EP1038016A2 - Nadelloses injektion von formulierte nukleinsäure moleküle - Google Patents

Nadelloses injektion von formulierte nukleinsäure moleküle

Info

Publication number
EP1038016A2
EP1038016A2 EP98964020A EP98964020A EP1038016A2 EP 1038016 A2 EP1038016 A2 EP 1038016A2 EP 98964020 A EP98964020 A EP 98964020A EP 98964020 A EP98964020 A EP 98964020A EP 1038016 A2 EP1038016 A2 EP 1038016A2
Authority
EP
European Patent Office
Prior art keywords
needle
nucleic acid
delivering
acid molecule
mammal
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.)
Withdrawn
Application number
EP98964020A
Other languages
English (en)
French (fr)
Inventor
Michael Barry
Russ Mumper
Lou Smith
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.)
Baylor College of Medicine
Urigen Pharmaceuticals Inc
Original Assignee
Baylor College of Medicine
Urigen Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Baylor College of Medicine, Urigen Pharmaceuticals Inc filed Critical Baylor College of Medicine
Publication of EP1038016A2 publication Critical patent/EP1038016A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH

Definitions

  • the present invention relates to products and methods useful for delivering formulated nucleic acid molecules by needle-free injection.
  • nucleic acids in vivo have been pursued by a variety of methods. These include lipofectin/liposome fusion: Proc. Natl. Acad. Sci., Volume 84, pp. 7413-7417 (1993); polylysine condensation with and without adenovirus enhancement: Human Gene Therapy, Volume 3, pp. 147-154 (1992); and transfer ⁇ n:transferrin receptor delivery of nucleic acid to cells: Proc. Natl. Acad. Sci., Volume 87, pp. 3410-3414 (1990).
  • the use of a specific composition consisting of polyacrylic acid has been disclosed in WO 94/24983. Naked DNA has been administered as disclosed in International Patent Publication No.
  • Aerosol injection has been use to introduce human cytomegalovirus immediate early gene 1 enhancer/ promoter sequences, bacterial chloramphenicol acetyltransferase, whey acidic protein promoter sequences, and the bacterial -galactosidase gene through the skin surface so as to transfect skin, muscle, fat and mammary tissue of living animals (Furth et al., Analytical biochemistry, 205, 365- 368, 1992). It has been stated that suspending aerosol-injected DNA constructs in solutions which enhance cellular uptake of DNA may increase the number of cells transfected in a single injection (Furth et al., supra).
  • DNA bound by precipitation methods to inert particles have been delivered by needle-free injection.
  • Coating inert particles, such as gold microprojectiles, and injecting. them by needle-free devices offers direct injection into the cells due to the projectile carrier penetrating the cell.
  • Tang et al. inoculated mice with DNA encoding human growth hormone coated gold microprojectiles and stated that this method was superior to needle injection of plasmids encoding human growth hormone. Tang et al, reports that this technique simplifies the procedure and shortens the time required to produce antibodies to particular proteins by eliminating steps for protein purification and adjuvant administration. (Tang et al., Nature, Vol. 356:152-154 1992)
  • This invention features compositions and methods for enhancing the administration to and uptake of nucleic acids in a mammal.
  • An efficient strategy for enhancing needle-free delivery of nucleic acids in vivo is to protect the nucleic acid from degradation, thereby maintaining the administered nucleic acid at the target site in order to further increase its cellular uptake.
  • the data presented herein demonstrates that the combination of formulated nucleic acid molecules and needle free injection methods are synergistic, providing the desired antibody response to the resulting expressed protein that is unexpectedly high when compared with either needle-free injection of non-formulated nucleic acids or needle injection of formulated nucleic acids.
  • the invention provides a method to deliver formulated nucleic acid molecules through and/or to the skin of a mammal by using an apparatus configured and arranged to administer molecules by air, or mechanical pressure through and/or to the skin of a mammal.
  • the present invention allows for superior delivery of nucleic acid molecules into cells in vivo by the combination of a needle free device and formulated nucleic acid molecules.
  • the present invention also allows for treatment of diseases, vaccination, and treatment of muscle disorder and serum protein deficiencies.
  • the present invention features a method for delivering a nucleic acid molecule formulated with a transfection facilitating agent through and/or to the skin of a mammal by the use of a needle-free injection device.
  • the needle-free device is configured and arranged to cause aerosol delivery of the formulated nucleic acid through and/or to the skin of the mammal.
  • nucleic acid molecules By “delivery” or “delivering” is meant transportation of nucleic acid molecules to desired cells or any cells.
  • the nucleic acid molecules will be delivered to multiple cell lines, including the desired target. Delivery results in the nucleic acid molecules coming in contact with the cell surface, cell membrane, cell endosome, within the cell membrane, nucleus or within the nucleus, or any other desired area of the cell from which transfection can occur within a variety of cell lines which can include but are not limited to; epithelial cells, Langerhan cells, Langhans' cells, littoral cells, keratinocytes, dendritic cells, macrophage cells, kupffer cells, lymphocytes.and lymph nodes.
  • the nucleic acid molecule can be delivered through and/or to the skin by aerosol pressure and is not significantly sheared.
  • nucleic acid refers to both RNA and DNA including: cDNA, genomic DNA, plasmid DNA or condensed nucleic acid, nucleic acid formulated with cationic Iipids, nucleic acid formulated with peptides, antisense molecule, cationic substances, RNA or mRNA.
  • the nucleic acid administered is plasmid DNA which comprises a "vector".
  • the nucleic acid can be, but is not limited to, a plasmid DNA vector with a eukaryotic promoter which expresses Human Growth Hormone, such as in the example provided.
  • transfection facilitating agent refers to an agent that forms a complex with the nucleic acid. This molecular complex is associated with nucleic acid, molecule in either a covalent or a non-covalent manner.
  • the transfection facilitating agent should be capable of transporting nucleic acid molecules in a stable state and of releasing the bound nucleic acid molecules into the cellular interior.
  • the transfection facilitating agent should also be capable of being bound to nucleic acid molecules and lyopholized or freeze dried and either rehydrated prior to needle-free delivery or delivered as a fine powder via needle-free delivery.
  • transfection facilitating agent may prevent lysosomal degradation of the nucleic acid molecules by endosomal lysis.
  • the transfection facilitating agent allows for efficient transport of the nucleic acid molecule through the cytoplasm of the cell to the nuclear membrane and into the nucleus and provide protection.
  • transfection facilitating agents are non-condensing polymers, oils and surfactants.
  • nucleic acid polyvinylpyrrolidones; polyvinylaicohols; propylene glycols; polyethylene glycols; polyvinylacetates; poloxamers (Pluronics)(block copolymers of propylene oxide and ethylene oxide, relative amounts of the two subunits may vary in different poloxamers); poloxamines (Tetronics); ethylene vinyl acetates; celluloses, including salts of carboxymethylcelluloses, methylcelluloses, hydroxypropylcelluloses, hydroxypropylmethylcelluloses; salts of hyaluronates; salts of alginates; heteropolysaccharides (pectins); dextrans; chitosans; phosphatidylcholines (lecithins); miglyols; polylactic acid; polyhydroxybutyric acid.
  • Some of these compounds may be used as protective, interactive, non-condensing
  • cationic condensing agents such as cationic Iipids, peptides, or lipopetides may associate with the nucleic acid molecule and may facilitate transfection.
  • some of these compounds may be covalently attached to gold particles and thereby bind with nucleic acid molecules of the present invention.
  • Gold particles coated with a polymer or polymers of the present invention can deliver nucleic acid molecules into cells by penetrating the cell when delivered by needle-free injection device.
  • the term “protects” or “protective” or “protected” as used herein refers to an effect of the interaction between such a compound and a nucleic acid such that the rate of degradation of the nucleic acid is decreased in a particular environment, thereby prolonging the localized bioavailability of the nucleic acid molecule. Such degradation may be due a variety of different of factors, which specifically include the enzymatic action of a nuclease.
  • the protective action may be provided in different ways, for example, by exclusion of the nuclease molecules or by exclusion of water.
  • the compounds which protect the nucleic acid and/or prolong the localized bioavailability of a nucleic acid may achieve one or more of the following effects, due to their physical, chemical or rheological properties: (1) Protect nucleic acid, for example plasmid DNA, from nucleases due to steric, viscosity, or other effects such as shearing; (2) increase the area of contact between nucleic acid, such as plasmid DNA, through extracellular matrices and over cellular membranes, into which the nucleic acid is to be taken up; (3) concentrate nucleic acid, such as plasmid DNA, at cell surfaces due to water exclusion; (4) indirectly facilitate uptake of nucleic acid, such as plasmid DNA, by disrupting cellular membranes due to osmotic, hydrophobic or lytic effects; and (5) indirectly facilitate uptake of nucleic acids by allowing diffusion of protected nucleic acid chains through tissue at the administration site.
  • nucleic acid By “prolonging the localized bioavailability of a nucleic acid” is meant that a nucleic acid when administered to an organism in a composition comprising such a compound will be available for uptake by cells for a longer period of time than if administered in a composition without such a compound, for example when administered in a saline solution. This increased availability of nucleic acid to cells could occur, for example, due to increased duration of contact between the composition containing the nucleic acid and a cell or due to protection of the nucleic acid from attack by nucleases.
  • the compounds which prolong the localized bioavailability of a nucleic acid are suitable for internal administration.
  • suitable for internal administration is meant that the compounds are suitable to be administered within the tissue of an organism, for example within a muscle or within a joint space, epidermally, intradermally or subcutaneously.
  • Properties making a compound suitable for internal administration can include, for example, the absence of a high level of toxicity to the organism as a whole.
  • needle-free injection device as used herein relates to an apparatus that is capable of injecting an aerosol through and/or to the skin of a mammal into the tissue by air and or mechanical pressure. It is understood that conventional devices of this type are calibrated to allow one of ordinary skill in the art to select and/or adjust the desired injection depth and therefore it is expected that future devices that perform this function will also be calibrated in the same manner. It is also understood that devices of this type may have a needle which is only used IO collect a solution which is subsequently aerosolized, and delivered by needle-free means. The type of injection device is not considered a limiting aspect of the present invention.
  • the primary importance of a needle-free device is, in fact, the capability of the device to deliver an aerosol of formulated nucleic acid molecules through and/or to the skin of a mammal.
  • the needle-free injection device can include, for example, a Gene Gun or a Needle-Less Injector as described in U.S. Patent 5,480,381 or a powder delivery device such as in PCT WO/O97/134652.
  • the term "apparatus” as used herein relates to the set of components that upon combination allow the delivery of an aerosol through and/or to the skin of a mammal.
  • The-. components can be a nozzle or needle-free syringe with which one can collect and/or administer the formulated nucleic acid molecules, and a pump or spring for creating air pressure which forcibly evacuates the formulated nucleic acid molecules from the nozzle or needle-free syringe in a manner that creates an aerosol capable of penetrating the skin of a mammal.
  • the apparatus can employ gas pressure, gas spring or spring force.
  • the apparatus is capable of being calibrated to allow selection of depth of delivery. Hence, delivery can occur through the skin or to the skin.
  • aerosol as used herein is a suspension of formulated nucleic acid molecules in the form of a particulate mist. Aerosols have been defined as colloidal systems consisting of very finely subdivided liquid or solid particles dispersed in and surrounded by a gas. The aerosol of the present invention can depend upon the power of a liquified or compressed gas or mechanical spring to generate the fine mist of formulated nucleic acid molecules. Particles of an aerosol can range from less than 1 to 50 m. The particles are said to remain suspended in the air for relatively long periods of time.
  • the size of the particles can be measured by conventional methods such as the Milken Oil Drop Experiment for measuring aerosol particle size, but the need to determine specific size is falling into disuse (Sciarra, et al., in Remington's Pharmaceutical Sciences, 18th ed. chapter 92, 1990).
  • the aerosol can be a liquid, a powder, or a heterogeneous mist comprising both a liquid and solid phase.
  • skin refers to the outer covering of a mammal consisting of epidermal and dermal tissue and appendages such as sweat ducts and hair folicles. Skin can comprise the hair of a mammal in cases where the mammal has an epidermis which is covered by hair.
  • the method results in an immune response, preferably a humoral immune response targeted for the protein product encoded by the nucleic acid molecule, such as an antibody response that is preferably at least 3 times greater than an antibody response caused by needle injection of a protein product encoded by a nucleic acid molecule suspended in saline, and at least 10 times greater than an antibody response caused by needle injection of a nucleic acid molecule formulated with a transfection facilitating agent.
  • the immune response preferably is a cytotoxic T- lymphocyte response.
  • the term "immune response" as used herein refers to the mammalian natural defense mechanism which can occur when foreign material is internalized.
  • the immune response can be a global immune response involving the immune system components in their entirety.
  • the immune response results from the protein product encoded by the formulated nucleic acid molecule.
  • the immune response can be, but is not limited to; antibody production, T-cell proliferation /differentiation, activation of cytotoxic T- lymphocytes, and or activation of natural killer cells.
  • the immune response is a humoral immune response.
  • the immune response preferably, is a cytotoxic T-lymphocyte response.
  • the term "humoral immune response” refers to the production of antibodies in response to internalized foreign material.
  • the foreign material is the protein product encoded by a formulated nucleic acid molecule internalized by injection with a needle free device.
  • the needle-free device is selected from the group consisting of: Mediject, Bioject, Gene Gun, Mesoflash, Ped-O-ject and Powder- Ject. Generally, it is understood that such a device is accompanied by directions for usage.
  • the invention features a kit.
  • the kit includes a provider for providing a nucleic acid molecule formulated with a transfection facilitating agent and a needle-free means for delivering the nucleic acid molecule through and/or to the skin of a mammal.
  • the "provider” can be instructions furnished to allow one of ordinary skill in the art to make formulated nucleic acid molecules.
  • the instructions will furnish steps to make the compounds used for formulating nucleic acid molecules. Additionally, the instructions will include methods for testing the formulated nucleic acid molecules that entail establishing if the formulated nucleic acid molecules are damaged upon injection from the needle-free device.
  • the provider can also be the formulated nucleic acid molecules themselves.
  • transfection refers to the process of introducing DNA
  • transfected DNA may: (1) recombine with that of the host; (2) replicate independently as a plasmid or temperate phage; or (3) be maintained as an episome without replication prior to elimination.
  • transformation relates to transient or permanent changes in the characteristics (expressed phenotype) of a cell induced by the uptake of a vector by that cell. Genetic material is introduced into a cell in a form where it expresses a specific gene product or alters the expression or effect of endogenous gene products.
  • Transformation of the cell may be associated with production of a variety of gene products including protein and RNA. These products may function as intracellular or extracellular structural elements, ligands, hormones, neurotransmitters, growth regulating factors, enzymes, chemotaxins, serum proteins, receptors, carriers for small molecular weight compounds, drugs, immunomodulators, oncogenes, cytokines, tumor suppressors, toxins, tumor antigens, antigens, antisense inhibitors, triple strand forming inhibitors, ribozymes, or as a ligand recognizing specific structural determinants on cellular structures for the purpose of modifying their activity. This list is only an example and is not meant to be limiting.
  • the invention features a method for making a kit.
  • the method involves the step of combining a provider for providing a nucleic acid formulated with a transfection facilitating agent and a needle-free means for delivering the nucleic acid to a mammal.
  • the provider for providing a nucleic acid can be instructions for formulating nucleic acid molecules or simply the formulated nucleic acid molecules.
  • the invention also features a method for treating a mammal that is suffering from a disorder conventionally treated by administering human growth hormone.
  • the method requires administering a nucleic acid molecule encoding human growth hormone and formulated with a transfection facilitating agent through and/or to the skin of the mammal by use of a needle-free device.
  • the invention features a method for treating a mammal that is suffering from a cancer by administering a nucleic acid molecule encoding the appropriate-. cancer antigen.
  • the method requires administering a nucleic acid molecule encoding a cancer antigen and formulated with a transfection facilitating agent through and/or to the skin of the mammal by use of a needle-free device.
  • the cancer is melanoma and the appropriate cancer antigen is MAGE 1.
  • the invention also features a method for treating a mammal that is suffering from an infectious disease by administering a nucleic acid molecule encoding an antigen for the infectious disease.
  • the method requires administering a nucleic acid molecule encoding an antigen for the infectious disease and formulated with a transfection facilitating agent through and/or to the skin of the mammal by use of a needle- free device.
  • the infectious disease is chronic hepatitis and the antigen is HBV core antigen.
  • Administration refers to the route of introducing the formulated nucleic acid molecules of the invention into the body of cells or organisms. Administration includes the use of aerosol pressure as provided by a needle free device to targeted areas of the mammalian body such as the muscle and the lymph nodes.
  • the nucleic acid molecules of the invention can be formulated with at least one other type of molecule.
  • the molecular complexes can be formulated with other molecules such as polyvinyl-pyrrolidone as described herein. Formulation techniques are provided herein by example.
  • administration is directed through the epidermis, intradermis, and subcutaneous layer to the muscle tissue.
  • the present invention administers formulated nucleic acid molecules in a manner which causes contact with various mammalian cell types which are not contacted by conventional needle injection techniques during one injection.
  • Cell lines contacted by injection with the present invention are, but are not limited to, epithelial cells, langerhan cells, keratinocytes, dendritic cells, macrophage cells, kupfer cells, lymphocytes and lymph nodes.
  • Another aspect of the present invention features a method for modifying inert particles by first washing the inert particles in fuming nitric acid solutions or in solutions of nitric acid mixed with sulfuric acid, and then combining the inert particles with cysteine terminated cationic DNA binding peptides to form a monolayer peptide coating.
  • the DNA binding peptide recognizes and binds a specific nucleic acid sequence of a designed plasmid. Hence, one molecule of DNA binding peptide binds with an inert particle and a plasmid, therefore reducing the amount of crosslinking between inert particles.
  • the inert particles can be but are not limited to gold particles.
  • the inert particles are combined with nucleic acid molecules and delivered through the skin of a mammal to cells by a needle-free device.
  • inactive particles refers to biologically inactive particles which can be but are not limited to particles from the group consisting of, ferrite crystals, gold particles or beads, tungsten spheres, other metals or biologically inactive compounds which are of a density of roughly 15 - 20g/cm and a size of roughly 1-3 m. Generally, the optimum particle is small enough to produce minimal cell damage.
  • the size of particles is not a limiting aspect. It is known by those of ordinary skill in the art that the particle must be large enough to acquire sufficient momentum to penetrate the cell: momentum being a function of size density and velocity, and by convention particle size should be roughly 10 times smaller than the target cell.
  • a further aspect of the invention features a method for delivering a nucleic acid molecule formulated with a modified inert particle through and/or to the skin of a mammal by the use of a needle-free injection device.
  • the needle-free device is configured and arranged to cause aerosol delivery of the formulated nucleic acid through and/or to the skin of the mammal.
  • Figure 1 shows the humoral immune response to hGH in dogs after needle and needle-free injection of transfection facilitating agent formulated pCMV-hGH in dogs.
  • the antibody titer for each data point is an average for two dogs.
  • Figure 2 shows a comparison between pCMV-hGH suspended in saline and transfection facilitating agent formulated pC ⁇ vTV-hGH for the elicitation of humoral immune response to hGH in dogs after needle-free injection.
  • the antibody titer for each data point is an average for two dogs.
  • Figure 3 shows the amounts of expressed Luciferase in conditions where a plasmid encoding CMV-Luciferase is formulated with modified gold particles that are coated or uncoated with Cys-Tyr-Lys-ala- (Lys) 8 -Trp-Lys (CK8).
  • the formulations are then loaded on a Kapton carrier membrane in ethanol, water or precipitated with Ca2+ and then loaded in ethanol.
  • the delivery of formulated nucleic acid molecules by the use of a needle free device represents a novel approach to gene delivery.
  • the present invention offers a nucleic acid delivery apparatus that provides an increased immune response when compared to previous methods.
  • the invention provides the advantage of allowing the uptake of formulated nucleic acid molecules by a wide variety of cell types simultaneously. Injecting formulated nucleic acid molecules by needle free device results in the formulated nucleic acid molecules directly contacting many more cell types than in conventional needle injection.
  • the present invention provides an enhanced delivery of nucleic acid molecules and also provides a more efficient gene delivery system which can be used to generate an immune response. Needle-free delivery of formulated nucleic acid molecules through and/or to the skin of a mammal, depends on several factors which are discussed below, including transfection efficiency and the composition of the formulated nucleic acid molecule.
  • the intensity of an immune response and the level of gene delivery and expression achieved with the present invention can be optimized (>5-fold effect over controls) by altering the following variables.
  • the variables are: the formulation (composition, plasmid topology), the technique and protocol for injection (angle of injection, state of muscle), and, the pretreatment of the muscle with myotoxic agents.
  • An immune response can be measured by, but is not limited to, the amount of antibodies produced for a protein encoded and expressed by the injected nucleic acid molecule.
  • injection variables that can be used to significantly effect the levels of antibodies and/or cytotoxic T-lymphocytes produced in response to the protein encoded by the formulated nucleic acid molecule provided by the needle-free injection method of the present invention are the state of the muscle being injected and injection technique. Examples of the variables include muscle stimulation, muscle contraction, muscle massage, delivery angle, and apparatus manipulation. Massaging the muscle may force plasmid out of the muscle either directly or via lymphatic drainage. By altering the depth of penetration and/or the angle at which the needle-free device is placed in relation to muscle fibers the present invention improves the plasmid distribution throughout the injection area which subsequently increases the antibody response to the protein which is encoded and expressed by the plasmid.
  • Needle free injection systems provide an attractive method for administration of plasmid DNA for the purpose of intramuscular immunization. Not only do they provide the general benefit of avoiding needle-stick injury, but they may produce better distribution of injected substances in the muscle. Direct gene transfer was first demonstrated on mammary tissue using the Ped-O-jet system, but this apparatus was designed largely for intradermal or subcutaneous injections (Furth et al., Anal, biochem. 205:365-368., 1992).
  • the present invention can be used to deliver nucleic acid vaccines in a more efficient manner than is conventionally done at the present time.
  • Nucleic acid vaccines or the use of plasmid encoding antigens or therapeutic molecules such as Human Growth Hormone, has become an area of intensive research and development in the last half decade.
  • Comprehensive reviews on nucleic acid based vaccines have been published [M.A. Liu, et al.(Eds.), 1995, DNA Vaccines: A new era in vaccinology, Vol. 772, Ann. NY. Acad. Sci., New York; Kumar, V., and Sercarz, E., 1996, Nat. Med.
  • nucleic acid based vaccines are an attractive alternative vaccination strategy to subunit vaccines, purified viral protein vaccines, or viral vector vaccines.
  • Each of the traditional approaches has limitations that are overcome if the antigen(s) is expressed directly in cells of the body. Furthermore, these traditional vaccines are only protective in a strain-specific fashion. Thus, it is very difficult, and even impossible using traditional vaccine approaches to obtain long lasting immunity to viruses that have several sera types.-. or viruses that are prone to mutation.
  • Nucleic acid based vaccines offer the potential to produce long lasting immunity against viral epitopes that are highly conserved, such as with the nucleoprotein of viruses. Injecting plasmids encoding specific proteins by the present invention results in increased immune responses, as measured by antibody production.
  • the present invention includes new methods of providing nucleic acid vaccines by delivering a formulated nucleic acid molecule with a needle-free device as described herein.
  • nucleic acid vaccines are enhanced by one of at least three methods: (1) the use of delivery systems to increase the stability and distribution of plasmid within the muscle, (2) by the expression (or delivery) of molecules to stimulate antigen presentation transfer, or (3) by the use of adjuvants that may modulate the immune response.
  • the present invention provides polymeric and non-polymeric formulations which address problems associated with injection of nucleic acids suspended in saline. Plasmids suspended in saline have poor bioavailability in muscle due to rapid degradation of plasmid by extracellular nucleases.
  • One possible approach to overcome the poor bioavailability is to protect plasmid from rapid nuclease degradation by condensing the plasmid with commonly used cationic complexing agents.
  • the use of rigid condensed particles containing plasmid for efficient transfection of a larger number of muscle cells has not been successful to date.
  • nucleic acids Delivery and expression of nucleic acids is limited due to degradation of the nucleic acids by components of organisms, such as nucleases. Thus, protection of the nucleic acids when delivered in vivo can greatly enhance the resulting expression, thereby enhancing a desired pharmacological or therapeutic effect. It was found that certain types of compounds which interact with a nucleic acid (e.g., DNA) in solution but do not condense the nucleic acid provide in vivo protection to the nucleic acid, and correspondingly enhance the expression of an encoded gene product.
  • a detailed description of the formulations that can be used in the present invention can be found in PCT Application No. PCT/US96/05679 which is hereby incorporated as a reference in its entirety including any drawings. As noted above, preferably such formulations are delivered by using a needle free device as described herein.
  • Inert particles coated with DNA have been used to deliver genes to cells.
  • An advantage of coating inert particles, for instance gold beads, with nucleic acids is that the particle carrier actually penetrates the cell. Hence, the nucleic acid is delivered to the interior of the cell and should become incorporated and expressed in a more efficient manner.
  • the conventional procedure for preparation of DNA-coated gold particles results in heterogeneous distribution of DNA from particle to particle, which produces variable expression from cell to cell (Butow et al., Meth Enzymol. 264:265-278, 1996).
  • the present invention provides gold particles uniformly coated with DNA by covalently attaching cysteine terminated DNA binding peptides to the gold surface. Furthermore, the invention enhances the intracellular transport of DNA released from the gold particles by non-covalent association of high affinity DNA binding peptides, which contain nuclear localization sequences, with the plasmid.
  • the formulation provides the most reproducible amount of plasmid on gold particles, independent of the diameter of the particle. Preferably such modified gold particle formulations are delivered by using a needle-free device as described herein.
  • the present invention described herein can be utilized for the delivery and expression of many different coding sequences.
  • the demonstrated effectiveness for the PINC systems (PCT Application No. PCT/US96/05679) for delivery to muscle indicate that such formulations are effective for delivery of a large variety of coding sequences to muscle by needle free injection.
  • Specific suggestions for delivery of coding sequences to muscle with the needle free device of the present invention include those summarized in Table 1 below.
  • Table 1 Applications for Plasmid-Based Gene Therapy by Intramuscular Injection
  • PVP Formulated Nucleic Acid Molecules Concentrated pDNA stock solutions were made by lyophilizing and rehydrating pDNA with water to a final pDNA concentration of 3-5mg/ml. Formulations were made by aliquoting appropriate volumes of sterile stock solutions of pDNA, 5M NaCL, and polymer to obtain a final pDNA concentration in an isotonic polymer solution. Stock solutions were added in the following order: water, plasmid, polymer, and 5M NaCl. The plasmid and polymers were allowed to incubate at room temperature for 15 minutes prior to adding salt or lactose for ionicity adjustments.
  • Na-citrate buffers in 0.9% NaCl were added after incubating the plasmid and polymers for 15 minutes at room temperature.
  • the pH of all formulations was measured using.
  • an Accumet Model 15 pH Meter and the viscosity of all formulations was measured using a Programmable Rheometer Model DV-III.
  • Dynamic dialysis was used with various interactive polymer formulations to measure binding between PVP and plasmid DNA.
  • One ml of formulations and corresponding controls were place in prewashed dialysis sacs.
  • the dialysis sacs were closed and suspended in stirred saline solutions (100 ml) at 25°C.
  • stirred saline solutions 100 ml
  • One ml aliquots were taken from the acceptor compartment over time and replaced with fresh media.
  • the concentration of PVP in the diffused samples collected over time was measured spectroscopically at 220 nm.
  • Formulated plasmids were prepared as follows in a sterile manner in 2ml single dose vials.
  • Plasmid/Liposomes (DOTMA:DOPE 1:1 m m) 1:3 -/+ 20 g DNA ml
  • DNA formulations were injected by needle-free device into 50ml polypropylene tubes indicative of a "high impact” worst case scenario. Additionally the needle free device was set at maximum penetration as described in the product instruction manual.
  • DNA stability was determined by agarose gel electrophoresis of the samples before and after injection. Measurement of stability was determined by the amount of visible degradation, i.e. smearing, and by quantitating supercoiled and open circular DNA. Our data indicates that even in this extreme example the formulated DNA samples only exhibit slight degradation.
  • Example 3
  • Formulated nucleic acid molecules were prepared as follows in a sterile manner. 0.1 mg pCT0129/mL in DOTMA:Chol (1:1 m m) 1 :3 (-/+); 10%lactose
  • Measurement of stability was determined by the amount of visible degradation, i.e., smearing, and by quantitating supercoiled and open circular DNA. Our data indicates that even in this example which uses the most extreme injection parameters the nozzle size did not effect the state of the formulated DNA.
  • hGH Human Growth Hormone
  • the expression plasmid was injected via a needle free device or by a needle to examine if the magnitude of the immune response is affected by the injection method or formulation.
  • pCMV-hGH plasmid was suspended in saline or formulated with PVP and injected into biceps femoris and semitendinosus muscles of dogs using a needle free device or 22 gauge needle. Blood was collected before plasmid injection and once a week after the injection. Blood samples were kept overnight at 4° C, centrifuged at 2000 g for 15 min and serum was collected for detection of anti-hGH antibodies by ELISA. Significant levels of hGH antibodies were detectable in jet (aerosol) injected animals 14 days after a single i.m. dose of hGH plasmid. The levels of hGH antibodies increased over time reaching a plateau by 21 days and remained elevated throughout the 112 day pre-boost periods. Repeating the plasmid dose on day 112 augmented the antibody response by 100 fold.
  • hGH plasmid formulated with PVP did not produce significantly detectable levels of hGH antibodies.
  • a booster dose was required to achieve significant antibody response from needle injection. Peak antibody levels from aerosol injections were about 20 times higher than from needle injections.
  • Control animals received CMV-C AT plasmid formulated with PVP without the hGH gene, and the animals did not produce anti-hGH antibodies.
  • Needle-free injection of hGH plasmid suspended in saline also leads to the production of anti-hGH antibodies.
  • a single dose of naked DNA was sufficient to achieve significant levels of hGH antibodies when administered via the needle-free device.
  • multiple doses were required to elicit antibody response with needle injection.
  • the antibody response from needle-free injection was 5 times higher compared to needle injection.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Endocrinology (AREA)
  • Communicable Diseases (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Diabetes (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Infusion, Injection, And Reservoir Apparatuses (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
EP98964020A 1997-12-16 1998-12-16 Nadelloses injektion von formulierte nukleinsäure moleküle Withdrawn EP1038016A2 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US6975497P 1997-12-16 1997-12-16
US69754P 1997-12-16
PCT/US1998/026823 WO1999031262A2 (en) 1997-12-16 1998-12-16 Needle-free injection of formulated nucleic acid molecules

Publications (1)

Publication Number Publication Date
EP1038016A2 true EP1038016A2 (de) 2000-09-27

Family

ID=22091009

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98964020A Withdrawn EP1038016A2 (de) 1997-12-16 1998-12-16 Nadelloses injektion von formulierte nukleinsäure moleküle

Country Status (5)

Country Link
EP (1) EP1038016A2 (de)
JP (1) JP2003528024A (de)
AU (1) AU1922999A (de)
CA (1) CA2315256A1 (de)
WO (1) WO1999031262A2 (de)

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7393529B2 (en) 1998-04-09 2008-07-01 Idexx Laboratories, Inc. Methods and compositions for inhibiting binding of IgE to a high affinity receptor
KR100750695B1 (ko) * 1999-07-20 2007-08-22 파멕사 에이/에스 Gdf-8 활성을 하향-조절하는 방법
EP1196617A2 (de) 1999-07-21 2002-04-17 Immunoporation Ltd. Methode zum einführen einer substanz in eine zelle
US7196066B1 (en) * 1999-11-03 2007-03-27 Powderject Vaccines, Inc. DNA-vaccines based on constructs derived from the genomes of human and animal pathogens
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
US8273866B2 (en) 2002-02-20 2012-09-25 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SINA)
US6875748B2 (en) 2000-04-21 2005-04-05 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US20050148530A1 (en) 2002-02-20 2005-07-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
FR2835749B1 (fr) 2002-02-08 2006-04-14 Inst Nat Sante Rech Med Composition pharmaceutique ameliorant le transfert de gene in vivo
EP1432724A4 (de) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Durch rna-interferenz vermittelte inhibierung von map-kinase-genen
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7923547B2 (en) 2002-09-05 2011-04-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
CA2535799A1 (en) 2003-08-15 2005-03-03 University Of Florida Identification of porphyromonas gingivalis virulence polynucleotides for diagnosis, treatment, and monitoring of periodontal diseases
EP1671655A4 (de) * 2003-08-29 2009-08-12 Anges Mg Inc Gentherapie für hauterkrankungen mit nadelfreier spritze
EP2395112B1 (de) 2003-12-31 2017-02-15 The Penn State Research Foundation Verfahren zur Vorhersage und Überwindung des Widerstands auf eine Chemotherapie bei Eierstockkrebs und zur Vorhersage des Kolonkrebsvorkommens
JP2005335054A (ja) * 2004-04-27 2005-12-08 Japan Science & Technology Agency 金属ナノワイヤー及びその製造方法
US20060040882A1 (en) 2004-05-04 2006-02-23 Lishan Chen Compostions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
EP2298896A1 (de) 2004-06-22 2011-03-23 The Board of Trustees of the University of Illinois Verfahren zur Inhibierung von Tumorzellwachstum mit FOXM1 siRNS
CA2587950C (en) * 2004-12-01 2014-02-11 Wlt Distributors Inc. Needle-free injector
EP1973946B1 (de) 2006-01-20 2015-03-25 Cell Signaling Technology, Inc. Translokation und mutations-ros-kinase bei menschlichem nicht-kleinzelligem lungenkrebs
US20120208824A1 (en) 2006-01-20 2012-08-16 Cell Signaling Technology, Inc. ROS Kinase in Lung Cancer
HUE035732T2 (en) 2006-04-14 2018-05-28 Cell Signaling Technology Inc Gene defects and mutant ALK kinase in human solid tumors
CA2658550C (en) 2006-07-21 2018-06-19 Silence Therapeutics Ag Means for inhibiting the expression of protein kinase 3
US8188214B2 (en) 2007-02-28 2012-05-29 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Brachyury polypeptides and methods for use
EP2157982B1 (de) 2007-05-04 2014-12-17 Marina Biotech, Inc. Aminosäurelipide und ihre verwendungen
EP3741851A1 (de) 2007-10-18 2020-11-25 Cell Signaling Technology, Inc. Translokation und mutante ros-kinase in humanem nichtkleinzelligem lungenkarzinom
AU2009236270B2 (en) 2008-04-15 2014-06-26 Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Compositions and methods for delivering inhibitory oligonucleotides
JP2011521649A (ja) 2008-05-30 2011-07-28 イェール ユニバーシティー 遺伝子発現を改変するための標的化オリゴヌクレオチド組成物
EP2902013A1 (de) 2008-10-16 2015-08-05 Marina Biotech, Inc. Verfahren und Zusammensetzungen zur liposomalen und effizienten Abgabe von Gen-Silencing-Therapeutika
ES2637174T3 (es) 2009-02-12 2017-10-11 Cell Signaling Technology, Inc. Expresión de ROS mutante en el cáncer de hígado humano
JP2012523225A (ja) 2009-04-10 2012-10-04 アソシアシオン・アンスティテュ・ドゥ・ミオロジー 疾患の処置のためのトリシクロ−dnaアンチセンスオリゴヌクレオチド、組成物及び方法
US8710209B2 (en) 2009-12-09 2014-04-29 Nitto Denko Corporation Modulation of HSP47 expression
WO2011120023A1 (en) 2010-03-26 2011-09-29 Marina Biotech, Inc. Nucleic acid compounds for inhibiting survivin gene expression uses thereof
WO2011133584A2 (en) 2010-04-19 2011-10-27 Marina Biotech, Inc. Nucleic acid compounds for inhibiting hras gene expression and uses thereof
WO2011139842A2 (en) 2010-04-28 2011-11-10 Marina Biotech, Inc. Nucleic acid compounds for inhibiting fgfr3 gene expression and uses thereof
WO2011146938A1 (en) 2010-05-21 2011-11-24 NanoOncology, Inc. Reagents and methods for treating cancer
US9045755B2 (en) 2010-06-24 2015-06-02 Quark Pharmaceuticals, Inc. Double stranded RNA compounds to RhoA and use thereof
WO2012019132A2 (en) 2010-08-06 2012-02-09 Cell Signaling Technology, Inc. Anaplastic lymphoma kinase in kidney cancer
EP2609198B8 (de) 2010-08-24 2018-03-28 Sirna Therapeutics, Inc. Einsträngige rnai-mittel mit einem internen nukleinsäurefreien spacer
US20140134231A1 (en) 2010-10-11 2014-05-15 Sanford-Burnham Medical Research Institute Mir-211 expression and related pathways in human melanoma
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
AU2011323508B2 (en) 2010-11-01 2017-04-27 Peptimed, Inc. Compositions of a peptide targeting system for treating cancer
JP6000287B2 (ja) 2011-03-03 2016-09-28 クォーク ファーマシューティカルズ インコーポレーティッドQuark Pharmaceuticals,Inc. 肺疾患および損傷を治療するための組成物および方法
DE102011018499A1 (de) 2011-04-23 2012-10-25 Emc Microcollections Gmbh Topische Nanopartikel-Vakzine zur Immunstimulation der dendritischen Zellen in der Haut
EP2581448B1 (de) 2011-10-13 2015-01-28 Association Institut de Myologie Tricyclo-phosphorothioat-DNA
KR20140111673A (ko) 2012-01-12 2014-09-19 쿠아크 파마수티칼스 인코퍼레이티드 청력 및 균형 장애를 치료하기 위한 조합요법
PL2838998T3 (pl) 2012-04-18 2018-04-30 Cell Signaling Technology, Inc. EGFR i ROS1 w nowotworze
EP3453762B1 (de) 2012-05-02 2021-04-21 Sirna Therapeutics, Inc. Sina-zusammensetzungen
ES2704855T3 (es) 2012-09-12 2019-03-20 Quark Pharmaceuticals Inc Moléculas de oligonucleótido de doble cadena para p53 y métodos de uso de las mismas
US9932578B2 (en) 2012-09-12 2018-04-03 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to P53 and methods of use thereof
US9884886B2 (en) 2012-12-06 2018-02-06 Merck Sharp & Dohme Disulfide masked prodrug compositions and methods
WO2015020960A1 (en) 2013-08-09 2015-02-12 Novartis Ag Novel lncrna polynucleotides
AU2015307186A1 (en) 2014-08-27 2017-04-13 Peptimed, Inc. Anti-tumor compositions and methods
US20190255143A1 (en) 2016-04-18 2019-08-22 The Trustees Of Columbia University In The City Of New York Therapeutic targets involved in the progression of nonalcoholic steatohepatitis (nash)
WO2022149550A1 (ja) * 2021-01-05 2022-07-14 株式会社ダイセル 注入器

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1026253B2 (de) * 1989-03-21 2012-12-19 Vical Incorporated Expression von exogenen Polynukleotidsequenzen in Wirbeltieren
TW404844B (en) * 1993-04-08 2000-09-11 Oxford Biosciences Ltd Needleless syringe
EP0695186A1 (de) * 1993-04-28 1996-02-07 Ribozyme Pharmaceuticals, Inc. Verabreichung von nukleinsäuren an das auge
EP0800403A2 (de) * 1994-12-30 1997-10-15 Chiron Corporation Nichttraumatische erteilung von genbeförderungsmitteln
US5766901A (en) * 1995-05-04 1998-06-16 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and method for delivering a nucleotide into cell nuclei
EP0832269A1 (de) * 1995-06-07 1998-04-01 Baylor College Of Medicine Nukleinsäure transporter zur abgabe von nukleinsäure in zellen

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9931262A2 *

Also Published As

Publication number Publication date
JP2003528024A (ja) 2003-09-24
AU1922999A (en) 1999-07-05
CA2315256A1 (en) 1999-06-24
WO1999031262A3 (en) 1999-09-10
WO1999031262A2 (en) 1999-06-24

Similar Documents

Publication Publication Date Title
WO1999031262A2 (en) Needle-free injection of formulated nucleic acid molecules
Pardi et al. mRNA vaccines—a new era in vaccinology
US7491537B2 (en) Nucleic acid formulations for gene delivery and methods of use
Golombek et al. Intradermal delivery of synthetic mRNA using hollow microneedles for efficient and rapid production of exogenous proteins in skin
AU2001241958A1 (en) Improved poloxamer and poloxamine compositions for nucleic acid delivery
Dileo et al. Gene transfer to subdermal tissues via a new gene gun design
CZ475599A3 (cs) Použití nukleové kyseliny k výrobě léčiva pro genovou terapii, které se injikuje do příčně pruhovaného svalstva
US20020102729A1 (en) Formulations for electroporation
Stanton Current status of messenger RNA delivery systems
Somvanshi et al. Peptide-based DNA delivery system
Fang et al. Applications and challenges of biomaterial mediated mRNA delivery
CA2328180A1 (en) Needleless administration of polynucleotide formulations
Chamarthy et al. A cationic peptide consists of ornithine and histidine repeats augments gene transfer in dendritic cells
Manthorpe et al. Plasmid vaccines and therapeutics: from design to applications
Loehr et al. Immunization of livestock with DNA vaccines: current studies and future prospects
CN111603553A (zh) 一种靶向免疫系统治疗肿瘤的dna药物及其应用
WO2001008709A1 (en) Sonoporation of tumors
Xin et al. mRNA-Based Cancer Therapy and Challenges
Lin et al. An Overview of Nanoparticle-Based Delivery Platforms for Vaccines
Yoshikawa et al. Fusogenic liposomes and their suitability for gene delivery
WO2002020060A1 (en) Vp22 protein/nucleic acid aggregates, uses thereof
Teng Yap et al. Navigating the horizon of mRNA vaccines: Tracing their evolution, ensuring safety, and unveiling therapeutic potential.
Singh et al. The Junction of Biomaterials and Gene Therapy–Current Strategies and Future Directions
Sullivan Non-Viral Gene Therapy
Volc-Platzer et al. Nonviral Gene Transfer into the Skin

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000714

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20030701