAn 3:1 0 1 1:3 0 1 EMCLacZ ⁇ GAn 3:1 74 70 1:3 45 15
  • 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 ⁇ l, 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. The results are as follows:
  • the newborn mouse was injected with 1 ⁇ l ⁇ gLuc ⁇ gA n (3.6 ⁇ g/ ⁇ l; 20% sucrose) into the bilateral forebrain and tissues were similarly processed and analyzed.
  • 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.
  • 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 3: 189-202 (1988); Hoffman, E. P. and Kunkel, E.
  • 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-60kd antibody.

Abstract

A method for delivering an isolated polynucleotide to the interior of a cell in a vertebrate, comprising the interstitial introduction of an isolated 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

This application divisional of U.S. patent application Ser. No. 07/496,991, filed Mar. 21, 1990, now abandoned which is a continuation in part of Ser. No. 07/467,881, filed Jan. 19, 1990, abandoned, which is a continuation in part of Ser. No. 07/326,305, filed Mar. 21, 1989, abandoned.
This application is a continuation-in-part of U.S. application Ser. No. 467,881 filed Jan. 19, 1990, which is a continuation-in-part of U.S. application Ser. No. 326,305 filed Mar. 21, 1989.
BACKGROUND OF THE INVENTION
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.
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., 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.
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.
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.
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.
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.
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.
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).
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.
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.
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.
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.
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.
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.
These above-described problems associated with gene therapy; immunization, and delivery of therapeutic peptides to cells are addressed by the present invention.
DESCRIPTION OF THE DRAWINGS
FIG. 1 comprises autoradiograms of chromatographic studies showing the expression of the CAT gene in mouse muscle.
FIGS. 2A-2F comprise photomicrographs of muscle tissue stained for beta-galactosidase activity following injection with the pRSVLac-Z DNA vector.
FIGS. 3A, 3B, and 3C present data for luciferase activity in muscle following the injection of βgLucBgAn 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.
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.
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.
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.
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.
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.
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.
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.
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.
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. 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.
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. Also contemplated is such use, wherein 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. 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. 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.
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.
In preferred embodiments, the polynucleotide is introduced into muscle tissue; in other embodiments the polynucleotide is incorporated into tissuess 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.
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. 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.
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.
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.
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.
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.
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.
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. 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.
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.
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.
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.
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.
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.
Polynucleotide Materials
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 CaPO4.
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. 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)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. 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; alternatively, it 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.
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.
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.
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).
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. Alternatively, a 5′ untranslated sequence such as EMC can be used which does not require a 5′ cap.
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.
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, 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. 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.
Transient Gene Therapy
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, 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.
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. 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. 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 ColEl (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. 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.
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.
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. 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.
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. 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, 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, 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.
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. 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.
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. 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.
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.
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-fornix 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 lost of function of specific neuronal groups.
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. 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. 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.
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.
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 possible DOPA decarboxylase as well. Tyrosine is readily available within the CNS.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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.
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. Alternatively, an anti-sense DNA may encode an RNA the 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.
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.
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.
DNA and mRNA Vaccines
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.
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.
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.
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.
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. In any case, this method provides a way to stimulate and modulate the immune response in a way which has not previously been possible.
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.
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.
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.
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 eliminate the humoral epitomes which enhance infectivity.
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.
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 an infections. However, 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. 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 an cellular response to proteins of the pathogen.
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.
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.
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 antigen can be screened for antigenicity at the DNA or RNA level by TGT, the vaccine development program could be made to proceed much faster.
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.
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.
Therapeutic Formulations
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., 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.
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.
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 indivdual procedures are well known to those in the art.
Dosage and Route of 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 tisues or lungs.
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.
Regulation of TGT
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.
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, Nucleic Acids Res. 15: 8125 (1987)) and the 5G 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 dependant translation and translational controls and mediate ann 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, 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
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:
Figure US06214804-20010410-C00001
wherein R1 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:
Figure US06214804-20010410-C00002
wherein R1 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., Biochemistry 27: 3917-3925 (1988); H. Eibl, et al., Biophysical Chemistry 10: 261-271 (1979).
Liposome Preparation
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.
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., 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 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.
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.
EXAMPLE 1 Preparation of Liposome-Forming DOTAP
The cationic liposome-forming 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, an d1 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 Rf 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., 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.
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 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 Aectyltransferase
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-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.
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, 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.
Next, a transcription buffer was prepared, comprising 400 mM Tris HCl (pH 8.0), 80 mM MgCl2, 50 m 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; 7meG(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.
Following generation of the mRNA, 2 U RQl 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 (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:
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. (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.
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 the an aqueous formulation having the following components: 20 mM Tris, pH 7.6; 2 mM MgCl2; 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 μl of each sample (the supernatant prepared above) was incubated for two hours at 37° C. with 5 μl C14 chloramphenicol (Amersham); 20 μl 4 mM Acetyl CoA; and 50 μl 1 M Tris, pH 7.8. Thereafter, 20 μl 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)
DOTMA: (Lipofectin brand; Bethesda Research Labs, Gaithersburg, Md.)
CAT RNA: From Example 5
All formulations made up in DEPC-treated RNAse-free water (International Biotechnologies, Inc., New Haven, Conn. 06535).
EXAMPLE 8 mRNA Vaccination of Mice to Produce the gp120Protein 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 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:
HT4-6C cells (CD4+ HeLa cells) are obtained from Dr. Bruce Chesebro, (Rocky Mountain National Lab, Mont.) 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.
EXAMPLE 9 mRNA Vaccination of Human Stem Cell-Bearing SCID Mice with Nef mRNA Followed by HIV Challenge
Severe combined imunodeficient 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.
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 ug/ml NEF RNA and 500 ug/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 ug/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., 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 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. 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 μl 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 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 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 μgrams of pRSVCAT DNA plasmid or 100 μgrams of βgCATβgAn 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.
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. 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 βgCATβgAn 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 μl of a lysis solution (20 mM Tris, pH 7.4, 2 mM MgCl2 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 C14-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. Lane numbers appear at the top of the autoradiograms and the % chloramphenicol conversions are at the bottom. Sample locations are as follows:
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 μg of βgCATβgAn RNA in 5% sucrose
Lanes 11, 12, and 17 to 20: Muscle injected with 100 μgrams pRSVCAT DNA in 5% sucrose
Lanes 9 and 10: 20 μgrams of βgCATβgAn RNA, lipofected, with 60 μgrams of DOTMA, into a 70% confluent 60 mm plate of 3T3 cells (106)
Lanes 21, 22: 20 μgrams of pRSVCAT lipofected, with 60 μg of DOTMA, into a 50% confluent 60 mm plate of 3T3 cells
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 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 Molec. Cell Biol. 5: 281-290 (1985)) expressing the E. coil β-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 N2-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:
(A) and (B): Cross-sections of a muscle injected with pRSVLacZ at 25×and 160×, magnification, respectively.
(C): A longitudinal section of another muscle injected with pRSVLacZ, 160×.
(D) (E) and (F): Serial cross-sections of the same muscle that are 0.6 mm apart.
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 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 μm (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.
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.
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. Molec. Cell Biol. 7: 725-737(1987)) using an LKB 1251 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 3C illustrate the results of the following:
3(A) Luciferase activity measured 18 hours following the injection of varying amounts of βgLUCβgAn RNA in 20% sucrose and 4 days following the injection of varying amounts of pRSVL in 20% sucrose
3(B) Luciferase activity assayed at varying times after 20 μg of βgLUCβgAn 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.
A. Level of Gene Expression
A dose-response effect was observed when quadriceps muscles were injected with various amounts of βgLucβgAn 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.
B. Time Course of Expression
The time course was also investigated (FIGS. 3B and 3C). Luciferase activity was assayed at varying times after 25 μg of βgLucβgAn 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.
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 N2 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, New York), using a vacublot apparatus (LKB) and hybridized with multiprimed 32P-luciferase probe (the HindIII-BamH1 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:
Lane 1: 0.05 ng of undigested pRSVL plasmid
Lane 2: 0.05 ng of BamH1 digested pRSVL
Lane 3: Blank
Lane 4: BamH1 digest of HIRT supernatant from control muscle
Lane 5: BamH1 digest of cellular DNA from control, uninjected muscle
Lanes 6,7: BamH1 digest of HIRT supernatant from two different pools of pRSVL injected muscles
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
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 (λ/HindIII) are shown at the left).
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, 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:
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 MgCl2 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 21.2 21.5 21.8 21.6 21.9 21.2
alone
 25 μg DNA 21.7 21.4 21.3 22.2 21.5
alone
250 μg DNA 21.7 23.2 21.9 28.5 22.6 22.0 21.3
alone
250 μg DNA 22.9 22.5 33.3 23.0 25.4 24.3 21.5
alone
250 μg DNA 21.8 21.5 21.8 20.4 20.7 20.8 20.7
alone
 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
3 × 104 l.u. 105.3  77.1 98.7 80.0 86.3 89.6 178.9 
Blank 22.5
Mock: Values are those for an animal that received 25 μg of DNA in 0.3 ml 20% sucrose into the esophagus. (A sample containing only water yields 22.5 l.u.)
25 μg DNA alone: represent separate animals that received intratracheal injections of 25 μg of pPGKLuc in 0.3 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 ml 5% sucrose.
The above animals were sacrificed and lung extracts prepared 2 days after injection.
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 μg 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 1251 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 (IH) 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 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.
Luciferase Activity
Mice Intrahepatic (Light Units, LU)
Liver Injection Lower Upper
Blank (20.2 LU)
Control: 20% Sucrose Only 20.8 23.8
 50 μg pPGKLuc + 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 says after similar preparations of pPGKLuc (+ and −Lipofectin™) were injected into the portal circulation of rats.
EXAMPLE 18 Expression of Growth Hormone Gene Injected into Liver and Muscle
Mice were injected with the pXGH5 (metalothionien 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.
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.
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:
Animal Growth Hormone (ng/ml):
No. Day 1 Day 2
1 0.6 0.6
2 0.8 1.0
3  0.95 0.8
Background: 0.5 ng/ml
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 retains its signal sequence, and the protein is efficiently excreted from cells.
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 I2 (FIG. 6A) was injected with the transfected cells. Mouse I1 (FIG. 6A) received an identical number of untransfected cells. Blood samples were obtained prior to the injection (Day 0) and at the times indicated in FIG. 6B. The serum samples were processed as in the preceding example. Both IgG and IgM antibodies were detected as indicated in FIG. 6.
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 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 EcoR1/Nco1 fragment from the encephalomyocarditis virus (EMCV). (See construction of plasmid pE5LVPO from DNA of plasmid pE3T11 in Parks, G. et al., J. Virology 60: 376-384, at 378 (1986)). These EMC 5′ untranslated sequences had previously been shown to be able to initiate efficient translation in vitro in reticulocyte 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 βGLacZβGAn 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, O. 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., 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.
(b) pSVNU-G1-A: pAR3132-SV40 promotor driving expression of T7 polymerase protein which is directed to the cytoplasm.
Each of these two plasmids were co-lipofected with pEMCLacZβGAn at 1:3 and 3:1 ratios into a 60 mm plates of 3T3 cells. The number of blue β-galactosidase cells were counted and scored as indicated below.
β-gal Ratio:template/ Co-Lipofectant:
template polymerase vector pSV-G1-A pSVNU-G1-A
βGLacZβGAn 3:1  0  1
1:3  0  1
EMCLacZβGAn 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 βgLucβgAn 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 μl, 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:
Animal Hemisphere:
Treatment I.D. Left Right
Sham Injection AMra   649   629
βgLucβgAn AMrb 1,734 1,911
The newborn mouse was injected with 1 μl βgLucβgAn (3.6 μg/μl; 20% sucrose) into the bilateral forebrain and tissues were similarly processed and analyzed.
Animal Hemisphere:
Treatment 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” Genomics 3: 189-202 (1988); Hoffman, E. P. and Kunkel, E. M., “Dystrophin abnormalities of Duchenne's/Becher Muscular Dystrophy” Neuron 2: 1019-1029 (1989); Koenig, M., Monaco, A. P., Kunkel, L. M. “The Complete Sequence of Dystrophin Predicts a Rod-Shaped Cytoskeletal Protein” Cell 53 (2): 219-226 (1988)). 200 μg of the plasmid in 100 μl 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 Jun. 30, 1988; 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-60kd antibody.
EXAMPLE 25 Admistration of the Correcting Dystrophin Gene Directly into the Muscle of Patients with Duchenne's Muscular Dystrophy
Patients with muscular dystrophy are given multiple 200 ug injections of plasmid containing the functional dystrophin gene (see previous example) in 100 ul 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 accessed by a surgical procedure to expose the muscle to direct injection of plasmid DNA.
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.

Claims (79)

What is claimed is:
1. A method of producing antibodies directed to gp-120 protein of HIV virus in a mouse, comprising injecting into muscle of said mouse a DNA plasmid constructed of sequence encoding said gp-120 operably linked to a CMV promoter in an amount sufficient to generate said antibodies; wherein said plasmid is free from association with transfection-facilitating proteins, viral particles, liposomes, cationic lipids, and calcium phosphate precipitating agents.
2. A process for generating a detectable antibody response in a mammal, comprising:
administering in vivo into a tissue of the mammal a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the mammal occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with transfection-facilitating proteins, viral particles, liposomal formulations and charged lipids.
3. The process of claim 2, wherein said construct is introduced into mucous membrane tissue.
4. The process of claim 2, wherein the mammal is human.
5. The process of claim 2, wherein said construct is further free from association with transfection facilitating calcium phosphate.
6. The process of claim 2, wherein said DNA comprises a plasmid.
7. The process of claim 2, wherein said administration is intravenous or intranasal.
8. The process of claim 7, wherein said construct is injected.
9. The process of claim 7, wherein said administration is intravenous.
10. The process of claim 7, wherein said administration is intranasal.
11. The process of claim 2, wherein said immunogen is viral.
12. The process of claim 2, wherein said uptake is into tissue cells.
13. A process for inducing an effective immune response in a mammal to a pathogen comprising:
administering in vivo into a tissue of the mammal a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the mammal occurs, and sufficient expression results, to induce the effective immune response to a pathogen,
wherein said construct is free from association with transfection-facilitating proteins, viral particles, liposomal formulations and charged lipids.
14. The process of claim 13, wherein said construct is introduced into mucous membrane tissue.
15. The process of claim 13, wherein the mammal is human.
16. The process of claim 13, wherein said construct is further free from association with transfection facilitating calcium phosphate.
17. The process of claim 13, wherein said DNA comprises a plasmid.
18. The process of claim 13, wherein said administration is intravenous or intranasal.
19. The process of claim 18, wherein said construct is injected.
20. The process of claim 18, wherein said administration is intravenous.
21. The process of claim 18, wherein said administration is intranasal.
22. The process of claim 13, wherein said immunogen is viral.
23. The process of claim 13, wherein said uptake is into tissue cells.
24. A process for generating a detectable antibody response in a vertebrate, comprising:
administering in vivo into a tissue of the vertebrate a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the vertebrate occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with transfection-facilitating proteins, viral particles, liposomal formulations and charged lipids.
25. The process of claim 24, wherein said construct is introduced into mucous membrane tissue.
26. The process of claim 24, wherein said construct is introduced into muscle.
27. The process of claim 24, wherein said construct is introduced into skin.
28. The process of claim 24, wherein said construct is further free from association with transfection facilitating calcium phosphate.
29. The process of claim 24, wherein said DNA comprises a plasmid.
30. The process of claim 24, wherein said administration is intramuscular, intravenous, intradermal subcutaneous, or intranasal.
31. The process of claim 30, wherein said construct is injected.
32. The process of claim 30, wherein said administration is intramuscular.
33. The process of claim 30, wherein said administration is intravenous.
34. The process of claim 30, wherein said administration is intradermal.
35. The process of claim 30, wherein said administration is subcutaneous.
36. The process of claim 30, wherein said administration is intranasal.
37. The process of claim 24, wherein said immunogen is viral.
38. The process of claim 24, wherein said uptake is into tissue cells.
39. The process of claim 24, wherein the vertebrate is a bird.
40. The process of claim 24, wherein the vertebrate is a fish.
41. A process for generating a detectable antibody response in a mammal, comprising:
injecting into muscle of the mammal a construct comprising a noninfectious, nonintegrating mRNA sequence encoding an immunogen in an amount sufficient that uptake of said construct occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with transfection-facilitating proteins, viral particles, liposomal formulations and charged lipids.
42. The process of claim 41, wherein the mamnal is human.
43. The process of claim 41, wherein said construct is further free from association with transfection including calcium phosphate.
44. The process of claim 41, wherein said immunogen is viral.
45. The process of claim 41, wherein said uptake is into said muscle.
46. A method of generating an effective immune response against a pathogen in a mammal, comprising:
administering in vivo into a tissue of a mammal in need of said immune response a composition comprising a DNA plasmid which directs synthesis of an immunogenic peptide or polypeptide in mammalian cells through association with a promoter, and a pharmaceutically acceptable carrier;
wherein said DNA plasmid is free from association with liposomal formulations and charged lipids;
wherein said DNA plasmid incorporates into the cells of said mammal; and
wherein sufficient expression of said immunogenic peptide or polypeptide occurs to generate said effective immune response against said pathogen in said mammal.
47. The method of claim 46, wherein said immunogenic peptide or polypeptide metabolically incorporates into the cell membranes of said mammal by binding to a class I major histocompatibility molecule, a class II major histocompatibility molecule, or a combination of class I and class II major histocompatibility molecules.
48. The method of claim 46, wherein said promoter is selected from the group consisting of a Rous sarcoma virus long terminal repeat (RSVLTR), a myeloproliferative sarcoma virus long terminal repeat (MPSVLTR), a simian virus 40 immediate early promoter (SV40IEP), a metallothionein promoter, and a human cytomegalovirus immediate early promoter (CMVIEP).
49. The method of claim 48, wherein said promoter is a human cytomegalovirus immediate early IEP promoter.
50. The method of claim 46, wherein said tissue is selected from the group consisting of muscle, skin, and blood.
51. The method of claim 46, wherein said composition is injected.
52. The method of claim 46, wherein said tissue is muscle.
53. The method of claim 46, wherein said immunogenic peptide or polypeptide is a viral protein.
54. A method of generating an effective immune response against a pathogen in a mammal, comprising:
administering in vivo into a tissue of a mammal in need of said immune response a composition consisting essentially of a DNA plasmid operably encoding an immunogenic peptide or polypeptide through association with a promoter, and a pharmaceutically acceptable carrier;
wherein said plasmid incorporates into the cells of said mammal; and
wherein sufficient expression of said immunogenic peptide or polypeptide occurs to generate said effective immune response against said pathogen in said mammal.
55. The method of claim 54, wherein said immunogenic peptide or polypeptide metabolically incorporates into the cell membranes of said mammal by binding to a class I major histocompatibility molecule, a class II major histocompatibility molecule, or a combination of class I and class II major histocompatibility molecules.
56. The method of claim 54, wherein said promoter is selected from the group consisting of a Rous sarcoma virus long terminal repeat (RSVLTR), a myeloproliferative sarcoma virus long terminal repeat (MPSVLTR), a simian virus 40 immediate early promoter (SV40IEP), a metallothionein promoter, and a human cytomegalovirus immediate early promoter (CMVIEP).
57. The method of claim 56, wherein said promoter is a human cytomegalovirus immediate early promoter.
58. The method of claim 54, wherein said tissue is selected from the group consisting of muscle, skin, and blood.
59. The method of claim 54, wherein said composition is injected.
60. The method of claim 54, wherein said tissue is muscle.
61. The method of claim 54, wherein said immunogenic peptide or polypeptide is a viral protein.
62. A method of generating an effective immune response against a pathogen in a mammal, comprising:
administering in vivo into a tissue of a mammal in need of said immune response a composition consisting of a DNA plasmid operably encoding an immunogenic peptide or polypeptide through association with a promoter, and a pharmaceutically acceptable carrier;
wherein said plasmid incorporates into the cells of said mammal; and
wherein sufficient expression of said immunogenic peptide or polypeptide occurs to generate said effective immune response against said pathogen in said mammal.
63. The method of claim 62, wherein said immunogenic peptide or polypeptide metabolically incorporates into the cell membranes of said mammal by binding to a class I major histocompatibility molecule, a class II major histocompatibility molecule, or a combination of class I and class II major histocompatibility molecules.
64. The method of claim 62, wherein said promoter is selected from the group consisting of a Rous sarcoma virus long terminal repeat (RSVLTR), a myeloproliferative sarcoma virus long terminal repeat (MPSVLTR), a simian virus 40 immediate early promoter (SV40IEP), a metallothionein promoter, and a human cytomegalovirus immediate early promoter (CMVIEP).
65. The method of claim 64, wherein said promoter is a human cytomegalovirus immediate early promoter.
66. The method of claim 62, wherein said tissue is selected from the group consisting of muscle, skin, and blood.
67. The method of claim 62, wherein said composition is injected.
68. The method of claim 62, wherein said tissue is muscle.
69. The method of claim 62, wherein said immunogenic peptide or polypeptide is a viral protein.
70. A method for immunizing a mammal against infection by a pathogen, comprising:
obtaining an injectable preparation consisting of a pharmaceutically acceptable injectable carrier containing an expressible polynucleotide coding for an immunogenic peptide or polypeptide, wherein said polynucleotide is a DNA plasmid operably encoding said immunogenic peptide or polypeptide through association with a promoter;
introducing said injectable preparation into a mammal in need of said immunization, whereby said polynucleotide incorporates into a cell where an immunogenic translation product of said polynucleotide is formed, and said product is processed and presented by said cell in the context of the major histocompatibility complex, thereby eliciting an effective immune response against said pathogen.
71. A method of generating an effective immune response against a pathogen in a mammal, comprising:
administering in vivo into muscle tissue of a mammal in need of said immune response a composition comprising a polynucleotide which directs synthesis of an immunogenic peptide or polypeptide in mammalian cells, and a pharmaceutically acceptable carrier;
wherein said polynucleotide is selected from the group consisting of a messenger RNA and a DNA plasmid operably encoding said immunogenic peptide or polypeptide through association with a promoter;
wherein said polynucleotide is free from association with liposomal formulations and charged lipids;
wherein said polynucleotide incorporates into the cells of said mammal; and
wherein sufficient expression of said immunogenic peptide or polypeptide occurs to generate said effective immune response against said pathogen in said mammal.
72. The method of claim 71, wherein said polynucleotide is messenger RNA.
73. The method of claim 72, wherein said immunogenic peptide or polypeptide metabolically incorporates into the cell membranes of said mammal by binding to a class I major histocompatibility molecule, a class II major histocompatibility molecule, or a combination of class I and class II major histocompatibility molecules.
74. The method of claim 72, wherein said composition is injected.
75. The method of claim 72, wherein said immunogenic peptide or polypeptide is a viral protein.
76. A process for generating a detectable antibody response in a mammal, comprising:
administering in vivo into a tissue of the mammal a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the mammal occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with any transfection-facilitating material.
77. A process for inducing an effective immune response in a mammal to a pathogen comprising:
administering in vivo into a tissue of the mammal a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the mammal occurs, and sufficient expression results, to induce the effective immune response to a pathogen,
wherein said construct is free from association with any transfection-facilitating material.
78. A process for generating a detectable antibody response in a vertebrate, comprising:
administering in vivo into a tissue of the vertebrate a construct comprising a noninfectious, nonintegrating DNA sequence encoding an immunogen, wherein said DNA is operably linked to a promoter, in an amount sufficient that uptake of said construct into cells of the vertebrate occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with any transfection-facilitating material.
79. A process for generating a detectable antibody response in a mammal, comprising:
injecting into muscle of the mammal a construct comprising a noninfectious, nonintegrating mRNA sequence encoding an immunogen in an amount sufficient that uptake of said construct occurs, and sufficient expression results, to generate the detectable antibody response,
wherein said construct is free from association with transfection-facilitating material.
US08/481,919 1989-03-21 1995-06-07 Induction of a protective immune response in a mammal by injecting a DNA sequence Expired - Lifetime US6214804B1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
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

Applications Claiming Priority (4)

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

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US49699190A Continuation 1989-03-21 1990-03-21
US49699190A Division 1989-03-21 1990-03-21

Related Child 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
US6214804B1 true US6214804B1 (en) 2001-04-10

Family

ID=46255738

Family Applications (1)

Application Number Title Priority Date Filing Date
US08/481,919 Expired - Lifetime US6214804B1 (en) 1989-03-21 1995-06-07 Induction of a protective immune response in a mammal by injecting a DNA sequence

Country Status (1)

Country Link
US (1) US6214804B1 (en)

Cited By (181)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002008448A2 (en) * 2000-07-25 2002-01-31 The Board Of Trustees Of The Leland Stanford Junior University Non-viral linear dna vectors and methods for using the same
US20020019358A1 (en) * 2000-04-21 2002-02-14 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US6348449B1 (en) * 1993-09-21 2002-02-19 The Trustees Of The University Of Pennsylvania Methods of inducing mucosal immunity
WO2002098443A2 (en) * 2001-06-05 2002-12-12 Curevac Gmbh Stabilised mrna with an increased g/c content and optimised codon for use in gene therapy
US20030032615A1 (en) * 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US20030049801A1 (en) * 2001-09-10 2003-03-13 Chung-Hsiun Wu 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
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US20030073142A1 (en) * 2001-08-13 2003-04-17 Chen Swey-Shen Alex Immunoglobulin E vaccines and methods of use thereof
US20030083272A1 (en) * 1997-09-19 2003-05-01 Lahive & Cockfield, Llp Sense mrna therapy
US20030091544A1 (en) * 2001-03-13 2003-05-15 Vical Incorporated Interferon-Beta polynucleotide therapy for autoimmune and inflammatory diseases
US6602705B1 (en) 1998-12-31 2003-08-05 Chiron Corporation Expression of HIV polypeptides and production of virus-like particles
US20030170614A1 (en) * 2001-08-31 2003-09-11 Megede Jan Zur Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20030175292A1 (en) * 2000-03-02 2003-09-18 Robinson Harriet L. Compositions and methods for generating an immune response
US20030176377A1 (en) * 2002-03-02 2003-09-18 Rong Xiang DNA vaccines encoding CEA and a CD40 ligand and methods of use thereof
US20030186913A1 (en) * 1990-03-21 2003-10-02 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate
US20030185802A1 (en) * 2002-03-02 2003-10-02 Reisfeld Ralph A. DNA vaccine against proliferating endothelial cells and methods of use thereof
US20030194800A1 (en) * 2001-08-31 2003-10-16 Megede Jan Zur Polynucleotides encoding antigenic HIV type B 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
US20030198626A1 (en) * 2002-04-22 2003-10-23 Antigen Express, Inc. Inhibition of Ii expression in mammalian cells
US20030203863A1 (en) * 1994-11-28 2003-10-30 Vical Incorporated Plasmids suitable for IL-2 expression
US20030223961A1 (en) * 2000-07-05 2003-12-04 Megede Jan Zur Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US6689879B2 (en) 1998-12-31 2004-02-10 Chiron Corporation Modified HIV Env polypeptides
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs
US6716882B2 (en) 1993-12-20 2004-04-06 Invitrogen Corporation Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US20040142468A1 (en) * 2001-03-16 2004-07-22 Pardoll Drew M Modulation of systemic immune responses by transplantation of hematopoietic stem cells transduced with genes encoding antigens and antigen presenting cell regulatory molecules
US20040146528A1 (en) * 2001-03-08 2004-07-29 Bernard Moss MVA expressing modified HIV envelope, gag, and pol genes
US20040152656A1 (en) * 1994-07-15 2004-08-05 University Of Iowa Research Foundation Immunomodulatory oligonucleotides
WO2004065575A2 (en) 2003-01-15 2004-08-05 Research Institute For Genetic And Human Therapy (Right) Dna compositon and uses thereof
US20040176282A1 (en) * 2003-01-09 2004-09-09 Brian Dalby Cellular delivery and activation of polypeptide-nucleic acid complexes
US20040266719A1 (en) * 1998-05-22 2004-12-30 Mccluskie Michael J. Methods and products for inducing mucosal immunity
US6849455B1 (en) 2000-08-22 2005-02-01 New York University Enhanced recovery of transformed cells
US20050054601A1 (en) * 1997-01-23 2005-03-10 Coley Pharmaceutical Gmbh Pharmaceutical composition comprising a polynucleotide and optionally an antigen especially for vaccination
US20050059624A1 (en) * 2001-12-19 2005-03-17 Ingmar Hoerr Application of mRNA for use as a therapeutic against tumour diseases
US6890554B2 (en) * 1993-06-01 2005-05-10 Invitrogen Corporation Genetic immunization with cationic lipids
US20050164971A1 (en) * 1998-11-12 2005-07-28 Yongliang Chu New transfection reagents
US6936464B1 (en) 1992-10-02 2005-08-30 Cancer Research Technology Limited Immune responses to fusion proteins
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US20050260757A1 (en) * 1994-02-11 2005-11-24 Invitrogen Coroporation Novel reagents for intracellular delivery of macromolecules
US20050277127A1 (en) * 2003-11-26 2005-12-15 Epitomics, Inc. High-throughput method of DNA immunogen preparation and immunization
US6977074B2 (en) 1997-07-10 2005-12-20 Mannkind Corporation Method of inducing a CTL response
US20050287118A1 (en) * 2003-11-26 2005-12-29 Epitomics, Inc. Bacterial plasmid with immunological adjuvant function and uses thereof
US20060003316A1 (en) * 2002-07-15 2006-01-05 John Simard Immunogenic compositions derived from poxviruses and methods of using same
US20060008448A1 (en) * 1996-06-11 2006-01-12 Minzhen Xu Inhibition of li expression in mammalian cells
US20060024670A1 (en) * 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
US6994851B1 (en) 1997-07-10 2006-02-07 Mannkind Corporation Method of inducing a CTL response
US7001759B1 (en) * 1993-01-26 2006-02-21 The Trustees Of The University Of Pennsylvania Compositions and methods for delivery of genetic material
WO2006024518A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immunostimulation
US20060153844A1 (en) * 2004-12-29 2006-07-13 Thomas Kundig Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
KR100600988B1 (en) 2002-03-13 2006-07-13 주식회사 엘지생명과학 Method for enhancing immune responses by codelivering influenza NP DNA in DNA immunization
US20060222629A1 (en) * 1993-01-26 2006-10-05 Weiner David B Compositions and methods for delivery of genetic material
WO2006122828A2 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized injection formulation for rna
WO2006130581A2 (en) 2005-05-31 2006-12-07 Avigen, Inc. Methods for delivering genes
US20070048861A1 (en) * 2000-03-02 2007-03-01 Robinson Harriet L Compositions and methods for generating an immune response
US7211659B2 (en) 2001-07-05 2007-05-01 Chiron Corporation Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20070105193A1 (en) * 2003-05-16 2007-05-10 Vical Incorporated Severe acute respiratory syndrome DNA vaccine compositions and methods of use
US7268120B1 (en) 1997-11-20 2007-09-11 Vical Incorporated Methods for treating cancer using cytokine-expressing polynucleotides
US20080131455A1 (en) * 2004-04-30 2008-06-05 Pds Biotechnology Corporation Antigen Delivery Compositions And Methods Of Use
US20080153166A1 (en) * 1995-01-23 2008-06-26 Leaf Huang Stable lipid-comprising drug delivery complexes and methods for their production
WO2008074678A1 (en) 2006-12-18 2008-06-26 F. Hoffmann-La Roche Ag Novel use of inhibitors of soluble epoxide hydrolase
US20090017057A1 (en) * 2007-03-22 2009-01-15 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
US20090047339A1 (en) * 1998-12-31 2009-02-19 Barnett Susan W Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20090068214A1 (en) * 2005-04-15 2009-03-12 Jiahua Qian Methods and Compositions for Producing an Enhanced Immune Response to a Human Papillomavirus Immunogen
US7534772B2 (en) 2000-06-22 2009-05-19 University Of Iowa Research Foundation Methods for enhancing antibody-induced cell lysis and treating cancer
US7648468B2 (en) 2002-04-19 2010-01-19 Pelikon Technologies, Inc. Method and apparatus for penetrating tissue
US7666149B2 (en) 1997-12-04 2010-02-23 Peliken Technologies, Inc. Cassette of lancet cartridges for sampling blood
US7674232B2 (en) 2002-04-19 2010-03-09 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7682318B2 (en) 2001-06-12 2010-03-23 Pelikan Technologies, Inc. Blood sampling apparatus and method
US7699791B2 (en) 2001-06-12 2010-04-20 Pelikan Technologies, Inc. Method and apparatus for improving success rate of blood yield from a fingerstick
US7708701B2 (en) 2002-04-19 2010-05-04 Pelikan Technologies, Inc. Method and apparatus for a multi-use body fluid sampling device
US7717863B2 (en) 2002-04-19 2010-05-18 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7731729B2 (en) 2002-04-19 2010-06-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7749174B2 (en) 2001-06-12 2010-07-06 Pelikan Technologies, Inc. Method and apparatus for lancet launching device intergrated onto a blood-sampling cartridge
US7780631B2 (en) 1998-03-30 2010-08-24 Pelikan Technologies, Inc. Apparatus and method for penetration with shaft having a sensor for sensing penetration depth
US7795017B2 (en) 2000-03-02 2010-09-14 Emory University DNA expression vectors and methods of use
US7822454B1 (en) 2005-01-03 2010-10-26 Pelikan Technologies, Inc. Fluid sampling device with improved analyte detecting member configuration
US7833171B2 (en) 2002-04-19 2010-11-16 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
EP2258841A1 (en) 2003-06-23 2010-12-08 The Regents of the University of Colorado Methods for treating pain
US7850621B2 (en) 2003-06-06 2010-12-14 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US7850622B2 (en) 2001-06-12 2010-12-14 Pelikan Technologies, Inc. Tissue penetration device
US7862520B2 (en) 2002-04-19 2011-01-04 Pelikan Technologies, Inc. Body fluid sampling module with a continuous compression tissue interface surface
US7874994B2 (en) 2002-04-19 2011-01-25 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7892185B2 (en) 2002-04-19 2011-02-22 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US7892183B2 (en) 2002-04-19 2011-02-22 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US7901365B2 (en) 2002-04-19 2011-03-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7901362B2 (en) 2002-04-19 2011-03-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7909778B2 (en) 2002-04-19 2011-03-22 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7909777B2 (en) 2002-04-19 2011-03-22 Pelikan Technologies, Inc Method and apparatus for penetrating tissue
US7914465B2 (en) 2002-04-19 2011-03-29 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7976476B2 (en) 2002-04-19 2011-07-12 Pelikan Technologies, Inc. Device and method for variable speed lancet
US7988645B2 (en) 2001-06-12 2011-08-02 Pelikan Technologies, Inc. Self optimizing lancing device with adaptation means to temporal variations in cutaneous properties
US8007446B2 (en) 2002-04-19 2011-08-30 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8079960B2 (en) 2002-04-19 2011-12-20 Pelikan Technologies, Inc. Methods and apparatus for lancet actuation
US8197421B2 (en) 2002-04-19 2012-06-12 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8221334B2 (en) 2002-04-19 2012-07-17 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
EP2484770A1 (en) 2007-09-04 2012-08-08 CureVac GmbH Complexes of RNA and cationic peptides for transfection and for immunostimulation
WO2012106281A2 (en) 2011-01-31 2012-08-09 The General Hospital Corporation Multimodal trail molecules and uses in cellular therapies
US8262614B2 (en) 2003-05-30 2012-09-11 Pelikan Technologies, Inc. Method and apparatus for fluid injection
US8267870B2 (en) 2002-04-19 2012-09-18 Sanofi-Aventis Deutschland Gmbh Method and apparatus for body fluid sampling with hybrid actuation
WO2012130941A2 (en) 2011-03-31 2012-10-04 Schaefer Konstanze Perfluorinated compounds for the non-viral transfer of nucleic acids
US8282576B2 (en) 2003-09-29 2012-10-09 Sanofi-Aventis Deutschland Gmbh Method and apparatus for an improved sample capture device
WO2012170765A2 (en) 2011-06-10 2012-12-13 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
US8337421B2 (en) 2001-06-12 2012-12-25 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8344019B2 (en) 2004-03-19 2013-01-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods for the production of biliverdin
CN102879566A (en) * 2012-10-12 2013-01-16 武汉康苑生物医药科技有限公司 Enzyme-linked immunosorbent assay (ELISA) kit for human immunodeficiency virus (HIV) P24 antigen
US8360992B2 (en) 2002-04-19 2013-01-29 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
EP2568289A2 (en) 2011-09-12 2013-03-13 International AIDS Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
EP2586461A1 (en) 2011-10-27 2013-05-01 Christopher L. Parks Viral particles derived from an enveloped virus
US8435190B2 (en) 2002-04-19 2013-05-07 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8524680B2 (en) 2002-02-01 2013-09-03 Applied Biosystems, Llc High potency siRNAS for reducing the expression of target genes
US8556829B2 (en) 2002-04-19 2013-10-15 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8574895B2 (en) 2002-12-30 2013-11-05 Sanofi-Aventis Deutschland Gmbh Method and apparatus using optical techniques to measure analyte levels
EP2679596A1 (en) 2012-06-27 2014-01-01 Simon Hoffenberg HIV-1 env glycoprotein variant
US8641644B2 (en) 2000-11-21 2014-02-04 Sanofi-Aventis Deutschland Gmbh Blood testing apparatus having a rotatable cartridge with multiple lancing elements and testing means
US8652831B2 (en) 2004-12-30 2014-02-18 Sanofi-Aventis Deutschland Gmbh Method and apparatus for analyte measurement test time
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
WO2014035474A1 (en) 2012-08-30 2014-03-06 The General Hospital Corporation Compositions and methods for treating cancer
US8668656B2 (en) 2003-12-31 2014-03-11 Sanofi-Aventis Deutschland Gmbh Method and apparatus for improving fluidic flow and sample capture
US8702624B2 (en) 2006-09-29 2014-04-22 Sanofi-Aventis Deutschland Gmbh Analyte measurement device with a single shot actuator
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US8721671B2 (en) 2001-06-12 2014-05-13 Sanofi-Aventis Deutschland Gmbh Electric lancet actuator
US8784335B2 (en) 2002-04-19 2014-07-22 Sanofi-Aventis Deutschland Gmbh Body fluid sampling device with a capacitive sensor
US8815821B2 (en) 2002-02-01 2014-08-26 Life Technologies Corporation Double-stranded oligonucleotides
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8828203B2 (en) 2004-05-20 2014-09-09 Sanofi-Aventis Deutschland Gmbh Printable hydrogels for biosensors
WO2014145042A1 (en) 2013-03-15 2014-09-18 Loma Linda University Treatment of autoimmune diseases
US20140294939A1 (en) * 2011-06-08 2014-10-02 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mrna
WO2014172560A1 (en) 2013-04-17 2014-10-23 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
WO2014190040A1 (en) 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
US8965476B2 (en) 2010-04-16 2015-02-24 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP2848937A1 (en) 2013-09-05 2015-03-18 International Aids Vaccine Initiative Methods of identifying novel HIV-1 immunogens
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
EP2873423A2 (en) 2013-10-07 2015-05-20 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
US9061021B2 (en) 2010-11-30 2015-06-23 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
WO2015120309A1 (en) 2014-02-06 2015-08-13 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9144401B2 (en) 2003-06-11 2015-09-29 Sanofi-Aventis Deutschland Gmbh Low pain penetrating member
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US9226699B2 (en) 2002-04-19 2016-01-05 Sanofi-Aventis Deutschland Gmbh Body fluid sampling module with a continuous compression tissue interface surface
US9248267B2 (en) 2002-04-19 2016-02-02 Sanofi-Aventis Deustchland Gmbh Tissue penetration device
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9314194B2 (en) 2002-04-19 2016-04-19 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9351680B2 (en) 2003-10-14 2016-05-31 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a variable user interface
US9375169B2 (en) 2009-01-30 2016-06-28 Sanofi-Aventis Deutschland Gmbh Cam drive for managing disposable penetrating member actions with a single motor and motor and control system
US9386944B2 (en) 2008-04-11 2016-07-12 Sanofi-Aventis Deutschland Gmbh Method and apparatus for analyte detecting device
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9427532B2 (en) 2001-06-12 2016-08-30 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
EP3069730A2 (en) 2015-03-20 2016-09-21 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
EP3072901A1 (en) 2015-03-23 2016-09-28 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2016168601A1 (en) 2015-04-17 2016-10-20 Khalid Shah Agents, systems and methods for treating cancer
WO2016180430A1 (en) 2015-05-08 2016-11-17 Curevac Ag Method for producing rna
EP2101823B1 (en) 2007-01-09 2016-11-23 CureVac AG Rna-coded antibody
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP3187585A1 (en) 2010-03-25 2017-07-05 Oregon Health&Science University Cmv glycoproteins and recombinant vectors
US9775553B2 (en) 2004-06-03 2017-10-03 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a fluid sampling device
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9795747B2 (en) 2010-06-02 2017-10-24 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US9820684B2 (en) 2004-06-03 2017-11-21 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a fluid sampling device
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
EP3354644A1 (en) * 2011-06-08 2018-08-01 Translate Bio, Inc. Cleavable lipids
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10780052B2 (en) 2013-10-22 2020-09-22 Translate Bio, Inc. CNS delivery of MRNA and uses thereof
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10898584B2 (en) 2013-11-01 2021-01-26 Curevac Ag Modified RNA with decreased immunostimulatory properties
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11612652B2 (en) 2015-11-13 2023-03-28 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
US11801257B2 (en) 2008-04-17 2023-10-31 Pds Biotechnology Corporation Stimulation of an immune response by enantiomers of cationic lipids
US11904015B2 (en) 2012-09-21 2024-02-20 Pds Biotechnology Corporation Vaccine compositions and methods of use
US11951179B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR7781M (en) 1967-06-19 1970-03-23
US3931397A (en) 1971-11-05 1976-01-06 Beecham Group Limited Biologically active material
US4224404A (en) 1976-12-16 1980-09-23 The International Institute Of Differentiation Limited 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
WO1986000930A1 (en) 1984-07-20 1986-02-13 Worcester Foundation For Experimental Biology Retroviral vaccines and vectors and methods for their construction
EP0187702A1 (en) 1985-01-07 1986-07-16 Syntex (U.S.A.) Inc. N-(omega,omega-1-dialkoxy)- and N-(omega,omega-1-dialkenoxy)-alk-1-yl-N,N,N-trisubstituted ammonium surfactants, their preparation and pharmaceutical formulations containing them
US4689320A (en) 1983-10-17 1987-08-25 Akira Kaji Method for inhibiting propagation of virus and anti-viral agent
US4699880A (en) 1984-09-25 1987-10-13 Immunomedics, Inc. Method of producing monoclonal anti-idiotype antibody
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
US4738927A (en) 1982-03-31 1988-04-19 Ajinomoto Co. Inc. Gene coded for interleukin-2 polypeptide, recombinant DNA carrying the said gene, a living cell line possessing the recombinant DNA, and method for producing interleukin-2 using the said cell
WO1988005077A1 (en) 1986-12-29 1988-07-14 Battelle Memorial Institute A method of modifiying the metabolism of eukaryotic cells upon incorporation therein of foreign sequences of nucleic acids by means of an internalizing vector
US4761375A (en) 1984-05-08 1988-08-02 Genetics Institute, Inc. Human interleukin-2 cDNA sequence
JPS63285738A (en) 1987-05-19 1988-11-22 Canon Inc Magneto-optical recording medium
US4806463A (en) 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
JPS6447381A (en) 1987-08-19 1989-02-21 Vitamin Kenkyusho Kk Preparation of liposome containing sealed gene
WO1990001543A1 (en) 1988-07-29 1990-02-22 Intracel Corporation Method for the genetic expression of heterologous proteins by cells transfected in vivo
US4945050A (en) 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
JPH03102682A (en) 1989-09-14 1991-04-30 Sony Corp Disk driving device
WO1991007487A1 (en) 1989-11-16 1991-05-30 Duke University Particle-mediated transformation of animal tissue cells
WO1991016024A1 (en) 1990-04-19 1991-10-31 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
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
WO1997037966A1 (en) 1996-04-09 1997-10-16 Vical Incorporated Quaternary cytofectins
JP3102682B2 (en) 1997-02-20 2000-10-23 大淀小松株式会社 Station platform snow removal equipment

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR7781M (en) 1967-06-19 1970-03-23
US3931397A (en) 1971-11-05 1976-01-06 Beecham Group Limited Biologically active material
US4224404A (en) 1976-12-16 1980-09-23 The International Institute Of Differentiation Limited 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
US4738927A (en) 1982-03-31 1988-04-19 Ajinomoto Co. Inc. Gene coded for interleukin-2 polypeptide, recombinant DNA carrying the said gene, a living cell line possessing the recombinant DNA, and method for producing interleukin-2 using the said cell
US4689320A (en) 1983-10-17 1987-08-25 Akira Kaji Method for inhibiting propagation of virus and anti-viral agent
US4761375A (en) 1984-05-08 1988-08-02 Genetics Institute, Inc. Human interleukin-2 cDNA sequence
EP0188574A1 (en) 1984-07-20 1986-07-30 Worcester Foundation For Experimental Biology Retroviral vaccines and vectors and methods for their construction
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
EP0187702A1 (en) 1985-01-07 1986-07-16 Syntex (U.S.A.) Inc. N-(omega,omega-1-dialkoxy)- and N-(omega,omega-1-dialkenoxy)-alk-1-yl-N,N,N-trisubstituted ammonium surfactants, their preparation and pharmaceutical formulations containing them
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
WO1988005077A1 (en) 1986-12-29 1988-07-14 Battelle Memorial Institute A method of modifiying the metabolism of eukaryotic cells upon incorporation therein of foreign sequences of nucleic acids by means of an internalizing vector
JPS63285738A (en) 1987-05-19 1988-11-22 Canon Inc Magneto-optical recording medium
JPS6447381A (en) 1987-08-19 1989-02-21 Vitamin Kenkyusho Kk Preparation of liposome containing sealed gene
WO1990001543A1 (en) 1988-07-29 1990-02-22 Intracel Corporation Method for the genetic expression of heterologous proteins by cells transfected in vivo
JPH03102682A (en) 1989-09-14 1991-04-30 Sony Corp Disk driving device
WO1991007487A1 (en) 1989-11-16 1991-05-30 Duke University Particle-mediated transformation of animal tissue cells
WO1991016024A1 (en) 1990-04-19 1991-10-31 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
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
WO1997037966A1 (en) 1996-04-09 1997-10-16 Vical Incorporated Quaternary cytofectins
JP3102682B2 (en) 1997-02-20 2000-10-23 大淀小松株式会社 Station platform snow removal equipment

Non-Patent Citations (178)

* Cited by examiner, † Cited by third party
Title
Adrian, et al. Mol. Cell. Biol. 4 (9): 1712-1717 (1984).
Agadjanyan, M.G. et al., "DNA Inoculation with an HTLV-I Envelope DNA Construct Elicits Immune Responses in Rabbits," Vaccines 94:47-53 (1994).
Alpaugh, V., "Uncovering Naked DNA," Merck World (1993).
Anderson, E. D. et al., "Genetic Immunization of Rainbow Trout (Oncorhynchus mykiss) Against Infectious Hematopoietic Necrosis Virus," Molecular Marine Biology and Biotechnology 5:114-122 (1996).
Anker, P. et al. "Nude Mice Injected with DNA Excreted by Antigen-Stimulated Human T Lymphocytes Synthesize Specific Human Antibodies," Expl. Cell. Biol. 53:133-136 (1984).
Anker, P. et al. "Spontaneous Release of DNA by Human Blood Lymphocytes as Shown in an in Vitro System," Cancer Res. 35:2375-2382 (1975).
Anker, P. et al. "The Role of Extracellular DNA in the Transfer of Information from T to B Human Lymphocytes in the Course of an Immune Response," J. Immunogen. 7:475-481 (1980).
Anker, P. et al. "Transfert d'information de lymphocytes T à B au cours d'une réponse immune: rô ;le de l'AND extracellulaire," Schweiz med. Wschr. 110:1444-1446 (1980).
Anker, P. et al., "Anticorps porteurs d'allotypes humains synthét{overscore (i)}sés par des souris nues après injection de DNA relâ ;chè par des lymphocytes T humains," Schweiz. Med. Wschr. 112:1438-1439 (1982).
Ascadi, et al. The New Biologist 3(1): 71-81 (1991).
Ausubel, Current Protocols in Nol. Biol., John Wiley & Sons, New York (1988) §1.5.2 §§ 9.1.1-9.1.4.
Bains, W. Biotechnology from A to Z. 2nd ed. Oxford University Press, New York, NY pp. 17-19 (1998).
Beardsley, et al. Scientific American 261(5): 28-30 (1989).
Beardsley, T., "Better Than a Cure," Scientific American, 88-95 (1995).
Been, et al. Cell 47: 206-216 (1986).
Benvenisty, et al. Proc. Natl. Acad. Sci. USA 83: 9551-9555 (1986).
Berge, et al. J. Pharm. Sciences 66: 1-19 (1977).
Bhoopalam, et al. Clin. Exp. Immunol. 23: 139-148 (1976).
Bouchard, et al. Virology 135: 53-64 (1984).
Boynton, et al. Science 240: 1534-1538 (1988).
Brock, et al. Cell 34: 207-214 (1983).
Brown, et al. J. of Virology 62(12): 4828-4831 (1988).
Brown, et al. Science 232: 34-47 (1986).
Burmeister, et al. Cytogen. Cell. Genet. 46(1-4): 589 (1988).
Buttrick, P. M. et al., "Behavior of Genes Directly Injected Into the Rat Hear in Vivo," Circulation Research 70:193-198 (1992).
Chan, H.W. et al. "Molecular Cloning of Polyoma Virus DNA in Escherichia coli: Lambda Phage Vector System," Science 203:887-892 (1979).
Chelly, et al. Nature 333: 858-860 (1988).
Chen, et al. Mol. and Cell. Biol. 7: 2745-2752 (1987).
Clements-Mann, M.L. et al. "Safety and Immunogenicity of Influenza Hemagglutinin DNA Vaccine Alone or With Aluminum Adjuvant in Adult Volunteers," American Soc. Virol. Annual Meeting (1997).
Cohen, J., "Naked DNA Points Way to Vaccines," Science 259:1691-1692 (1993).
Coney, L. et al., "Facilitated DNA Inoculation Induces Anti-HIV-1 Immunity in Vivo," Vaccine 12:1545-1550 (1994).
Conry, R. M. et al., "Characterization of a Messenger RNA Polynucleotide Vaccine Vector," Cancer Research 55:1397-1400 (1995).
Cox, G. J. M. et al., "Bovine Herpesvirus 1: Immune Responses in Mice and Cattle Injected with Plasmid DNA," Journal of Virology 67:5664-5667 (1993).
Dalemans, W. et al., "Protection against Homologous Influenza Challenge by Genetic Immunization with SFV-RNA Encoding Flu-HA," Annals of the New York Academy of Sciences 772:255-256 (1995).
Daniell, et al. Proc. Natl. Acad. Sci. USA 87: 88-92 (1990).
Davey, J. et al., "Localization of Influenza Virus M, NP, and NS1 Proteins in Microinjected Cells," J. gen. Virol. 66:2319-2334 (1985).
Davis, H. L. et al., "Direct gene transfer in skeletal muscle: plasmid DNA-based immunization against the hepatitis B virus surface antigen," Vaccine 12:1503-1509 (1994).
Davis, H. L. et al., "Direct gene transfer into mouse diaphragm," FEBS Letters 333:146-150 (1993).
de Wet, et al. Mol. Cell Biol. 7: 725-737 (1987).
Dean, et al. J. Cell. Biol. 106: 2159-2170 (1988).
Deck, R.R. et al. "Characterization of humoral immune responses induced by an influenza hemagglutinin DNA vaccine," Vaccine 15:71-78 (Jan. 1997).
DeNoto, F.M. et al. "Human growth hormone DNA sequence and mRNA structure: possible alternative splicing," Nucl. Acids Res. 9:3719-3730 (1981).
Dialog File 351, Accession No. 67-04439H/196800, Derwent WPI English language abstract for French Patent No. 7 781 M, Document No. AN1.
Dixon, B. "The Third Vaccine Revolution," Biotechnol. 13:420 (May 1995).
Dolph, et al. J. of Virol. 62(6): 2059-2066 (1988).
Donnelly, J. J. et al., "Polynucleotide Vaccination against Influenza," Vaccines 94:55-59 (1994).
Donnelly, J.J. et al. "Preclinical efficacy of a prototype DNA vaccine: Enhanced protection against antigenic drift in influenza virus," Nat. Med. 1:583-597 (Jun. 1995).
Drummond, et al. Nucl. Acids Res. 13: 7375 (1985).
Dubensky, et al. Proc. Natl. Acad. Sci. USA 81: 5849-5852 (1984).
Dunn, et al. Gene 68: 259-266 (1988).
Eibl, et al. Biophys. Chem. 10: 261-271 (1979).
Elroy-Stein, et al. Proc. Natl. Acad. Sci. USA 86: 6126-6130.
Engerlhard, V. H., "How Cells Process Antigens," Scientific American, 54-61 (1994).
Fainboim, L. et al., "Transfer of experimental allergic orchitis with immune RNA. Studies in vivo," Clin. exp. Immunol. 34:92-99 (1978).
Feitelson, M.A. et al. "A Chronic Carrierlike State Is Established in Nude Mice Injected with Clones Hepatitis B Virus DNA," J. Virol. 62:1408-1415 (Apr. 1988).
Felgner, et al. Proc. Natl. Acad. Sci. USA 84: 6730-6734, 7413-7417 (1987).
Fields, B.N. et al., Eds., Fields Virology 2nd ed., Raven Press, New York, pp. 1596-1614 (1989).
Friedman, et al. Science 244: 1275-1281 (1989).
Fung, et al. Proc. Natl. Acad. Sci. USA 80: 353-357 (1983).
Fynan, E. F. et al., "DNA vaccines: Protective immunizations by parenteral, mucosal, and gene-gun inoculations," Proc. Natl. Acad. Sci. USA 90:11478-11482 (1993).
Gal, D. et al., "Direct Myocardial Transfection In Two Animal Models," Laboratory Investigation 68:18-25 (1993).
Gélinas, C. et al. "Tumorigenic activity of cloned polyoma virus DNA in newborn rats," Experientia 37:1074-1075 (1981).
Gillies, et al. Biotechol. 7: 799-804 (1989).
Goldman, C.K. et al. "In vitro and in vivo gene delivery mediated by a synthetic polycationic amino polymer," Nat. Biotech. 15:462-466 (May 1997).
Goodfellow, et al. Nature 341(6238): 102-103 (1989).
Gorman, C.M. et al. "Recombinant Genomes Which Express Chloramphenicol Acetyltransferase in Mammalian Cells," Mol. Cell. Biol. 2:1044-1051 (1982).
Gorman, C.M. et al. "The Rous sarcoma virus long terminal repeat is a strong promoter when introduced into a variety of eukaryotic cells by DNA-mediated transfection," Proc. Natl. Acad. Sci. USA 79:6777-6781 (1982).
Graves, et al. Cell 48: 615-626 (1987).
Hansen, E. et al., "Strong expression of foreign genes following direct injection into fish muscle," FEBS Letters 290:7376 (1991).
Hardman, J.G. et al. Eds., Goodman & Gillman's The Pharmacological Basis of Therapeutics, 9th ed., McGraw-Hill, p.8 (1996).
Harland, et al. Development 102: 837-852 (1988).
Haynes, J. R. et al., "Gene-gun-mediated DNA Immunization Elicits Humoral, Cytotoxic, and Protective Immune Responses," Vaccines 94:65-70 (1994).
Hentze, et al. Proc. Natl. Acad. Sci. USA 84: 6730-6734 (1987).
Hoffman, et al. Neuron 2: 1019-1029 (1989).
Hoffman, et al. Science 254: 1455-1456 (1991).
Hoffman, S. L. et al., "Protection against malaria by immunization with a Plasmodium yoelii circumsporozoite protein nucleic acid vaccine," Vaccine 12:1529-1533 (1994).
Holt, et al. Neuron 4: 203-214 (1990).
Huang, et al. J. of Virol. 50: 417-424 (1984).
Israel, M., et al. (1979) Biological activity of polyoma viral DNA in mice and hamsters. Journal of Virology 29(3):990-996.
Israel, M.A. et al. "Molecular Cloning of Polyoma Virus DNA in Escherichia coli: Plasmid Vector System," Science 203:883-887 (1979).
Jacherts, D. and Drescher, J. "Antibody Response in Rhesus Monkeys and Guinea Pigs to Inoculation with RNA Derived from Antigenetically Stimulated Cell-Free Systems," J. Immunol. 104:746-752 (1970).
Jachertz, D. and Egger, M. "Treatment of P815 Mastocytoma in DBA/2 Mice with RNA," J. Immunogen. 1:355-362 (1974).
Jachertz, D. et al. "Information carried by the DNA released by antigen-stimulated lymphocytes," Immunol. 37:753-763 (1979).
Jiao, S. et al., "Direct Gene Transfer into Nonhuman Primate Myofibers In Vivo," Human Gene Therapy 3:21-33 (1992).
Jiao, S. et al., "Particle Bombardment-Mediated Gene Transfer and Expression in Rat Brain Tissues," Bio/Technology 11:497-502 (1993).
Johanning, F. W. et al., "A sindbis virus mRNA polynucleotide vector achieves prolonged and high level heterologous gene expression in vivo," Nucleic Acids Research 23:1495-1501 (1995).
Johnston, et al. Science 240: 1538-1541 (1988).
Kabnick, et al. Mol. and Cell. Biol. 8: 3244-3250 (1988).
Kaneda, et al. Science 243: 375-378 (1989).
Kitsis, R. N. et al., "Hormonal modulation of a gene injected into rat heart in vivo," Proc. Natl. Acad. Sci. USA 88:4138-4142 (1991).
Klemenz, et al. EMBO Journal 4(8): 2053-2060 (1985).
Koenig, et al. Cell 53(2): 219-226 (1988).
Kozak, et al. Nucl. Acids Res. 15(20): 8125 (1987).
Kreig, A.M. et al. "Sequence motifs in adenoviral DNA block immune activation by stimulatory CpG motifs," Proc. Natl. Acad. Sci. USA 95:12631-12636 (Oct. 1998).
Kreig, et al. Nucl. Acids Res. 12(18): 7057-7070 (1984).
Kruczek, I. and Doerfler, W. "Expression of the chloramphenicol acetyltransferase gene in mammalian cells under the control of adenovirus type 12 promoters: Effect of promoter methylation on gene expression," Proc. Natl. Acad. Sci. USA 80:7586-7590 (1983).
Kuklin, W. et al., "Induction of Mucosal Immunity against Herpes Simplex Virus by Plasmid DNA Immunization," Journal of Virology 71:3138-3145 (1997).
Laub, O. et al. "Synthesis of Hepatitis B Surface Antigen in Mammalian Cells: Expression of the Entire Gene and the Coding Region," J. Virol. 48:271-280 (1983).
Lewin, B., Genes IV, Oxford University Press, pp. 178 (1990).
Lin, et al. Circulation 82: 2217-2221 (1990).
Loyter, et al. Exp. Cell Res. 139: 223-234 (1982).
Magee, et al. Cancer Res. 38: 1173-1176 (1978).
Malone, et al. Proc. Natl. Acad. Sci. USA 86: 6077-6081 (1989).
Mandi, C.W. et al. "In vitro-synthesized infectious RNA as an attenuated live vaccine in a flavivirus model," Natl. Med. 4:1438-1440 (Dec. 1998).
Mannino, et al. Biotechniques 6: 682-690 (1988).
McCrae, et al. Eur. J. of Biochem. 116: 467-470 (1981).
Mosier, et al. Nature 355: 256-259 (1988).
Muesing, et al. Cell 48: 691 (1987).
Mullner, et al. Cell 53: 815-825 (1988).
Nakatani, et al. Biotechnology 7: 805-810 (1989).
Namikawa, et al. Science 242: 1684-1686 (1988).
New England Biolabs 1986/87 Catalog, 32 Tozer Rd., Beverly, MA 01915-0990 USA, p. 45.
Nicolau, et al. Methods in Enzymology 149: 157-176 (1987).
Nicolau, et al. Proc. Natl. Acad. Sci. USA 80: 1068-1072 (1983).
Norton, et al. Mol. Cell Biol. 5: 281-290 (1985).
Ostro, et al. Nature 274: 921-923 (1979).
Oudrhiri, N. et al., "Gene transfer by guanidine-cholestrol cationic lipids into airway epithelial cells in vitro and in vivo," Proc. Natl. Acad. Sci. USA 94:1651-1656 (Mar. 1997).
Parks, et al. J. Virol. 60: 376-384 (1986).
Partial English translation of Japanese Patent Application No. 63-285738.
Pelletier, et al. Nature 334: 320-325 (1988).
Poyet, et al. Mol. endocrinoloay 3(12): 1961-1968 (1989).
Price, et al. Proc. Natl. Acad. Sci. USA 84: 156-160 (1987).
Qui, P. et al., "Gene gun delivery of mRNA in situ results in efficient transgene expression and genetic immunization," Gene Therapy 3:262-268 (1996).
Rao, et al. Mol. and Cell. Biol. 8: 284 (1988).
Raz, E. et al., "Intradermal gene immunization: The possible role of DNA uptake in the induction of cellular immunity to viruses," Proc. Natl. Acad. Sci. USA 91:9519-9523 (1994).
Raz, E. et al., "Systemic immunological effects of cytokine genes injected into skeletal muscle," Proc. Natl. Acad. Sci. USA 90:4523-4527 (1993).
Rhodes, G. H. et al., "Characterization of Humoral Immunity after DNA Injection," Dev Biol Stand 82:229-236 (1994).
Robinson, et al. Science 22: 417-419 (1984).
Robinson, H. L. et al., "Protection against a lethal influenza virus challenge by immunization with a haemagglutinin-expressing plasmid DNA," Vaccine 11 (1993).
Robinson, H. L. et al., "Use of Direct DNA Inoculations to Elicit Protective Immune Responses," Abstracts of papers, p. 92 (Cold Spring Harbor Laboratory 1994).
Rommens, et al. Science 245 (4922): 1059-1065 (1989).
Rosenberg, S.A. et al. "Biological Activity of Recombinant Human Interleukin-2 Produced in Escherichia coli," Science 223:1412-1415 (1984).
Ross, et al. Mol. Biol. Med. 5: 1-14 (1988).
Sato, Y. et al. "Immunostimulatory DNA Sequences Necessary for Effective Intradermal Gene Immunization," Science 273:352-354 (Jul. 1996).
Satz, M.L. et al. "Mechanism of immune transfer by RNA extracts," Mol. Cell. Biochem. 33:105-113 (1980).
Sedegah, M. et al., "Protection against malaria by immunization with plasmid DNA encoding circumsporozoite protein," Proc. Natl. Acad. Sci. USA 91:9866-9870 (1994).
Seeger, C., et al. (1984) The cloned genome of ground squirrel hepatitis virus is infectious in the animal. Proc. Natl. Acad. Sci. 81:5849-5852.
Selden, et al. Mol. Cell. Biol. 6: 3173-3179 (1986).
Selden, et al. Proc. Natl. Acad. Sci. USA 85: 8241-8245 (1988).
Sell, S. and Mendelsohn, J. "Transfer of Specific Immunity With RNA," Arch. Path. Lab Med. 102:217-222 (1978).
Shaw, et al. Cell 46: 659-667 (1986).
Sizemore, D. R. et al., "Attenuated Shigella as a DNA Delivery Vehicle for DNA-Mediated Immunization," Science 270:299-302 (1995).
Soares, M.B. et al. "RNA-Mediated Gene Duplication: the Rat Preproinsulin I Gene Is a Functional Retroposon," Mol. Cell. Biol. 5:2090-2103 (1985).
Sol, C.J.A. and Van der Noordaa, J. "Oncogenecity of SV40 DNA in the Syrian Hamster," J. Gen. Virol. 37:635-638 (1977).
Spooner, R. A. et al., "DNA vaccination for cancer treatment," Gene Therapy 2:173-180 (1995).
Stamatatos, et al. Biochemistry 27: 3917-3925 (1988).
Straubinger, et al. Methods in enzymology 101: 512-527 (1983).
Taniguchi, T. et al. "Structure and expression of a cloned cDNA for human interleukin-2," Nature 302:305-310 (1983).
Taylor, J. et al. "Recombinant fowlpox virus inducing protective immunity in non-avian species," Vaccine 6:497-503 (Dec. 1988).
Travis, J., "Tracing the Immune System's Evolutionary History," Science 261:164-165 (1993).
Tripathy, S. K. et al., "Long-term expression erythropoietin in the systemic circulation of mice after intramuscular injection of a plasmid DNA vector," Proc. Natl. Acad. Sci. USA 93:10876-10880 (1996).
Ulmer, J. B. et al., "DNA vaccines," Current Opinion in Immunology 8:531-536 (1996).
Ulmer, J. B. et al., "Heterologous Protection Against Influenza by Injection of DNA encoding a Viral Protein," Science 259:1745-1749 (1993).
Ulmer, J. B. et al., "Protective immunity by intramuscular injection of low doses of influenza virus DNA vaccines," Vaccine 12:1541-1544 (1994).
Valerio, et al. Gene 31: 147-153 (1984).
von Harsdorf, R. et al., "Gene Injection Into Canine Myocardium as a Useful Model for Studying Gene Expression in the Heart of Large Mammals," Circulation Research 72:688-695 (1993).
Wang, B. et al., "Gene inoculation generates immune responses against human immunodeficiency virus type 1," Proc. Natl. Acad. Sci. USA 90:4156-4160 (1993).
Wang, C. et al., "Direct Gene Delivery of Human Tissue Kallikrein Reduces Blood Pressure in Spontaneously Hypertensive Rats," J. Clin. Invest 95:1710-1716 (1995).
Wang, C-Y and Huang, L. "pH-sensitive immunoliposomes mediate target-cell-specific delivery and controlled expression of a foreign gene in mouse," Proc. Natl. Acad. Sci. USA 84:7851-7855 (1987).
Ward, et al. Nature 341: 544-546 (1989).
Watkins, et al. Nature 6176: 863-866 (1988).
Webster, R. G. et al., "Protection of ferrets against influenza challenge with a DNA vaccine to the haemagglutinin," Vaccine 12:1495-1498 (1994).
Weeratna, R. et al. "Reduction of Antigen Expression from DNA Vaccines by Coadministered Oligodeoxynucleotides," Antisense & Nucl. Acid Drug Develop. 8:351-356 (Aug. 1998).
Wickner, et al. Science 230: 400-407 (1985).
Will, H. et al. "Infectious hepatitis B virus from cloned DNA of known nucleotide sequence," Proc. Natl. Acad. Sci. USA 82:891-895 (1985).
Will, H., et al. (1982) Cloned HBV DNA causes hepatitis in chimpanzees. Nature 299:740-742.
Wolff, et al. Nature, Jan. (1990).
Wolff, J. A. et al., "Direct Gene Transfer into Mouse Muscle in Vivo," Science 247:1465-1468 (1990).
Wu, et al. J. Biol. Chem. 263(29): 14621-14624 (1986).
Wu, et al. J. of Biol. Chem. 264: 16985-16987 (1989).
Wynshaw-Boris, A. et al. "Identification of a cAMP Regulatory Region in the Gene for Rate Cytosolic Phosphoenolpyruvate Carboxykinase (GTP)" J. Biol. Chem. 259:12161-12169 (1984).
Xiang, Z. et al., "Manipulation of the Immune Response to a Plasmid-Encoded Viral Antigen by Coinoculation with Plasmids Expressing Cytokines," Immunity 2:129-135 (1995).
Xiang, Z. et al., "Vaccination with a Plasmid Vector Carrying the Rabies Virus Glycoprotein Gene Induces Protective Immunity against Rabies Virus," Virology 199:132-140 (1994).
Xu, D. et al., "Genetic vaccination against leishmaniasis," Vaccine 12:1534-1536 (1994).
Yakubov, et al. Proc. Natl. Acad. Sci. USA 86: 6454-6458 (1989).
Yang, J. P. et al., "Direct gene transfer to mouse melanoma by intratumor injection of free DNA," Gene Therapy 3:542-548 (1996).
Yankauckas, M. A. et al., "Long-Term Anti-Nucleoprotein Cellular and Humoral Immunity Is Induced by Intramuscular Injection of Plasmid DNA Containing NP Gene," DNA and Cell Biology 12:771-776 (1993).
Zhou, X. et al., "Self-replicating Semliki Forest virus RNA as recombinant vaccine," Vaccine 12:1510-1513 (1994).

Cited By (438)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030032615A1 (en) * 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US7250404B2 (en) 1989-03-21 2007-07-31 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US20030186913A1 (en) * 1990-03-21 2003-10-02 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate
US6936464B1 (en) 1992-10-02 2005-08-30 Cancer Research Technology Limited Immune responses to fusion proteins
US7001759B1 (en) * 1993-01-26 2006-02-21 The Trustees Of The University Of Pennsylvania Compositions and methods for delivery of genetic material
US20060222629A1 (en) * 1993-01-26 2006-10-05 Weiner David B Compositions and methods for delivery of genetic material
US8304234B2 (en) * 1993-01-26 2012-11-06 The Trustees Of The Universtiy Of Pennsylvania Compositions and methods for delivery of genetic material
US6890554B2 (en) * 1993-06-01 2005-05-10 Invitrogen Corporation Genetic immunization with cationic lipids
US20050124039A1 (en) * 1993-06-01 2005-06-09 Invitrogen Corporation Genetic Immunization with Cationic Lipids
US7166298B2 (en) 1993-06-01 2007-01-23 Invitrogen Corporation Genetic immunization with cationic lipids
US6348449B1 (en) * 1993-09-21 2002-02-19 The Trustees Of The University Of Pennsylvania Methods of inducing mucosal immunity
US6716882B2 (en) 1993-12-20 2004-04-06 Invitrogen Corporation Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US7501542B2 (en) 1993-12-20 2009-03-10 Invitrogen Corporation Highly-packed polycationic ammonium, sulfonium and phosphonium lipids
US20090317908A1 (en) * 1993-12-20 2009-12-24 Life Technologies Corporation Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US7687070B2 (en) 1994-02-11 2010-03-30 Life Technologies Corporation Reagents for intracellular delivery of macromolecules
US6989434B1 (en) 1994-02-11 2006-01-24 Invitrogen Corporation Reagents for intracellular delivery of macromolecules
US20050260757A1 (en) * 1994-02-11 2005-11-24 Invitrogen Coroporation Novel reagents for intracellular delivery of macromolecules
US20040152656A1 (en) * 1994-07-15 2004-08-05 University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US20050065107A1 (en) * 1994-11-28 2005-03-24 Hobart Peter M. Plasmids suitable for IL-2 expression
US20030203863A1 (en) * 1994-11-28 2003-10-30 Vical Incorporated Plasmids suitable for IL-2 expression
US20080139494A1 (en) * 1994-12-16 2008-06-12 Weiner David B Methods of inducing mucosal immunity
US7220728B2 (en) 1994-12-16 2007-05-22 The Trustees Of The University Of Pennsylvania Methods of inducing mucosal immunity
US8536145B2 (en) 1994-12-16 2013-09-17 The Trustees Of The University Of Pennsylvania Methods of inducing mucosal immunity
US20080153166A1 (en) * 1995-01-23 2008-06-26 Leaf Huang Stable lipid-comprising drug delivery complexes and methods for their production
US7655468B2 (en) 1995-01-23 2010-02-02 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US8771728B2 (en) 1995-01-23 2014-07-08 University of Pittsburgh—of the Commonwealth System of Higher Education Stable lipid-comprising drug delivery complexes and methods for their production
US7993672B2 (en) 1995-01-23 2011-08-09 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US20100184953A1 (en) * 1995-01-23 2010-07-22 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US20030144230A1 (en) * 1995-06-07 2003-07-31 Pamela Hawley-Nelson Peptide-enhanced transfections
US8058068B2 (en) 1995-06-07 2011-11-15 Life Technologies Corporation Peptide-enhanced transfections
US20060008448A1 (en) * 1996-06-11 2006-01-12 Minzhen Xu Inhibition of li expression in mammalian cells
US7001890B1 (en) 1997-01-23 2006-02-21 Coley Pharmaceutical Gmbh Pharmaceutical compositions comprising a polynucleotide and optionally an antigen especially for vaccination
US20050054601A1 (en) * 1997-01-23 2005-03-10 Coley Pharmaceutical Gmbh Pharmaceutical composition comprising a polynucleotide and optionally an antigen especially for vaccination
US20090035252A1 (en) * 1997-07-10 2009-02-05 Kundig Thomas M Method of inducing a CTL response
US7364729B2 (en) 1997-07-10 2008-04-29 Mannkind Corporation Method of inducing a CTL response
US8372393B2 (en) 1997-07-10 2013-02-12 Mannkind Corporation Method of inducing a CTL response
US20060153858A1 (en) * 1997-07-10 2006-07-13 Kundig Thomas M Method of inducing a CTL response
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
US20030083272A1 (en) * 1997-09-19 2003-05-01 Lahive & Cockfield, Llp Sense mrna therapy
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
US7666149B2 (en) 1997-12-04 2010-02-23 Peliken Technologies, Inc. Cassette of lancet cartridges for sampling blood
US20060229246A1 (en) * 1998-03-16 2006-10-12 Pamela Hawley-Nelson Peptide-enhanced transfections
US20030069173A1 (en) * 1998-03-16 2003-04-10 Life Technologies, Inc. Peptide-enhanced transfections
US8439872B2 (en) 1998-03-30 2013-05-14 Sanofi-Aventis Deutschland Gmbh Apparatus and method for penetration with shaft having a sensor for sensing penetration depth
US7780631B2 (en) 1998-03-30 2010-08-24 Pelikan Technologies, Inc. Apparatus and method for penetration with shaft having a sensor for sensing penetration depth
US20040266719A1 (en) * 1998-05-22 2004-12-30 Mccluskie Michael J. Methods and products for inducing mucosal immunity
US8574599B1 (en) 1998-05-22 2013-11-05 Ottawa Hospital Research Institute Methods and products for inducing mucosal immunity
US20090143583A1 (en) * 1998-11-12 2009-06-04 Life Technologies Corporation Transfection reagents
US20050164971A1 (en) * 1998-11-12 2005-07-28 Yongliang Chu New transfection reagents
US8785200B2 (en) 1998-11-12 2014-07-22 Life Technologies Corporation Transfection reagents
US7915450B2 (en) 1998-11-12 2011-03-29 Life Technologies Corporation Transfection reagents
US9358300B2 (en) 1998-11-12 2016-06-07 Life Technologies Corporation Transfection reagents
US20100159593A1 (en) * 1998-11-12 2010-06-24 Life Technologies Corporation Transfection reagents
US8158827B2 (en) 1998-11-12 2012-04-17 Life Technologies Corporation Transfection reagents
US20070202598A1 (en) * 1998-11-12 2007-08-30 Invitrogen Corporation New transfection reagents
US7943375B2 (en) 1998-12-31 2011-05-17 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20110212164A1 (en) * 1998-12-31 2011-09-01 Novartis Vaccines & Diagnostics, Inc. Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
US7935805B1 (en) 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
US20090004733A1 (en) * 1998-12-31 2009-01-01 Novartis Vaccines & Diagnostics, Inc. Polynucleotides encoding antigenic hiv type b polypeptides, polypeptides, and uses thereof
US20060057115A1 (en) * 1998-12-31 2006-03-16 Zur Megede Jan Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20090047339A1 (en) * 1998-12-31 2009-02-19 Barnett Susan W Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US8541230B2 (en) 1998-12-31 2013-09-24 Novartis Vaccines And Diagnostics, Inc Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20080261271A1 (en) * 1998-12-31 2008-10-23 Novartis Vaccines & Diagnostics, Inc. Expression of hiv polypeptides and production of virus-like particles
US7718401B2 (en) 1998-12-31 2010-05-18 Novartis Vaccines And Diagnostics, Inc. Expression of HIV polypeptides and production of virus-like particles
US6602705B1 (en) 1998-12-31 2003-08-05 Chiron Corporation Expression of HIV polypeptides and production of virus-like particles
US20100316698A1 (en) * 1998-12-31 2010-12-16 Novartis Vaccines And Diagnostics, Inc. Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
US8263394B2 (en) 1998-12-31 2012-09-11 Novartis Vaccines & Diagnostics Inc. Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides, and uses thereof
US7662916B2 (en) 1998-12-31 2010-02-16 Novartis Vaccines & Diagnostics, Inc Modified HIV Env polypeptides
US8168418B2 (en) 1998-12-31 2012-05-01 Susan W Barnett Expression of HIV polypeptides and production of virus-like particles
US6689879B2 (en) 1998-12-31 2004-02-10 Chiron Corporation Modified HIV Env polypeptides
US7348177B2 (en) 1998-12-31 2008-03-25 Novartis Vaccines And Diagnostics, Inc. Expression of HIV polypeptides and production of virus-like particles
US20100092502A1 (en) * 1998-12-31 2010-04-15 Novartis Vaccines And Diagnostics, Inc. Modified hiv env polypeptides
US7776344B2 (en) 1999-09-27 2010-08-17 University Of Iowa Research Foundation Methods related to immunostimulatory nucleic acid-induced interferon
US20060286070A1 (en) * 1999-09-27 2006-12-21 Coley Pharmaceutical Gmbh Methods related to immunostimulatory nucleic acid-induced interferon
US6949520B1 (en) 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
US7795017B2 (en) 2000-03-02 2010-09-14 Emory University DNA expression vectors and methods of use
US20070048861A1 (en) * 2000-03-02 2007-03-01 Robinson Harriet L Compositions and methods for generating an immune response
US20030175292A1 (en) * 2000-03-02 2003-09-18 Robinson Harriet L. Compositions and methods for generating an immune response
US8623379B2 (en) 2000-03-02 2014-01-07 Emory University Compositions and methods for generating an immune response
US20020019358A1 (en) * 2000-04-21 2002-02-14 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US6875748B2 (en) 2000-04-21 2005-04-05 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US7534772B2 (en) 2000-06-22 2009-05-19 University Of Iowa Research Foundation Methods for enhancing antibody-induced cell lysis and treating cancer
US20030223961A1 (en) * 2000-07-05 2003-12-04 Megede Jan Zur Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
WO2002008448A3 (en) * 2000-07-25 2002-07-25 Univ Leland Stanford Junior Non-viral linear dna vectors and methods for using the same
US20050079615A1 (en) * 2000-07-25 2005-04-14 Kay Mark A. Non-viral linear DNA vectors and methods for using the same
WO2002008448A2 (en) * 2000-07-25 2002-01-31 The Board Of Trustees Of The Leland Stanford Junior University Non-viral linear dna vectors and methods for using the same
US6849455B1 (en) 2000-08-22 2005-02-01 New York University Enhanced recovery of transformed cells
US8641644B2 (en) 2000-11-21 2014-02-04 Sanofi-Aventis Deutschland Gmbh Blood testing apparatus having a rotatable cartridge with multiple lancing elements and testing means
US20040146528A1 (en) * 2001-03-08 2004-07-29 Bernard Moss MVA expressing modified HIV envelope, gag, and pol genes
US20090074726A1 (en) * 2001-03-08 2009-03-19 Bernard Moss Mva expressing modified hiv envelope, gag, and pol genes
US8916172B2 (en) 2001-03-08 2014-12-23 Emory University MVA expressing modified HIV envelope, gag, and pol genes
US7867982B2 (en) 2001-03-08 2011-01-11 Emory University MVA expressing modified HIV envelope, gag, and pol genes
US20110104199A1 (en) * 2001-03-08 2011-05-05 Bernard Moss MVA Expressing Modified HIV Envelope, GAG, and POL Genes
US20030091544A1 (en) * 2001-03-13 2003-05-15 Vical Incorporated Interferon-Beta polynucleotide therapy for autoimmune and inflammatory diseases
US20040142468A1 (en) * 2001-03-16 2004-07-22 Pardoll Drew M Modulation of systemic immune responses by transplantation of hematopoietic stem cells transduced with genes encoding antigens and antigen presenting cell regulatory molecules
US7767202B2 (en) 2001-03-16 2010-08-03 The Johns Hopkins University Modulation of systemic immune responses by transplantation of hematopoietic stem cells transduced with genes encoding antigens and antigen presenting cell regulatory molecules
US20110077287A1 (en) * 2001-06-05 2011-03-31 Curevac Gmbh Pharmaceutical composition containing a stabilised mrna optimised for translation in its coding regions
US20050032730A1 (en) * 2001-06-05 2005-02-10 Florian Von Der Mulbe Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
EP1832603A2 (en) * 2001-06-05 2007-09-12 CureVac GmbH Stabilised mRNA with increased G/C-content and optimised codon usage for gene therapy
WO2002098443A2 (en) * 2001-06-05 2002-12-12 Curevac Gmbh Stabilised mrna with an increased g/c content and optimised codon for use in gene therapy
EP1857122A3 (en) * 2001-06-05 2008-11-19 CureVac GmbH Pharmaceutical compound, containing a stabilised mRNA which is optimised for translation in its coded areas
EP1832603A3 (en) * 2001-06-05 2007-10-31 CureVac GmbH Stabilised mRNA with increased G/C-content and optimised codon usage for gene therapy
EP2305699A1 (en) 2001-06-05 2011-04-06 CureVac GmbH Stabilised mRNA with increased G/C content which is optimised for translation in its coded areas
US10188748B2 (en) 2001-06-05 2019-01-29 Curevac Ag Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
EP1800697A2 (en) * 2001-06-05 2007-06-27 CureVac GmbH Pharmaceutical compound, containing a stabilised mRNA which is optimised for translation in its coded areas
EP1800697A3 (en) * 2001-06-05 2007-10-31 CureVac GmbH Pharmaceutical compound, containing a stabilised mRNA which is optimised for translation in its coded areas
EP1903054A3 (en) * 2001-06-05 2008-07-23 CureVac GmbH Pharmaceutical compound containing a stabilised mRNA which is optimised for translation in its coded areas
US11369691B2 (en) 2001-06-05 2022-06-28 Curevac Ag Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
WO2002098443A3 (en) * 2001-06-05 2003-11-13 Curevac Gmbh Stabilised mrna with an increased g/c content and optimised codon for use in gene therapy
EP1857122A2 (en) * 2001-06-05 2007-11-21 CureVac GmbH Pharmaceutical compound, containing a stabilised mRNA which is optimised for translation in its coded areas
EP1604688A1 (en) 2001-06-05 2005-12-14 CureVac GmbH Stabilised mRNA with increased G/C-content and optimised codon usage for gene therapy
EP1903054A2 (en) 2001-06-05 2008-03-26 CureVac GmbH Pharmaceutical compound containing a stabilised mRNA which is optimised for translation in its coded areas
US11135312B2 (en) 2001-06-05 2021-10-05 Curevac Ag Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
US20100239608A1 (en) * 2001-06-05 2010-09-23 Curevac Gmbh PHARMACEUTICAL COMPOSITION CONTAINING A STABILISED mRNA OPTIMISED FOR TRANSLATION IN ITS CODING REGIONS
US10568972B2 (en) 2001-06-05 2020-02-25 Curevac Ag Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
EP2842964A1 (en) 2001-06-05 2015-03-04 Curevac GmbH Virtual method of determining a modified mRNA sequence
US8382683B2 (en) 2001-06-12 2013-02-26 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8206319B2 (en) 2001-06-12 2012-06-26 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8206317B2 (en) 2001-06-12 2012-06-26 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US7682318B2 (en) 2001-06-12 2010-03-23 Pelikan Technologies, Inc. Blood sampling apparatus and method
US7699791B2 (en) 2001-06-12 2010-04-20 Pelikan Technologies, Inc. Method and apparatus for improving success rate of blood yield from a fingerstick
US9694144B2 (en) 2001-06-12 2017-07-04 Sanofi-Aventis Deutschland Gmbh Sampling module device and method
US9427532B2 (en) 2001-06-12 2016-08-30 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9802007B2 (en) 2001-06-12 2017-10-31 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US8211037B2 (en) 2001-06-12 2012-07-03 Pelikan Technologies, Inc. Tissue penetration device
US8845550B2 (en) 2001-06-12 2014-09-30 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US7981055B2 (en) 2001-06-12 2011-07-19 Pelikan Technologies, Inc. Tissue penetration device
US7749174B2 (en) 2001-06-12 2010-07-06 Pelikan Technologies, Inc. Method and apparatus for lancet launching device intergrated onto a blood-sampling cartridge
US8123700B2 (en) 2001-06-12 2012-02-28 Pelikan Technologies, Inc. Method and apparatus for lancet launching device integrated onto a blood-sampling cartridge
US8216154B2 (en) 2001-06-12 2012-07-10 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9937298B2 (en) 2001-06-12 2018-04-10 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8282577B2 (en) 2001-06-12 2012-10-09 Sanofi-Aventis Deutschland Gmbh Method and apparatus for lancet launching device integrated onto a blood-sampling cartridge
US8016774B2 (en) 2001-06-12 2011-09-13 Pelikan Technologies, Inc. Tissue penetration device
US7909775B2 (en) 2001-06-12 2011-03-22 Pelikan Technologies, Inc. Method and apparatus for lancet launching device integrated onto a blood-sampling cartridge
US8360991B2 (en) 2001-06-12 2013-01-29 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8622930B2 (en) 2001-06-12 2014-01-07 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8641643B2 (en) 2001-06-12 2014-02-04 Sanofi-Aventis Deutschland Gmbh Sampling module device and method
US8721671B2 (en) 2001-06-12 2014-05-13 Sanofi-Aventis Deutschland Gmbh Electric lancet actuator
US8679033B2 (en) 2001-06-12 2014-03-25 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US7850622B2 (en) 2001-06-12 2010-12-14 Pelikan Technologies, Inc. Tissue penetration device
US8337421B2 (en) 2001-06-12 2012-12-25 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US7988645B2 (en) 2001-06-12 2011-08-02 Pelikan Technologies, Inc. Self optimizing lancing device with adaptation means to temporal variations in cutaneous properties
US9598469B2 (en) 2001-07-05 2017-03-21 Novartis Vaccines And Diagnostics, Inc. HIV-1 south african subtype C env proteins
US7211659B2 (en) 2001-07-05 2007-05-01 Chiron Corporation Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US20080089908A1 (en) * 2001-07-05 2008-04-17 Megede Jan Z Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US8133494B2 (en) 2001-07-05 2012-03-13 Novartis Vaccine & Diagnostics Inc Expression cassettes endcoding HIV-1 south african subtype C modified ENV proteins with deletions in V1 and V2
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
US7604955B2 (en) 2001-08-13 2009-10-20 Swey-Shen Alex Chen Immunoglobulin E vaccines and methods of use thereof
US20030073142A1 (en) * 2001-08-13 2003-04-17 Chen Swey-Shen Alex Immunoglobulin E vaccines and methods of use thereof
US20080095833A1 (en) * 2001-08-31 2008-04-24 Novartis Vaccines And Diagnostics, Inc. Polynucleotides encoding antigenic hiv type b polypeptides, polypeptides, and uses thereof
US7282364B2 (en) 2001-08-31 2007-10-16 Novartis Vaccines And Diagnostics, Inc. Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20030194800A1 (en) * 2001-08-31 2003-10-16 Megede Jan Zur Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20050214256A1 (en) * 2001-08-31 2005-09-29 Chiron Corporation Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20030170614A1 (en) * 2001-08-31 2003-09-11 Megede Jan Zur Polynucleotides encoding antigenic HIV type B polypeptides, polypeptides and uses thereof
US20030049801A1 (en) * 2001-09-10 2003-03-13 Chung-Hsiun Wu 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
US6800462B2 (en) 2001-09-10 2004-10-05 Abgenomics Corporation Production of recombinant proteins in vivo and use for generating antibodies
US9560993B2 (en) 2001-11-21 2017-02-07 Sanofi-Aventis Deutschland Gmbh Blood testing apparatus having a rotatable cartridge with multiple lancing elements and testing means
US9439956B2 (en) 2001-12-19 2016-09-13 Curevac Ag Application of mRNA for use as a therapeutic against tumour diseases
US9463228B2 (en) 2001-12-19 2016-10-11 Curevac Ag Application of mRNA for use as a therapeutic against tumour diseases
US8217016B2 (en) 2001-12-19 2012-07-10 Curevac Gmbh Application of mRNA for use as a therapeutic agent for tumorous diseases
US9433669B2 (en) 2001-12-19 2016-09-06 Curevac Ag Application of mRNA for use as a therapeutic against tumor diseases
US9155788B2 (en) 2001-12-19 2015-10-13 Curevac Gmbh Application of mRNA for use as a therapeutic against tumour diseases
US9655955B2 (en) 2001-12-19 2017-05-23 Curevac Ag Application of mRNA for use as a therapeutic against tumour diseases
US9433670B2 (en) 2001-12-19 2016-09-06 Curevac Ag Application of mRNA for use as a therapeutic against tumour diseases
US20050059624A1 (en) * 2001-12-19 2005-03-17 Ingmar Hoerr Application of mRNA for use as a therapeutic against tumour diseases
US10036025B2 (en) 2002-02-01 2018-07-31 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
US8815821B2 (en) 2002-02-01 2014-08-26 Life Technologies Corporation Double-stranded oligonucleotides
US10196640B1 (en) 2002-02-01 2019-02-05 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9796978B1 (en) 2002-02-01 2017-10-24 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10626398B2 (en) 2002-02-01 2020-04-21 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US8524680B2 (en) 2002-02-01 2013-09-03 Applied Biosystems, Llc High potency siRNAS for reducing the expression of target genes
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
US20030176377A1 (en) * 2002-03-02 2003-09-18 Rong Xiang DNA vaccines encoding CEA and a CD40 ligand and methods of use thereof
US6923958B2 (en) 2002-03-02 2005-08-02 The Scripps Research Institute DNA vaccines encoding CEA and a CD40 ligand and methods of use thereof
US7094410B2 (en) 2002-03-02 2006-08-22 The Scripps Research Institute DNA vaccine against proliferating endothelial cells and methods of use thereof
US20030185802A1 (en) * 2002-03-02 2003-10-02 Reisfeld Ralph A. DNA vaccine against proliferating endothelial cells and methods of use thereof
KR100600988B1 (en) 2002-03-13 2006-07-13 주식회사 엘지생명과학 Method for enhancing immune responses by codelivering influenza NP DNA in DNA immunization
US8430828B2 (en) 2002-04-19 2013-04-30 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a multi-use body fluid sampling device with sterility barrier release
US9089678B2 (en) 2002-04-19 2015-07-28 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8197421B2 (en) 2002-04-19 2012-06-12 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8197423B2 (en) 2002-04-19 2012-06-12 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8157748B2 (en) 2002-04-19 2012-04-17 Pelikan Technologies, Inc. Methods and apparatus for lancet actuation
US9339612B2 (en) 2002-04-19 2016-05-17 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9314194B2 (en) 2002-04-19 2016-04-19 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US8221334B2 (en) 2002-04-19 2012-07-17 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8235915B2 (en) 2002-04-19 2012-08-07 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7674232B2 (en) 2002-04-19 2010-03-09 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7648468B2 (en) 2002-04-19 2010-01-19 Pelikon Technologies, Inc. Method and apparatus for penetrating tissue
US9498160B2 (en) 2002-04-19 2016-11-22 Sanofi-Aventis Deutschland Gmbh Method for penetrating tissue
US8079960B2 (en) 2002-04-19 2011-12-20 Pelikan Technologies, Inc. Methods and apparatus for lancet actuation
US9248267B2 (en) 2002-04-19 2016-02-02 Sanofi-Aventis Deustchland Gmbh Tissue penetration device
US8267870B2 (en) 2002-04-19 2012-09-18 Sanofi-Aventis Deutschland Gmbh Method and apparatus for body fluid sampling with hybrid actuation
US9226699B2 (en) 2002-04-19 2016-01-05 Sanofi-Aventis Deutschland Gmbh Body fluid sampling module with a continuous compression tissue interface surface
US9186468B2 (en) 2002-04-19 2015-11-17 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8062231B2 (en) 2002-04-19 2011-11-22 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7708701B2 (en) 2002-04-19 2010-05-04 Pelikan Technologies, Inc. Method and apparatus for a multi-use body fluid sampling device
US8007446B2 (en) 2002-04-19 2011-08-30 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US9724021B2 (en) 2002-04-19 2017-08-08 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7988644B2 (en) 2002-04-19 2011-08-02 Pelikan Technologies, Inc. Method and apparatus for a multi-use body fluid sampling device with sterility barrier release
US9795334B2 (en) 2002-04-19 2017-10-24 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US9089294B2 (en) 2002-04-19 2015-07-28 Sanofi-Aventis Deutschland Gmbh Analyte measurement device with a single shot actuator
US8360992B2 (en) 2002-04-19 2013-01-29 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7981056B2 (en) 2002-04-19 2011-07-19 Pelikan Technologies, Inc. Methods and apparatus for lancet actuation
US8366637B2 (en) 2002-04-19 2013-02-05 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8372016B2 (en) 2002-04-19 2013-02-12 Sanofi-Aventis Deutschland Gmbh Method and apparatus for body fluid sampling and analyte sensing
US7976476B2 (en) 2002-04-19 2011-07-12 Pelikan Technologies, Inc. Device and method for variable speed lancet
US8382682B2 (en) 2002-04-19 2013-02-26 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7938787B2 (en) 2002-04-19 2011-05-10 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8388551B2 (en) 2002-04-19 2013-03-05 Sanofi-Aventis Deutschland Gmbh Method and apparatus for multi-use body fluid sampling device with sterility barrier release
US9072842B2 (en) 2002-04-19 2015-07-07 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8403864B2 (en) 2002-04-19 2013-03-26 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8414503B2 (en) 2002-04-19 2013-04-09 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US7914465B2 (en) 2002-04-19 2011-03-29 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US9839386B2 (en) 2002-04-19 2017-12-12 Sanofi-Aventis Deustschland Gmbh Body fluid sampling device with capacitive sensor
US8435190B2 (en) 2002-04-19 2013-05-07 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7909777B2 (en) 2002-04-19 2011-03-22 Pelikan Technologies, Inc Method and apparatus for penetrating tissue
US8491500B2 (en) 2002-04-19 2013-07-23 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US8496601B2 (en) 2002-04-19 2013-07-30 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US7909778B2 (en) 2002-04-19 2011-03-22 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7901362B2 (en) 2002-04-19 2011-03-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US7901365B2 (en) 2002-04-19 2011-03-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8556829B2 (en) 2002-04-19 2013-10-15 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8562545B2 (en) 2002-04-19 2013-10-22 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9907502B2 (en) 2002-04-19 2018-03-06 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7892183B2 (en) 2002-04-19 2011-02-22 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US8579831B2 (en) 2002-04-19 2013-11-12 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7713214B2 (en) 2002-04-19 2010-05-11 Pelikan Technologies, Inc. Method and apparatus for a multi-use body fluid sampling device with optical analyte sensing
US7892185B2 (en) 2002-04-19 2011-02-22 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US7874994B2 (en) 2002-04-19 2011-01-25 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8636673B2 (en) 2002-04-19 2014-01-28 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US7875047B2 (en) 2002-04-19 2011-01-25 Pelikan Technologies, Inc. Method and apparatus for a multi-use body fluid sampling device with sterility barrier release
US7862520B2 (en) 2002-04-19 2011-01-04 Pelikan Technologies, Inc. Body fluid sampling module with a continuous compression tissue interface surface
US7717863B2 (en) 2002-04-19 2010-05-18 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8905945B2 (en) 2002-04-19 2014-12-09 Dominique M. Freeman Method and apparatus for penetrating tissue
US8202231B2 (en) 2002-04-19 2012-06-19 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US7731729B2 (en) 2002-04-19 2010-06-08 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US8845549B2 (en) 2002-04-19 2014-09-30 Sanofi-Aventis Deutschland Gmbh Method for penetrating tissue
US8808201B2 (en) 2002-04-19 2014-08-19 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for penetrating tissue
US8690796B2 (en) 2002-04-19 2014-04-08 Sanofi-Aventis Deutschland Gmbh Method and apparatus for penetrating tissue
US8784335B2 (en) 2002-04-19 2014-07-22 Sanofi-Aventis Deutschland Gmbh Body fluid sampling device with a capacitive sensor
US7833171B2 (en) 2002-04-19 2010-11-16 Pelikan Technologies, Inc. Method and apparatus for penetrating tissue
US20030198626A1 (en) * 2002-04-22 2003-10-23 Antigen Express, Inc. Inhibition of Ii expression in mammalian cells
US20060003316A1 (en) * 2002-07-15 2006-01-05 John Simard Immunogenic compositions derived from poxviruses and methods of using same
US20040029275A1 (en) * 2002-08-10 2004-02-12 David Brown Methods and compositions for reducing target gene expression using cocktails of siRNAs or constructs expressing siRNAs
US9034639B2 (en) 2002-12-30 2015-05-19 Sanofi-Aventis Deutschland Gmbh Method and apparatus using optical techniques to measure analyte levels
US8574895B2 (en) 2002-12-30 2013-11-05 Sanofi-Aventis Deutschland Gmbh Method and apparatus using optical techniques to measure analyte levels
US20040176282A1 (en) * 2003-01-09 2004-09-09 Brian Dalby Cellular delivery and activation of polypeptide-nucleic acid complexes
WO2004065575A2 (en) 2003-01-15 2004-08-05 Research Institute For Genetic And Human Therapy (Right) Dna compositon and uses thereof
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
US8262614B2 (en) 2003-05-30 2012-09-11 Pelikan Technologies, Inc. Method and apparatus for fluid injection
US8251921B2 (en) 2003-06-06 2012-08-28 Sanofi-Aventis Deutschland Gmbh Method and apparatus for body fluid sampling and analyte sensing
US7850621B2 (en) 2003-06-06 2010-12-14 Pelikan Technologies, Inc. Method and apparatus for body fluid sampling and analyte sensing
US9144401B2 (en) 2003-06-11 2015-09-29 Sanofi-Aventis Deutschland Gmbh Low pain penetrating member
US10034628B2 (en) 2003-06-11 2018-07-31 Sanofi-Aventis Deutschland Gmbh Low pain penetrating member
EP2258841A1 (en) 2003-06-23 2010-12-08 The Regents of the University of Colorado Methods for treating pain
US8282576B2 (en) 2003-09-29 2012-10-09 Sanofi-Aventis Deutschland Gmbh Method and apparatus for an improved sample capture device
US8945910B2 (en) 2003-09-29 2015-02-03 Sanofi-Aventis Deutschland Gmbh Method and apparatus for an improved sample capture device
US9351680B2 (en) 2003-10-14 2016-05-31 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a variable user interface
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
US8668656B2 (en) 2003-12-31 2014-03-11 Sanofi-Aventis Deutschland Gmbh Method and apparatus for improving fluidic flow and sample capture
US9561000B2 (en) 2003-12-31 2017-02-07 Sanofi-Aventis Deutschland Gmbh Method and apparatus for improving fluidic flow and sample capture
US8296918B2 (en) 2003-12-31 2012-10-30 Sanofi-Aventis Deutschland Gmbh Method of manufacturing a fluid sampling device with improved analyte detecting member configuration
US8344019B2 (en) 2004-03-19 2013-01-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods for the production of biliverdin
US20080131455A1 (en) * 2004-04-30 2008-06-05 Pds Biotechnology Corporation Antigen Delivery Compositions And Methods Of Use
US8128938B1 (en) 2004-05-18 2012-03-06 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
US20070286869A1 (en) * 2004-05-18 2007-12-13 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
US20060024670A1 (en) * 2004-05-18 2006-02-02 Luke Catherine J Influenza virus vaccine composition and methods of use
US9261476B2 (en) 2004-05-20 2016-02-16 Sanofi Sa Printable hydrogel for biosensors
US8828203B2 (en) 2004-05-20 2014-09-09 Sanofi-Aventis Deutschland Gmbh Printable hydrogels for biosensors
US9775553B2 (en) 2004-06-03 2017-10-03 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a fluid sampling device
US9820684B2 (en) 2004-06-03 2017-11-21 Sanofi-Aventis Deutschland Gmbh Method and apparatus for a fluid sampling device
US20080025944A1 (en) * 2004-09-02 2008-01-31 Cure Vac Gmbh Combination Therapy for Immunostimulation
WO2006024518A1 (en) 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immunostimulation
US20060153844A1 (en) * 2004-12-29 2006-07-13 Thomas Kundig Methods to trigger, maintain and manipulate immune responses by targeted administration of biological response modifiers into lymphoid organs
US8652831B2 (en) 2004-12-30 2014-02-18 Sanofi-Aventis Deutschland Gmbh Method and apparatus for analyte measurement test time
US7822454B1 (en) 2005-01-03 2010-10-26 Pelikan Technologies, Inc. Fluid sampling device with improved analyte detecting member configuration
US20090068214A1 (en) * 2005-04-15 2009-03-12 Jiahua Qian Methods and Compositions for Producing an Enhanced Immune Response to a Human Papillomavirus Immunogen
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
WO2006122828A2 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized injection formulation for rna
EP3153179A1 (en) 2005-05-19 2017-04-12 CureVac AG Optimised injection formulation for mrna
EP3583953A1 (en) 2005-05-19 2019-12-25 CureVac AG Optimised injection formulation for mrna
WO2006130581A2 (en) 2005-05-31 2006-12-07 Avigen, Inc. Methods for delivering genes
EP2816118A1 (en) 2005-05-31 2014-12-24 The Regents of the University of Colorado, A Body Corporate Methods for delivering genes
US8702624B2 (en) 2006-09-29 2014-04-22 Sanofi-Aventis Deutschland Gmbh Analyte measurement device with a single shot actuator
WO2008074678A1 (en) 2006-12-18 2008-06-26 F. Hoffmann-La Roche Ag Novel use of inhibitors of soluble epoxide hydrolase
US11421038B2 (en) 2007-01-09 2022-08-23 Curevac Ag RNA-coded antibody
EP3115064B1 (en) 2007-01-09 2021-11-24 CureVac AG Rna-coded antibody
EP2101823B1 (en) 2007-01-09 2016-11-23 CureVac AG Rna-coded antibody
US8877206B2 (en) 2007-03-22 2014-11-04 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
US11911359B2 (en) 2007-03-22 2024-02-27 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
US20090017057A1 (en) * 2007-03-22 2009-01-15 Pds Biotechnology Corporation Stimulation of an immune response by cationic lipids
EP2484770A1 (en) 2007-09-04 2012-08-08 CureVac GmbH Complexes of RNA and cationic peptides for transfection and for immunostimulation
US9386944B2 (en) 2008-04-11 2016-07-12 Sanofi-Aventis Deutschland Gmbh Method and apparatus for analyte detecting device
US11801257B2 (en) 2008-04-17 2023-10-31 Pds Biotechnology Corporation Stimulation of an immune response by enantiomers of cationic lipids
US9375169B2 (en) 2009-01-30 2016-06-28 Sanofi-Aventis Deutschland Gmbh Cam drive for managing disposable penetrating member actions with a single motor and motor and control system
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10143758B2 (en) 2009-12-01 2018-12-04 Translate Bio, Inc. Liver specific delivery of messenger RNA
EP3187585A1 (en) 2010-03-25 2017-07-05 Oregon Health&Science University Cmv glycoproteins and recombinant vectors
US8965476B2 (en) 2010-04-16 2015-02-24 Sanofi-Aventis Deutschland Gmbh Tissue penetration device
US9795747B2 (en) 2010-06-02 2017-10-24 Sanofi-Aventis Deutschland Gmbh Methods and apparatus for lancet actuation
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11135274B2 (en) 2010-11-30 2021-10-05 Translate Bio, Inc. MRNA for use in treatment of human genetic diseases
US9956271B2 (en) 2010-11-30 2018-05-01 Translate Bio, Inc. mRNA for use in treatment of human genetic diseases
US9061021B2 (en) 2010-11-30 2015-06-23 Shire Human Genetic Therapies, Inc. mRNA for use in treatment of human genetic diseases
WO2012106281A2 (en) 2011-01-31 2012-08-09 The General Hospital Corporation Multimodal trail molecules and uses in cellular therapies
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
WO2012130941A2 (en) 2011-03-31 2012-10-04 Schaefer Konstanze Perfluorinated compounds for the non-viral transfer of nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US10350303B1 (en) 2011-06-08 2019-07-16 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10507183B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Cleavable lipids
US11185595B2 (en) 2011-06-08 2021-11-30 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US11291734B2 (en) 2011-06-08 2022-04-05 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11338044B2 (en) 2011-06-08 2022-05-24 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
EP3354644A1 (en) * 2011-06-08 2018-08-01 Translate Bio, Inc. Cleavable lipids
US11052159B2 (en) 2011-06-08 2021-07-06 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10888626B2 (en) 2011-06-08 2021-01-12 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US20140294939A1 (en) * 2011-06-08 2014-10-02 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mrna
EP3674292A1 (en) * 2011-06-08 2020-07-01 Translate Bio, Inc. Cleavable lipids
US9597413B2 (en) * 2011-06-08 2017-03-21 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mRNA
US10507249B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11234936B2 (en) 2011-06-08 2022-02-01 Translate Bio, Inc. Cleavable lipids
US10413618B2 (en) 2011-06-08 2019-09-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
EP4212514A1 (en) * 2011-06-08 2023-07-19 Translate Bio, Inc. Cleavable lipids
US10238754B2 (en) 2011-06-08 2019-03-26 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
WO2012170765A2 (en) 2011-06-10 2012-12-13 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
EP2568289A2 (en) 2011-09-12 2013-03-13 International AIDS Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
EP2586461A1 (en) 2011-10-27 2013-05-01 Christopher L. Parks Viral particles derived from an enveloped virus
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
EP2679596A1 (en) 2012-06-27 2014-01-01 Simon Hoffenberg HIV-1 env glycoprotein variant
WO2014035474A1 (en) 2012-08-30 2014-03-06 The General Hospital Corporation Compositions and methods for treating cancer
US11911465B2 (en) 2012-09-21 2024-02-27 Pds Biotechnology Corporation Vaccine compositions and methods of use
US11904015B2 (en) 2012-09-21 2024-02-20 Pds Biotechnology Corporation Vaccine compositions and methods of use
CN102879566A (en) * 2012-10-12 2013-01-16 武汉康苑生物医药科技有限公司 Enzyme-linked immunosorbent assay (ELISA) kit for human immunodeficiency virus (HIV) P24 antigen
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US10420791B2 (en) 2013-03-14 2019-09-24 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
US11510937B2 (en) 2013-03-14 2022-11-29 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US11692189B2 (en) 2013-03-14 2023-07-04 Translate Bio, Inc. Methods for purification of messenger RNA
US9713626B2 (en) 2013-03-14 2017-07-25 Rana Therapeutics, Inc. CFTR mRNA compositions and related methods and uses
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US11820977B2 (en) 2013-03-14 2023-11-21 Translate Bio, Inc. Methods for purification of messenger RNA
US10876104B2 (en) 2013-03-14 2020-12-29 Translate Bio, Inc. Methods for purification of messenger RNA
WO2014145042A1 (en) 2013-03-15 2014-09-18 Loma Linda University Treatment of autoimmune diseases
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10640771B2 (en) 2013-04-17 2020-05-05 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
EP3741385A1 (en) 2013-04-17 2020-11-25 Genzyme Corporation Compositions for use in a method of treating and preventing macular degeneration
WO2014172560A1 (en) 2013-04-17 2014-10-23 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
EP3848045A1 (en) 2013-05-21 2021-07-14 President and Fellows of Harvard College Engineered heme-binding compositions and uses thereof
WO2014190040A1 (en) 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
EP2848937A1 (en) 2013-09-05 2015-03-18 International Aids Vaccine Initiative Methods of identifying novel HIV-1 immunogens
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
EP2873423A2 (en) 2013-10-07 2015-05-20 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
US10780052B2 (en) 2013-10-22 2020-09-22 Translate Bio, Inc. CNS delivery of MRNA and uses thereof
US11377642B2 (en) 2013-10-22 2022-07-05 Translate Bio, Inc. mRNA therapy for phenylketonuria
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US10208295B2 (en) 2013-10-22 2019-02-19 Translate Bio, Inc. MRNA therapy for phenylketonuria
US10898584B2 (en) 2013-11-01 2021-01-26 Curevac Ag Modified RNA with decreased immunostimulatory properties
WO2015120309A1 (en) 2014-02-06 2015-08-13 Genzyme Corporation Compositions and methods for treating and preventing macular degeneration
US10155785B2 (en) 2014-04-25 2018-12-18 Translate Bio, Inc. Methods for purification of messenger RNA
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US11059841B2 (en) 2014-04-25 2021-07-13 Translate Bio, Inc. Methods for purification of messenger RNA
US11884692B2 (en) 2014-04-25 2024-01-30 Translate Bio, Inc. Methods for purification of messenger RNA
US11872285B2 (en) 2014-07-15 2024-01-16 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10195280B2 (en) 2014-07-15 2019-02-05 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
US10792362B2 (en) 2014-07-15 2020-10-06 Life Technologies Corporation Compositions and methods for efficient delivery of molecules to cells
EP3069730A2 (en) 2015-03-20 2016-09-21 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
EP3072901A1 (en) 2015-03-23 2016-09-28 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
WO2016168601A1 (en) 2015-04-17 2016-10-20 Khalid Shah Agents, systems and methods for treating cancer
WO2016180430A1 (en) 2015-05-08 2016-11-17 Curevac Ag Method for producing rna
US11638753B2 (en) 2015-11-13 2023-05-02 PDS Biotechnology Corporalion Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
US11612652B2 (en) 2015-11-13 2023-03-28 Pds Biotechnology Corporation Lipids as synthetic vectors to enhance antigen processing and presentation ex-vivo in dendritic cell therapy
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11951179B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951180B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951181B2 (en) 2023-04-03 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery

Similar Documents

Publication Publication Date Title
US6214804B1 (en) Induction of a protective immune response in a mammal by injecting a DNA sequence
US6710035B2 (en) Generation of an immune response to a pathogen
US7250404B2 (en) Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US6673776B1 (en) Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
CA2049287C (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
AU781356B2 (en) Viral core protein-cationic lipid-nucleic acid-delivery complexes
JP4085231B2 (en) Protein hollow nanoparticles, substance carrier using the same, and method for introducing substance into cells
EP0902682A2 (en) Cationic virosomes as transfer system for genetic material
US6706694B1 (en) Expression of exogenous polynucleotide sequences in a vertebrate
US20030186913A1 (en) Expression of exogenous polynucleotide sequences in a vertebrate
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
US20050220806A1 (en) Method of transporting physiological polymer using protein having rxp repeated sequence
JP2006517799A (en) A non-immunogenic selectable marker resistant to cardiac glycosides
WO1996029422A1 (en) Nucleic acid carrier

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Free format text: PAYER NUMBER DE-ASSIGNED (ORIGINAL EVENT CODE: RMPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 12