US20040137071A1 - Nanocapsule encapsulation system and method - Google Patents

Nanocapsule encapsulation system and method Download PDF

Info

Publication number
US20040137071A1
US20040137071A1 US10/652,814 US65281403A US2004137071A1 US 20040137071 A1 US20040137071 A1 US 20040137071A1 US 65281403 A US65281403 A US 65281403A US 2004137071 A1 US2004137071 A1 US 2004137071A1
Authority
US
United States
Prior art keywords
composition
poly
surfactant
gly
thr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/652,814
Inventor
Gretchen Unger
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.)
GeneSegues Inc
Original Assignee
GeneSegues 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 GeneSegues Inc filed Critical GeneSegues Inc
Priority to US10/652,814 priority Critical patent/US20040137071A1/en
Publication of US20040137071A1 publication Critical patent/US20040137071A1/en
Assigned to GENESEGUES, INC. reassignment GENESEGUES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UNGER, GRETCHEN M
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J13/00Colloid chemistry, e.g. the production of colloidal materials or their solutions, not otherwise provided for; Making microcapsules or microballoons
    • B01J13/02Making microcapsules or microballoons
    • B01J13/06Making microcapsules or microballoons by phase separation
    • B01J13/08Simple coacervation, i.e. addition of highly hydrophilic material
    • 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
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • A61K9/1075Microemulsions or submicron emulsions; Preconcentrates or solids thereof; Micelles, e.g. made of phospholipids or block copolymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5138Organic macromolecular compounds; Dendrimers obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J13/00Colloid chemistry, e.g. the production of colloidal materials or their solutions, not otherwise provided for; Making microcapsules or microballoons
    • B01J13/02Making microcapsules or microballoons
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/773Nanoparticle, i.e. structure having three dimensions of 100 nm or less
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/788Of specified organic or carbon-based composition
    • Y10S977/795Composed of biological material
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/84Manufacture, treatment, or detection of nanostructure
    • Y10S977/895Manufacture, treatment, or detection of nanostructure having step or means utilizing chemical property
    • Y10S977/896Chemical synthesis, e.g. chemical bonding or breaking
    • Y10S977/897Polymerization
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/915Therapeutic or pharmaceutical composition
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/92Detection of biochemical

Definitions

  • the present invention generally relates to a field of controlled-release delivery systems for macromolecules, particularly those for nucleic acids and gene therapy. More specifically, the present invention relates to nanocapsules having a diameter of less than about 50 nanometers, in which a bioactive component is located in a core of the nanocapsule, and to methods of forming these nanocapsules.
  • the organic solvent when included in the manufacturing process for nanoparticles, the organic solvent may denature the bioactive agent which reduces most, if not all, efficacy of the bioactive agent.
  • denaturation of the bioactive agent may promote a toxic response upon administration of the nanoparticle, to a human subject, for example.
  • organic solvent when used to prepare nanoparticles, the organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore
  • organic solvents are typically removed during the manufacturing process of nanoparticles.
  • inclusion of one or more organic solvent removal techniques generally increases the costs and complexity of forming nanoparticles.
  • Entangling or embedding the bioactive agent in the polymeric matrix of the nanoparticle may also reduce the efficacy of the bioactive agent by permitting premature release of the bioactive agent prior to reaching a target cell. Premature release of the bioactive agent typically promotes cytotoxicity or cell death during administration of the nanoparticle.
  • nanoparticles that reach the target cell are typically transported into the target cell via endosomal regulated pathways that results in lysosomal degradation of the bioactive agent and the nanoparticle. Therefore, functional activity of the bioactive agent inside the target cell may not occur since the bioactive agent and the nanoparticle undergoes degradation.
  • the term “functional activity” refers to an ability of a bioactive agent to function within a target cell for purposes of providing a therapeutic effect on the target cell.
  • the present invention generally relates to nanocapsules and methods of preparing these nanocapsules.
  • the present invention includes a method of forming a surfactant micelle and dispersing the surfactant micelle into an aqueous composition having a hydrophilic polymer to form a stabilized dispersion of surfactant micelles.
  • the method further includes mechanically forming droplets of the stabilized dispersion of surfactant micelles, precipitating the hydrophilic polymer to form precipitated nanocapsules, incubating the nanocapsules to reduce a diameter of the nanocapsules, and filtering or centrifuging the nanocapsules.
  • FIG. 1 is a schematic of a method of the present invention for preparing nanocapsules.
  • FIG. 1A illustrates atomic force microscopy of nanocapsule formulations prepared under different dispersion conditions.
  • FIG. 1B illustrates results from an experiment documenting quantitative recovery of small amounts of DNA from releasing solutions.
  • FIG. 1C illustrates cumulative release over 72 hours for nanocapsules prepared under different dispersion conditions.
  • FIG. 2 illustrates relative pinocytotic activity of HacaT keratinocyte cultures treated with DNA complexes, nanocapsules containing DNA or no treatment.
  • FIG. 3 illustrates the results of western blotting of total protein from rat fibroblast cultures.
  • FIG. 4A illustrates
  • FIG. 4B illustrates immunofluorescence microscopy of porcine dermal tissue sections from the experiment of FIG. 4.
  • FIG. 5 shows incorporation of nanocapsules into a solid dosage form.
  • FIG. 6A illustrates polyvinylpyrolidone nanocapsule uptake and Green Fluorescent Protein (GFP) expression in 35 mm human dermal fibroblast and immortalized keratinocyte cultures.
  • GFP Green Fluorescent Protein
  • FIG. 6B illustrates tumor targeting of GFP plasmid DNA by Tenascin nanocapsules.
  • FIG. 6C illustrates an effect of nanocapsules that are coated with Tenascin and nanocapsules that are not coated with Tenascin on growth inhibition of squamous cell carcinoma and human dermal fibroblast (HDF) cultures.
  • FIG. 7 shows uptake of HDF cultures treated with nanocapsules containing 20 mer Fitc-labeled O-methyl RNA oligonucleotides.
  • the present invention generally relates to nanocapsules having a diameter of less than about 50 nanometers (nm).
  • the present invention also relates to a method of preparing these nanocapsules.
  • a nanocapsule is formed by partitioning a bioactive component within a core of surfactant molecules, and surrounding the surfactant molecules with a biocompatible polymer shell.
  • a method for producing the nanocapsule is generally depicted at 10 in FIG. 1.
  • a bioactive component 12 is homogeneously dispersed into a first aqueous composition 14 to form a hydrophilic composition (not shown).
  • a surfactant composition 16 including a surfactant component (not shown) that contains a plurality of surfactant molecules, and an optional biocompatible oil component 18 , are introduced into a first dispersing apparatus 20 along with the hydrophilic composition.
  • the surfactant composition 16 is subjected to conditions in the first dispersing apparatus 20 that initiate at least partial adsorption of the surfactant molecules onto a surface of the bioactive component 12 .
  • Partial adsorption of surfactant molecules onto the surface of the bioactive component 12 initiates partitioning of the bioactive component 12 into a core of a shell formed from the surfactant molecules in the first aqueous composition 14 .
  • Adsorption of the surfactant molecules onto the surface of the bioactive component 12 may proceed until an entire surface of the bioactive component 12 is covered by the surfactant molecules to complete partitioning of the bioactive component 12 into the core of surfactant molecules and form a surfactant micelle 22 .
  • a biocompatible polymer component 24 is added to the surfactant micelle 22 to stabilize the surfactant micelle 22 located in the first aqueous composition 14 .
  • the biocompatible polymer component 24 surrounds the surfactant micelle 22 in a stabilizing apparatus 26 to form a stabilized surfactant micelle 28 .
  • the stabilized surfactant micelle 28 is transferred from the stabilizing apparatus 26 into a second aqueous composition 30 located in a second dispersing apparatus 32 .
  • the second aqueous composition 30 includes a solute (not shown) that is capable of precipitating the biocompatible polymer component 24 that coats the stabilized surfactant micelle 28 .
  • dispersed, optionally atomized precipitated nanocapsules 36 are formed.
  • a surfactant composition that includes a surfactant component having a hydrophile-lipophile-balance (HLB) value of less than about 6.0 units
  • HLB hydrophile-lipophile-balance
  • stabilizing the surfactant micelles by adding a biocompatible polymer coats the surfactant micelles to form nanocapsules having a diameter of less than about 50 nm.
  • nanoparticle refers a particle having a matrix-type structure with a size of less than about 1,000 nanometers.
  • the bioactive component is entangled or embedded in the matrix-type structure of the nanoparticle.
  • nanosphere refers to a particle having a solid spherical-type structure with a size of less than about 1,000 nanometers.
  • the bioactive component is adsorbed onto the surface of the nanosphere or embedded in the nanosphere.
  • nanocore refers to a particle having a solid core with a size of less than about 1,000 nanometers.
  • the nanocore includes a bioactive component, the bioactive component is entangled in the nanocore.
  • nanocapsule refers to a particle having a hollow core that is surrounded by a shell, such that the particle has a size of less than about 1,000 nanometers.
  • a nanocapsule includes a bioactive component
  • the bioactive component is located in the core that is surrounded by the shell of the nanocapsule.
  • nanocapsule is not meant to encompass, and generally does not include, a particle having a size of less than about 1,000 nanometers, in which a bioactive component is entangled or embedded in the matrix of the nanocapsule or adsorbed onto the surrounding shell of the nanocapsule.
  • the bioactive component 12 may be included into the first aqueous composition 14 as a liquid, vapor or in granular form.
  • the form of the bioactive component 12 that is selected preferably permits the bioactive component 12 to (1) remain stable prior to dissolving or dispersing into the first aqueous composition 14 , (2) be homogeneously dispersed into the first aqueous composition 14 , (3) be optionally condensed to reduce a size of the bioactive component 12 , (4) be partitioned into the core of the surfactant micelles 22 , (5) be released upon degradation of the biocompatible polymer shell 24 of the nanocapsule 36 , and (6) be functionally active upon release from the nanocapsule 36 .
  • the bioactive component 12 may be characterized as “hydrophilic” or “hydrophobic”.
  • the term “hydrophilic” and “hydrophilicity” refers to an ability of a molecule to adsorb water or form one or more hydrogen-bond(s) with water. All references to “hydrophilic” is also understood as encompassing any portion of the molecule that is capable of adsorbing water or forming one or more hydrogen-bond(s) with water.
  • the term “hydrophobic” and “hydrophobicity” refers to an ability of a molecule to not adsorb water nor form one or more hydrogen-bond(s) with water. All references to “hydrophobic” is also understood as encompassing any portion of the molecule that is not capable of adsorbing water nor forming one or more hydrogen-bond(s) with water.
  • the hydrophilic bioactive component 12 is a hydrophilic bioactive component
  • the hydrophilic bioactive component maybe directly added to the first aqueous composition 14 .
  • the hydrophilic bioactive component 12 may be optionally dissolved or dispersed in one or more solvents, such as water, a nonpolar solvent, a polar solvent, or any combination of any of these.
  • nonpolar solvent refers to a solvent that does not have a permanent electric dipole moment, and therefore has no ability for an intramolecular association with a polar solvent. Additionally, a nonpolar solvent may be characterized as a solvent that includes molecules having a dielectric constant of less than about 20 units. Similarly, the term “immiscible”, as used herein, refers to an inability of two or more substances, such as two or more liquids, solids, vapors, or any combination of any of these, to form an intramolecular association with another substance. Some non-exhaustive examples of nonpolar solvents may be found in Perry's Chemical Engineer's Handbook, Sixth Edition, which is incorporated herein by reference.
  • polar solvent refers to a solvent that has a permanent electrical dipole moment, and therefore has an ability to form an intramolecular association with another polar substance, such as a liquid, a solid, a vapor or any combination of any of these. Additionally, a polar solvent may be characterized as a solvent that includes molecules having a dielectric constant of more than about 20 units. Likewise, the term “miscible”, as used herein, refers to an ability of two or more substances to form an intramolecular association with each other. Some non-exhaustive examples of polar solvents may be found in Perry's Chemical Engineer's Handbook, Sixth Edition, which has been incorporated herein by reference.
  • the hydrophobic bioactive component may be dispersed or dissolved in a solvent that is capable of dispersing or dissolving the hydrophobic molecule, such as the above-mentioned water, a nonpolar solvent, a polar solvent, or any combination of any of these.
  • the hydrophobic bioactive component 12 is dissolved or dispersed in a water-miscible solvent, such as, acetone, acetonitrile, ethanol, dimethyl acetamide (DMA), tetrahydrofuran (THF), dioxane, dimethylsulfoxide (DMSO), and dimethylformamide (DMF).
  • a water-miscible solvent such as, acetone, acetonitrile, ethanol, dimethyl acetamide (DMA), tetrahydrofuran (THF), dioxane, dimethylsulfoxide (DMSO), and dimethylformamide (DMF).
  • a water-miscible solvent such as, acetone, acetonitrile, ethanol, dimethyl acetamide (DMA), tetrahydrofuran (THF), dioxane, dimethylsulfoxide (DMSO), and dimethylformamide (DMF).
  • DMA dimethyl acetamide
  • THF
  • the bioactive component 12 may be optionally condensed in the first aqueous composition 14 prior to forming the surfactant micelle 16 .
  • the bioactive component is a polynucleotide
  • the polynucleotide may be condensed using a DNA-condensing agent.
  • a “DNA-Condensing Agent” is a molecule that facilitates condensation or a size reduction of DNA.
  • condensation of the bioactive component 12 is not critical to the present invention, condensation of the bioactive component 12 maybe practiced to reduce the size of the bioactive component 12 .
  • Condensation of the bioactive component 12 generally reduces the size of the bioactive component 12 prior to partitioning into the core of the surfactant micelle 16 . Reducing the size of the bioactive component 12 may permit maximum incorporation of the bioactive component 12 into the surfactant micelle 22 or may assist a reduction in the overall size of the nanocapsule 36 .
  • Increasing the amount of the bioactive component 12 that may be included as part of the nanocapsule 36 permits incorporation of macromolecules having a large number of monomers, such as a large number of base pairs or amino acids, for example.
  • the bioactive component 12 may further include additional components that are compatible with, and that do not interfere with solvation or dispersion of the bioactive component 12 .
  • additional components that may be added to the bioactive component 12 include a DNA-associating moiety, which refers to a molecule, or portions thereof, that interact in a non-covalent fashion with nucleic acids.
  • DNA-associating moieties may include, but are not limited to, a major-and minor-groove binder, a DNA intercalator, a polycation, a DNA-masking component, a membrane-permeabilizing component, a subcellular-localization component, or the like.
  • Major- and minor-groove binders as used herein, are molecules thought to interact with DNA by associating with the major or minor groove of double-stranded DNA.
  • DNA intercalator refers to a planar molecule or planar portion of a molecule thought to intercalate into DNA by inserting themselves between, and parallel to, a nucleotide base pair.
  • a “polycation” is thought to associate with the negative charges on the DNA backbone.
  • the DNA-associating moiety may be covalently linked through a “reactive group” to a functional component of this invention. The reactive group is easily reacted with a nucleophile on the functional component.
  • reactive groups include, but are not limited to N-hydroxysuccinimide (e.g., amine), maleimide and maleimidophenyl (e.g., sulfhydryl), pyridyl disulfide (e.g., sulfhydryl), hydrazide (e.g., carbohydrate), and phenylglyoxal (e.g., arginine).
  • N-hydroxysuccinimide e.g., amine
  • maleimide and maleimidophenyl e.g., sulfhydryl
  • pyridyl disulfide e.g., sulfhydryl
  • hydrazide e.g., carbohydrate
  • phenylglyoxal e.g., arginine
  • DNA-masking component refers to a molecule capable of masking all or part of a polynucleotide following release from a nanocapsule to increase its circulatory half-life by inhibiting attack by degrading reagents, such as nucleases, present in the circulation and/or interfering with uptake by the reticuloendothelial system.
  • degrading reagents such as nucleases
  • membrane-permeabilizing component refers to any component that aids in the passage of a polynucleotide or encapsulated polynucleotide across a membrane.
  • membrane permeabilizing component encompasses in part a charge-neutralizing component, usually a polycation, that neutralizes the large negative charge on a polynucleotide, and enables the polynucleotide to traverse the hydrophobic interior of a membrane.
  • membrane-permeabilizers Many charge-neutralizing components can act as membrane-permeabilizers. Membrane-permeabilization may also arise from amphipathic molecules.
  • a “membrane permeabilizer”, as used herein, is a molecule that can assist a normally impermeable molecule to traverse a cellular membrane and gain entrance to the cytoplasm of the cell.
  • the membrane permeabilizer may be a peptide, bile salt, glycolipid, phospholipid or detergent molecule.
  • Membrane permeabilizers often have amphipathic properties such that one portion is hydrophobic and another is hydrophilic, permitting them to interact with membranes.
  • subcellular-localization component refers to a molecule capable of recognizing a subcellular component in a targeted cell. Recognized subcellular components include the nucleus, ribosomes, mitochondria, and chloroplasts. Particular subcellular-localization components include the “nuclear-localization components” that aid in carrying molecules into the nucleus and are known to include the nuclear localization peptides and amino acid sequences.
  • the bioactive component 12 may be included at an amount that is therapeutically effective to transform a plurality of cells, such as in vitro, in vivo or ex vivo cells.
  • transformation refers to a presence and/or functional activity of the bioactive component in the plurality of cells after exposing the nanocapsules to the plurality of cells.
  • the amount of the bioactive component 12 may vary depending upon the bioactive component 12 , the temperature, pH, osmolarity, any solutes, any additional component or optional solvents present in the second aqueous component 30 , the surfactant composition 16 , a type or an amount of the surfactant micelle 22 , the biocompatible polymer component 24 , any desired characteristics of the stabilized surfactant micelle 28 , any desired characteristics of the nanocapsules 36 , or any combination of any of these.
  • the bioactive component 12 of the nanocapsule 36 maybe supplied as an individual macromolecule or supplied in various prepared mixtures of two or more macromolecules that are subsequently combined to form the bioactive component 12 .
  • Some non-exhaustive examples of hydrophilic macromolecules that may be suitable for inclusion as part of the bioactive component 12 include, but are not limited to polynucleotides, polypeptides, genetic material, peptide nucleic acids, aptamers, carbohydrates, mini-chromosomes, molecular polymers, aggregates or associations of an inorganic or organic nature, genes, any other hydrophilic macromolecule or any combination of any of these.
  • hydrophobic macromolecules that may be included part of the bioactive component 12 include, but are not limited to, adregergic, adrenocotical steroid, adrenocortical suppressant, aldosterone antagonist, and anabolic agents; analeptic, analgesic, anesthetic, anorectic, and anti-acne agents; anti-adrenergic, anti-allergic, anti-amebic, anti-anemic, and anti-anginal agents; anti-arthritic, anti-asthmatic, anti-atherosclerotic, antibacterial, and anticholinergic agents; anticoagulant, anticonvulsant, antidepressant, antidiabetic, and antidiarrheal agents; antidiuretic, anti-emetic, anti-epileptic, antifibrionlytic, and antifungal agent; antihemorrhagic, inflammatory, antimicrobial, antimigraine, and
  • polypeptide refers to a polymer of amino acids not limited by the number of amino acids. It is also to be understood that the term “polypeptide” is meant to encompass an oligopeptide, a peptide, or a protein, for example.
  • polynucleotide refers to RNA or DNA sequences of more than 1 nucleotide in either single chain, duplex or multiple chain form.
  • polynucleotide is also meant to encompass polydeoxyribonucleotides containing 2′-deoxy-D-ribose or modified forms thereof, RNA and any other type of polynucleotide which is an N-glycoside or C-glycoside of a purine or pyrimidine base, or modified purine or pyrimidine base or basic nucleotide.
  • the polynucleotide may encode promoter regions, operator regions, structural regions, termination regions, combinations thereof or any other genetically relevant material.
  • the term “genetic” as used herein refers to any material capable of modifying gene expression.
  • the first aqueous composition 14 may be included in the method of the present invention as a gel, liquid, or in vapor form.
  • the form of the first aqueous composition 14 that is selected preferably permits the first aqueous composition 14 to (1) remain stable prior to dissolving or dispersing the bioactive component, the surfactant composition 16 , the surfactant micelle 22 , or optionally the stabilizer surfactant micelle 28 , (2) homogeneously disperse the bioactive component 12 , the surfactant composition 16 , the surfactant micelle 22 , or optionally the stabilizer surfactant 28 , (3) function as a continuous phase in an oil-in-water emulsion, (4) not interfere with, or mask the functional activity of the bioactive component 12 , and (5) not modify or degrade the bioactive component 12 , the surfactant composition 16 , the surfactant micelle 22 , or optionally the stabilized surfactant micelle 28 .
  • the first aqueous composition 14 may include only water, or may optionally include additional solutes or solvents that do not interfere with the method of forming the nanocapsules 36 nor mask the functional activity of the bioactive component 12 .
  • an amount of the first aqueous composition 14 used to prepare the nanocapsules 36 may vary depending upon the bioactive component 12 , the surfactant composition 16 , the temperature, pH, osmolarity, optional solutes or optional solvents, the surfactant micelle 22 , the biocompatible polymer component 24 , any desired characteristics of the stabilized surfactant micelle 28 or the nanocapsules 36 .
  • the bioactive component 12 may be added to the first aqueous composition 14 or the first aqueous composition 14 may be added to the bioactive component 12 . While the order of addition of the bioactive component 12 and the first aqueous composition 14 is not critical to the present invention, the hydrophilic composition (not shown) that is formed when the bioactive component 12 is dissolved or dispersed in the first aqueous composition 14 is preferably capable of maintaining a homogeneous solution or dispersion in the hydrophilic composition.
  • the first aqueous composition 14 may be supplied as an individual component or supplied in various prepared mixtures of two or more components that are subsequently combined to form the first aqueous composition 14 .
  • Some non-exhaustive examples of the first aqueous composition 14 include, but are not limited to, the above-mentioned water, nonpolar solvents, polar solvents, or any combination of any of these.
  • water is the first aqueous composition 14 .
  • the surfactant composition 16 maybe introduced into the bioactive component 12 , the first aqueous composition 14 , the hydrophilic composition as a liquid, vapor or in granular form.
  • the form of the surfactant composition 16 that is selected preferably permits the surfactant composition 16 to (1) remain stable prior to introducing into the bioactive component 12 , the first aqueous composition 14 , or the hydrophilic composition, (2) be homogeneously dispersed into the bioactive component 12 , the first aqueous composition 14 , or the hydrophilic composition, (3) form a micellar structure, (4) be adsorbed onto a surface of the bioactive component 12 , the first aqueous composition 14 , the hydrophilic composition (5) displace the first aqueous composition that is located on the surface of the bioactive component 12 , (6) partition the bioactive component 12 or the hydrophilic composition into a core of the micellar structure to form the surfactant micelle 22 , and (7) provide a thermodynamic driving force that is effective to reduce
  • a “surfactant” refers to any molecule containing a polar portion that thermodynamically prefers to be solvated by a polar solvent, and a hydrocarbon portion that thermodynamically prefers to be solvated by a non-polar solvent.
  • surfactant is also meant to encompass anionic, cationic, or non-ionic surfactants.
  • anionic surfactant refers to a surfactant with a polar portion that ionizes to form an anion in aqueous solution.
  • a “cationic surfactant” refers to a surfactant having a cationic polar portion that ionizes to form a cation in aqueous solution.
  • a “non-ionic” surfactant refers to a surfactant having a polar portion that does not ionize in aqueous solution.
  • a surfactant refers to a molecule that is effective to reduce a surface or an interfacial tension between a first substance dispersed in a second substance such that the first substance is solvated and any molecular groups of the first substance are dispersed.
  • a hydrodynamic diameter of the first substance increases after addition of the surfactant.
  • the surfactant composition 16 is believed to be effective to reduce the size or diameter of the surfactant micelles 22 in the first aqueous composition 14 , to thereby reduce the size of the nanocapsule 36 when practicing the present invention.
  • the surfactant composition 16 may include the surfactant component only (not shown), or may optionally include the biocompatible oil component 18 .
  • the surfactant component may be characterized on the HLB scale that ranges from less than about 1 to more than about 13 units.
  • a surfactant component having an HLB value of less than about 6.0 units may be described as being poorly, or not dispersable in an aqueous or water-based composition.
  • a surfactant component having an HLB value of less than about 6.0 units may be characterized as a hydrophobic or non-ionic surfactant.
  • a surfactant component having an HLB value of more than about 7.0 units may be described as being capable of forming a milky to translucent to clear dispersion when the surfactant having an HLB value of more than about 7.0 units is dispersed in an aqueous or water-based composition.
  • the surfactant component of the surfactant composition 16 has an HLB value of less than about 6.0 units when practicing the method of the present invention. Still more preferably the surfactant component of the surfactant composition 16 has an HLB value of less than about 5.0 units to facilitate preparation of nanocapsules having a diameter of less than about 50 nm.
  • the surfactant component may also be characterized in terms of a critical micelle concentration (CMC) value.
  • CMC critical micelle concentration
  • the surfactant component of the surfactant composition 16 has a CMC value of less than about 300 micromolars ( ⁇ m). Still more preferably, the surfactant component has a CMC value of less than about 200 ⁇ m.
  • the surfactant component of the surfactant composition 16 adsorbs onto the surface of the bioactive component 12 when introduced into the first aqueous composition 14 to minimize exposure of a surface of the hydrophobic surfactant component to a thermodynamically unfavorable environment created by the first aqueous composition 14 . Therefore, the surfactant component adsorbs onto the surface of the bioactive component to reduce the surface area of the surfactant component that may be exposed to the first aqueous composition 14 . Adsorption of the surfactant component onto the bioactive component 12 is believed to facilitate the size reduction of the bioactive component 12 and/or the surfactant micelle 22 .
  • the surfactant component of the surfactant composition 16 may be supplied as individual surfactants or supplied in various prepared mixtures of two or more surfactants that are subsequently combined to form the surfactant composition 16.
  • suitable surfactants having an HLB value of less than about 6.0 units or a CMC value of less than about 200 ⁇ m be listed in Dermatological Formulations (Barry, B., Marcel Dekker, (1983)), or in Percutaneous absorption: drug, cosmetics, mechanisms, methodology, 3 rd ed., Bronough, R. ed., 1999, or the Handbook of Industrial Surfactants (Ash, M., Ed., Gower Pub. (1993), which are incorporated herein by reference.
  • the surfactant component maybe 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol(TM-diol), blends of 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol(TM-diol), molecules having one or more acetylenic diol groups, cetyl alcohol or any combination of any of these.
  • the optional biocompatible oil component 18 of the surfactant composition 16 may be combined with the surfactant component as a liquid, vapor or in granular form.
  • the form of the optional biocompatible oil component 18 that is selected preferably permits the optional biocompatible oil component 18 to (1) remain stable prior to introduction into the surfactant composition 16 , (2) be homogeneously blended into the surfactant composition 16 , (3) dissolve or disperse the surfactant component, and (4) increase the hydrophobicity of the surfactant composition 16 , and therefore, the degree to which the size of the bioactive component 12 , the surfactant micelle 22 , the stabilizer surfactant micelle 28 , or the nanocapsule 36 may be reduced when practicing the present invention.
  • the concentration of the optional biocompatible oil component 18 in the surfactant composition 16 ranges from about 10 ⁇ 7 weight percent to about 10 weight percent, based upon a total volume of the stabilized surfactant micelles 28 in the first aqueous composition 14 .
  • Concentrations of the optional biocompatible oil component 18 higher than about 10 weight percent, based upon the total volume of the surfactant composition 18 may be less desirable because such higher concentrations increase a phase volume of the biocompatible oil, and consequently may cause difficulties in preparing, dispersing and/or handling the surfactant micelles 22 , the stabilized surfactant micelles 28 or the nanocapsules 36 .
  • Concentrations of the optional biocompatible oil component lower than about 10 ⁇ 7 weight percent in the surfactant composition 16 may be less preferred, because such lower concentrations would not be effective to solvate the surfactant component, or increase the hydrophobicity of the surfactant composition 16 , and may ultimately increase the diameter of the nanocapsules 36 .
  • the optional biocompatible oil component 18 of the surfactant composition 16 may be supplied as an individual biocompatible oil or supplied in various prepared mixtures of two or more biocompatible oils that are subsequently combined to form the optional biocompatible oil component 18 .
  • suitable biocompatible oils that may be included as part of the biocompatible oil component 18 may be found in Dermatological Formulations (Barry, B., Marcel Dekker, (1983)), or in Percutaneous absorption: drug, cosmetics, mechanisms, methodology, 3 rd ed., Bronough, R. ed. , 1999, or in the Handbook of Industrial Surfactants (Ash, M., Ed., Gower Pub. (1993), which have been incorporated herein by reference.
  • food or USP grade oils such as DMSO, DMF, castor oil, or any combination thereof, are used to practice the present method.
  • the surfactant composition 16 may be included at an amount that is effective to form the micellar structure that partitions the bioactive component 12 , the first aqueous composition 14 or the hydrophilic composition into the core of the micellar structure when forming the surfactant micelle 22 . Still more preferably, the surfactant composition 16 is included at an amount that is effective to provide a maximum thermodynamic driving force that minimizes the size of the bioactive component 12 , the surfactant micelle 22 , and ultimately, the size of the nanocapsule 36 when practicing the present invention.
  • the amount of the surfactant composition 16 may be varied based upon the bioactive component 12 , the first aqueous composition 14 , a ratio of the surfactant component to the optional biocompatible oil 18 , any desired characteristics of the surfactant micelles 22 , the stabilized surfactant micelles 28 or the nanocapsules 36 .
  • a surfactant composition containing a surfactant component having an HLB value of about 6.0 units mixed with a nonpolar biocompatible oil like castor oil may provide the same degree of a thermodynamic driving force as a second surfactant composition containing a surfactant component of about 4.0 units mixed with DMSO.
  • the amount of the surfactant composition 16 may range up to about 0.5 weight percent, based upon a total volume of the stabilized surfactant micelles dispersed in the first aqueous composition 14 . Still more preferably, the amount of the surfactant composition 16 is less than about 0.25 weight percent, based upon the total volume of the stabilized surfactant micelles 28 dispersed in the first aqueous composition 14 . Most preferably, the surfactant composition 16 is present at an amount of less than about 0.05 weight percent, based upon the total volume of the stabilized surfactant micelles 28 dispersed in the first aqueous composition 14 .
  • the surfactant composition 16 may be added to the total volume of the hydrophilic composition at a concentration of about 500 ppm, based on the total volume of the stabilized surfactant micelles 28 in the first aqueous composition 14 .
  • the first dispersing apparatus 20 initiates and promotes formation of the micellar structures that are based on the bioactive component 12 , the first aqueous composition 14 and the surfactant composition 16 .
  • Adsorption of surfactant component onto the surface of the bioactive component 12 , or hydrophilic composition continues until all of the surfactant molecules cover, and therefore, entrap the bioactive component 12 or hydrophilic composition in the core of the micellar structure to form surfactant micelles 22 .
  • Formation of a plurality of surfactant micelles 22 in the first aqueous composition 14 forms a dispersion of surfactant micelles 22 .
  • any conventional dispersing apparatus 20 that is capable of homogenously blending or dispersing may be suitable for use in forming the dispersion of surfactant micelles in accordance with the present invention.
  • the first dispersing apparatus 20 may vary depending upon the desired characteristics of the nanocapsules 36 .
  • the first dispersing apparatus 20 may include any device, such as a sonicating or a vortexing apparatus (not shown), or the like to disperse the bioactive component 12 in the hydrophilic composition, and the formation of the surfactant micelles 22 after addition of the surfactant composition 16 .
  • the first dispersing apparatus 20 may include a sonicating or a vortexing apparatus, the sonicating or the vortexing apparatus is not critical when practicing the method of the present invention.
  • a “surfactant micelle” may be characterized as a close packed mono-molecular barrier of surfactant molecules at an interface between the bioactive composition 12 and the surfactant composition 16 , such that the barrier encapsulates the bioactive component 12 , the first aqueous composition 14 or the hydrophilic composition. It is also to be understood that the term “surfactant micelle” encompasses partial or hemi-surfactant micelles that partially enclose the bioactive component 12 , the first aqueous composition 14 or the hydrophilic composition.
  • the bioactive component 12 is a hydrophilic bioactive component
  • the polar portion of the surfactant molecule associates with a surface of the hydrophilic bioactive component.
  • the bioactive component 12 is a hydrophobic bioactive component
  • the hydrocarbon portion of the surfactant micelle associates with a surface of the hydrophobic bioactive component.
  • surfactant micelle typically occurs at a well defined concentration known as the critical micelle concentration.
  • surfactant components having a CMC value of less than about 200 micromolars are preferred when practicing the present invention.
  • the dispersion of surfactant micelles 22 is transferred into the stabilizing apparatus 26 where a biocompatible polymer component 24 is added to stabilize the dispersion of surfactant micelles 22 .
  • the biocompatible polymer component 24 may be added to the dispersion of surfactant micelles 22 in the first dispersing apparatus 20 which obviates the need for the stabilizing apparatus 26 .
  • the biocompatible polymer component 24 stabilizes the dispersion of surfactant micelles 22 to form stabilized surfactant micelles 28 within the first aqueous composition 14 . Therefore, a dispersion of stabilized surfactant micelles 28 are present within the first aqueous composition 14 after addition of the biocompatible polymer component 24 .
  • biocompatible refers to a material that is capable of interacting with a biological system without causing cytotoxicity, undesired protein or nucleic acid modification or activation of an immune response.
  • the biocompatible polymer component 24 may be introduced into the dispersion of surfactant micelles 22 as a liquid, vapor or in granular form.
  • the form of the biocompatible polymer component 24 that is selected preferably permits the biocompatible polymer component 24 to (1) remain stable prior to addition into the dispersion of surfactant micelles 22 , (2) be homogeneously dispersed into the dispersion of surfactant micelles 22 , (3) increase a viscosity of the first aqueous composition 14 , (4) form a boundary layer at an interface of the surfactant micelle 22 and the first aqueous composition 14 , (5) be absorbed onto a surface of the surfactant micelles 22 , (6) be capable of iontophoretic exchange, (7) be capable of being precipitated upon addition of a solute, (8) be capable of enzymatic degradation, surface and/or bulk erosion, (9) not interfere with or mask the functional activity of the bioactive component 12 , (10) prevent aggregation and/or agglomeration of
  • the biocompatible polymer component 24 may be included at an amount that is effective to coat and therefore stabilize the surfactant micelle 22 .
  • the amount of the biocompatible polymer component 24 used to stabilize the surfactant micelles 22 may vary depending upon the bioactive component 12 , the first aqueous composition 14 , the surfactant composition 16 , the temperature, pH, osmolarity, presence of any optional solutes or optional solvents, the surfactant micelle 22 , any desired characteristics of the stabilized surfactant micelle 28 , the nanocapsules 36 , or a desired dissolution profile.
  • the concentration of the biocompatible polymer component 24 is not critical to the present invention, the concentration of the biocompatible polymer component 24 is preferably based upon the bioactive component and on the desired dissolution profile.
  • the concentration of the biocompatible polymer component 24 is to high, the shell of the nanocapsule 36 may not dissolve. If the concentration of the biocompatible polymer component 24 is to low, the shell of the nanocapsule 36 may dissolve rapidly in a manner that promotes cytotoxicity, for example. In addition, too low a concentration of the biopolymer component 24 may not provide an effective degree of mechanical force to stabilize the surfactant micelles 28 .
  • Concentrations of the biocompatible polymer component 24 that are to high may also be less desirable because such higher concentrations may increase the viscosity of the first aqueous composition 14 , and consequently may cause difficulties in preparing, mixing and/or transferring the stabilizer surfactant micelles 28 .
  • Concentrations of the biocompatible polymer component 24 that are to low may be less preferred, because lower concentrations may not provide the needed viscosity to stabilize the surfactant micelles, nor be capable of effectively coating the surfactant micelles 22 to prevent aggregation of the surfactant micelles 22 in the first aqueous composition 14 .
  • the biocompatible polymer component 24 may be supplied as individual biocompatible polymers or supplied in various prepared mixtures of two or more biocompatible polymers that are subsequently combined to form the biocompatible polymer component 18 .
  • biocompatible polymers include polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose,
  • biocompatible polymers that have been modified for enzymatic degradation, or change upon application of light, ultrasonic energy, radiation, a change in temperature, pH, osmolarity, solute or solvent concentration may also be included as part of the biocompatible polymer component 24 .
  • the biocompatible polymer component 24 is a hydrophilic polymer that is capable of substantially coating, and preferably continuously coating the surfactant micelle 22 .
  • the hydrophilic biocompatible polymer component 24 is capable of ionotophoretic exchange.
  • hydrophilic biocompatible polymer component 24 any other biocompatible polymer, such as hydrophobic biocompatible polymers may be substituted in place of the hydrophilic biocompatible polymer, in accordance with the present invention, while still realizing benefits of the present invention.
  • any combination of any biocompatible polymer may be included in accordance with the present invention, while still realizing benefits of the present invention.
  • any conventional apparatus and technique that is suitable for permitting the biocompatible polymer component 24 to stabilize the surfactant micelles 22 may be used as the stabilizing apparatus 26 in accordance with the present invention.
  • any other device, such as high pressure homogenization or high ultrasound sonication is preferably not included during stabilization.
  • the stabilized surfactant micelles 28 may be transferred into a second aqueous composition 30 located in a second dispersing apparatus 32 .
  • the stabilized surfactant micelles 28 may be transferred by mechanically forming droplets of the stabilized surfactant micelle 28 that are subsequently introduced into the second aqueous composition 30 .
  • the second aqueous composition 30 may include water only, or may optionally include a solute to precipitate the biocompatible polymer component 24 surrounding the stabilized surfactant micelle 28 .
  • solutes include ionic species derived from elements listed in the periodic table.
  • the second aqueous composition 30 includes a solute in an amount that is effective to precipitate the biocompatible polymer component 24 and form the dispersed, and optionally atomized nanocapsules 36 of the present invention.
  • a solute refers to a solidifying or a hardening of the biocompatible polymer component 24 that surrounds the stabilized surfactant micelles 28 . It is also to be understood that the term “precipitation” is also meant to encompass any crystallization of the biocompatible polymer 24 that may occur when the biocompatible polymer component 24 is exposed to the solute.
  • any other component that is capable of modulating the efficacy the nanocapsules 36 may be included as part of the second aqueous composition to thereby modulate the functional activity of the nanocapsule 36 .
  • the second aqueous composition may include additional coating excipients, such as a cell recognition component or various ionic species, such as Mn 2+ , Mg 2+ , Ca 2+ , Al 3+ , Be 2+ , Li 30 , Ba 2+ , Gd 3+ , or any other ionic species that is capable of interacting with the biocompatible polymer component 24.
  • cell recognition component refers to a molecule capable of recognizing a component on a surface of a targeted cell.
  • Cell recognition components may include an antibody to a cell surface antigen, a ligand for a cell surface receptor, such as cell surface receptors involved in receptor-mediated endocytosis, peptide hormones, and the like.
  • a flocculated suspension refers to the formation of a loose aggregation of discrete particles held together in a network-like structure either by physical absorption of bioactive components, bridging during chemical interaction (precipitation), or when longer range van der Waals forces of attraction exceed shorter range forces of repulsion.
  • the flocculated suspension of nanocapsules 36 may entrap varying amounts of the first aqueous composition 14 or the second aqueous composition 30 within the network-like structure. Additionally, the flocculated suspension of nanocapsules may be gently tapped to disperse the nanocapsules 36 .
  • the stabilized surfactant micelles 28 may be transferred into the second aqueous composition 30 via atomization through a nozzle (not shown) having a particular orifice size or through an aerosolizing apparatus (not shown).
  • Atomizing or aerosolizing the stabilized surfactant micelles 28 typically includes the application of a shear force that may be capable of further dispersing the stabilized surfactant micelles 28 .
  • the application of the shear force during transfer may also be effective to (1) reduce the size of the nanocapsules 36 , or (2) break up any agglomerates or associations between stabilized surfactant micelles 28 that may have formed in the stabilizing apparatus 26 .
  • Nozzle pressures of less than about 100 psi, for example, may be used to atomize the stabilized surfactant micelles 28 .
  • the diameter of the nanocapsules 36 may also be varied depending upon the orifice size of the nozzle that may be used to transfer the stabilized surfactant micelles 28 into the second aqueous composition.
  • the stabilized surfactant micelles 28 may be added to the second aqueous composition 30 containing the solute that precipitates the biocompatible polymer 24 to form a dispersion of nanocapsules 36 for purposes of providing the dispersion for sub-cutaneous delivery of the nanocapsules, for example.
  • the nanocapsules 36 may be filtered, centrifuged or dried to obtain separate and discrete nanocapsules 36 .
  • the nanocapsules 36 may be frozen or reconstituted for later use or may be delivered to a target cell or tissue by such routes of administration as oral, intravenous, subcutaneous, intraperitoneal, intrathecal, intramuscular, inhalational, topical, transdermal, suppository (rectal), pessary (vaginal), intra urethral, intraportal, intrahepatic, intra-arterial, intra-ocular, transtympanic, intraumoral, intrathecal, or any combination of any of these.
  • nanocapsules 36 having a diameter of less than about 50 nm are advantageous in the delivery of bioactive components to target cells for several reasons.
  • nanocapsules 36 having a diameter of less than about 50 nm enhances delivery of bioactive components by protecting the bioactive components against degradation during transport to the target cell.
  • nanocapsules 36 having a diameter of less than about 50 nm promotes efficient cellular uptake. Efficient cellular uptake into the target cell typically occurs when a particle has a diameter of less than about 50 nm, as opposed to when a particle has a diameter of more than about 50 nm.
  • the nanocapsules 36 have a biocompatible polymer shell that is separate from the bioactive component.
  • the bioactive component is not entangled in, embedded in, or adsorbed onto the biocompatible polymer shell of the nanocapsules 36 .
  • the cell that incorporate the nanocapsules 36 avoid apoptosis or cell death.
  • enclosing the bioactive component within a core surrounded by the biocompatible polymer shell when preparing the nanocapsules 36 in accordance with the present method is advantageous in avoiding premature degradation of the nanocapsules 36 , or a cytotoxic response during in vivo transport of the nanocapsule. Enclosing the bioactive component within the core results in a linear release rate of the bioactive component without any zero burst phenomenon during release from the nanocapsules 36 .
  • the linear release rate of the bioactive component from the nanocapsule without any zero burst phenomenon is also an advantageous feature as the linear release rate allows rational design of coating dissolution profiles to minimize cytotoxicity.
  • dissolution profile refers to a rate at which the biocompatible polymer shell is dissolved or degraded to release a bioactive agent from a core of a nanocapsule.
  • Another benefit of the nanocapsules 36 prepared by the method of the present invention is that little, if any, addition of an organic solvent is required to form the nanocapsules 36 . Eliminating the use of most, if not all, organic solvents from the method of the present invention is beneficial since organic solvents may damage the bioactive component 12 , destroy the target cells, or be toxic during preparation of the nanocapsule 36 . The elimination of most, if not all, use of organic solvents eliminates the need for complex solvent removal techniques, such as solvent dilution, vacuum evaporation, or the like, and obviates any associated costs or complex process strategies during preparation of the nanocapsules 36 .
  • the nanocapsules 36 of the present invention further permits stable encapsulation of a bioactive component, and in particular, hydrophilic bioactive components, such as polynucleotides and polypeptides.
  • “Stable encapsulation”, as used herein, refers to maintenance of the encapsulated bioactive component's structure. For nucleic acids, the appearance of low molecular weight nucleic acid breakdown products, which may be assayed for by electrophoresis, is substantially eliminated.
  • the nanocapsules 36 may also be used to encapsulate any bioactive component regardless of water solubility or charge density.
  • the nanocapsules 36 may be combined with additional polymeric binders, surfactants, fillers, and other excipients to incorporate the nanocapsules 36 into solid dosage forms such as granules, tablets, pellets, films or coatings for use in enhanced bioactive component 12 delivery. In this way, design of the dissolution profile, control of the particle size, and cellular uptake remains at the level of the nanocapsule.
  • Such applications include, but are not limited to, creation of rapidly dissolving nanocapsule pellets for pulmonary delivery or nanocapsule films for device-mediated delivery.
  • the nanocapsules 36 may be designed for specific cellular or tissue uptake by polymer selection and/or inclusion of cell-recognition components in the nanocapsule biocompatible polymer shell or coating.
  • Such coatings will have utility for specific or increased delivery of the bioactive agent to the target cell.
  • Such applications include, but are not limited to tumor-targeting of chemotherapeutic agents or anti-sense DNA, antigen delivery to antigen-presenting cells, ocular delivery of ribozymes to retinal cells, transdermal delivery of protein antibodies, or transtympanic membrane delivery of peptide nucleic acids.
  • FIG. 1A Samples were prepared on freshly cleaved mica as dispensed, dried in air and imaged using a Nanoscope II multimode AFM (Digital Instruments) with a J type scanner and ambient tapping mode holder. 125 ⁇ m long silicon cantilevers type IBMSC were from IBM and have resonant frequencies of 250-450 kHz. All imaging was in tapping mode, images were 512 ⁇ 512 pixels and scanning frequency was 1 kHz. Height, amplitude and phase images were collected. Images were processed in DI software and analyzed in NIH Image SXM.
  • Nanoscope II multimode AFM Digital Instruments
  • IBMSC 125 ⁇ m long silicon cantilevers type IBMSC were from IBM and have resonant frequencies of 250-450 kHz. All imaging was in tapping mode, images were 512 ⁇ 512 pixels and scanning frequency was 1 kHz. Height, amplitude and phase images were collected. Images were processed in DI software and analyzed in NIH Image SXM.
  • A Formula Q from 2-phase system, low HLB surfactant
  • B Formula S from 2-phase system, high HLB surfactant
  • C Formula T from 1-phase system, high HLB surfactant
  • D Formula V from 2-phase system, surfactant below CMC.
  • FIG. 1C Nominal 300 ng samples of DNA were aliquoted from a master batch containing surfactant and processed through commercial miniprep columns. Eluate was recycled through Qiaquik columns and collected either 3 times (4, 5) or twice (6,7) or recycled through Zymoclean columns and collected twice (8,9).
  • FIG. 2 Endocytic activity was assessed by immunosignal levels of clathrin (Chemicon). Potocytotic activity was assessed by immunosignal for caveolin-1 as described in the literature (Transduction Laboratories). Lysosomal activity was detected by an monoclonal antibody to Lamp-1 (Transduction Laboratories). Nanocapsule localization was detected by streptavidin-biotin immunocomplexes directed against sheep IgG (Jackson Laboratories). Nanocapsule coatings were spiked with ovine IgG to enable this detection strategy.
  • FIG. 3 immortalized Rt-1 fibroblast cultures at 70% confluence were treated for 4 days with increasing amounts of nanocapsule formula K and transiently treated (3 hours) with an optimized liposomal formula (dose, 500 ng) Results are expressed as a percentage of cellular actin integrated intensity and compared to the liposomal formula.
  • Expression vector was code 448: pEF/myc-his/GFP (Invitrogen).
  • FIG. 4A Radiated porcine biopsies were snapfrozen 7 days after treatment with saline or 6 ⁇ g of controlled release nanocapsules, then homogenized in RIPA. 100 ⁇ g lysate samples were electrophoresed on SDS-Page gradient gels, transferred to nitocellulose membranes and detected for either ⁇ -galactosidase (121 Kd) or involucrin ( ⁇ 100 kD) using chemiluminescence. Results were normalized to the post-transfer gel stained with Coomassie due to interference at 100 kD from a gel defect.
  • a component of the cornified membrane, manufactured by suprabasal cells can be detected in radiated porcine skin and will be used for future normalization purposes.
  • FIG. 4B The ⁇ -galactosidase reporter protein was detected by a monoclonal antibody directed at an incorporated fusion protein tag.
  • FIG. 5 Nanocapsules were incorporated into an aqueous suture coating and sutures were applied to pigskin biopsies in organ culture. Nanocapsules were detected with Cy3 conjugated-streptavidin-biotin complexes to incorporated ovine IgG and GFP transgene expression was detected by rabbit polyclonal antibodies to GFP (Abcom) in combination with Fitc-conjugated polyclonal antibodies to rabbit IgG and Alexa 488-conjugated polyclonal antibodies to Fitc (Molecular Probes). Controls omitting primary antibodies were included for signal-to-background level estimation.
  • FIG. 6A Nanocapsules were detected as previously described and GFP transgene expression was detected by rabbit polyclonal antibodies to GFP in combination with Cy3-conjugated antibodies to rabbit IgG (Jackson Laboratories).
  • FIG. 6B GFP expression was detected as described in FIG. 5 and cell nuclei were counterstained with 10 ⁇ g/ml bisbenzamide.
  • FIG. 6C Carcinoma cells and HDF's were seeded overnight into 96 well plates at 2000 and 6000 cells per well respectively. Cisplatin preparations were added to wells for 18 hours as noted on the graph than washed out. After 72 hours cell viability of assessed by a commercial MTT assay (WST assay, Boehringer Mannheim). Wells were executed in duplicate.
  • WST assay Boehringer Mannheim
  • FIG. 7 Colocalization with lysosomes was detected using a monoclonal antibody to Lamp-1 (Transduction Laboratories). AFM images are included of O-methyl RNA formulated by nanoencapsulation or complexation with 27 KD polyethyleneimine.
  • PEI Poly(ethylenimine) at 27 KiloDalton (kD). PEI was used at optimized conditions (90% charge neutralization)
  • PLL Polylysine at 70-150,000 molecular weight.
  • PLL condensing materials were conjugated with nuclear signal localization peptides, either SV-40 T antigen or cys-gly-tyr-gly-pro-lys-lys-lys-arg-lys-val-gly-gly using carboxiimide chemistry available from Pierce Chemical (Rockford, Ill.).
  • NMP nuclear matrix proteins
  • Hyaluronan derived from human umbilical cord, about 4MMKD and not conjugated.
  • Povidone polyvinylpyrolidone, PVP
  • Povidone 10,000 kD MW and not bioconjugated
  • Povidone polyvinylpyrolidone, PVP
  • Kd polyvinylpyrolidone
  • PVP polyvinylpyrolidone
  • 334 pcDNA/His/LacZ, produces galactosidase, incorporates CMV promoter, based on pcDNA 3.1. (Invitrogen), 8.6 kB
  • 425 pEGFP-c/farn, enhanced GFP (green fluorescent protein) expression vector modified with a farnasyl moiety to improve microscopy, CMV promoter, 4.6 kB
  • CCRL 1764 Immortalized rat neonatal fibroblast cell line
  • HaCaT immortalized human keratinocyte cell line
  • Ca9 human tumor cells derived from a squamous cell carcinoma of tongue origin.
  • Formulae were produced by i) predispersing 25 ⁇ g of DNA (425) on ice using a bath sonicator, ii) condensing DNA in a small amount of water by vortexing then incubating on ice for 20 minutes, iii) adding surfactant then oil followed by 30 seconds of probe sonication at 10 Watts, iv) diffusion dilution to 3 milliliters (mL) by first adding saline then 1 MM kD hyaluronan polymer (1%) as a protective colloid, v) mechanically shearing emulsion into droplets by pumping through a 250 micrometer ( ⁇ m) orifice into 22 mL of PBS, 10 milliMolar (mM) Ca 2+ , 200 mM Li + , vi) incubating overnight end over end and vii) centrifuging to recover nanoparticles for resuspension
  • the condenser-to-DNA weight ratio was determined by dye exclusion at 90% charge neutralization.
  • TM-diols were used in this experiment to represent water-immiscible surfactants, while Tergitol NP40 and Tween 80 were used to represent water-soluble and even more water-soluble emulsifiers/dispersing aids.
  • Dispersion conditions were systematically varied to discourage micelle formation in aqueous media by i) choosing water-soluble surfactants (Formulae S,T,U, and V), ii) removing the dispersed phase (Formula T) and iii) decreasing surfactant loading below that required for micelle formation.
  • Formula U featured use of a water-miscible oil (silicone oil).
  • Nanocapsule sizing was determined by tapping mode AFM and images are illustrated in FIG. 1A.
  • the data indicate average nanocapsule sizes less than 50 nm are achievable only with multi-phase systems in combination with low water solubility surfactants (Table 1A: Formulae Q,R vs. S,T,U,V, and W).
  • Table 1A Formulae Q,R vs. S,T,U,V, and W.
  • Effective dispersion also corresponded with decreased aggregation and enhanced DNA stability (as indicated by decreased electrophoretic breakdown products).
  • the starting DNA was partially relaxed (76% supercoiled by electrophoresis). Using this value as a basis, supercoil retention in DNA still encapsulated following 100 hrs of release testing, was excellent in multi-phase systems.
  • FIG. 1C illustrates that small amounts of DNA (in this case 300 nanograms of DNA) can be recovered accurately in a procedure comprising butanol extraction of 10% butanol/saline releasing fluid followed by isolation on a miniprep column and measurement of absorbance at 260 nm excitation. Results obtained from UV spectroscopy are confirmed by electrophoresis of recovered DNA following alcohol coprecipitation with a commercial coprecipitant aid.
  • Experiment 1A demonstrates the importance of a multi-phase system in creating coated particles from the micellar solution, defines surfactant requirements and validates method for measuring in vitro release profiles.
  • Aqueous dispersion of DNA condensates with poorly soluble surfactants in the inventive method produced average nanocapsule diameters under 50 nm.
  • a 30 number of successful operating regimes were feasible with varying effects on encapsulation yield.
  • under condensation resulted in an average particle diameter increase from 20 to 12 nm (Table 1B: F1vs. F2).
  • the same decrease in condenser weight ratio induced a particle size increase from 24 to 36 nm, while still maintaining nanocapsule functionality for trangene delivery, when using a TM-diol/DMSO surfactant system for initial micelle formation (Table 1B: F4 vs. F5).
  • This finding teaches surfactant selection impacts final average nanocapsule diameters.
  • nanocapsules increased cellular pinocytotic activity compared to standard formulations, and smaller nanocapsules shifted pinocytotic activity to caveolae from clathrin-coated pits (Table 2: Formula O vs. pei-dna and lipoplex pDNA). It was further observed that nanocapsules avoided lysosome co-localization at 10 hours post-addition with smaller nanocapsules being particularly effective (see Table 2: Formula vs. pei-dna and lipoplex pDNA). These results are illustrated further in FIG. 2. This improvement is further emphasized by comparison with published uptake studies for HacaT keratinocytes.
  • nanocapsule design plays a role in maintaining nanocapsule integrity and that size effects and dissolution profiles can contribute to observed cytotoxicity and functionality.
  • application of nanocapsule formulations increased dosing to useful efficiency levels without induction of an apoptotic program.
  • Table 3B exemplifies this improvement with a dose response of Formula K.35 measured in fibroblast lysates.
  • GFP production was measured in fibroblast lysates after 4 days of treatment with increasing doses of nanocapsules.
  • a 9.5 ⁇ g dose of nanocapsules equaled the production of a liposomal formulation without any evidence of cytotoxicity.
  • Example 4 Nanocapsule delivery of macromolecules to porcine tissue across keratinized barrier epithelia bv transdermal and subcutaneous means
  • tissue sections were analyzed for ⁇ -galactosidase expression in double-label experiments using antibodies to cell-specific epitopes (see FIG. 4B).
  • Digital image analysis of these sections indicated that radiated keratinocytes and endothelial cells are readily transduced in organ culture 7 days after treatment with a 10 day releasing formula.
  • Specific quantitation of fibroblastic cells was not possible without inclusion of a cell-specific marker, however, an 11-fold increase in area of expression was measured in N biopsy dermis (see FIG. 4B).
  • Example 5 Integration of inventive nanocapsules into a solid dosage form for additional utility in physical targeting
  • Nanocapsules containing a nuclear GFP transgene or empty vector were incorporated into a suture coating by vortexing the following components: i) 50 ⁇ g of nanocapsules containing plasmid DNA, ii) 200 ⁇ g of bovine mucin, and iii) 75 ⁇ g of sucrose (60% w/w) in a 1000 ⁇ l volume. Sutures were aseptically coated by drawing sutures 5 ⁇ through punctured microcentrifuge tubes. Coating functionality for gene transfer was tested by applying sutures in cultured porcine skin biopsies. Biopsies were cultured on a mesh such that the biopsy bottom was in contact with cell culture media. Biopsies were treated for 7 days, then snap-frozen and sectioned for immunofluorescence microscopy to assess nanocapsule penetration and transgene delivery.
  • FIG. 5A shows distribution of sheep IgG signal throughout porcine dermal tissue with accumulation on capillaries.
  • primary antibody is omitted during slide processing to determine level of background fluorescence.
  • a suture is visible in this view.
  • Sutures were identifiable as smooth objects without positive nuclear counterstain.
  • GFP expression was confirmed using a polyclonal GFP antibody to obviate the effect of nonspecific tissue green fluorescence.
  • FIG. 5B shows GFP expression throughout the suture-treated dermis using a GFP polyclonal antibody. A suture was visible 750 microns away.
  • FIG. 5C shows the lack of GFP expression in a biopsy treated with empty vector coating. This example demonstrates the usefulness of nanocapsules for use in physically targeted macromolecule delivery.
  • Example 6 Ultratility of nanocapsules for local targeting by design of nanocapsule coating
  • GFP nanocapsules were prepared by dispersion atomization as described in Example 1.
  • Polyvinylpyrylodone (PVP, MW 10,000) was used as the coating basis.
  • a coating weight ratio of 1:40 was used and rod-shaped nanocapsules of 23 ⁇ 2 nm were produced.
  • 1 ⁇ g of PVP nanocapsules were applied to both human dermal fibroblasts (HDF) and HacaT keratinocyte cultures for 4 hours then fixed for detection for nanocapsule uptake by streptavidin-bioting immunocomplexes to sheep IgG. Nanocapsule coatings are spiked with ovine IgG to enable this detection strategy.
  • FIG. 6 illustrates positive nuclear localization of PVP nanocapsules in HDF's and negative colocalization of PVP nanocapsules in keratinocytes (FIG. 6: 6 a vs. 6 b ). Views of untreated cultures are included for comparison ( 6 a ′, 6 b ′). Cultures were also treated with 5 ⁇ g of PVP nanocapsules for 5 days then tested for GFP transgene production. Consistent with uptake studies results, only the fibroblast cultures showed production of GFP transgene (FIG. 6: 6 a ′′ vs. 6 b ′′).
  • GFP nanocapsules were prepared by dispersion atomization as described in example 1. Tenascin (TN, MW 200,000) was used as the coating basis. A coating weight ratio of 1:20 was used and spherical nanocapsules of 19 ⁇ 0.9 nm were produced. 500 ng of TN nanocapsules were applied topically in successive small aliquots to pig biopsies maintained in organ culture. Biopsies were rinsed in media after 3 minutes of topical application and culture media was changed to preclude any delivery other than topical.
  • FIG. 6B To simulate tumor nests of epithelial-derived origin, biopsies had been seeded 12 hours previously with 50,000 human squamous carcinoma cells. 7 days later biopsies were snapfrozen and sectioned for immunological detection of GFP production.
  • view “a” shows intense GFP fluorescence in the tumor center
  • view “b” confirms this GFP expression with polyclonal antibodies to GFP
  • view “c” shows cell positioning in the section using a counterstain for cell nuclei
  • view “d” shows the level of background fluorescence by omission of GFP antibodies.
  • Tumor origin was confirmed by positive detection with antibody to keratin 10/1, an epithelial marker.
  • Comparison of view “b” and view “c” indicates that GFP expression is limited to cells within the tumor. As already demonstrated in example 5, expression throughout a tissue is also feasible and can modulated by coating design. This example demonstrates that nanocapsule delivery can be productively targeted.
  • Nanocapsules were prepared as described in Example 1 to encapsulate cisplatin, a hydrophobic molecule and a common cancer chemotherapeutic with serious side effects. A coating weight ratio of 1:100 was used and irregular nanocapsules of 29 ⁇ 3 nm were produced. Targeting efficacy was demonstrated by changes in the dose response for cell growth inhibition in fibroblast vs. squamous cell carcinoma cultures. Cells were seeded overnight into 96 well plates, treated for 18 hours with increasing amounts of encapsulated or unencapsulated drug, then assessed for cell growth inhibition using an MTT assay 48 hours later for total growth time of 72 hours. Results are illustrated in FIG. 6C.
  • TN nanocapsules protected nontarget cells from cell death (zero death) at drug levels that killed using unencapsulated drug (FIG. 6Aa: open vs. closed circles).
  • TN nanocapsules productively decreased the inhibition concentration (IC50) an estimated 300% from 525 to 160 ⁇ g/ml.
  • Example 6 demonstrates the usefulness of nanocapsules for use in coating-targeted macromolecule delivery.
  • Nanocapsules containing either 500 kD Fitc-labelled dextran, 20 mer Fitc-labelled mer O-methylated RNA oligonucleotide and 16 mer phosphodiester DNA oligonucleotide were prepared as described in Example 1. A 1:40 coating weight ratio was used and 1 MM kD hyaluronan was used a coating basis. PEI was used to condense the phosphodiester DNA oligonucleotide, but no PEI was included in the dextran or RNA oligonucleotide formulas. Nanocapsule functionality for drug delivery was tested by evaluating changes in relative pinocytotic activity and cellular uptake in 35 mm cultures of human dermal fibroblast.
  • Nanocapsule formulas were compared to naked species or species formulated as complexes. Quantitative results are summarized in Table 7. TABLE 6 Nanoencapsulation improves cellular uptake of other species used as pharmaceutical, nutraceutical, research or cosmetic agents. At 18 hours post-addition, lysosomes are only evident in conventionally formulated species.
  • Table 7 shows that average diameters for all nanocapsules were below 50 nm by AFM.
  • PEI complexes of DNA oligonucleotides were measured at 67 nm and PEI complexes of uncharged RNA O-methyl oligonucleotides were measured at 236 nm.
  • nanocapsules stimulate pinocytotic activity as indicated by increased signal levels of clathrin and caveolin-1. In the 500 kD dextran case, pinocytotic activity shifts productively towards caveolae with nanoencapsulation (Table 7, 500 kD Dextran).
  • FIG. 7B That distribution is exclusively nuclear for the nanoencapsules of RNA oligonucelotides (FIG. 7B: a vs. a′). Punctate inclusion are visible in the cultures treated with the complexed RNA oligonucleotides that co-localize with an immunological marker for lysosomes (FIG. 7A: a vs. a′). Particle sizing results from AFM microscopy are included to demonstrate dramatic difference in sizing following encapsulation. (FIG. 7A, 7 B:b vs.b, b′).
  • Formulas encapsulating lower molecular weight dextrans and unstabilized RNA were also prepared with analagous uptake, nanocapsule size and yield to demonstrate that encapsulation can provide not only a targeting function but aid in stabilizing molecules sensitive to chemical or enzymatic degradation. These examples demonstrates the usefulness of nanocapsules 36 for use in delivery of a broad range of macromolecules.

Abstract

The present invention generally relates to nanocapsules and methods of preparing these nanocapsules. The present invention includes a method of forming a surfactant micelle and dispersing the surfactant micelle into an aqueous composition having a hydrophilic polymer to form a stabilized dispersion of surfactant micelles. The method further includes mechanically forming droplets of the stabilized dispersion of surfactant micelles, precipitating the hydrophilic polymer to form precipitated nanocapsules, incubating the nanocapsules to reduce a diameter of the nanocapsules, and filtering or centrifuging the nanocapsules.

Description

    CROSS-REFERENCE TO RELATED APPLICATION(S)
  • This application is a continuation of application Ser. No. 09/796,575 filed Feb. 28, 2001, which claims the benefit of U.S. Provisional Application No. 60/185,282 filed Feb. 28, 2000.[0001]
  • BACKGROUND OF THE INVENTION
  • The present invention generally relates to a field of controlled-release delivery systems for macromolecules, particularly those for nucleic acids and gene therapy. More specifically, the present invention relates to nanocapsules having a diameter of less than about 50 nanometers, in which a bioactive component is located in a core of the nanocapsule, and to methods of forming these nanocapsules. [0002]
  • Over the past several decades, active and extensive research into the use of nanoparticles in the delivery of bioactive agents has generated a number approaches in the preparation of nanoparticles. These approaches typically include the use of heat, high pressure homogenization, or high intensity ultrasound sonication to prepare nanoparticles having a diameter of more than 100 nanometers, or high amounts of solvents or oils, cytotoxic chemicals, such as cross-linking agents, adjuvants, catalysts or any combination of any of these, to prepare nanoparticles having a diameter of less than 100 nanometers. Furthermore, these approaches are challenging due to a number of variables. [0003]
  • For example, when organic solvents are included in the manufacturing process for nanoparticles, the organic solvent may denature the bioactive agent which reduces most, if not all, efficacy of the bioactive agent. In fact, denaturation of the bioactive agent may promote a toxic response upon administration of the nanoparticle, to a human subject, for example. [0004]
  • In addition, when an organic solvent is used to prepare nanoparticles, the organic solvent may undergo degradation to form a low pH environment that destroys the efficacy of the bioactive agent. Therefore, organic [0005]
  • As a result, organic solvents are typically removed during the manufacturing process of nanoparticles. However, inclusion of one or more organic solvent removal techniques generally increases the costs and complexity of forming nanoparticles. [0006]
  • The incorporation of high pressure homogenization or high intensity ultrasound sonication to prepare nanoparticles typically results in entangling or embedding the bioactive agent in a polymeric matrix of the nanoparticle. Entangling or embedding the bioactive agent in the polymeric matrix may also denature the bioactive agent to thereby reduce the efficacy of the bioactive agent. [0007]
  • Entangling or embedding the bioactive agent in the polymeric matrix of the nanoparticle may also reduce the efficacy of the bioactive agent by permitting premature release of the bioactive agent prior to reaching a target cell. Premature release of the bioactive agent typically promotes cytotoxicity or cell death during administration of the nanoparticle. [0008]
  • Furthermore, nanoparticles that reach the target cell are typically transported into the target cell via endosomal regulated pathways that results in lysosomal degradation of the bioactive agent and the nanoparticle. Therefore, functional activity of the bioactive agent inside the target cell may not occur since the bioactive agent and the nanoparticle undergoes degradation. As used herein, the term “functional activity” refers to an ability of a bioactive agent to function within a target cell for purposes of providing a therapeutic effect on the target cell. [0009]
  • Additionally, high pressure homogenization or high intensity ultrasound sonication techniques often require complex and expensive equipment that generally increases costs in preparing nanoparticles. Therefore, an urgent need exists to prepare nanoparticles without the use of cytotoxic chemicals like organic solvents or the use of complex and expensive equipment. Furthermore, an urgent need exists to prepare nanoparticles that do not entangle nor embed the bioactive agent in the nanoparticle so that cytotoxic responses are minimized. Additionally, an urgent need exists to develop a nanoparticle that maybe transported into a target cell where the bioactive agent is released to accomplish therapeutic delivery of the bioactive agent. [0010]
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention generally relates to nanocapsules and methods of preparing these nanocapsules. The present invention includes a method of forming a surfactant micelle and dispersing the surfactant micelle into an aqueous composition having a hydrophilic polymer to form a stabilized dispersion of surfactant micelles. The method further includes mechanically forming droplets of the stabilized dispersion of surfactant micelles, precipitating the hydrophilic polymer to form precipitated nanocapsules, incubating the nanocapsules to reduce a diameter of the nanocapsules, and filtering or centrifuging the nanocapsules.[0011]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic of a method of the present invention for preparing nanocapsules. [0012]
  • FIG. 1A illustrates atomic force microscopy of nanocapsule formulations prepared under different dispersion conditions. [0013]
  • FIG. 1B illustrates results from an experiment documenting quantitative recovery of small amounts of DNA from releasing solutions. [0014]
  • FIG. 1C illustrates cumulative release over 72 hours for nanocapsules prepared under different dispersion conditions. [0015]
  • FIG. 2 illustrates relative pinocytotic activity of HacaT keratinocyte cultures treated with DNA complexes, nanocapsules containing DNA or no treatment. [0016]
  • FIG. 3 illustrates the results of western blotting of total protein from rat fibroblast cultures. [0017]
  • FIG. 4A illustrates [0018]
  • FIG. 4B illustrates immunofluorescence microscopy of porcine dermal tissue sections from the experiment of FIG. 4. [0019]
  • FIG. 5 shows incorporation of nanocapsules into a solid dosage form. [0020]
  • FIG. 6A illustrates polyvinylpyrolidone nanocapsule uptake and Green Fluorescent Protein (GFP) expression in 35 mm human dermal fibroblast and immortalized keratinocyte cultures. [0021]
  • FIG. 6B illustrates tumor targeting of GFP plasmid DNA by Tenascin nanocapsules. [0022]
  • FIG. 6C illustrates an effect of nanocapsules that are coated with Tenascin and nanocapsules that are not coated with Tenascin on growth inhibition of squamous cell carcinoma and human dermal fibroblast (HDF) cultures. [0023]
  • FIG. 7 shows uptake of HDF cultures treated with nanocapsules containing 20 mer Fitc-labeled O-methyl RNA oligonucleotides.[0024]
  • DETAILED DESCRIPTION
  • The present invention generally relates to nanocapsules having a diameter of less than about 50 nanometers (nm). The present invention also relates to a method of preparing these nanocapsules. According to the method of the present invention, a nanocapsule is formed by partitioning a bioactive component within a core of surfactant molecules, and surrounding the surfactant molecules with a biocompatible polymer shell. [0025]
  • A method for producing the nanocapsule is generally depicted at [0026] 10 in FIG. 1. In the method 10, a bioactive component 12 is homogeneously dispersed into a first aqueous composition 14 to form a hydrophilic composition (not shown). Next, a surfactant composition 16, including a surfactant component (not shown) that contains a plurality of surfactant molecules, and an optional biocompatible oil component 18, are introduced into a first dispersing apparatus 20 along with the hydrophilic composition. The surfactant composition 16 is subjected to conditions in the first dispersing apparatus 20 that initiate at least partial adsorption of the surfactant molecules onto a surface of the bioactive component 12.
  • Partial adsorption of surfactant molecules onto the surface of the [0027] bioactive component 12 initiates partitioning of the bioactive component 12 into a core of a shell formed from the surfactant molecules in the first aqueous composition 14. Adsorption of the surfactant molecules onto the surface of the bioactive component 12 may proceed until an entire surface of the bioactive component 12 is covered by the surfactant molecules to complete partitioning of the bioactive component 12 into the core of surfactant molecules and form a surfactant micelle 22.
  • Next, a [0028] biocompatible polymer component 24 is added to the surfactant micelle 22 to stabilize the surfactant micelle 22 located in the first aqueous composition 14. Preferably, the biocompatible polymer component 24 surrounds the surfactant micelle 22 in a stabilizing apparatus 26 to form a stabilized surfactant micelle 28.
  • After stabilization, the stabilized [0029] surfactant micelle 28 is transferred from the stabilizing apparatus 26 into a second aqueous composition 30 located in a second dispersing apparatus 32. Preferably, the second aqueous composition 30 includes a solute (not shown) that is capable of precipitating the biocompatible polymer component 24 that coats the stabilized surfactant micelle 28. After precipitating the biocompatible polymer component 24 of the stabilized surfactant micelle 28, dispersed, optionally atomized precipitated nanocapsules 36, hereinafter referred to as nanocapsules 36, are formed.
  • It has been discovered that dispersing a surfactant composition, that includes a surfactant component having a hydrophile-lipophile-balance (HLB) value of less than about 6.0 units, into an aqueous composition that contains a bioactive component forms surfactant micelles that surround the bioactive component. It has further been discovered that stabilizing the surfactant micelles by adding a biocompatible polymer coats the surfactant micelles to form nanocapsules having a diameter of less than about 50 nm. [0030]
  • As used herein, the term “nanoparticle” refers a particle having a matrix-type structure with a size of less than about 1,000 nanometers. When the nanoparticle includes a bioactive component, the bioactive component is entangled or embedded in the matrix-type structure of the nanoparticle. [0031]
  • The term “nanosphere”, as used herein, refers to a particle having a solid spherical-type structure with a size of less than about 1,000 nanometers. When the nanosphere includes a bioactive component, the bioactive component is adsorbed onto the surface of the nanosphere or embedded in the nanosphere. [0032]
  • Similarly, the term “nanocore”, as used herein, refers to a particle having a solid core with a size of less than about 1,000 nanometers. When the nanocore includes a bioactive component, the bioactive component is entangled in the nanocore. [0033]
  • As used herein, the term “nanocapsule” refers to a particle having a hollow core that is surrounded by a shell, such that the particle has a size of less than about 1,000 nanometers. When a nanocapsule includes a bioactive component, the bioactive component is located in the core that is surrounded by the shell of the nanocapsule. The term “nanocapsule” is not meant to encompass, and generally does not include, a particle having a size of less than about 1,000 nanometers, in which a bioactive component is entangled or embedded in the matrix of the nanocapsule or adsorbed onto the surrounding shell of the nanocapsule. [0034]
  • The [0035] bioactive component 12 may be included into the first aqueous composition 14 as a liquid, vapor or in granular form. The form of the bioactive component 12 that is selected preferably permits the bioactive component 12 to (1) remain stable prior to dissolving or dispersing into the first aqueous composition 14, (2) be homogeneously dispersed into the first aqueous composition 14, (3) be optionally condensed to reduce a size of the bioactive component 12, (4) be partitioned into the core of the surfactant micelles 22, (5) be released upon degradation of the biocompatible polymer shell 24 of the nanocapsule 36, and (6) be functionally active upon release from the nanocapsule 36.
  • The [0036] bioactive component 12 may be characterized as “hydrophilic” or “hydrophobic”. As used herein, the term “hydrophilic” and “hydrophilicity” refers to an ability of a molecule to adsorb water or form one or more hydrogen-bond(s) with water. All references to “hydrophilic” is also understood as encompassing any portion of the molecule that is capable of adsorbing water or forming one or more hydrogen-bond(s) with water. As used herein, the term “hydrophobic” and “hydrophobicity” refers to an ability of a molecule to not adsorb water nor form one or more hydrogen-bond(s) with water. All references to “hydrophobic” is also understood as encompassing any portion of the molecule that is not capable of adsorbing water nor forming one or more hydrogen-bond(s) with water.
  • When the [0037] bioactive component 12 is a hydrophilic bioactive component, the hydrophilic bioactive component maybe directly added to the first aqueous composition 14. As an alternative, the hydrophilic bioactive component 12 may be optionally dissolved or dispersed in one or more solvents, such as water, a nonpolar solvent, a polar solvent, or any combination of any of these.
  • As used herein, the term “nonpolar solvent” refers to a solvent that does not have a permanent electric dipole moment, and therefore has no ability for an intramolecular association with a polar solvent. Additionally, a nonpolar solvent may be characterized as a solvent that includes molecules having a dielectric constant of less than about 20 units. Similarly, the term “immiscible”, as used herein, refers to an inability of two or more substances, such as two or more liquids, solids, vapors, or any combination of any of these, to form an intramolecular association with another substance. Some non-exhaustive examples of nonpolar solvents may be found in Perry's Chemical Engineer's Handbook, Sixth Edition, which is incorporated herein by reference. [0038]
  • As used herein, the term “polar solvent” refers to a solvent that has a permanent electrical dipole moment, and therefore has an ability to form an intramolecular association with another polar substance, such as a liquid, a solid, a vapor or any combination of any of these. Additionally, a polar solvent may be characterized as a solvent that includes molecules having a dielectric constant of more than about 20 units. Likewise, the term “miscible”, as used herein, refers to an ability of two or more substances to form an intramolecular association with each other. Some non-exhaustive examples of polar solvents may be found in Perry's Chemical Engineer's Handbook, Sixth Edition, which has been incorporated herein by reference. [0039]
  • When the [0040] bioactive component 12 is a hydrophobic bioactive component, the hydrophobic bioactive component may be dispersed or dissolved in a solvent that is capable of dispersing or dissolving the hydrophobic molecule, such as the above-mentioned water, a nonpolar solvent, a polar solvent, or any combination of any of these. Preferably, when the bioactive component 12 is a hydrophobic bioactive component 12, the hydrophobic bioactive component 12 is dissolved or dispersed in a water-miscible solvent, such as, acetone, acetonitrile, ethanol, dimethyl acetamide (DMA), tetrahydrofuran (THF), dioxane, dimethylsulfoxide (DMSO), and dimethylformamide (DMF). Other suitable non-exhaustive examples of water-miscible solvents may be found in Perry's Chemical Engineer's Handbook, Sixth Edition, which has been incorporated herein by reference.
  • As noted, the [0041] bioactive component 12 may be optionally condensed in the first aqueous composition 14 prior to forming the surfactant micelle 16. For example, when the bioactive component is a polynucleotide, the polynucleotide may be condensed using a DNA-condensing agent. As used herein, a “DNA-Condensing Agent” is a molecule that facilitates condensation or a size reduction of DNA.
  • While condensation of the [0042] bioactive component 12 is not critical to the present invention, condensation of the bioactive component 12 maybe practiced to reduce the size of the bioactive component 12. Condensation of the bioactive component 12 generally reduces the size of the bioactive component 12 prior to partitioning into the core of the surfactant micelle 16. Reducing the size of the bioactive component 12 may permit maximum incorporation of the bioactive component 12 into the surfactant micelle 22 or may assist a reduction in the overall size of the nanocapsule 36. Increasing the amount of the bioactive component 12 that may be included as part of the nanocapsule 36 permits incorporation of macromolecules having a large number of monomers, such as a large number of base pairs or amino acids, for example. Some non-exhaustive examples of condensing agents have been reviewed in Rolland, A. P. (1998). Crit. Rev. Therapeutic Drug. Carr. Syst. 15:143-198, and is incorporated herein by reference.
  • The [0043] bioactive component 12 may further include additional components that are compatible with, and that do not interfere with solvation or dispersion of the bioactive component 12. Some non-exhaustive examples of additional components that may be added to the bioactive component 12 include a DNA-associating moiety, which refers to a molecule, or portions thereof, that interact in a non-covalent fashion with nucleic acids. DNA-associating moieties may include, but are not limited to, a major-and minor-groove binder, a DNA intercalator, a polycation, a DNA-masking component, a membrane-permeabilizing component, a subcellular-localization component, or the like. Major- and minor-groove binders, as used herein, are molecules thought to interact with DNA by associating with the major or minor groove of double-stranded DNA.
  • Similarly, the term “DNA intercalator”, as used herein, refer to a planar molecule or planar portion of a molecule thought to intercalate into DNA by inserting themselves between, and parallel to, a nucleotide base pair. As used herein, a “polycation” is thought to associate with the negative charges on the DNA backbone. The DNA-associating moiety may be covalently linked through a “reactive group” to a functional component of this invention. The reactive group is easily reacted with a nucleophile on the functional component. Some non-exhaustive examples of reactive groups (with their corresponding reactive nucleophiles) include, but are not limited to N-hydroxysuccinimide (e.g., amine), maleimide and maleimidophenyl (e.g., sulfhydryl), pyridyl disulfide (e.g., sulfhydryl), hydrazide (e.g., carbohydrate), and phenylglyoxal (e.g., arginine). [0044]
  • The term “DNA-masking component”, as used herein, refers to a molecule capable of masking all or part of a polynucleotide following release from a nanocapsule to increase its circulatory half-life by inhibiting attack by degrading reagents, such as nucleases, present in the circulation and/or interfering with uptake by the reticuloendothelial system. Similarly, the term “membrane-permeabilizing component”, as used herein, refers to any component that aids in the passage of a polynucleotide or encapsulated polynucleotide across a membrane. Therefore, “membrane permeabilizing component” encompasses in part a charge-neutralizing component, usually a polycation, that neutralizes the large negative charge on a polynucleotide, and enables the polynucleotide to traverse the hydrophobic interior of a membrane. [0045]
  • Many charge-neutralizing components can act as membrane-permeabilizers. Membrane-permeabilization may also arise from amphipathic molecules. A “membrane permeabilizer”, as used herein, is a molecule that can assist a normally impermeable molecule to traverse a cellular membrane and gain entrance to the cytoplasm of the cell. The membrane permeabilizer may be a peptide, bile salt, glycolipid, phospholipid or detergent molecule. Membrane permeabilizers often have amphipathic properties such that one portion is hydrophobic and another is hydrophilic, permitting them to interact with membranes. [0046]
  • The term “subcellular-localization component”, as used herein, refers to a molecule capable of recognizing a subcellular component in a targeted cell. Recognized subcellular components include the nucleus, ribosomes, mitochondria, and chloroplasts. Particular subcellular-localization components include the “nuclear-localization components” that aid in carrying molecules into the nucleus and are known to include the nuclear localization peptides and amino acid sequences. [0047]
  • The [0048] bioactive component 12 may be included at an amount that is therapeutically effective to transform a plurality of cells, such as in vitro, in vivo or ex vivo cells. As used herein, “transform” refers to a presence and/or functional activity of the bioactive component in the plurality of cells after exposing the nanocapsules to the plurality of cells.
  • Furthermore, those of ordinary skill in the art will recognize that the amount of the [0049] bioactive component 12 may vary depending upon the bioactive component 12, the temperature, pH, osmolarity, any solutes, any additional component or optional solvents present in the second aqueous component 30, the surfactant composition 16, a type or an amount of the surfactant micelle 22, the biocompatible polymer component 24, any desired characteristics of the stabilized surfactant micelle 28, any desired characteristics of the nanocapsules 36, or any combination of any of these.
  • The [0050] bioactive component 12 of the nanocapsule 36 maybe supplied as an individual macromolecule or supplied in various prepared mixtures of two or more macromolecules that are subsequently combined to form the bioactive component 12. Some non-exhaustive examples of hydrophilic macromolecules that may be suitable for inclusion as part of the bioactive component 12 include, but are not limited to polynucleotides, polypeptides, genetic material, peptide nucleic acids, aptamers, carbohydrates, mini-chromosomes, molecular polymers, aggregates or associations of an inorganic or organic nature, genes, any other hydrophilic macromolecule or any combination of any of these.
  • Some non-exhaustive examples of hydrophobic macromolecules that may be included part of the bioactive component [0051] 12 include, but are not limited to, adregergic, adrenocotical steroid, adrenocortical suppressant, aldosterone antagonist, and anabolic agents; analeptic, analgesic, anesthetic, anorectic, and anti-acne agents; anti-adrenergic, anti-allergic, anti-amebic, anti-anemic, and anti-anginal agents; anti-arthritic, anti-asthmatic, anti-atherosclerotic, antibacterial, and anticholinergic agents; anticoagulant, anticonvulsant, antidepressant, antidiabetic, and antidiarrheal agents; antidiuretic, anti-emetic, anti-epileptic, antifibrionlytic, and antifungal agent; antihemorrhagic, inflammatory, antimicrobial, antimigraine, and antimiotic agents; antimycotic, antinauseant, antineoplastic, antineutropenic, and antiparasitic agents; antiproliferative, antipsychotic, antirheumatic, antiseborrheic, and antisecretory agents; antipasmodic, antihrombotic, antiulcerative, antiviral, and appetite suppressant agents; blood glucose regulator, bone resorption inhibitor, bronchodilator, cardiovascular, and cholinergic agents; fluorescent, free oxygen radical scavenger, gastrointestinal motility effector, glucocorticoid, and hair growth stimulant agent; hemostatic, histamine H2 receptor antagonists; hormone; hypocholesterolemic, and hypoglycemic agents; hypolipidemic, hypotensive, and imaging agents, immunizing and agonist agents; mood regulators, mucolytic, mydriatic, or nasal decongestant; neuromuscular blocking agents; neuroprotective, NMDA antagonist, non-hormonal sterol derivative, plasminogen activator, and platelet activating factor antagonist agent; platelet aggregation inhibitor, psychotropic, radioactive, scabicide, and sclerosing agents; sedative, sedative-hypnotic, selective adenosine A1 antagonist, serotonin antagonist, and serotonin inhibitor agent; serotonin receptor antagonist, steroid, thyroid hormone, thyroid hormone, and thyroid inhibitor agent; thyromimetic, tranquilizer, amyotrophic lateral sclerosis, cerebral ischemia, and Paget's disease agent; unstable angina, vasoconstrictor, vasodilator, wound healing, and xanthine oxidase inhibitor agent; immunological agents, antigens from pathogens, such as viruses, bacteria, fungi and parasites, optionally in the form of whole inactivated organisms, peptides, proteins, glycoproteins, carbohydrates, or combinations thereof, any examples of pharmacological or immunological agents that fall within the above-mentioned categories and that have been approved for human use that may be found in the published literature, any other hydrophobic bioactive component, or any combination of any of these.
  • As used herein, the term “polypeptide” refers to a polymer of amino acids not limited by the number of amino acids. It is also to be understood that the term “polypeptide” is meant to encompass an oligopeptide, a peptide, or a protein, for example. [0052]
  • As used herein, the term “polynucleotide” refers to RNA or DNA sequences of more than 1 nucleotide in either single chain, duplex or multiple chain form. The term “polynucleotide” is also meant to encompass polydeoxyribonucleotides containing 2′-deoxy-D-ribose or modified forms thereof, RNA and any other type of polynucleotide which is an N-glycoside or C-glycoside of a purine or pyrimidine base, or modified purine or pyrimidine base or basic nucleotide. The polynucleotide may encode promoter regions, operator regions, structural regions, termination regions, combinations thereof or any other genetically relevant material. Similarly, the term “genetic” as used herein, refers to any material capable of modifying gene expression. [0053]
  • The first [0054] aqueous composition 14 may be included in the method of the present invention as a gel, liquid, or in vapor form. The form of the first aqueous composition 14 that is selected preferably permits the first aqueous composition 14 to (1) remain stable prior to dissolving or dispersing the bioactive component, the surfactant composition 16, the surfactant micelle 22, or optionally the stabilizer surfactant micelle 28, (2) homogeneously disperse the bioactive component 12, the surfactant composition 16, the surfactant micelle 22, or optionally the stabilizer surfactant 28, (3) function as a continuous phase in an oil-in-water emulsion, (4) not interfere with, or mask the functional activity of the bioactive component 12, and (5) not modify or degrade the bioactive component 12, the surfactant composition 16, the surfactant micelle 22, or optionally the stabilized surfactant micelle 28.
  • The first [0055] aqueous composition 14 may include only water, or may optionally include additional solutes or solvents that do not interfere with the method of forming the nanocapsules 36 nor mask the functional activity of the bioactive component 12. Furthermore, those of ordinary skill in the art will recognize that an amount of the first aqueous composition 14 used to prepare the nanocapsules 36 may vary depending upon the bioactive component 12, the surfactant composition 16, the temperature, pH, osmolarity, optional solutes or optional solvents, the surfactant micelle 22, the biocompatible polymer component 24, any desired characteristics of the stabilized surfactant micelle 28 or the nanocapsules 36.
  • The [0056] bioactive component 12 may be added to the first aqueous composition 14 or the first aqueous composition 14 may be added to the bioactive component 12. While the order of addition of the bioactive component 12 and the first aqueous composition 14 is not critical to the present invention, the hydrophilic composition (not shown) that is formed when the bioactive component 12 is dissolved or dispersed in the first aqueous composition 14 is preferably capable of maintaining a homogeneous solution or dispersion in the hydrophilic composition.
  • The first [0057] aqueous composition 14 may be supplied as an individual component or supplied in various prepared mixtures of two or more components that are subsequently combined to form the first aqueous composition 14. Some non-exhaustive examples of the first aqueous composition 14 include, but are not limited to, the above-mentioned water, nonpolar solvents, polar solvents, or any combination of any of these. Preferably, water is the first aqueous composition 14.
  • The surfactant composition [0058] 16 maybe introduced into the bioactive component 12, the first aqueous composition 14, the hydrophilic composition as a liquid, vapor or in granular form. The form of the surfactant composition 16 that is selected preferably permits the surfactant composition 16 to (1) remain stable prior to introducing into the bioactive component 12, the first aqueous composition 14, or the hydrophilic composition, (2) be homogeneously dispersed into the bioactive component 12, the first aqueous composition 14, or the hydrophilic composition, (3) form a micellar structure, (4) be adsorbed onto a surface of the bioactive component 12, the first aqueous composition 14, the hydrophilic composition (5) displace the first aqueous composition that is located on the surface of the bioactive component 12, (6) partition the bioactive component 12 or the hydrophilic composition into a core of the micellar structure to form the surfactant micelle 22, and (7) provide a thermodynamic driving force that is effective to reduce a size of the bioactive component 12, surfactant micelle 22, the stabilized surfactant 28 or the nanocapsule 36.
  • As used herein, a “surfactant” refers to any molecule containing a polar portion that thermodynamically prefers to be solvated by a polar solvent, and a hydrocarbon portion that thermodynamically prefers to be solvated by a non-polar solvent. The term “surfactant” is also meant to encompass anionic, cationic, or non-ionic surfactants. As used herein, the term “anionic surfactant” refers to a surfactant with a polar portion that ionizes to form an anion in aqueous solution. Similarly, a “cationic surfactant” refers to a surfactant having a cationic polar portion that ionizes to form a cation in aqueous solution. Likewise, a “non-ionic” surfactant refers to a surfactant having a polar portion that does not ionize in aqueous solution. [0059]
  • While not wanting to be bound to theory, it is generally believed that a surfactant refers to a molecule that is effective to reduce a surface or an interfacial tension between a first substance dispersed in a second substance such that the first substance is solvated and any molecular groups of the first substance are dispersed. Typically, a hydrodynamic diameter of the first substance increases after addition of the surfactant. Nonetheless, the surfactant composition [0060] 16 is believed to be effective to reduce the size or diameter of the surfactant micelles 22 in the first aqueous composition 14, to thereby reduce the size of the nanocapsule 36 when practicing the present invention.
  • The surfactant composition [0061] 16 may include the surfactant component only (not shown), or may optionally include the biocompatible oil component 18. The surfactant component may be characterized on the HLB scale that ranges from less than about 1 to more than about 13 units.
  • A surfactant component having an HLB value of less than about 6.0 units may be described as being poorly, or not dispersable in an aqueous or water-based composition. In addition, a surfactant component having an HLB value of less than about 6.0 units may be characterized as a hydrophobic or non-ionic surfactant. A surfactant component having an HLB value of more than about 7.0 units may be described as being capable of forming a milky to translucent to clear dispersion when the surfactant having an HLB value of more than about 7.0 units is dispersed in an aqueous or water-based composition. [0062]
  • Preferably, the surfactant component of the surfactant composition [0063] 16 has an HLB value of less than about 6.0 units when practicing the method of the present invention. Still more preferably the surfactant component of the surfactant composition 16 has an HLB value of less than about 5.0 units to facilitate preparation of nanocapsules having a diameter of less than about 50 nm.
  • The surfactant component may also be characterized in terms of a critical micelle concentration (CMC) value. Preferably, the surfactant component of the surfactant composition [0064] 16 has a CMC value of less than about 300 micromolars (μm). Still more preferably, the surfactant component has a CMC value of less than about 200 μm.
  • While not wanting to be bound to theory, it is believed that the surfactant component of the surfactant composition [0065] 16 adsorbs onto the surface of the bioactive component 12 when introduced into the first aqueous composition 14 to minimize exposure of a surface of the hydrophobic surfactant component to a thermodynamically unfavorable environment created by the first aqueous composition 14. Therefore, the surfactant component adsorbs onto the surface of the bioactive component to reduce the surface area of the surfactant component that may be exposed to the first aqueous composition 14. Adsorption of the surfactant component onto the bioactive component 12 is believed to facilitate the size reduction of the bioactive component 12 and/or the surfactant micelle 22.
  • The surfactant component of the surfactant composition [0066] 16 may be supplied as individual surfactants or supplied in various prepared mixtures of two or more surfactants that are subsequently combined to form the surfactant composition 16. Some non-exhaustive examples of suitable surfactants having an HLB value of less than about 6.0 units or a CMC value of less than about 200 μm be listed in Dermatological Formulations (Barry, B., Marcel Dekker, (1983)), or in Percutaneous absorption: drug, cosmetics, mechanisms, methodology, 3rd ed., Bronough, R. ed., 1999, or the Handbook of Industrial Surfactants (Ash, M., Ed., Gower Pub. (1993), which are incorporated herein by reference. As an example, the surfactant component maybe 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol(TM-diol), blends of 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol(TM-diol), molecules having one or more acetylenic diol groups, cetyl alcohol or any combination of any of these.
  • The optional [0067] biocompatible oil component 18 of the surfactant composition 16 may be combined with the surfactant component as a liquid, vapor or in granular form. The form of the optional biocompatible oil component 18 that is selected preferably permits the optional biocompatible oil component 18 to (1) remain stable prior to introduction into the surfactant composition 16, (2) be homogeneously blended into the surfactant composition 16, (3) dissolve or disperse the surfactant component, and (4) increase the hydrophobicity of the surfactant composition 16, and therefore, the degree to which the size of the bioactive component 12, the surfactant micelle 22, the stabilizer surfactant micelle 28, or the nanocapsule 36 may be reduced when practicing the present invention.
  • Preferably, the concentration of the optional [0068] biocompatible oil component 18 in the surfactant composition 16 ranges from about 10−7 weight percent to about 10 weight percent, based upon a total volume of the stabilized surfactant micelles 28 in the first aqueous composition 14. Concentrations of the optional biocompatible oil component 18 higher than about 10 weight percent, based upon the total volume of the surfactant composition 18, may be less desirable because such higher concentrations increase a phase volume of the biocompatible oil, and consequently may cause difficulties in preparing, dispersing and/or handling the surfactant micelles 22, the stabilized surfactant micelles 28 or the nanocapsules 36. Concentrations of the optional biocompatible oil component lower than about 10−7 weight percent in the surfactant composition 16 may be less preferred, because such lower concentrations would not be effective to solvate the surfactant component, or increase the hydrophobicity of the surfactant composition 16, and may ultimately increase the diameter of the nanocapsules 36.
  • The optional [0069] biocompatible oil component 18 of the surfactant composition 16 may be supplied as an individual biocompatible oil or supplied in various prepared mixtures of two or more biocompatible oils that are subsequently combined to form the optional biocompatible oil component 18. Some non-exhaustive examples of suitable biocompatible oils that may be included as part of the biocompatible oil component 18 may be found in Dermatological Formulations (Barry, B., Marcel Dekker, (1983)), or in Percutaneous absorption: drug, cosmetics, mechanisms, methodology, 3rd ed., Bronough, R. ed. , 1999, or in the Handbook of Industrial Surfactants (Ash, M., Ed., Gower Pub. (1993), which have been incorporated herein by reference. Preferably, food or USP grade oils, such as DMSO, DMF, castor oil, or any combination thereof, are used to practice the present method.
  • The surfactant composition [0070] 16 may be included at an amount that is effective to form the micellar structure that partitions the bioactive component 12, the first aqueous composition 14 or the hydrophilic composition into the core of the micellar structure when forming the surfactant micelle 22. Still more preferably, the surfactant composition 16 is included at an amount that is effective to provide a maximum thermodynamic driving force that minimizes the size of the bioactive component 12, the surfactant micelle 22, and ultimately, the size of the nanocapsule 36 when practicing the present invention.
  • Furthermore, those of ordinary skill in the art will recognize that the amount of the surfactant composition [0071] 16 may be varied based upon the bioactive component 12, the first aqueous composition 14, a ratio of the surfactant component to the optional biocompatible oil 18, any desired characteristics of the surfactant micelles 22, the stabilized surfactant micelles 28 or the nanocapsules 36. For example, a surfactant composition containing a surfactant component having an HLB value of about 6.0 units mixed with a nonpolar biocompatible oil like castor oil, may provide the same degree of a thermodynamic driving force as a second surfactant composition containing a surfactant component of about 4.0 units mixed with DMSO.
  • The amount of the surfactant composition [0072] 16 may range up to about 0.5 weight percent, based upon a total volume of the stabilized surfactant micelles dispersed in the first aqueous composition 14. Still more preferably, the amount of the surfactant composition 16 is less than about 0.25 weight percent, based upon the total volume of the stabilized surfactant micelles 28 dispersed in the first aqueous composition 14. Most preferably, the surfactant composition 16 is present at an amount of less than about 0.05 weight percent, based upon the total volume of the stabilized surfactant micelles 28 dispersed in the first aqueous composition 14. As one non-exhaustive example, the surfactant composition 16 may be added to the total volume of the hydrophilic composition at a concentration of about 500 ppm, based on the total volume of the stabilized surfactant micelles 28 in the first aqueous composition 14.
  • The [0073] first dispersing apparatus 20 initiates and promotes formation of the micellar structures that are based on the bioactive component 12, the first aqueous composition 14 and the surfactant composition 16. Adsorption of surfactant component onto the surface of the bioactive component 12, or hydrophilic composition continues until all of the surfactant molecules cover, and therefore, entrap the bioactive component 12 or hydrophilic composition in the core of the micellar structure to form surfactant micelles 22. Formation of a plurality of surfactant micelles 22 in the first aqueous composition 14 forms a dispersion of surfactant micelles 22.
  • In general, any [0074] conventional dispersing apparatus 20 that is capable of homogenously blending or dispersing may be suitable for use in forming the dispersion of surfactant micelles in accordance with the present invention. Furthermore, those of ordinary skill in the art will recognize that the first dispersing apparatus 20 may vary depending upon the desired characteristics of the nanocapsules 36. For example, the first dispersing apparatus 20 may include any device, such as a sonicating or a vortexing apparatus (not shown), or the like to disperse the bioactive component 12 in the hydrophilic composition, and the formation of the surfactant micelles 22 after addition of the surfactant composition 16. Nonetheless, while the first dispersing apparatus 20 may include a sonicating or a vortexing apparatus, the sonicating or the vortexing apparatus is not critical when practicing the method of the present invention.
  • As used herein, a “surfactant micelle” may be characterized as a close packed mono-molecular barrier of surfactant molecules at an interface between the [0075] bioactive composition 12 and the surfactant composition 16, such that the barrier encapsulates the bioactive component 12, the first aqueous composition 14 or the hydrophilic composition. It is also to be understood that the term “surfactant micelle” encompasses partial or hemi-surfactant micelles that partially enclose the bioactive component 12, the first aqueous composition 14 or the hydrophilic composition.
  • When the [0076] bioactive component 12 is a hydrophilic bioactive component, the polar portion of the surfactant molecule associates with a surface of the hydrophilic bioactive component. When the bioactive component 12 is a hydrophobic bioactive component, the hydrocarbon portion of the surfactant micelle associates with a surface of the hydrophobic bioactive component.
  • The formation of a surfactant micelle typically occurs at a well defined concentration known as the critical micelle concentration. As noted, surfactant components having a CMC value of less than about 200 micromolars are preferred when practicing the present invention. [0077]
  • After forming the dispersion of [0078] surfactant micelles 22, the dispersion of surfactant micelles 22 is transferred into the stabilizing apparatus 26 where a biocompatible polymer component 24 is added to stabilize the dispersion of surfactant micelles 22. Alternatively, the biocompatible polymer component 24 may be added to the dispersion of surfactant micelles 22 in the first dispersing apparatus 20 which obviates the need for the stabilizing apparatus 26.
  • The [0079] biocompatible polymer component 24 stabilizes the dispersion of surfactant micelles 22 to form stabilized surfactant micelles 28 within the first aqueous composition 14. Therefore, a dispersion of stabilized surfactant micelles 28 are present within the first aqueous composition 14 after addition of the biocompatible polymer component 24.
  • As used herein, the term “biocompatible” refers to a material that is capable of interacting with a biological system without causing cytotoxicity, undesired protein or nucleic acid modification or activation of an immune response. [0080]
  • The [0081] biocompatible polymer component 24 may be introduced into the dispersion of surfactant micelles 22 as a liquid, vapor or in granular form. The form of the biocompatible polymer component 24 that is selected preferably permits the biocompatible polymer component 24 to (1) remain stable prior to addition into the dispersion of surfactant micelles 22, (2) be homogeneously dispersed into the dispersion of surfactant micelles 22, (3) increase a viscosity of the first aqueous composition 14, (4) form a boundary layer at an interface of the surfactant micelle 22 and the first aqueous composition 14, (5) be absorbed onto a surface of the surfactant micelles 22, (6) be capable of iontophoretic exchange, (7) be capable of being precipitated upon addition of a solute, (8) be capable of enzymatic degradation, surface and/or bulk erosion, (9) not interfere with or mask the functional activity of the bioactive component 12, (10) prevent aggregation and/or agglomeration of the dispersion of surfactant micelles 22, and (11) be capable of obtaining a particular dissolution profile.
  • The [0082] biocompatible polymer component 24 may be included at an amount that is effective to coat and therefore stabilize the surfactant micelle 22. Furthermore, those of ordinary skill in the art will recognize that the amount of the biocompatible polymer component 24 used to stabilize the surfactant micelles 22 may vary depending upon the bioactive component 12, the first aqueous composition 14, the surfactant composition 16, the temperature, pH, osmolarity, presence of any optional solutes or optional solvents, the surfactant micelle 22, any desired characteristics of the stabilized surfactant micelle 28, the nanocapsules 36, or a desired dissolution profile.
  • While the concentration of the [0083] biocompatible polymer component 24 is not critical to the present invention, the concentration of the biocompatible polymer component 24 is preferably based upon the bioactive component and on the desired dissolution profile. When the concentration of the biocompatible polymer component 24 is to high, the shell of the nanocapsule 36 may not dissolve. If the concentration of the biocompatible polymer component 24 is to low, the shell of the nanocapsule 36 may dissolve rapidly in a manner that promotes cytotoxicity, for example. In addition, too low a concentration of the biopolymer component 24 may not provide an effective degree of mechanical force to stabilize the surfactant micelles 28.
  • Concentrations of the [0084] biocompatible polymer component 24 that are to high may also be less desirable because such higher concentrations may increase the viscosity of the first aqueous composition 14, and consequently may cause difficulties in preparing, mixing and/or transferring the stabilizer surfactant micelles 28. Concentrations of the biocompatible polymer component 24 that are to low may be less preferred, because lower concentrations may not provide the needed viscosity to stabilize the surfactant micelles, nor be capable of effectively coating the surfactant micelles 22 to prevent aggregation of the surfactant micelles 22 in the first aqueous composition 14.
  • The [0085] biocompatible polymer component 24 may be supplied as individual biocompatible polymers or supplied in various prepared mixtures of two or more biocompatible polymers that are subsequently combined to form the biocompatible polymer component 18. Some non-exhaustive examples of biocompatible polymers include polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetage phthalate, carboxylethyl cellulose, cellulose triacetate, cellulose sulphate sodium salt, poly(methyl methacrylate), poly(ethylmethacrylate), poly(butylmethacrylate), poly(isobutylmethacrylate), poly(hexlmethacrylate), poly(isodecylmethacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, polypropylene poly(ethylene glycol), poly(ethylene oxide), poly(ethylene terephthalate), poly(vinyl alcohols), poly(vinyl acetate, poly vinyl chloride polystyrene, polyvinylpryrrolidone, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutylmethacrylate), poly(hexlmethacrylate), poly(isodecl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecl acrylate) and combination of any of these.
  • Additionally, biocompatible polymers that have been modified for enzymatic degradation, or change upon application of light, ultrasonic energy, radiation, a change in temperature, pH, osmolarity, solute or solvent concentration may also be included as part of the [0086] biocompatible polymer component 24. Preferably, the biocompatible polymer component 24 is a hydrophilic polymer that is capable of substantially coating, and preferably continuously coating the surfactant micelle 22. Still more preferably, the hydrophilic biocompatible polymer component 24 is capable of ionotophoretic exchange.
  • Though descriptions of the present invention are primarily made in terms of a hydrophilic [0087] biocompatible polymer component 24, it is to be understood that any other biocompatible polymer, such as hydrophobic biocompatible polymers may be substituted in place of the hydrophilic biocompatible polymer, in accordance with the present invention, while still realizing benefits of the present invention. Likewise, it is to be understood that any combination of any biocompatible polymer may be included in accordance with the present invention, while still realizing benefits of the present invention.
  • In general, any conventional apparatus and technique that is suitable for permitting the [0088] biocompatible polymer component 24 to stabilize the surfactant micelles 22 may be used as the stabilizing apparatus 26 in accordance with the present invention. Furthermore, any other device, such as high pressure homogenization or high ultrasound sonication is preferably not included during stabilization.
  • After stabilizing the [0089] surfactant micelles 22, the stabilized surfactant micelles 28 may be transferred into a second aqueous composition 30 located in a second dispersing apparatus 32. The stabilized surfactant micelles 28 may be transferred by mechanically forming droplets of the stabilized surfactant micelle 28 that are subsequently introduced into the second aqueous composition 30.
  • The second [0090] aqueous composition 30 may include water only, or may optionally include a solute to precipitate the biocompatible polymer component 24 surrounding the stabilized surfactant micelle 28. Some non-exhaustive examples of solutes that may be used to precipitate the biocompatible polymer 24 include ionic species derived from elements listed in the periodic table.
  • Preferably, the second [0091] aqueous composition 30 includes a solute in an amount that is effective to precipitate the biocompatible polymer component 24 and form the dispersed, and optionally atomized nanocapsules 36 of the present invention. As used herein, the term “precipitate” refers to a solidifying or a hardening of the biocompatible polymer component 24 that surrounds the stabilized surfactant micelles 28. It is also to be understood that the term “precipitation” is also meant to encompass any crystallization of the biocompatible polymer 24 that may occur when the biocompatible polymer component 24 is exposed to the solute.
  • Additionally, any other component that is capable of modulating the efficacy the [0092] nanocapsules 36 may be included as part of the second aqueous composition to thereby modulate the functional activity of the nanocapsule 36. For example, the second aqueous composition may include additional coating excipients, such as a cell recognition component or various ionic species, such as Mn2+, Mg2+, Ca2+, Al3+, Be2+, Li30 , Ba2+, Gd3+, or any other ionic species that is capable of interacting with the biocompatible polymer component 24.
  • The term “cell recognition component”, as used herein, refers to a molecule capable of recognizing a component on a surface of a targeted cell. Cell recognition components may include an antibody to a cell surface antigen, a ligand for a cell surface receptor, such as cell surface receptors involved in receptor-mediated endocytosis, peptide hormones, and the like. [0093]
  • It has been observed that when the stabilized [0094] surfactant micelles 28 are allowed to incubate in the second aqueous composition 30 that includes the solute to precipitate the biocompatible polymer component 24, the nanocapsules 36 undergo a reduction in size. Furthermore, the formation of a flocculated suspension of the nanocapsules 36 has also been observed after incubating the stabilized surfactant micelles 28 in the second aqueous composition.
  • As used herein, “a flocculated suspension” refers to the formation of a loose aggregation of discrete particles held together in a network-like structure either by physical absorption of bioactive components, bridging during chemical interaction (precipitation), or when longer range van der Waals forces of attraction exceed shorter range forces of repulsion. The flocculated suspension of [0095] nanocapsules 36 may entrap varying amounts of the first aqueous composition 14 or the second aqueous composition 30 within the network-like structure. Additionally, the flocculated suspension of nanocapsules may be gently tapped to disperse the nanocapsules 36.
  • The stabilized [0096] surfactant micelles 28 may be transferred into the second aqueous composition 30 via atomization through a nozzle (not shown) having a particular orifice size or through an aerosolizing apparatus (not shown). Atomizing or aerosolizing the stabilized surfactant micelles 28 typically includes the application of a shear force that may be capable of further dispersing the stabilized surfactant micelles 28. Furthermore, the application of the shear force during transfer may also be effective to (1) reduce the size of the nanocapsules 36, or (2) break up any agglomerates or associations between stabilized surfactant micelles 28 that may have formed in the stabilizing apparatus 26. Nozzle pressures of less than about 100 psi, for example, may be used to atomize the stabilized surfactant micelles 28.
  • The diameter of the [0097] nanocapsules 36 may also be varied depending upon the orifice size of the nozzle that may be used to transfer the stabilized surfactant micelles 28 into the second aqueous composition. Alternatively, the stabilized surfactant micelles 28 may be added to the second aqueous composition 30 containing the solute that precipitates the biocompatible polymer 24 to form a dispersion of nanocapsules 36 for purposes of providing the dispersion for sub-cutaneous delivery of the nanocapsules, for example.
  • After precipitating and/or optionally incubating the [0098] nanocapsules 36 in the second aqueous composition 30, the nanocapsules 36 may be filtered, centrifuged or dried to obtain separate and discrete nanocapsules 36. The nanocapsules 36 may be frozen or reconstituted for later use or may be delivered to a target cell or tissue by such routes of administration as oral, intravenous, subcutaneous, intraperitoneal, intrathecal, intramuscular, inhalational, topical, transdermal, suppository (rectal), pessary (vaginal), intra urethral, intraportal, intrahepatic, intra-arterial, intra-ocular, transtympanic, intraumoral, intrathecal, or any combination of any of these.
  • The [0099] nanocapsules 36 having a diameter of less than about 50 nm are advantageous in the delivery of bioactive components to target cells for several reasons. First, nanocapsules 36 having a diameter of less than about 50 nm enhances delivery of bioactive components by protecting the bioactive components against degradation during transport to the target cell.
  • Second, nanocapsules [0100] 36 having a diameter of less than about 50 nm promotes efficient cellular uptake. Efficient cellular uptake into the target cell typically occurs when a particle has a diameter of less than about 50 nm, as opposed to when a particle has a diameter of more than about 50 nm.
  • Third, it is believed that uptake of the [0101] nanocapsules 36 by the target cell occurs via transport systems, such as a non-endosomal pathway, that prevents lysosomal degradation of the nanocapsules 36. Indeed, it is believed that the nanocapsules 36 of the present invention are efficiently exported into a cell via a caveolin-regulated pathway that circumvents most, if not all, endosomal-regulated pathways that typically degrade nanocapsules 36.
  • Fourth, the [0102] nanocapsules 36 have a biocompatible polymer shell that is separate from the bioactive component. In fact, the bioactive component is not entangled in, embedded in, or adsorbed onto the biocompatible polymer shell of the nanocapsules 36. When the bioactive component is not entangled in, embedded in, or adsorbed onto the biocompatible polymer shell, the cell that incorporate the nanocapsules 36 avoid apoptosis or cell death.
  • Fifth, enclosing the bioactive component within a core surrounded by the biocompatible polymer shell when preparing the [0103] nanocapsules 36 in accordance with the present method is advantageous in avoiding premature degradation of the nanocapsules 36, or a cytotoxic response during in vivo transport of the nanocapsule. Enclosing the bioactive component within the core results in a linear release rate of the bioactive component without any zero burst phenomenon during release from the nanocapsules 36.
  • The linear release rate of the bioactive component from the nanocapsule without any zero burst phenomenon is also an advantageous feature as the linear release rate allows rational design of coating dissolution profiles to minimize cytotoxicity. As used herein, the term “dissolution profile” refers to a rate at which the biocompatible polymer shell is dissolved or degraded to release a bioactive agent from a core of a nanocapsule. [0104]
  • Another benefit of the [0105] nanocapsules 36 prepared by the method of the present invention is that little, if any, addition of an organic solvent is required to form the nanocapsules 36. Eliminating the use of most, if not all, organic solvents from the method of the present invention is beneficial since organic solvents may damage the bioactive component 12, destroy the target cells, or be toxic during preparation of the nanocapsule 36. The elimination of most, if not all, use of organic solvents eliminates the need for complex solvent removal techniques, such as solvent dilution, vacuum evaporation, or the like, and obviates any associated costs or complex process strategies during preparation of the nanocapsules 36.
  • The [0106] nanocapsules 36 of the present invention further permits stable encapsulation of a bioactive component, and in particular, hydrophilic bioactive components, such as polynucleotides and polypeptides. “Stable encapsulation”, as used herein, refers to maintenance of the encapsulated bioactive component's structure. For nucleic acids, the appearance of low molecular weight nucleic acid breakdown products, which may be assayed for by electrophoresis, is substantially eliminated. The nanocapsules 36 may also be used to encapsulate any bioactive component regardless of water solubility or charge density.
  • APPLICATIONS
  • The [0107] nanocapsules 36 may be combined with additional polymeric binders, surfactants, fillers, and other excipients to incorporate the nanocapsules 36 into solid dosage forms such as granules, tablets, pellets, films or coatings for use in enhanced bioactive component 12 delivery. In this way, design of the dissolution profile, control of the particle size, and cellular uptake remains at the level of the nanocapsule. Such applications include, but are not limited to, creation of rapidly dissolving nanocapsule pellets for pulmonary delivery or nanocapsule films for device-mediated delivery.
  • In another application, the [0108] nanocapsules 36 may be designed for specific cellular or tissue uptake by polymer selection and/or inclusion of cell-recognition components in the nanocapsule biocompatible polymer shell or coating. Such coatings will have utility for specific or increased delivery of the bioactive agent to the target cell. Such applications include, but are not limited to tumor-targeting of chemotherapeutic agents or anti-sense DNA, antigen delivery to antigen-presenting cells, ocular delivery of ribozymes to retinal cells, transdermal delivery of protein antibodies, or transtympanic membrane delivery of peptide nucleic acids.
  • Property Determination and Characterization Techniques [0109]
  • Various analytical techniques are employed herein. An explanation of these techniques follows: [0110]
  • FIG. 1A: Samples were prepared on freshly cleaved mica as dispensed, dried in air and imaged using a Nanoscope II multimode AFM (Digital Instruments) with a J type scanner and ambient tapping mode holder. 125 μm long silicon cantilevers type IBMSC were from IBM and have resonant frequencies of 250-450 kHz. All imaging was in tapping mode, images were 512×512 pixels and scanning frequency was 1 kHz. Height, amplitude and phase images were collected. Images were processed in DI software and analyzed in NIH Image SXM. A: Formula Q from 2-phase system, low HLB surfactant, B: Formula S from 2-phase system, high HLB surfactant, C: Formula T from 1-phase system, high HLB surfactant, D: Formula V from 2-phase system, surfactant below CMC. [0111]
  • FIG. 1B: Nanocapsules were released into a solution of 10% isobutanol in Phosphate-buffered Saline (PBS), pH=7.2. Samples were run in duplicate. FIG. 1C: Nominal 300 ng samples of DNA were aliquoted from a master batch containing surfactant and processed through commercial miniprep columns. Eluate was recycled through Qiaquik columns and collected either 3 times (4, 5) or twice (6,7) or recycled through Zymoclean columns and collected twice (8,9). Samples were alcohol precipitated using a commercial coprecipitant, electrophoresed on 1.5% agarose gels modified with Synergel, stained with SybrGold dye, digitized on a Storm 860 and compared to unmodified but reprecipitated samples from the same master batch (10,11). Lanes 1-3: 100, 50 and 5 ng of ?_DNA. [0112]
  • FIG. 2: Endocytic activity was assessed by immunosignal levels of clathrin (Chemicon). Potocytotic activity was assessed by immunosignal for caveolin-1 as described in the literature (Transduction Laboratories). Lysosomal activity was detected by an monoclonal antibody to Lamp-1 (Transduction Laboratories). Nanocapsule localization was detected by streptavidin-biotin immunocomplexes directed against sheep IgG (Jackson Laboratories). Nanocapsule coatings were spiked with ovine IgG to enable this detection strategy. [0113]
  • FIG. 3: immortalized Rt-1 fibroblast cultures at 70% confluence were treated for 4 days with increasing amounts of nanocapsule formula K and transiently treated (3 hours) with an optimized liposomal formula (dose, 500 ng) Results are expressed as a percentage of cellular actin integrated intensity and compared to the liposomal formula. Expression vector was code 448: pEF/myc-his/GFP (Invitrogen). [0114]
  • FIG. 4A: Radiated porcine biopsies were snapfrozen 7 days after treatment with saline or 6 μg of controlled release nanocapsules, then homogenized in RIPA. 100 μg lysate samples were electrophoresed on SDS-Page gradient gels, transferred to nitocellulose membranes and detected for either §-galactosidase (121 Kd) or involucrin (˜100 kD) using chemiluminescence. Results were normalized to the post-transfer gel stained with Coomassie due to interference at 100 kD from a gel defect. Involucrin, a component of the cornified membrane, manufactured by suprabasal cells can be detected in radiated porcine skin and will be used for future normalization purposes. Lane A: N, topical, biopsy oc-2; B: N, topical, biopsy oc-3; C: 0, topical, biopsy 1-1; D: PBS only, biopsy 1-5; E: N, subcutaneous injection, biopsy 1-6. [0115]
  • FIG. 4B: The β-galactosidase reporter protein was detected by a monoclonal antibody directed at an incorporated fusion protein tag. A: N, topical, biopsy oc-1, detection with anti-Xpress[0116] a; B: Matching view to A with detection for anti-von Willenbrand factor (Sigma);C: untreated biopsy, detection with anti-Xpressa(Invitrogen).
  • FIG. 5: Nanocapsules were incorporated into an aqueous suture coating and sutures were applied to pigskin biopsies in organ culture. Nanocapsules were detected with Cy3 conjugated-streptavidin-biotin complexes to incorporated ovine IgG and GFP transgene expression was detected by rabbit polyclonal antibodies to GFP (Abcom) in combination with Fitc-conjugated polyclonal antibodies to rabbit IgG and Alexa 488-conjugated polyclonal antibodies to Fitc (Molecular Probes). Controls omitting primary antibodies were included for signal-to-background level estimation. [0117]
  • FIG. 6A: Nanocapsules were detected as previously described and GFP transgene expression was detected by rabbit polyclonal antibodies to GFP in combination with Cy3-conjugated antibodies to rabbit IgG (Jackson Laboratories). [0118]
  • FIG. 6B: GFP expression was detected as described in FIG. 5 and cell nuclei were counterstained with 10 μg/ml bisbenzamide. [0119]
  • FIG. 6C: Carcinoma cells and HDF's were seeded overnight into 96 well plates at 2000 and 6000 cells per well respectively. Cisplatin preparations were added to wells for 18 hours as noted on the graph than washed out. After 72 hours cell viability of assessed by a commercial MTT assay (WST assay, Boehringer Mannheim). Wells were executed in duplicate. [0120]
  • FIG. 7: Colocalization with lysosomes was detected using a monoclonal antibody to Lamp-1 (Transduction Laboratories). AFM images are included of O-methyl RNA formulated by nanoencapsulation or complexation with 27 KD polyethyleneimine. [0121]
  • EXAMPLES
  • The present invention is more particularly described in the following Examples which are intended as illustrations only since numerous modifications and variations within the scope of the present invention will be apparent to those skilled in the art. [0122]
  • Reagents: [0123]
  • A. Nucleic acid condensing agents [0124]
  • Poly(ethylenimine) (PEI) at 27 KiloDalton (kD). PEI was used at optimized conditions (90% charge neutralization) [0125]
  • Polylysine (PLL) at 70-150,000 molecular weight. PLL condensing materials were conjugated with nuclear signal localization peptides, either SV-40 T antigen or cys-gly-tyr-gly-pro-lys-lys-lys-arg-lys-val-gly-gly using carboxiimide chemistry available from Pierce Chemical (Rockford, Ill.). [0126]
  • Preparations of nuclear matrix proteins (NMP). NMP were collected from a rat fibroblast cell line, and a human keratinocyte cell line using a procedure described in Gerner et al. [0127] J. Cell. Biochem. 71 (1998):363-374 which is incorporated herein by reference. Protein preparations were conjugated with nuclear signal localization peptides as described.
  • B. Surfactants [0128]
  • 2, 4, 7, 9 - tetramethyl-5-decyn-4, 7 - diol (TM-diol): HLB=4-5, CMC is not determined [0129]
  • Poly(oxy-1, 2-ethanediol), a-(4-nonylphenol)-w-hydroxy, Tergitol NP-40 (NP40): HLB=17.8, CMC 180 μM, [0130]
  • [0131] Polyoxyethylene 20 sorbitan monooleate (Tween 80): HLB=10, CMC 920 μM, Cetyl Alcohol: HLB=4,CMC is not determined.
  • C. Polymers [0132]
  • Hyaluronan, bacterially-derived, 1 million kiloDalton (MM kD) and conjugated with nuclear localization signal peptides as described in U.S. Pat. No. 5,846,561, which is incorporated herein by reference. [0133]
  • Hyaluronan, derived from human umbilical cord, about 4MMKD and not conjugated. [0134]
  • Povidone (polyvinylpyrolidone, PVP) 10,000 kD MW and not bioconjugated Povidone (polyvinylpyrolidone, PVP) 40,000 Kd MW and not bioconjugated Povidone (polyvinylpyrolidone, PVP) 360,000 kD MW and not bioconjugated Tenascin, 220 kD and not bioconjugated. [0135]
  • D. Expression Vectors [0136]
  • 334: pcDNA/His/LacZ, produces galactosidase, incorporates CMV promoter, based on pcDNA 3.1. (Invitrogen), 8.6 kB [0137]
  • 425: pEGFP-c/farn, enhanced GFP (green fluorescent protein) expression vector modified with a farnasyl moiety to improve microscopy, CMV promoter, 4.6 kB [0138]
  • 423: pEGFP-c3/p57(Kpn/Sma) Clontech enhanced GFP (green fluorescent protein) expression vector modified with a nuclear localization tag from a cyclin dependent kinase to improve microscopy, 4.6 kB [0139]
  • E. Cells [0140]
  • CCRL 1764: Immortalized rat neonatal fibroblast cell line [0141]
  • HaCaT: immortalized human keratinocyte cell line [0142]
  • Ca9: human tumor cells derived from a squamous cell carcinoma of tongue origin. [0143]
  • Example 1A—Effect of changing dispersion conditions on hydrophillic nanocapsules.
  • The importance of appropriate dispersion conditions was investigated in the following series of formulations. Formulae were produced by i) predispersing 25 μg of DNA (425) on ice using a bath sonicator, ii) condensing DNA in a small amount of water by vortexing then incubating on ice for 20 minutes, iii) adding surfactant then oil followed by 30 seconds of probe sonication at 10 Watts, iv) diffusion dilution to 3 milliliters (mL) by first adding saline then 1 MM kD hyaluronan polymer (1%) as a protective colloid, v) mechanically shearing emulsion into droplets by pumping through a 250 micrometer (μm) orifice into 22 mL of PBS, 10 milliMolar (mM) Ca[0144] 2+, 200 mM Li+, vi) incubating overnight end over end and vii) centrifuging to recover nanoparticles for resuspension and filter sterilization. The condenser-to-DNA weight ratio was determined by dye exclusion at 90% charge neutralization. TM-diols were used in this experiment to represent water-immiscible surfactants, while Tergitol NP40 and Tween 80 were used to represent water-soluble and even more water-soluble emulsifiers/dispersing aids.
  • Dispersion conditions were systematically varied to discourage micelle formation in aqueous media by i) choosing water-soluble surfactants (Formulae S,T,U, and V), ii) removing the dispersed phase (Formula T) and iii) decreasing surfactant loading below that required for micelle formation. Formula U featured use of a water-miscible oil (silicone oil). [0145]
  • Formulas were characterized physically and tested for functionality in in vitro gene transfer. Quantitative results are summarized in Table 1A: [0146]
    TABLE 1A
    Effect of changing dispersion conditions on hydrophillic
    nanocapsules.
    Formula
    Q R S
    Experimental surf > CMC surf > CMC surf > CMC
    Modification:
    Critical Micelle ˜0 ˜0 360 ppm
    Concentration (CMC)
    Pre-aerosol surfactant 500 ppm 500 ppm 600 ppm
    Concentration (3 ml
    basis)
    HLB number 4-5 4 17.8
    Phases Water/ Water/ Water/
    misc. oil misc. oil misc. oil
    Formula
    Characteristics:
    Nucleic Acid 86 ± 8   67 ± 1.4 50.3 ± 12  
    Incorporation (%)
    Low MW DNA 15.00 76 93.00
    Appearance
    (% above bkground,
    Post nanocapsule
    digest by
    electrophoresis)
    Supercoil retention 87% 65% 66%
    (post
    100 hrs release)
    (area %, initial
    distribution = 76%
    supercoiled)
    Particle Size 42 ± 2  45 ± 3  73 ± 4 
    (mean ± SE)
    Secondary Structure(s) 25% 30% 70%
    Flocculation Status 100-200 nm 500 nm 300 nm
    stringy flocs stringy flocs spheroid
    aggregates
    Comments:
    Performance:
    Transduced GFP 420 340 0
    Protein Generation
    (pixel units, % of
    control liposome
    formula, 100 μg total
    protein, Day 11)
  • [0147]
    TABLE 1A
    Effect of changing dispersion conditions on hydrophillic
    nanocapsules.
    Formula
    T U W V
    Experimental surf > CMC surf > CMC surf > CMC surf < CMC
    Modification:
    Critical Micelle 360 ppm 360 ppm 1200 ppm 360 ppm
    Concentration
    (CMC)
    Pre-aerosol 600 ppm 600 ppm 4000 ppm  90 ppm
    surfactant
    Concentration (3 ml
    basis)
    HLB number 17.8 17.8 10 17.8
    Phases Water only Water/immisc. oil Water/misc. oil Water/misc. oil
    Formula
    Characteristics:
    Nucleic Acid  39 ± 1.7 32.8 ± 6     37 ± 1.41 57.6 ± 16  
    Incorporation (%)
    Low MW DNA 53.00 66 28 41.00
    Appearance
    (% above bkground,
    Post nanocapsule
    digest by
    electrophoresis)
    Supercoil retention 59% 43% 65% 80%
    (post 100 hrs
    release)
    (area %, initial
    distribution = 76%
    supercoiled)
    Particle Size (mean ± SE) 226 ± 11  291 ± 25  150 ± 7  199 ± 11 
    S < 10% S < 10% S > 40% S > 80%
    400 nm
    Secondary yeast-like aggregates
    Structure(s) aggregates
    Flocculation Status
    ppt. during ppt. during ppt. during
    Comments: aerosolization aerosolization aerosolization
    Performance: 0 0 0 0
    Transduced GFP
    Protein Generation
    (pixel units, % of
    control liposome
    formula, 100 μg
    total protein, Day
    11)
  • Nanocapsule sizing was determined by tapping mode AFM and images are illustrated in FIG. 1A. The data indicate average nanocapsule sizes less than 50 nm are achievable only with multi-phase systems in combination with low water solubility surfactants (Table 1A: Formulae Q,R vs. S,T,U,V, and W). Furthermore, only nanocapsules of less than 50 nm resulted in detectable transgene production in CRL-1764 rat fibroblast cells (Table 1A). Effective dispersion also corresponded with decreased aggregation and enhanced DNA stability (as indicated by decreased electrophoretic breakdown products). The starting DNA was partially relaxed (76% supercoiled by electrophoresis). Using this value as a basis, supercoil retention in DNA still encapsulated following 100 hrs of release testing, was excellent in multi-phase systems. [0148]
  • Release profiles for hydrophillic dispersed atomized nanocapsules were linear, showed no zero burst and resulted in about 60% release after 72 hours (See FIG. 1B). Formula W, manufactured with the most water-soluble surfactant in the series (Tween 80) failed to completely release loaded DNA. FIG. 1C illustrates that small amounts of DNA (in this case 300 nanograms of DNA) can be recovered accurately in a procedure comprising butanol extraction of 10% butanol/saline releasing fluid followed by isolation on a miniprep column and measurement of absorbance at 260 nm excitation. Results obtained from UV spectroscopy are confirmed by electrophoresis of recovered DNA following alcohol coprecipitation with a commercial coprecipitant aid. Experiment 1A demonstrates the importance of a multi-phase system in creating coated particles from the micellar solution, defines surfactant requirements and validates method for measuring in vitro release profiles. [0149]
  • Example 1B—Effect of process parameters on particle functionality
  • To investigate the effect of modulating process parameters on nanocapsule functionality for DNA delivery, a series of formulas (designed to release in 3 days) were prepared and measured transduction efficiency of these formulas for delivering a nuclear Green Fluorescent Protein (GFP) reporter transgene in [0150] rat fibroblast cultures 5 days later. Charge neutralization of the DNA molecule, the surfactant/oil system, total surfactant phase volume, the inclusion of probe sonication, the absolute requirement for atomization and receiving bath osmolality were modulated. Results for this experiment are summarized in the Table 1B:
    TABLE 1B
    Effective of process parameters on particle functionality
    Oil Receiving
    Nano charge Phase Atomize bath
    capsule Formula neutralization by Biocompatible Volume Emulsify by Diameter Osmolality
    Design Name condensor Surfactant Oil (%, 4.5 ml basis) sonication (μm) (mOs)
    1 q.co.2 + Cetyl Castor 4 + 250 220
    OH oil/Etoh
    2 q.co Cetyl Castor 4 + 250 220
    OH oil/Etoh
    3 o.35 + TM-diol DMSO 4 + 1.4 220
    4 ea0.2 + TM-diol DMSO 4 220
    5 ea0.1 TM-diol DMSO 4 220
    6 ed0.2 + TM-diol DMSO 0.05 250 220
    7 ed0a.12.di + TM-diol DMSO 0.05 250 0
    Transduction
    Nanocapsule Encapsulation Efficiency, (5
    Nanocapsule Formula diameter (nm)* yield days, rat
    Design name n = 20 (%, mean ± SE) fibroblasts)
    1 q.co.2 20 ± 3, rods 48.6 ± 11 87 ± 7%
    2 q.co 12 ± 0.7, irregular 48.6 ± 2 71 ± 28%
    3 o.35 17 ± 1.2, spheres 82.3 ± 7 (4) 86 ± 2%
    4 ea0.2 24 ± 2, s/r   32 ± 10 72 ± 2%
    5 ea0.1 36 ± 3, irregular   57 ± 2 85 ± 1%
    6 ed0.2 39 ± 3, r/e   39 ± 5  96%
    7 ed0a12.di 39 ± 3, ellipse   69 ± 2 100%
  • Aqueous dispersion of DNA condensates with poorly soluble surfactants in the inventive method produced average nanocapsule diameters under 50 nm. A 30 number of successful operating regimes were feasible with varying effects on encapsulation yield. In a cetyl alcohol/castor oil system, under condensation resulted in an average particle diameter increase from 20 to 12 nm (Table 1B: F1vs. F2). The same decrease in condenser weight ratio induced a particle size increase from 24 to 36 nm, while still maintaining nanocapsule functionality for trangene delivery, when using a TM-diol/DMSO surfactant system for initial micelle formation (Table 1B: F4 vs. F5). This finding teaches surfactant selection impacts final average nanocapsule diameters. [0151]
  • Moderate energy input was removed (dropped probe sonication, atomization but kept bath sonication) during nanocapsule formation and resulted in functional particles with decreased yield (Table 1B: F3 vs. F4). This finding indicates that optimal nanocapsule production is not dependent on any spontaneous micro-emulsification process. Cosolvent phase volume was reduced from 4 weight percent to 500 ppm without any negative effect on particle functionality (Table 1B: F4 vs. F6). This finding indicates that essentially solvent-free nanocapsules can be made by the inventive method. Finally, salt was removed from the atomization receiving bath without any negative effects on nanocapsule functionality (Table 1B: F6 vs. F7). [0152]
  • Example 2—Effect of nanocapsule sizing on a nanocapsule uptake in human keratinocytes
  • The effect of nanocapsule sizing on intracellular trafficking in immortalized HacaT human keratinocyte cultures (HacaT's) was investigated in this example. In this series of formulae, thee micellar dispersion were sheared by syringes of different orifice diameter. The coating weight was also lowered from 1:1 DNA:Polymer (w/w) to 1:40 to shorten the dissolution profile from 5 to 3 days. In these experiments, nanocapsule formulae were compared to standard polyplexes of DNA and PEI, and lipoplexed plasmid DNA. Table 2 summarizes the experimental design and results: [0153]
    TABLE 2
    Effect of particle size on nanocapsule functionality for gene transfer
    4 hr. 4 hr. 10 hr. Transduction
    Particle Size colocalization colocalization colocalization Efficiency.
    Formula (mean, nm; with with with (5 days, human
    Name morphology) caveolin-l* cathrin lysosomes keratinocytes)
    o.22 (64) 47 ± 3, rods 0 ++ + 16 ± 13
    o.27 (57) 21 ± 2, rods + + ND 81 ± 8
    o.35 (85) 17 ± 1.2, spheres +++ 0 0 78 ± 9
    pei- 67 ± 4, 0 +++ +++ 40 ± 15
    pDNA spheres, irreg
    Lipoplex
    48 ± 2 + + +++ 41 ± 27
    pDNA 200 nm
    aggregates
  • It was observed that compared to the unstimulated state, nanocapsules increased cellular pinocytotic activity compared to standard formulations, and smaller nanocapsules shifted pinocytotic activity to caveolae from clathrin-coated pits (Table 2: Formula O vs. pei-dna and lipoplex pDNA). It was further observed that nanocapsules avoided lysosome co-localization at 10 hours post-addition with smaller nanocapsules being particularly effective (see Table 2: Formula vs. pei-dna and lipoplex pDNA). These results are illustrated further in FIG. 2. This improvement is further emphasized by comparison with published uptake studies for HacaT keratinocytes. Compared to primary keratinocytes, uptake of naked DNA oligonucleotides (20 μm) were very poor in HacaT's and showed accumulation of oligonucleotides in punctate vesicles consistent lysosomes at 2 hours. Using hydrophillic dispersed atomized nanocapsules of the inventive method, complete avoidance of lysosomes at 10 hours post-addition was demonstrated. These results indicate that products of the inventive process will have increased and prolonged effectiveness. [0154]
  • Example 3—Effect of nanoparticle delivery on DNA and reagent-induced cytoxicity
  • To test whether soluble exogenous DNA released from liposomes or dendrimers induces apoptosis, Rt-1's were treated with loaded/unloaded liposome complexes, dendrimer complexes, nanoparticles and 1 μg/ml etoposide, a DNA intercalating agent as a positive control. Cultures were treated with standard formulas for 3 hours then assayed for [0155] gene product expression 30 hours later. Cultures were treated with nanocapsules for 4 days to ensure full DNA release during the experiment. Controls included as a positive control for apoptotic cell death, 1 μg/ml etoposide, a DNA intercalating agent was applied to cultures overnight before experiment termination. Other controls included standard PEI-DNA complexes, empty nanocapsules and nanocapsules containing empty vector plasmid DNA. Hydrophillic nanoparticles were produced for this experiment as described earlier using a 35-gage syringe.
  • One of the later steps in apoptosis is DNA fragmentation mediated by activation of endonucleases during the apoptic program. Therefore, DNA fragmentation was assayed by end-labeling of fragments using an exogenous enzyme and a substituted nucleotide (TUNEL: tdt-mediated uridine nucleotide and labeling. Results are expressed as a Fragmentation Index, or the percent of cells in the total culture exhibiting BRDU end-labeled DNA. Cultures were run in duplicate. The experimental design and results are summarized in Table 3: [0156]
    TABLE 3
    Effect of nanocapsule coating weight on nonspecific reagent and plasmid DNA-
    associated cytoxicity.
    Formula
    κ.35 ζ ο(Omicron) b.35
    Particle Design:
    DNA Condensing Denatured h. 100 Kd MW 27 kD 27 kD
    Agent keratinocyte Polylsine PEI PEI
    nuclear protein
    Coating Ratio 0.1 0.25 0.25 0.01
    (DNA/polymer)
    Performance:
    dose: (30 hrs for Std. 4.6 4.1 4 5
    Formulas, 100 hrs
    for nanocapsules)
    Cytotoxicity: ND  0.26 ± 0.15   2 ± 0.7 1.9 ± 0.6
    (Fragmentation
    Index, %)
    cytotoxicity controls:
    (1 μg etoposide (8 hr):
    25 ± 10%) (Pei-
    DNA polyplexes
    (100 hr): 24 ± 7%)
    (Empty vector
    nanocapsules: 1.25 ± 1.25%)
    (Empty
    vector
    nanocapsules: 0.9 ± 0.7%)
    Transduction 31 ± 2 ND 85 ± 7  32 ± 3 
    Efficiency: (% cells)
    120 hrs, dose as
    listed)
    Formula
    Characteristics:
    Nucleic: Acid  55 ± 10 27 ± 7 54 ± 5  65 ± 4 
    Incorporation: (%)
    Cumulative Release: 70 75 ± 8 83 ± 12 ND
    (%, 48 hr)
    Particle Size (mean ± SE, 26 ± 2 22 ± 2 20 ± 1  35 ± 2 
    nm)
    Agglomerates (as few 50% 80 ± 6 200 nm 200 nm
    dispensed)
    Formula
    Y.35 Lipoplex GP Lipoplex L+ Polyplex
    Particle Design:
    DNA Condensing 27 kD cationic cationic dendrimer
    Agent PEI lipid lipid
    Coating Ratio 0.0025
    (DNA/polymer)
    Performance:
    dose: (30 hrs for 5  1 μg 500 ng 2 μg
    Std. Formulas, 100 hrs 500 ng 250 ng 1 μg
    for  0 ng  0 ng 0 μg
    nanocapsules)
    Cytotoxicity:  9 ± 8 27 ± 8 9.3 ± 0.2 6.63 ± 1.4 
    (Fragmentation
    Index, %)
    cytotoxicity  6 ± 3 12.8 ± 1.5  5.7 ± 1.8
    controls:   4 ± 2.5 7.8 ± 0.1 3.1 ± 0.3
    (1 μg etoposide (8 hr):
    25 ± 10%)
    (Pei-DNA
    polyplexes (100 hr):
    24 ± 7%)
    (Empty vector
    nanocapsules:
    1.25 ± 1.25%)
    (Empty vector
    nanocapsules: 0.9 ± 0.7%)
    Transduction 24 ± 4 17 ± 2 dead dead
    Efficiency: (%
    cells) 120 hrs, dose
    as listed)
    Formula
    Characteristics:
    Nucleic: Acid  667 ± 0.2 ND ND ND
    Incorporation: (%)
    Cumulative ND ND ND ND
    Release: (%, 48 hr)
    Particle Size 57 ± 5 48 ± 2 ND 22.4 ± 2  
    (mean ± SE, nm)
    Agglomerates (as g.t. 50% 300 nm 25% 300 nm
    dispensed) 300 nm hard-fused
  • [0157]
    TABLE 3B
    Dose response of nanoencapsulated pDNA
    GFP/Actin Production
    Formula Dose (100 hr.) (density ratio, %)
    K.35   9 μg 94.8
    K.35 4.5 μg 83.5
    K.35 1.5 μg 83.3
    Lipoplex GP o.5 μg 94.9
  • It was observed that use of controlled-release nanocapsules reduced the fraction of apoptotic cells in [0158] fibroblast cultures 3 to 100 fold. Conventional reagents without DNA showed a 4-fold increase in FI over empty nanoparticles, but increased another 50-100% without additional reagents in the presence of additional DNA. Decreasing the coating weight from 1:40 to 1:400 resulted in an increase in average nanocapsule diameter from 20 to 57 nm and the appearance of regions of apoptotic induction in cultures (Table 3: F omicron vs. F upsilon 35). Decreasing the coating weight from 1:40 to an intermediate 1:100 reduced transduction efficiency without increasing particle size and the appearance of cytotoxicity. These findings indicate that nanocapsule design plays a role in maintaining nanocapsule integrity and that size effects and dissolution profiles can contribute to observed cytotoxicity and functionality. We concluded that application of nanocapsule formulations increased dosing to useful efficiency levels without induction of an apoptotic program.
  • Table 3B exemplifies this improvement with a dose response of Formula K.35 measured in fibroblast lysates. GFP production was measured in fibroblast lysates after 4 days of treatment with increasing doses of nanocapsules. A 9.5 μg dose of nanocapsules equaled the production of a liposomal formulation without any evidence of cytotoxicity. [0159]
  • Example 4—Nanocapsule delivery of macromolecules to porcine tissue across keratinized barrier epithelia bv transdermal and subcutaneous means
  • The utility of nanocapsules for nonviral nucleic acid delivery to tissue in a pig biopsy organ culture system was investigated. 6 and 8 mm circular biopsies were collected under sterile conditions from sedated research animals and cultured on meshes in partial contact with media containing 20% Fetal Calf Serum. Biopsies were either injected with 90 μl (6.3 μg) or treated topically with 3×30 μl aliquots. Biopsies were snapfrozen 7 days later and sectioned/homogenized for β-galactosidase production measurement. Formulation information and results from this experiment are summarized in Table 4: [0160]
    TABLE 4:
    Table 4: Functionality of dispersed atomized nanocapsules for
    macromolecule delivery across
    keratinized barrier membranes.
    Formula
    N O
    Exp. Modification coating wt. is 2.5x coating
    (from Formula Q) Polymer MW is 1x wt. is 2.5x
    Polymer
    MW is 4x
    Formula Characteristics:
    Nucleic Acid Incorporation (%) 70.00 70.50
    Cumulative Release 83 83. ± 1.5
    (%, 169 hr 2.5 μg
    sample
    Low MW DNA in postdigested 0 0
    Electrophoresis Samples
    Supercoil retention
    100% 100%
    (237 hr release,
    initial = 69.7% sc/releaxed)
    Particle Size (mean, SE, major 18.2 ± 0.2 nm ND
    species)
    Particle Description spherical
    Secondary structure: 20% 100 nm flocs
    Performance:
    Transduced Protein Production 312 ± 74 (topical)
    (pixel units, 142 (s.c.)
    % of neg control, 100 μg
    total protein, normalized by
    protein)
    Reporter (gene Product 100% 100%
    Distribution
    (6.3 μg dose,
    6 mm (N), 8 mm (O)
    porcine biopsy, 1 wk)
    keratinocytes (% cells), n = 2 73 ± 20 (pap) 13.8 ± 0.5 (pap)
    fields/200 cells, neg cntrl: 6%
    endothelial cells, (% vwf-+area)
    papillary and/or reticular, 32 ± 15 (ret) 8 ± 2 (ret)
    n = 2-4 fields,
    neg cntrl: 1.07 ± 0.72
    dermis (% area); 2.74 ± 0.96* 1.77 ± 0.49*
    negative cntrl:
    0.24 ± 0.03, n = 4/20x fields
  • Western blotting of radiated tissue lysates showed a 3-fold increase in β-galactosidase in duplicate biopsies treated topically with Formula N over an identically cultured 6mm biopsy treated with saline. Only a 2-fold increase was measured in a 8 mm biopsy treated topically with formula 0 nanoparticles (see FIG. 4B). Formula O was produced with a higher molecular weight analog of the N polymer suggesting a difference in particle morphology, a dose effect or differing in situ release profiles between the two formulations related to this difference. To identify initial cell type-specific differences in nanocapsule delivery effectiveness, tissue sections were analyzed for β-galactosidase expression in double-label experiments using antibodies to cell-specific epitopes (see FIG. 4B). Digital image analysis of these sections indicated that radiated keratinocytes and endothelial cells are readily transduced in [0161] organ culture 7 days after treatment with a 10 day releasing formula. Specific quantitation of fibroblastic cells was not possible without inclusion of a cell-specific marker, however, an 11-fold increase in area of expression was measured in N biopsy dermis (see FIG. 4B). Interestingly, for both the formulae N and O topically-treated biopsies examined, the area percentage of blood vessels transduced decreased about 50% in nearby fields between 100 μm and 300 μm of depth (Table 4: papillary vs. reticular endothelial cells). These data suggest that nanocapsules are penetrating the epidermis to enter the dermis.
  • Example 5—Incorporation of inventive nanocapsules into a solid dosage form for additional utility in physical targeting
  • Nanocapsules containing a nuclear GFP transgene or empty vector were incorporated into a suture coating by vortexing the following components: i) 50 μg of nanocapsules containing plasmid DNA, ii) 200 μg of bovine mucin, and iii) 75 μg of sucrose (60% w/w) in a 1000 μl volume. Sutures were aseptically coated by drawing [0162] sutures 5× through punctured microcentrifuge tubes. Coating functionality for gene transfer was tested by applying sutures in cultured porcine skin biopsies. Biopsies were cultured on a mesh such that the biopsy bottom was in contact with cell culture media. Biopsies were treated for 7 days, then snap-frozen and sectioned for immunofluorescence microscopy to assess nanocapsule penetration and transgene delivery.
  • Nanocapsule penetration was detected by streptavidin-biotin immunocomplexes directed at sheep IgG. Nanocapsule coatings are spiked with ovine IgG to enable this detection strategy. FIG. 5A shows distribution of sheep IgG signal throughout porcine dermal tissue with accumulation on capillaries. In FIG. 5A′, primary antibody is omitted during slide processing to determine level of background fluorescence. A suture is visible in this view. Sutures were identifiable as smooth objects without positive nuclear counterstain. GFP expression was confirmed using a polyclonal GFP antibody to obviate the effect of nonspecific tissue green fluorescence. FIG. 5B shows GFP expression throughout the suture-treated dermis using a GFP polyclonal antibody. A suture was visible 750 microns away. FIG. 5C shows the lack of GFP expression in a biopsy treated with empty vector coating. This example demonstrates the usefulness of nanocapsules for use in physically targeted macromolecule delivery. [0163]
  • Example 6—Utility of nanocapsules for local targeting by design of nanocapsule coating
  • Fibroblast targeting [0164]
  • GFP nanocapsules were prepared by dispersion atomization as described in Example 1. Polyvinylpyrylodone (PVP, MW 10,000) was used as the coating basis. A coating weight ratio of 1:40 was used and rod-shaped nanocapsules of 23±2 nm were produced. 1 μg of PVP nanocapsules were applied to both human dermal fibroblasts (HDF) and HacaT keratinocyte cultures for 4 hours then fixed for detection for nanocapsule uptake by streptavidin-bioting immunocomplexes to sheep IgG. Nanocapsule coatings are spiked with ovine IgG to enable this detection strategy. FIG. 6 illustrates positive nuclear localization of PVP nanocapsules in HDF's and negative colocalization of PVP nanocapsules in keratinocytes (FIG. 6: [0165] 6 a vs. 6 b). Views of untreated cultures are included for comparison (6 a′, 6 b′). Cultures were also treated with 5 μg of PVP nanocapsules for 5 days then tested for GFP transgene production. Consistent with uptake studies results, only the fibroblast cultures showed production of GFP transgene (FIG. 6: 6 a″ vs. 6 b″).
  • Tumor-targeting [0166]
  • GFP nanocapsules were prepared by dispersion atomization as described in example 1. Tenascin (TN, MW 200,000) was used as the coating basis. A coating weight ratio of 1:20 was used and spherical nanocapsules of 19±0.9 nm were produced. 500 ng of TN nanocapsules were applied topically in successive small aliquots to pig biopsies maintained in organ culture. Biopsies were rinsed in media after 3 minutes of topical application and culture media was changed to preclude any delivery other than topical. [0167]
  • To simulate tumor nests of epithelial-derived origin, biopsies had been seeded 12 hours previously with 50,000 human squamous carcinoma cells. 7 days later biopsies were snapfrozen and sectioned for immunological detection of GFP production. In FIG. 6B, view “a” shows intense GFP fluorescence in the tumor center, view “b” confirms this GFP expression with polyclonal antibodies to GFP, view “c” shows cell positioning in the section using a counterstain for cell nuclei and view “d” shows the level of background fluorescence by omission of GFP antibodies. Tumor origin was confirmed by positive detection with antibody to [0168] keratin 10/1, an epithelial marker. Comparison of view “b” and view “c” indicates that GFP expression is limited to cells within the tumor. As already demonstrated in example 5, expression throughout a tissue is also feasible and can modulated by coating design. This example demonstrates that nanocapsule delivery can be productively targeted.
  • Cell-specific delivery for enhanced drug therapeutic window [0169]
  • Nanocapsules were prepared as described in Example 1 to encapsulate cisplatin, a hydrophobic molecule and a common cancer chemotherapeutic with serious side effects. A coating weight ratio of 1:100 was used and irregular nanocapsules of 29±3 nm were produced. Targeting efficacy was demonstrated by changes in the dose response for cell growth inhibition in fibroblast vs. squamous cell carcinoma cultures. Cells were seeded overnight into 96 well plates, treated for 18 hours with increasing amounts of encapsulated or unencapsulated drug, then assessed for cell growth inhibition using an [0170] MTT assay 48 hours later for total growth time of 72 hours. Results are illustrated in FIG. 6C. The data shows that tenascin nanocapsules protected nontarget cells from cell death (zero death) at drug levels that killed using unencapsulated drug (FIG. 6Aa: open vs. closed circles). In carcinoma cultures, TN nanocapsules productively decreased the inhibition concentration (IC50) an estimated 300% from 525 to 160 μg/ml. Example 6 demonstrates the usefulness of nanocapsules for use in coating-targeted macromolecule delivery.
  • Example 7—Utility of nanoencapsulation for improved cellular uptake of other species used as pharmaceutical, nutraceutical, research or cosmetic agents
  • Nanocapsules containing either 500 kD Fitc-labelled dextran, 20 mer Fitc-labelled mer O-methylated RNA oligonucleotide and 16 mer phosphodiester DNA oligonucleotide were prepared as described in Example 1. A 1:40 coating weight ratio was used and 1 MM kD hyaluronan was used a coating basis. PEI was used to condense the phosphodiester DNA oligonucleotide, but no PEI was included in the dextran or RNA oligonucleotide formulas. Nanocapsule functionality for drug delivery was tested by evaluating changes in relative pinocytotic activity and cellular uptake in 35 mm cultures of human dermal fibroblast. Nanocapsule formulas were compared to naked species or species formulated as complexes. Quantitative results are summarized in Table 7. [0171]
    TABLE 6
    Nanoencapsulation improves cellular uptake of other
    species used as pharmaceutical, nutraceutical,
    research or cosmetic agents. At 18 hours
    post-addition, lysosomes are only evident in
    conventionally formulated species.
    4.5 hours post-addition 18 hours post-addition
    Increase in cellular Bioactive
    uptake activity, Nuclear component
    (% cells above Uptake Colocalization with Detection
    Particle size baseline, Efficiency lysosomes, persistence
    Bioactive (mean, SE, nm, caveolin-l/clathrin) (% cells, (% cells, human (% cell, human
    Component Formulation morphology) dose fibroblast) fibroblasts) fibroblast)
    500 kd nanocapsule  22 ± 2, s/r 89/20  25 μg* 95 ± 2  2 ± 2  5 μg 88 ± 11
    flic-dextran naked, Fitclabelled 75/18 100 μg  10 100 ± 10 100 μg 61 ± 20
     20 mer o- nanocapsule  13 ± 0.7, r 78/90  2 μg 74 ± 5  0 ± 0  5 μg 80 ± 6
    methylated naked, Fitclabelled —/73  5 μg 14 ± 7
    RNA oligo PEI/Fitclabelled 236 ± 26, r —/— 100 ± 0  5 μg 94 ± 10
     16 mer PO nanocapsule  17 ± 1, r 70/94  1 μg 34 ± 25  0 ± 0  5 μg 91 ± 8
    DNA oligo PEI/Fitclabelled  67 ± 4, s/r 72%  2 μg 95 ± 2  80 ± 7  5 μg 66
    lysosomes
    Nominal n 20 particles 70 cells 140 cells 50 cells 50 cells
  • Table 7 shows that average diameters for all nanocapsules were below 50 nm by AFM. PEI complexes of DNA oligonucleotides were measured at 67 nm and PEI complexes of uncharged RNA O-methyl oligonucleotides were measured at 236 nm. As discussed in Example 2 using keratinocyte cultures and plasmid DNA, nanocapsules stimulate pinocytotic activity as indicated by increased signal levels of clathrin and caveolin-1. In the 500 kD dextran case, pinocytotic activity shifts productively towards caveolae with nanoencapsulation (Table 7, 500 kD Dextran). At 4.5 hours post-addition, nuclear uptake is enhanced for encapsulated dextran and RNA relative to naked species. For the nanocapsules of DNA oligonucleotides, cellular uptake is decreased relative to complexed oligonucelotide, however, a majority of that DNA oligonucleotides is already nonproductively sequestered in lysosomes by that 4.5 hours (Table 7). At 18 hours post-addition, nanocapsules species show continued exclusion from lysosomes, while naked species show high levels of sequestration. These results are illustrated in FIG. 7A and 7B. Views “a” and “b” show Fitc detection in cultures at 18 hours. That distribution is exclusively nuclear for the nanoencapsules of RNA oligonucelotides (FIG. 7B: a vs. a′). Punctate inclusion are visible in the cultures treated with the complexed RNA oligonucleotides that co-localize with an immunological marker for lysosomes (FIG. 7A: a vs. a′). Particle sizing results from AFM microscopy are included to demonstrate dramatic difference in sizing following encapsulation. (FIG. 7A, [0172] 7B:b vs.b, b′). Formulas encapsulating lower molecular weight dextrans and unstabilized RNA were also prepared with analagous uptake, nanocapsule size and yield to demonstrate that encapsulation can provide not only a targeting function but aid in stabilizing molecules sensitive to chemical or enzymatic degradation. These examples demonstrates the usefulness of nanocapsules 36 for use in delivery of a broad range of macromolecules.
  • Although the present invention has been described with reference to preferred embodiments, workers skilled in the art will recognize that changes may be made in form and detail without departing from the spirit and scope of the invention. [0173]
  • 1 5 1 6232 DNA Artificial Sequence Supplied by Invitrogen of Carlsbad, California 1 gtaccgaatt caagcttcgt gaggctccgg tgcccgtcag tgggcagagc gcacatcgcc 60 cacagtcccc gagaagttgg ggggaggggt cggcaattga accggtgcct agagaaggtg 120 gcgcggggta aactgggaaa gtgatgtcgt gtactggctc cgcctttttc ccgagggtgg 180 gggagaaccg tatataagtg cagtagtcgc cgtgaacgtt ctttttcgca acgggtttgc 240 cgccagaaca caggtaagtg ccgtgtgtgg ttcccgcggg cctggcctct ttacgggtta 300 tggcccttgc gtgccttgaa ttacttccac ctggctccag tacgtgattc ttgatcccga 360 gctggagcca ggggcgggcc ttgcgcttta ggagcccctt cgcctcgtgc ttgagttgag 420 gcctggcctg ggcgctgggg ccgccgcgtg cgaatctggt ggcaccttcg cgcctgtctc 480 gctgctttcg ataagtctct agccatttaa aatttttgat gacctgctgc gacgcttttt 540 ttctggcaag atagtcttgt aaatgcgggc caggatctgc acactggtat ttcggttttt 600 gggcccgcgg ccggcgacgg ggcccgtgcg tcccagcgca catgttcggc gaggcggggc 660 ctgcgagcgc ggccaccgag aatcggacgg gggtagtctc aagctggccg gcctgctctg 720 gtgcctggcc tcgcgccgcc gtgtatcgcc ccgccctggg cggcaaggct ggcccggtcg 780 gcaccagttg cgtgagcgga aagatggccg cttcccggcc ctgctccagg gggctcaaaa 840 tggaggacgc ggcgctcggg agagcgggcg ggtgagtcac ccacacaaag gaaaagggcc 900 tttccgtcct cagccgtcgc ttcatgtgac tccacggagt accgggcgcc gtccaggcac 960 ctcgattagt tctggagctt ttggagtacg tcgtctttag gttgggggga ggggttttat 1020 gcgatggagt ttccccacac tgagtgggtg gagactgaag ttaggccagc ttggcacttg 1080 atgtaattct ccttggaatt tggccttttt gagtttggat cttggttcat tctcaagcct 1140 cagacagtgg ttcaaagttt ttttcttcca tttcaggtgt cgtgaacacg tggccaccat 1200 ggcccaggtg cagctgcaga tggctagcaa aggagaagaa cttttcactg gagttgtccc 1260 aattcttgtt gaattagatg gtgatgttaa tgggcacaaa ttttctgtca gtggagaggg 1320 tgaaggtgat gctacatacg gaaagcttac ccttaaattt atttgcacta ctggaaaact 1380 acctgttcca tggccaacac ttgtcactac tttctcttat ggtgttcaat gcttttcccg 1440 ttatccggat catatgaaac ggcatgactt tttcaagagt gccatgcccg aaggttatgt 1500 acaggaacgc actatatctt tcaaagatga cgggaactac aagacgcgtg ctgaagtcaa 1560 gtttgaaggt gatacccttg ttaatcgtat cgagttaaaa ggtattgatt ttaaagaaga 1620 tggaaacatt ctcggacaca aactcgagta caactataac tcacacaatg tatacatcac 1680 ggcagacaaa caaaagaatg gaatcaaagc taacttcaaa attcgccaca acattgaaga 1740 tggatccgtt caactagcag accattatca acaaaatact ccaattggcg atggccctgt 1800 ccttttacca gacaaccatt acctgtcgac acaatctgcc ctttcgaaag atcccaacga 1860 aaagcgtgac cacatggtcc ttcttgagtt tgtaactgct gctgggatta cacatggcat 1920 ggatgagctc tacaaagcgg ccgcagatcc aaaaaagaag agaaaggtag atccaaaaaa 1980 gaagagaaag gtagatccaa aaaagaagag aaaggtagat acggccgcag aacaaaaact 2040 catctcagaa gaggatctga atggggccgc atagtctaga agctcgctga tcagcctcga 2100 ctgtgccttc tagttgccag ccatctgttg tttgcccctc ccccgtgcct tccttgaccc 2160 tggaaggtgc cactcccact gtcctttcct aataaaatga ggaaattgca tcgcattgtc 2220 tgagtaggtg tcattctatt ctggggggtg gggtggggca ggacagcaag ggggaggatt 2280 gggaagacaa tagcaggcat gctggggatg gcccgggctc tatggcttct gaggcggaaa 2340 gaaccagctg gggctctagg gggtatcccc acgcgccctg tagcggcgca ttaagcgcgg 2400 cgggtgtggt ggttacgcgc agcgtgaccg ctacacttgc cagcgcccta gcgcccgctc 2460 ctttcgcttt cttcccttcc tttctcgcca cgttcgccgg ctttccccgt caagctctaa 2520 atcggggcat ccctttaggg ttccgattta gtgctttacg gcacctcgac cccaaaaaac 2580 ttgattaggg tgatggttca cgtagtgggc catcgccctg atagacggtt tttcgccctt 2640 tgacgttgga gtccacgttc tttaatagtg gactcttgtt ccaaactgga acaacactca 2700 accctatctc ggtctattct tttgatttat aagggatttt ggggatttcg gcctattggt 2760 taaaaaatga gctgatttaa caaaaattta acgcgaatta attctgtgga atgtgtgtca 2820 gttagggtgt ggaaagtccc caggctcccc aggcaggcag aagtatgcaa agcatgcatc 2880 tcaattagtc agcaaccagg tgtggaaagt ccccaggctc cccagcaggc agaagtatgc 2940 aaagcatgca tctcaattag tcagcaacca tagtcccgcc cctaactccg cccatcccgc 3000 ccctaactcc gcccagttcc gcccattctc cgcccctagg ctgactaatt ttttttattt 3060 atgcagaggc cgaggccgcc tctgcctctg agctattcca gaagtagtga ggaggctttt 3120 ttggaggcct aggcttttgc aaaaagctcc cgggaggtcc acaatgattg aacaagatgg 3180 attgcacgca ggttctccgg ccgcttgggt ggagaggcta ttcggctatg actgggcaca 3240 acagacaatc ggctgctctg atgccgccgt gttccggctg tcagcgcagg ggcgcccggt 3300 tctttttgtc aagaccgacc tgtccggtgc cctgaatgaa ctccaggacg aggcagcgcg 3360 gctatcgtgg ctggccacga cgggcgttcc ttgcgcagct gtgctcgacg ttgtcactga 3420 agcgggaagg gactggctgc tattgggcga agtgccgggg caggatctcc tgtcatctca 3480 ccttgctcct gccgagaaag tatccatcat ggctgatgca atgcggcggc tgcatacgct 3540 tgatccggct acctgcccat tcgaccacca agcgaaacat cgcatcgagc gagcacgtac 3600 tcggatggaa gccggtcttg tcgatcagga tgatctggac gaagagcatc aggggctcgc 3660 gccagccgaa ctgttcgcca ggctcaaggc gcgtatgccc gacggcgagg atctcgtcgt 3720 gactcatggc gatgcctgct tgccgaatat catggtggaa aatggccgct tttctggatt 3780 catcgactgt ggccggctgg gtgtggcgga ccgctatcag gacatagcgt tggctacccg 3840 tgatattgct gaagagcttg gcggcgaatg ggctgaccgc ttcctcgtgc tttacggtat 3900 cgccgctccc gattcgcagc gcatcgcctt ctatcgcctt cttgacgagt tcttctgagc 3960 gggactctgg ggttcgaaat gaccgaccaa gcgacgccca acctgccatc acgagatttc 4020 gattccaccg ccgccttcta tgaaaggttg ggcttcggaa tcgttttccg ggacgccggc 4080 tggatgatcc tccagcgcgg ggatctcatg ctggagttct tcgcccaccc caacttgttt 4140 attgcagctt ataatggtta caaataaagc aatagcatca caaatttcac aaataaagca 4200 tttttttcac tgcattctag ttgtggtttg tccaaactca tcaatgtatc ttatcatgtc 4260 tgtataccgg atctttccgc ttcctcgctc actgactcgc tgcgctcggt cgttcggctg 4320 cggcgagcgg tatcagctca ctcaaaggcg gtaatacggt tatccacaga atcaggggat 4380 aacgcaggaa agaacatgtg agcaaaaggc cagcaaaagg ccaggaaccg taaaaaggcc 4440 gcgttgctgg cgtttttcca taggctccgc ccccctgacg agcatcacaa aaatcgacgc 4500 tcaagtcaga ggtggcgaaa cccgacagga ctataaagat accaggcgtt tccccctgga 4560 agctccctcg tgcgctctcc tgttccgacc ctgccgctta ccggatacct gtccgccttt 4620 ctcccttcgg gaagcgtggc gctttctcaa tgctcacgct gtaggtatct cagttcggtg 4680 taggtcgttc gctccaagct gggctgtgtg cacgaacccc ccgttcagcc cgaccgctgc 4740 gccttatccg gtaactatcg tcttgagtcc aacccggtaa gacacgactt atcgccactg 4800 gcagcagcca ctggtaacag gattagcaga gcgaggtatg taggcggtgc tacagagttc 4860 ttgaagtggt ggcctaacta cggctacact agaaggacag tatttggtat ctgcgctctg 4920 ctgaagccag ttaccttcgg aaaaagagtt ggtagctctt gatccggcaa acaaaccacc 4980 gctggtagcg gtggtttttt tgtttgcaag cagcagatta cgcgcagaaa aaaaggatct 5040 caagaagatc ctttgatctt ttctacgggg tctgacgctc agtggaacga aaactcacgt 5100 taagggattt tggtcatgag attatcaaaa aggatcttca cctagatcct tttaaattaa 5160 aaatgaagtt ttaaatcaat ctaaagtata tatgagtaaa cttggtctga cagttaccaa 5220 tgcttaatca gtgaggcacc tatctcagcg atctgtctat ttcgttcatc catagttgcc 5280 tgactccccg tcgtgtagat aactacgata cgggagggct taccatctgg ccccagtgct 5340 gcaatgatac cgcgagaccc acgctcaccg gctccagatt tatcagcaat aaaccagcca 5400 gccggaaggg ccgagcgcag aagtggtcct gcaactttat ccgcctccat ccagtctatt 5460 aattgttgcc gggaagctag agtaagtagt tcgccagtta atagtttgcg caacgttgtt 5520 gccattgcta caggcatcgt ggtgtcacgc tcgtcgtttg gtatggcttc attcagctcc 5580 ggttcccaac gatcaaggcg agttacatga tcccccatgt tgtgcaaaaa agcggttagc 5640 tccttcggtc ctccgatcgt tgtcagaagt aagttggccg cagtgttatc actcatggtt 5700 atggcagcac tgcataattc tcttactgtc atgccatccg taagatgctt ttctgtgact 5760 ggtgagtact caaccaagtc attctgagaa tagtgtatgc ggcgaccgag ttgctcttgc 5820 ccggcgtcaa tacgggataa taccgcgcca catagcagaa ctttaaaagt gctcatcatt 5880 ggaaaacgtt cttcggggcg aaaactctca aggatcttac cgctgttgag atccagttcg 5940 atgtaaccca ctcgtgcacc caactgatct tcagcatctt ttactttcac cagcgtttct 6000 gggtgagcaa aaacaggaag gcaaaatgcc gcaaaaaagg gaataagggc gacacggaaa 6060 tgttgaatac tcatactctt cctttttcaa tattattgaa gcatttatca gggttattgt 6120 ctcatgagcg gatacatatt tgaatgtatt tagaaaaata aacaaatagg ggttccgcgc 6180 acatttcccc gaaaagtgcc acctgacgtc agatcgacgg atcgggagat cg 6232 2 2200 PRT Homo sapiens 2 Met Gly Ala Met Thr Gln Leu Leu Ala Gly Val Phe Leu Ala Phe Leu 1 5 10 15 Ala Leu Ala Thr Glu Gly Gly Val Leu Lys Lys Val Ile Arg His Lys 20 25 30 Arg Gln Ser Gly Val Asn Ala Thr Leu Pro Glu Glu Asn Gln Pro Val 35 40 45 Val Phe Asn His Val Tyr Asn Ile Lys Leu Pro Val Gly Ser Gln Cys 50 55 60 Ser Val Asp Leu Glu Ser Ala Ser Gly Glu Lys Asp Leu Ala Pro Pro 65 70 75 80 Ser Glu Pro Ser Glu Ser Phe Gln Glu His Thr Val Asp Gly Glu Asn 85 90 95 Gln Ile Val Phe Thr His Arg Ile Asn Ile Pro Arg Arg Ala Cys Gly 100 105 110 Cys Ala Ala Ala Pro Asp Val Lys Glu Leu Leu Ser Arg Leu Glu Glu 115 120 125 Leu Glu Asn Leu Val Ser Ser Leu Arg Glu Gln Cys Thr Ala Gly Ala 130 135 140 Gly Cys Cys Leu Gln Pro Ala Thr Gly Arg Leu Asp Thr Arg Pro Phe 145 150 155 160 Cys Ser Gly Arg Gly Asn Phe Ser Thr Glu Gly Cys Gly Cys Val Cys 165 170 175 Glu Pro Gly Trp Lys Gly Pro Asn Cys Ser Glu Pro Glu Cys Pro Gly 180 185 190 Asn Cys His Leu Arg Gly Arg Cys Ile Asp Gly Gln Cys Ile Cys Asp 195 200 205 Asp Gly Phe Thr Gly Glu Asp Cys Ser Gln Leu Ala Cys Pro Ser Asp 210 215 220 Cys Asn Asp Gln Gly Lys Cys Val Asn Gly Val Cys Ile Cys Phe Glu 225 230 235 240 Gly Tyr Ala Gly Ala Asp Cys Ser Arg Glu Ile Cys Pro Val Pro Cys 245 250 255 Ser Glu Glu His Gly Thr Cys Val Asp Gly Leu Cys Val Cys His Asp 260 265 270 Gly Phe Ala Gly Asp Asp Cys Asn Lys Pro Leu Cys Leu Asn Asn Cys 275 280 285 Tyr Asn Arg Gly Arg Cys Val Glu Asn Glu Cys Val Cys Asp Glu Gly 290 295 300 Phe Thr Gly Glu Asp Cys Ser Glu Leu Ile Cys Pro Asn Asp Cys Phe 305 310 315 320 Asp Arg Gly Arg Cys Ile Asn Gly Thr Cys Tyr Cys Glu Glu Gly Phe 325 330 335 Thr Gly Glu Asp Cys Gly Lys Pro Thr Cys Pro His Ala Cys His Thr 340 345 350 Gln Gly Arg Cys Glu Glu Gly Gln Cys Val Cys Asp Glu Gly Phe Ala 355 360 365 Gly Leu Asp Cys Ser Glu Lys Arg Cys Pro Ala Asp Cys His Asn Arg 370 375 380 Gly Arg Cys Val Asp Gly Arg Cys Glu Cys Asp Asp Gly Phe Thr Gly 385 390 395 400 Ala Asp Cys Gly Glu Leu Lys Cys Pro Asn Gly Cys Ser Gly His Gly 405 410 415 Arg Cys Val Asn Gly Gln Cys Val Cys Asp Glu Gly Tyr Thr Gly Glu 420 425 430 Asp Cys Ser Gln Leu Arg Cys Pro Asn Asp Cys His Ser Arg Gly Arg 435 440 445 Cys Val Glu Gly Lys Cys Val Cys Glu Gln Gly Phe Lys Gly Tyr Asp 450 455 460 Cys Ser Asp Met Ser Cys Pro Asn Asp Cys His Gln His Gly Arg Cys 465 470 475 480 Val Asn Gly Met Cys Val Cys Asp Asp Gly Tyr Thr Gly Glu Asp Cys 485 490 495 Arg Asp Arg Gln Cys Pro Arg Asp Cys Ser Asn Arg Gly Leu Cys Val 500 505 510 Asp Gly Gln Cys Val Cys Glu Asp Gly Phe Thr Gly Pro Asp Cys Ala 515 520 525 Glu Leu Ser Cys Pro Asn Asp Cys His Gly Gln Gly Arg Cys Val Asn 530 535 540 Gly Gln Cys Val Cys His Glu Gly Phe Met Gly Lys Asp Cys Lys Glu 545 550 555 560 Gln Arg Cys Pro Ser Asp Cys His Gly Gln Gly Arg Cys Val Asp Gly 565 570 575 Gln Cys Ile Cys His Glu Gly Phe Thr Gly Leu Asp Cys Gly Gln His 580 585 590 Ser Cys Pro Ser Asp Cys Asn Asn Leu Gly Gln Cys Val Ser Gly Arg 595 600 605 Cys Ile Cys Asn Glu Gly Tyr Ser Gly Glu Asp Cys Ser Glu Val Ser 610 615 620 Pro Pro Lys Asp Leu Val Val Thr Glu Val Thr Glu Glu Thr Val Asn 625 630 635 640 Leu Ala Trp Asp Asn Glu Met Arg Val Thr Glu Tyr Leu Val Val Tyr 645 650 655 Thr Pro Thr His Glu Gly Gly Leu Glu Met Gln Phe Arg Val Pro Gly 660 665 670 Asp Gln Thr Ser Thr Ile Ile Gln Glu Leu Glu Pro Gly Val Glu Tyr 675 680 685 Phe Ile Arg Val Phe Ala Ile Leu Glu Asn Lys Lys Ser Ile Pro Val 690 695 700 Ser Ala Arg Val Ala Thr Tyr Leu Pro Ala Pro Glu Gly Leu Lys Phe 705 710 715 720 Lys Ser Ile Lys Glu Thr Ser Val Glu Val Glu Trp Asp Pro Leu Asp 725 730 735 Ile Ala Phe Glu Thr Trp Glu Ile Ile Phe Arg Asn Met Asn Lys Glu 740 745 750 Asp Glu Gly Glu Ile Thr Lys Ser Leu Arg Arg Pro Glu Thr Ser Tyr 755 760 765 Arg Gln Thr Gly Leu Ala Pro Gly Gln Glu Tyr Glu Ile Ser Leu His 770 775 780 Ile Val Lys Asn Asn Thr Arg Gly Pro Gly Leu Lys Arg Val Thr Thr 785 790 795 800 Thr Arg Leu Asp Ala Pro Ser Gln Ile Glu Val Lys Asp Val Thr Asp 805 810 815 Thr Thr Ala Leu Ile Thr Trp Phe Lys Pro Leu Ala Glu Ile Asp Gly 820 825 830 Ile Glu Leu Thr Tyr Gly Ile Lys Asp Val Pro Gly Asp Arg Thr Thr 835 840 845 Ile Asp Leu Thr Glu Asp Glu Asn Gln Tyr Ser Ile Gly Asn Leu Lys 850 855 860 Pro Asp Thr Glu Tyr Glu Val Ser Leu Ile Ser Arg Arg Gly Asp Met 865 870 875 880 Ser Ser Asn Pro Ala Lys Glu Thr Phe Thr Thr Gly Leu Asp Ala Pro 885 890 895 Arg Asn Leu Arg Arg Val Ser Gln Thr Asp Asn Ser Ile Thr Leu Glu 900 905 910 Trp Arg Asn Gly Lys Ala Ala Ile Asp Ser Tyr Arg Ile Lys Tyr Ala 915 920 925 Pro Ile Ser Gly Gly Asp His Ala Glu Val Asp Val Pro Lys Ser Gln 930 935 940 Gln Ala Thr Thr Lys Thr Thr Leu Thr Gly Leu Arg Pro Gly Thr Glu 945 950 955 960 Tyr Gly Ile Gly Val Ser Ala Val Lys Glu Asp Lys Glu Ser Asn Pro 965 970 975 Ala Thr Ile Asn Ala Ala Thr Glu Leu Asp Thr Pro Lys Asp Leu Gln 980 985 990 Val Ser Glu Thr Ala Glu Thr Ser Leu Thr Leu Leu Trp Lys Thr Pro 995 1000 1005 Leu Ala Lys Phe Asp Arg Tyr Arg Leu Asn Tyr Ser Leu Pro Thr 1010 1015 1020 Gly Gln Trp Val Gly Val Gln Leu Pro Arg Asn Thr Thr Ser Tyr 1025 1030 1035 Val Leu Arg Gly Leu Glu Pro Gly Gln Glu Tyr Asn Val Leu Leu 1040 1045 1050 Thr Ala Glu Lys Gly Arg His Lys Ser Lys Pro Ala Arg Val Lys 1055 1060 1065 Ala Ser Thr Glu Gln Ala Pro Glu Leu Glu Asn Leu Thr Val Thr 1070 1075 1080 Glu Val Gly Trp Asp Gly Leu Arg Leu Asn Trp Thr Ala Ala Asp 1085 1090 1095 Gln Ala Tyr Glu His Phe Ile Ile Gln Val Gln Glu Ala Asn Lys 1100 1105 1110 Val Glu Ala Ala Arg Asn Leu Thr Val Pro Gly Ser Leu Arg Ala 1115 1120 1125 Val Asp Ile Pro Gly Leu Lys Ala Ala Thr Pro Tyr Thr Val Ser 1130 1135 1140 Ile Tyr Gly Val Ile Gln Gly Tyr Arg Thr Pro Val Leu Ser Ala 1145 1150 1155 Glu Ala Ser Thr Gly Glu Thr Pro Asn Leu Gly Glu Val Val Val 1160 1165 1170 Ala Glu Val Gly Trp Asp Ala Leu Lys Leu Asn Trp Thr Ala Pro 1175 1180 1185 Glu Gly Ala Tyr Glu Tyr Phe Phe Ile Gln Val Gln Glu Ala Asp 1190 1195 1200 Thr Val Glu Ala Ala Gln Asn Leu Thr Val Pro Gly Gly Leu Arg 1205 1210 1215 Ser Thr Asp Leu Pro Gly Leu Lys Ala Ala Thr His Tyr Thr Ile 1220 1225 1230 Thr Ile Arg Gly Val Thr Gln Asp Phe Ser Thr Thr Pro Leu Ser 1235 1240 1245 Val Glu Val Leu Thr Glu Glu Val Pro Asp Met Gly Asn Leu Thr 1250 1255 1260 Val Thr Glu Val Ser Trp Asp Ala Leu Arg Leu Asn Trp Thr Thr 1265 1270 1275 Pro Asp Gly Thr Tyr Asp Gln Phe Thr Ile Gln Val Gln Glu Ala 1280 1285 1290 Asp Gln Val Glu Glu Ala His Asn Leu Thr Val Pro Gly Ser Leu 1295 1300 1305 Arg Ser Met Glu Ile Pro Gly Leu Arg Ala Gly Thr Pro Tyr Thr 1310 1315 1320 Val Thr Leu His Gly Glu Val Arg Gly His Ser Thr Arg Pro Leu 1325 1330 1335 Ala Val Glu Val Val Thr Glu Asp Leu Pro Gln Leu Gly Asp Leu 1340 1345 1350 Ala Val Ser Glu Val Gly Trp Asp Gly Leu Arg Leu Asn Trp Thr 1355 1360 1365 Ala Ala Asp Asn Ala Tyr Glu His Phe Val Gln Val Gln Glu Val 1370 1375 1380 Asn Lys Val Glu Ala Ala Gln Asn Leu Thr Leu Pro Gly Ser Leu 1385 1390 1395 Arg Ala Val Asp Ile Pro Gly Leu Glu Ala Ala Thr Pro Tyr Arg 1400 1405 1410 Val Ser Ile Tyr Gly Val Ile Arg Gly Tyr Arg Thr Pro Val Leu 1415 1420 1425 Ser Ala Glu Ala Ser Thr Ala Lys Glu Pro Glu Ile Gly Asn Leu 1430 1435 1440 Asn Val Ser Asp Ile Thr Pro Glu Ser Phe Asn Leu Ser Trp Met 1445 1450 1455 Ala Thr Asp Gly Ile Phe Glu Thr Phe Thr Ile Glu Ile Ile Asp 1460 1465 1470 Ser Asn Arg Leu Leu Glu Thr Val Glu Tyr Asn Ile Ser Gly Ala 1475 1480 1485 Glu Arg Thr Ala His Ile Ser Gly Leu Pro Pro Ser Thr Asp Phe 1490 1495 1500 Ile Val Tyr Leu Ser Gly Leu Ala Pro Ser Ile Arg Thr Lys Thr 1505 1510 1515 Ile Ser Ala Thr Ala Thr Thr Glu Ala Leu Pro Leu Leu Glu Asn 1520 1525 1530 Leu Thr Ile Ser Asp Ile Asn Pro Tyr Gly Phe Thr Val Ser Trp 1535 1540 1545 Met Ala Ser Glu Asn Ala Phe Asp Ser Phe Leu Val Thr Val Val 1550 1555 1560 Asp Ser Gly Lys Leu Leu Asp Pro Gln Glu Phe Thr Leu Ser Gly 1565 1570 1575 Thr Gln Arg Lys Leu Glu Leu Arg Gly Leu Ile Thr Gly Ile Gly 1580 1585 1590 Tyr Glu Val Met Val Ser Gly Phe Thr Gln Gly His Gln Thr Lys 1595 1600 1605 Pro Leu Arg Ala Glu Ile Val Thr Glu Ala Glu Pro Glu Val Asp 1610 1615 1620 Asn Leu Leu Val Ser Asp Ala Thr Pro Asp Gly Phe Arg Leu Ser 1625 1630 1635 Trp Thr Ala Asp Glu Gly Val Phe Asp Asn Phe Val Leu Lys Ile 1640 1645 1650 Arg Asp Thr Lys Lys Gln Ser Glu Pro Leu Glu Ile Thr Leu Leu 1655 1660 1665 Ala Pro Glu Arg Thr Arg Asp Leu Thr Gly Leu Arg Glu Ala Thr 1670 1675 1680 Glu Tyr Glu Ile Glu Leu Tyr Gly Ile Ser Lys Gly Arg Arg Ser 1685 1690 1695 Gln Thr Val Ser Ala Ile Ala Thr Thr Ala Met Gly Ser Pro Lys 1700 1705 1710 Glu Val Ile Phe Ser Asp Ile Thr Glu Asn Ser Ala Thr Val Ser 1715 1720 1725 Trp Arg Ala Pro Thr Ala Gln Val Glu Ser Phe Arg Ile Thr Tyr 1730 1735 1740 Val Pro Ile Thr Gly Gly Thr Pro Ser Met Val Thr Val Asp Gly 1745 1750 1755 Thr Lys Thr Gln Thr Arg Leu Val Lys Leu Ile Pro Gly Val Glu 1760 1765 1770 Tyr Leu Val Ser Ile Ile Ala Met Lys Gly Phe Glu Glu Ser Glu 1775 1780 1785 Pro Val Ser Gly Ser Phe Thr Thr Ala Leu Asp Gly Pro Ser Gly 1790 1795 1800 Leu Val Thr Ala Asn Ile Thr Asp Ser Glu Ala Leu Ala Arg Trp 1805 1810 1815 Gln Pro Ala Ile Ala Thr Val Asp Ser Tyr Val Ile Ser Tyr Thr 1820 1825 1830 Gly Glu Lys Val Pro Glu Ile Thr Arg Thr Val Ser Gly Asn Thr 1835 1840 1845 Val Glu Tyr Ala Leu Thr Asp Leu Glu Pro Ala Thr Glu Tyr Thr 1850 1855 1860 Leu Arg Ile Phe Ala Glu Lys Gly Pro Gln Lys Ser Ser Thr Ile 1865 1870 1875 Thr Ala Lys Phe Thr Thr Asp Leu Asp Ser Pro Arg Asp Leu Thr 1880 1885 1890 Ala Thr Glu Val Gln Ser Glu Thr Ala Leu Leu Thr Trp Arg Pro 1895 1900 1905 Pro Arg Ala Ser Val Thr Gly Tyr Leu Leu Val Tyr Glu Ser Val 1910 1915 1920 Asp Gly Thr Val Lys Glu Val Ile Val Gly Pro Asp Thr Thr Ser 1925 1930 1935 Tyr Ser Leu Ala Asp Leu Ser Pro Ser Thr His Tyr Thr Ala Lys 1940 1945 1950 Ile Gln Ala Leu Asn Gly Pro Leu Arg Ser Asn Met Ile Gln Thr 1955 1960 1965 Ile Phe Thr Thr Ile Gly Leu Leu Tyr Pro Phe Pro Lys Asp Cys 1970 1975 1980 Ser Gln Ala Met Leu Asn Gly Asp Thr Thr Ser Gly Leu Tyr Thr 1985 1990 1995 Ile Tyr Leu Asn Gly Asp Lys Ala Gln Ala Leu Glu Val Phe Cys 2000 2005 2010 Asp Met Thr Ser Asp Gly Gly Gly Trp Ile Val Phe Leu Arg Arg 2015 2020 2025 Lys Asn Gly Arg Glu Asn Phe Tyr Gln Asn Trp Lys Ala Tyr Ala 2030 2035 2040 Ala Gly Phe Gly Asp Arg Arg Glu Glu Phe Trp Leu Gly Leu Asp 2045 2050 2055 Asn Leu Asn Lys Ile Thr Ala Gln Gly Gln Tyr Glu Leu Arg Val 2060 2065 2070 Asp Leu Arg Asp His Gly Glu Thr Ala Phe Ala Val Tyr Asp Lys 2075 2080 2085 Phe Ser Val Gly Asp Ala Lys Thr Arg Tyr Lys Leu Lys Val Glu 2090 2095 2100 Gly Tyr Ser Gly Thr Ala Gly Asp Ser Met Ala Tyr His Asn Gly 2105 2110 2115 Arg Ser Phe Ser Thr Phe Asp Lys Asp Thr Asp Ser Ala Ile Thr 2120 2125 2130 Asn Cys Ala Leu Ser Tyr Lys Gly Ala Phe Trp Tyr Arg Asn Cys 2135 2140 2145 His Arg Val Asn Leu Met Gly Arg Tyr Gly Asp Asn Asn His Ser 2150 2155 2160 Gln Gly Val Asn Trp Phe His Trp Lys Gly His Glu His Ser Ile 2165 2170 2175 Gln Phe Ala Glu Met Lys Leu Arg Pro Ser Asn Phe Arg Asn Leu 2180 2185 2190 Glu Gly Arg Arg Lys Arg Ala 2195 2200 3 8578 DNA Artificial Sequence Supplied by Invitrogen of Carlsbad, California 3 gacggatcgg gagatctccc gatcccctat ggtcgactct cagtacaatc tgctctgatg 60 ccgcatagtt aagccagtat ctgctccctg cttgtgtgtt ggaggtcgct gagtagtgcg 120 cgagcaaaat ttaagctaca acaaggcaag gcttgaccga caattgcatg aagaatctgc 180 ttagggttag gcgttttgcg ctgcttcgcg atgtacgggc cagatatacg cgttgacatt 240 gattattgac tagttattaa tagtaatcaa ttacggggtc attagttcat agcccatata 300 tggagttccg cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc 360 cccgcccatt gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc 420 attgacgtca atgggtggac tatttacggt aaactgccca cttggcagta catcaagtgt 480 atcatatgcc aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt 540 atgcccagta catgacctta tgggactttc ctacttggca gtacatctac gtattagtca 600 tcgctattac catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg 660 actcacgggg atttccaagt ctccacccca ttgacgtcaa tgggagtttg ttttggcacc 720 aaaatcaacg ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg 780 gtaggcgtgt acggtgggag gtctatataa gcagagctct ctggctaact agagaaccca 840 ctgcttactg gcttatcgaa attaatacga ctcactatag ggagacccaa gctggctagc 900 gtttaaactt aagcttacca tggggggttc tcatcatcat catcatcatg gtatggctag 960 catgactggt ggacagcaaa tgggtcggga tctgtacgac gatgacgata aggtacctaa 1020 ggatcagctt ggagttgatc ccgtcgtttt acaacgtcgt gactgggaaa accctggcgt 1080 tacccaactt aatcgccttg cagcacatcc ccctttcgcc agctggcgta atagcgaaga 1140 ggcccgcacc gatcgccctt cccaacagtt gcgcagcctg aatggcgaat ggcgctttgc 1200 ctggtttccg gcaccagaag cggtgccgga aagctggctg gagtgcgatc ttcctgaggc 1260 cgatactgtc gtcgtcccct caaactggca gatgcacggt tacgatgcgc ccatctacac 1320 caacgtaacc tatcccatta cggtcaatcc gccgtttgtt cccacggaga atccgacggg 1380 ttgttactcg ctcacattta atgttgatga aagctggcta caggaaggcc agacgcgaat 1440 tatttttgat ggcgttaact cggcgtttca tctgtggtgc aacgggcgct gggtcggtta 1500 cggccaggac agtcgtttgc cgtctgaatt tgacctgagc gcatttttac gcgccggaga 1560 aaaccgcctc gcggtgatgg tgctgcgttg gagtgacggc agttatctgg aagatcagga 1620 tatgtggcgg atgagcggca ttttccgtga cgtctcgttg ctgcataaac cgactacaca 1680 aatcagcgat ttccatgttg ccactcgctt taatgatgat ttcagccgcg ctgtactgga 1740 ggctgaagtt cagatgtgcg gcgagttgcg tgactaccta cgggtaacag tttctttatg 1800 gcagggtgaa acgcaggtcg ccagcggcac cgcgcctttc ggcggtgaaa ttatcgatga 1860 gcgtggtggt tatgccgatc gcgtcacact acgtctgaac gtcgaaaacc cgaaactgtg 1920 gagcgccgaa atcccgaatc tctatcgtgc ggtggttgaa ctgcacaccg ccgacggcac 1980 gctgattgaa gcagaagcct gcgatgtcgg tttccgcgag gtgcggattg aaaatggtct 2040 gctgctgctg aacggcaagc cgttgctgat tcgaggcgtt aaccgtcacg agcatcatcc 2100 tctgcatggt caggtcatgg atgagcagac gatggtgcag gatatcctgc tgatgaagca 2160 gaacaacttt aacgccgtgc gctgttcgca ttatccgaac catccgctgt ggtacacgct 2220 gtgcgaccgc tacggcctgt atgtggtgga tgaagccaat attgaaaccc acggcatggt 2280 gccaatgaat cgtctgaccg atgatccgcg ctggctaccg gcgatgagcg aacgcgtaac 2340 gcgaatggtg cagcgcgatc gtaatcaccc gagtgtgatc atctggtcgc tggggaatga 2400 atcaggccac ggcgctaatc acgacgcgct gtatcgctgg atcaaatctg tcgatccttc 2460 ccgcccggtg cagtatgaag gcggcggagc cgacaccacg gccaccgata ttatttgccc 2520 gatgtacgcg cgcgtggatg aagaccagcc cttcccggct gtgccgaaat ggtccatcaa 2580 aaaatggctt tcgctacctg gagagacgcg cccgctgatc ctttgcgaat acgcccacgc 2640 gatgggtaac agtcttggcg gtttcgctaa atactggcag gcgtttcgtc agtatccccg 2700 tttacagggc ggcttcgtct gggactgggt ggatcagtcg ctgattaaat atgatgaaaa 2760 cggcaacccg tggtcggctt acggcggtga ttttggcgat acgccgaacg atcgccagtt 2820 ctgtatgaac ggtctggtct ttgccgaccg cacgccgcat ccagcgctga cggaagcaaa 2880 acaccagcag cagtttttcc agttccgttt atccgggcaa accatcgaag tgaccagcga 2940 atacctgttc cgtcatagcg ataacgagct cctgcactgg atggtggcgc tggatggtaa 3000 gccgctggca agcggtgaag tgcctctgga tgtcgctcca caaggtaaac agttgattga 3060 actgcctgaa ctaccgcagc cggagagcgc cgggcaactc tggctcacag tacgcgtagt 3120 gcaaccgaac gcgaccgcat ggtcagaagc cgggcacatc agcgcctggc agcagtggcg 3180 tctggcggaa aacctcagtg tgacgctccc cgccgcgtcc cacgccatcc cgcatctgac 3240 caccagcgaa atggattttt gcatcgagct gggtaataag cgttggcaat ttaaccgcca 3300 gtcaggcttt ctttcacaga tgtggattgg cgataaaaaa caactgctga cgccgctgcg 3360 cgatcagttc acccgtgcac cgctggataa cgacattggc gtaagtgaag cgacccgcat 3420 tgaccctaac gcctgggtcg aacgctggaa ggcggcgggc cattaccagg ccgaagcagc 3480 gttgttgcag tgcacggcag atacacttgc tgatgcggtg ctgattacga ccgctcacgc 3540 gtggcagcat caggggaaaa ccttatttat cagccggaaa acctaccgga ttgatggtag 3600 tggtcaaatg gcgattaccg ttgatgttga agtggcgagc gatacaccgc atccggcgcg 3660 gattggcctg aactgccagc tggcgcaggt agcagagcgg gtaaactggc tcggattagg 3720 gccgcaagaa aactatcccg accgccttac tgccgcctgt tttgaccgct gggatctgcc 3780 attgtcagac atgtataccc cgtacgtctt cccgagcgaa aacggtctgc gctgcgggac 3840 gcgcgaattg aattatggcc cacaccagtg gcgcggcgac ttccagttca acatcagccg 3900 ctacagtcaa cagcaactga tggaaaccag ccatcgccat ctgctgcacg cggaagaagg 3960 cacatggctg aatatcgacg gtttccatat ggggattggt ggcgacgact cctggagccc 4020 gtcagtatcg gcggagttcc agctgagcgc cggtcgctac cattaccagt tggtctggtg 4080 tcaaaaataa taaagccgaa ttctgcagat atccagcaca gtggcggccg ctcgagtcta 4140 gagggcccgt ttaaacccgc tgatcagcct cgactgtgcc ttctagttgc cagccatctg 4200 ttgtttgccc ctcccccgtg ccttccttga ccctggaagg tgccactccc actgtccttt 4260 cctaataaaa tgaggaaatt gcatcgcatt gtctgagtag gtgtcattct attctggggg 4320 gtggggtggg gcaggacagc aagggggagg attgggaaga caatagcagg catgctgggg 4380 atgcggtggg ctctatggct tctgaggcgg aaagaaccag ctggggctct agggggtatc 4440 cccacgcgcc ctgtagcggc gcattaagcg cggcgggtgt ggtggttacg cgcagcgtga 4500 ccgctacact tgccagcgcc ctagcgcccg ctcctttcgc tttcttccct tcctttctcg 4560 ccacgttcgc cggctttccc cgtcaagctc taaatcgggg catcccttta gggttccgat 4620 ttagtgcttt acggcacctc gaccccaaaa aacttgatta gggtgatggt tcacgtagtg 4680 ggccatcgcc ctgatagacg gtttttcgcc ctttgacgtt ggagtccacg ttctttaata 4740 gtggactctt gttccaaact ggaacaacac tcaaccctat ctcggtctat tcttttgatt 4800 tataagggat tttggggatt tcggcctatt ggttaaaaaa tgagctgatt taacaaaaat 4860 ttaacgcgaa ttaattctgt ggaatgtgtg tcagttaggg tgtggaaagt ccccaggctc 4920 cccaggcagg cagaagtatg caaagcatgc atctcaatta gtcagcaacc aggtgtggaa 4980 agtccccagg ctccccagca ggcagaagta tgcaaagcat gcatctcaat tagtcagcaa 5040 ccatagtccc gcccctaact ccgcccatcc cgcccctaac tccgcccagt tccgcccatt 5100 ctccgcccca tggctgacta atttttttta tttatgcaga ggccgaggcc gcctctgcct 5160 ctgagctatt ccagaagtag tgaggaggct tttttggagg cctaggcttt tgcaaaaagc 5220 tcccgggagc ttgtatatcc attttcggat ctgatcaaga gacaggatga ggatcgtttc 5280 gcatgattga acaagatgga ttgcacgcag gttctccggc cgcttgggtg gagaggctat 5340 tcggctatga ctgggcacaa cagacaatcg gctgctctga tgccgccgtg ttccggctgt 5400 cagcgcaggg gcgcccggtt ctttttgtca agaccgacct gtccggtgcc ctgaatgaac 5460 tgcaggacga ggcagcgcgg ctatcgtggc tggccacgac gggcgttcct tgcgcagctg 5520 tgctcgacgt tgtcactgaa gcgggaaggg actggctgct attgggcgaa gtgccggggc 5580 aggatctcct gtcatctcac cttgctcctg ccgagaaagt atccatcatg gctgatgcaa 5640 tgcggcggct gcatacgctt gatccggcta cctgcccatt cgaccaccaa gcgaaacatc 5700 gcatcgagcg agcacgtact cggatggaag ccggtcttgt cgatcaggat gatctggacg 5760 aagagcatca ggggctcgcg ccagccgaac tgttcgccag gctcaaggcg cgcatgcccg 5820 acggcgagga tctcgtcgtg acccatggcg atgcctgctt gccgaatatc atggtggaaa 5880 atggccgctt ttctggattc atcgactgtg gccggctggg tgtggcggac cgctatcagg 5940 acatagcgtt ggctacccgt gatattgctg aagagcttgg cggcgaatgg gctgaccgct 6000 tcctcgtgct ttacggtatc gccgctcccg attcgcagcg catcgccttc tatcgccttc 6060 ttgacgagtt cttctgagcg ggactctggg gttcgaaatg accgaccaag cgacgcccaa 6120 cctgccatca cgagatttcg attccaccgc cgccttctat gaaaggttgg gcttcggaat 6180 cgttttccgg gacgccggct ggatgatcct ccagcgcggg gatctcatgc tggagttctt 6240 cgcccacccc aacttgttta ttgcagctta taatggttac aaataaagca atagcatcac 6300 aaatttcaca aataaagcat ttttttcact gcattctagt tgtggtttgt ccaaactcat 6360 caatgtatct tatcatgtct gtataccgtc gacctctagc tagagcttgg cgtaatcatg 6420 gtcatagctg tttcctgtgt gaaattgtta tccgctcaca attccacaca acatacgagc 6480 cggaagcata aagtgtaaag cctggggtgc ctaatgagtg agctaactca cattaattgc 6540 gttgcgctca ctgcccgctt tccagtcggg aaacctgtcg tgccagctgc attaatgaat 6600 cggccaacgc gcggggagag gcggtttgcg tattgggcgc tcttccgctt cctcgctcac 6660 tgactcgctg cgctcggtcg ttcggctgcg gcgagcggta tcagctcact caaaggcggt 6720 aatacggtta tccacagaat caggggataa cgcaggaaag aacatgtgag caaaaggcca 6780 gcaaaaggcc aggaaccgta aaaaggccgc gttgctggcg tttttccata ggctccgccc 6840 ccctgacgag catcacaaaa atcgacgctc aagtcagagg tggcgaaacc cgacaggact 6900 ataaagatac caggcgtttc cccctggaag ctccctcgtg cgctctcctg ttccgaccct 6960 gccgcttacc ggatacctgt ccgcctttct cccttcggga agcgtggcgc tttctcaatg 7020 ctcacgctgt aggtatctca gttcggtgta ggtcgttcgc tccaagctgg gctgtgtgca 7080 cgaacccccc gttcagcccg accgctgcgc cttatccggt aactatcgtc ttgagtccaa 7140 cccggtaaga cacgacttat cgccactggc agcagccact ggtaacagga ttagcagagc 7200 gaggtatgta ggcggtgcta cagagttctt gaagtggtgg cctaactacg gctacactag 7260 aaggacagta tttggtatct gcgctctgct gaagccagtt accttcggaa aaagagttgg 7320 tagctcttga tccggcaaac aaaccaccgc tggtagcggt ggtttttttg tttgcaagca 7380 gcagattacg cgcagaaaaa aaggatctca agaagatcct ttgatctttt ctacggggtc 7440 tgacgctcag tggaacgaaa actcacgtta agggattttg gtcatgagat tatcaaaaag 7500 gatcttcacc tagatccttt taaattaaaa atgaagtttt aaatcaatct aaagtatata 7560 tgagtaaact tggtctgaca gttaccaatg cttaatcagt gaggcaccta tctcagcgat 7620 ctgtctattt cgttcatcca tagttgcctg actccccgtc gtgtagataa ctacgatacg 7680 ggagggctta ccatctggcc ccagtgctgc aatgataccg cgagacccac gctcaccggc 7740 tccagattta tcagcaataa accagccagc cggaagggcc gagcgcagaa gtggtcctgc 7800 aactttatcc gcctccatcc agtctattaa ttgttgccgg gaagctagag taagtagttc 7860 gccagttaat agtttgcgca acgttgttgc cattgctaca ggcatcgtgg tgtcacgctc 7920 gtcgtttggt atggcttcat tcagctccgg ttcccaacga tcaaggcgag ttacatgatc 7980 ccccatgttg tgcaaaaaag cggttagctc cttcggtcct ccgatcgttg tcagaagtaa 8040 gttggccgca gtgttatcac tcatggttat ggcagcactg cataattctc ttactgtcat 8100 gccatccgta agatgctttt ctgtgactgg tgagtactca accaagtcat tctgagaata 8160 gtgtatgcgg cgaccgagtt gctcttgccc ggcgtcaata cgggataata ccgcgccaca 8220 tagcagaact ttaaaagtgc tcatcattgg aaaacgttct tcggggcgaa aactctcaag 8280 gatcttaccg ctgttgagat ccagttcgat gtaacccact cgtgcaccca actgatcttc 8340 agcatctttt actttcacca gcgtttctgg gtgagcaaaa acaggaaggc aaaatgccgc 8400 aaaaaaggga ataagggcga cacggaaatg ttgaatactc atactcttcc tttttcaata 8460 ttattgaagc atttatcagg gttattgtct catgagcgga tacatatttg aatgtattta 8520 gaaaaataaa caaatagggg ttccgcgcac atttccccga aaagtgccac ctgacgtc 8578 4 4748 DNA Artificial Sequence Provided by Dr. Brett Levay-Young of the University of Minnesota 4 tagttattaa tagtaatcaa ttacggggtc attagttcat agcccatata tggagttccg 60 cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 120 gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 180 atgggtggag tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 240 aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 300 catgacctta tgggactttc ctacttggca gtacatctac gtattagtca tcgctattac 360 catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg actcacgggg 420 atttccaagt ctccacccca ttgacgtcaa tgggagtttg ttttggcacc aaaatcaacg 480 ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg gtaggcgtgt 540 acggtgggag gtctatataa gcagagctgg tttagtgaac cgtcagatcc gctagcgcta 600 ccggtcgcca ccatggtgag caagggcgag gagctgttca ccggggtggt gcccatcctg 660 gtcgagctgg acggcgacgt aaacggccac aagttcagcg tgtccggcga gggcgagggc 720 gatgccacct acggcaagct gaccctgaag ttcatctgca ccaccggcaa gctgcccgtg 780 ccctggccca ccctcgtgac caccctgacc tacggcgtgc agtgcttcag ccgctacccc 840 gaccacatga agcagcacga cttcttcaag tccgccatgc ccgaaggcta cgtccaggag 900 cgcaccatct tcttcaagga cgacggcaac tacaagaccc gcgccgaggt gaagttcgag 960 ggcgacaccc tggtgaaccg catcgagctg aagggcatcg acttcaagga ggacggcaac 1020 atcctggggc acaagctgga gtacaactac aacagccaca acgtctatat catggccgac 1080 aagcagaaga acggcatcaa ggtgaacttc aagatccgcc acaacatcga ggacggcagc 1140 gtgcagctcg ccgaccacta ccagcagaac acccccatcg gcgacggccc cgtgctgctg 1200 cccgacaacc actacctgag cacccagtcc gccctgagca aagaccccaa cgagaagcgc 1260 gatcacatgg tcctgctgga gttcgtgacc gccgccggga tcactctcgg catggacgag 1320 ctgtacaagt actcagatct cgagctcaag cttaaccctc cggacgagag cggccctggc 1380 tgtatgtcct gcaagtgcgt gctgtcctga tcaccggatc tagataactg atcataatca 1440 gccataccac atttgtagag gttttacttg ctttaaaaaa cctcccacac ctccccctga 1500 acctgaaaca taaaatgaat gcaattgttg ttgttaactt gtttattgca gcttataatg 1560 gttacaaata aagcaatagc atcacaaatt tcacaaataa agcatttttt tcactgcatt 1620 ctagttgtgg tttgtccaaa ctcatcaatg tatcttaacg cgtaaattgt aagcgttaat 1680 attttgttaa aattcgcgtt aaatttttgt taaatcagct cattttttaa ccaataggcc 1740 gaaatcggca aaatccctta taaatcaaaa gaatagaccg agatagggtt gagtgttgtt 1800 ccagtttgga acaagagtcc actattaaag aacgtggact ccaacgtcaa agggcgaaaa 1860 accgtctatc agggcgatgg cccactacgt gaaccatcac cctaatcaag ttttttgggg 1920 tcgaggtgcc gtaaagcact aaatcggaac cctaaaggga gcccccgatt tagagcttga 1980 cggggaaagc cggcgaacgt ggcgagaaag gaagggaaga aagcgaaagg agcgggcgct 2040 agggcgctgg caagtgtagc ggtcacgctg cgcgtaacca ccacacccgc cgcgcttaat 2100 gcgccgctac agggcgcgtc aggtggcact tttcggggaa atgtgcgcgg aacccctatt 2160 tgtttatttt tctaaataca ttcaaatatg tatccgctca tgagacaata accctgataa 2220 atgcttcaat aatattgaaa aaggaagagt cctgaggcgg aaagaaccag ctgtggaatg 2280 tgtgtcagtt agggtgtgga aagtccccag gctccccagc aggcagaagt atgcaaagca 2340 tgcatctcaa ttagtcagca accaggtgtg gaaagtcccc aggctcccca gcaggcagaa 2400 gtatgcaaag catgcatctc aattagtcag caaccatagt cccgccccta actccgccca 2460 tcccgcccct aactccgccc agttccgccc attctccgcc ccatggctga ctaatttttt 2520 ttatttatgc agaggccgag gccgcctcgg cctctgagct attccagaag tagtgaggag 2580 gcttttttgg aggcctaggc ttttgcaaag atcgatcaag agacaggatg aggatcgttt 2640 cgcatgattg aacaagatgg attgcacgca ggttctccgg ccgcttgggt ggagaggcta 2700 ttcggctatg actgggcaca acagacaatc ggctgctctg atgccgccgt gttccggctg 2760 tcagcgcagg ggcgcccggt tctttttgtc aagaccgacc tgtccggtgc cctgaatgaa 2820 ctgcaagacg aggcagcgcg gctatcgtgg ctggccacga cgggcgttcc ttgcgcagct 2880 gtgctcgacg ttgtcactga agcgggaagg gactggctgc tattgggcga agtgccgggg 2940 caggatctcc tgtcatctca ccttgctcct gccgagaaag tatccatcat ggctgatgca 3000 atgcggcggc tgcatacgct tgatccggct acctgcccat tcgaccacca agcgaaacat 3060 cgcatcgagc gagcacgtac tcggatggaa gccggtcttg tcgatcagga tgatctggac 3120 gaagagcatc aggggctcgc gccagccgaa ctgttcgcca ggctcaaggc gagcatgccc 3180 gacggcgagg atctcgtcgt gacccatggc gatgcctgct tgccgaatat catggtggaa 3240 aatggccgct tttctggatt catcgactgt ggccggctgg gtgtggcgga ccgctatcag 3300 gacatagcgt tggctacccg tgatattgct gaagagcttg gcggcgaatg ggctgaccgc 3360 ttcctcgtgc tttacggtat cgccgctccc gattcgcagc gcatcgcctt ctatcgcctt 3420 cttgacgagt tcttctgagc gggactctgg ggttcgaaat gaccgaccaa gcgacgccca 3480 acctgccatc acgagatttc gattccaccg ccgccttcta tgaaaggttg ggcttcggaa 3540 tcgttttccg ggacgccggc tggatgatcc tccagcgcgg ggatctcatg ctggagttct 3600 tcgcccaccc tagggggagg ctaactgaaa cacggaagga gacaataccg gaaggaaccc 3660 gcgctatgac ggcaataaaa agacagaata aaacgcacgg tgttgggtcg tttgttcata 3720 aacgcggggt tcggtcccag ggctggcact ctgtcgatac cccaccgaga ccccattggg 3780 gccaatacgc ccgcgtttct tccttttccc caccccaccc cccaagttcg ggtgaaggcc 3840 cagggctcgc agccaacgtc ggggcggcag gccctgccat agcctcaggt tactcatata 3900 tactttagat tgatttaaaa cttcattttt aatttaaaag gatctaggtg aagatccttt 3960 ttgataatct catgaccaaa atcccttaac gtgagttttc gttccactga gcgtcagacc 4020 ccgtagaaaa gatcaaagga tcttcttgag atcctttttt tctgcgcgta atctgctgct 4080 tgcaaacaaa aaaaccaccg ctaccagcgg tggtttgttt gccggatcaa gagctaccaa 4140 ctctttttcc gaaggtaact ggcttcagca gagcgcagat accaaatact gtccttctag 4200 tgtagccgta gttaggccac cacttcaaga actctgtagc accgcctaca tacctcgctc 4260 tgctaatcct gttaccagtg gctgctgcca gtggcgataa gtcgtgtctt accgggttgg 4320 actcaagacg atagttaccg gataaggcgc agcggtcggg ctgaacgggg ggttcgtgca 4380 cacagcccag cttggagcga acgacctaca ccgaactgag atacctacag cgtgagctat 4440 gagaaagcgc cacgcttccc gaagggagaa aggcggacag gtatccggta agcggcaggg 4500 tcggaacagg agagcgcacg agggagcttc cagggggaaa cgcctggtat ctttatagtc 4560 ctgtcgggtt tcgccacctc tgacttgagc gtcgattttt gtgatgctcg tcaggggggc 4620 ggagcctatg gaaaaacgcc agcaacgcgg cctttttacg gttcctggcc ttttgctggc 4680 cttttgctca catgttcttt cctgcgttat cccctgattc tgtggataac cgtattaccg 4740 ccatgcat 4748 5 4992 DNA Artificial Sequence Supplied by BD Biosciences Clonetech of Palo Alto, California 5 tagttattaa tagtaatcaa ttacggggtc attagttcat agcccatata tggagttccg 60 cgttacataa cttacggtaa atggcccgcc tggctgaccg cccaacgacc cccgcccatt 120 gacgtcaata atgacgtatg ttcccatagt aacgccaata gggactttcc attgacgtca 180 atgggtggag tatttacggt aaactgccca cttggcagta catcaagtgt atcatatgcc 240 aagtacgccc cctattgacg tcaatgacgg taaatggccc gcctggcatt atgcccagta 300 catgacctta tgggactttc ctacttggca gtacatctac gtattagtca tcgctattac 360 catggtgatg cggttttggc agtacatcaa tgggcgtgga tagcggtttg actcacgggg 420 atttccaagt ctccacccca ttgacgtcaa tgggagtttg ttttggcacc aaaatcaacg 480 ggactttcca aaatgtcgta acaactccgc cccattgacg caaatgggcg gtaggcgtgt 540 acggtgggag gtctatataa gcagagctgg tttagtgaac cgtcagatcc gctagcgcta 600 ccggtcgcca ccatggtgag caagggcgag gagctgttca ccggggtggt gcccatcctg 660 gtcgagctgg acggcgacgt aaacggccac aagttcagcg tgtccggcga gggcgagggc 720 gatgccacct acggcaagct gaccctgaag ttcatctgca ccaccggcaa gctgcccgtg 780 ccctggccca ccctcgtgac caccctgacc tacggcgtgc agtgcttcag ccgctacccc 840 gaccacatga agcagcacga cttcttcaag tccgccatgc ccgaaggcta cgtccaggag 900 cgcaccatct tcttcaagga cgacggcaac tacaagaccc gcgccgaggt gaagttcgag 960 ggcgacaccc tggtgaaccg catcgagctg aagggcatcg acttcaagga ggacggcaac 1020 atcctggggc acaagctgga gtacaactac aacagccaca acgtctatat catggccgac 1080 aagcagaaga acggcatcaa ggtgaacttc aagatccgcc acaacatcga ggacggcagc 1140 gtgcagctcg ccgaccacta ccagcagaac acccccatcg gcgacggccc cgtgctgctg 1200 cccgacaacc actacctgag cacccagtcc gccctgagca aagaccccaa cgagaagcgc 1260 gatcacatgg tcctgctgga gttcgtgacc gccgccggga tcactctcgg catggacgag 1320 ctgtacaagt actcagatct cgagctcaag cttaccatgg ggggttctca tcatcatcat 1380 catcatggta tggctagcat gactggtgga cagcaaatgg gtcgggatct gtacgacgat 1440 gacgataagg ggactgctgc ggccaatgcg aacgacttct tcgccaagcg caagagaact 1500 gcgcaggaga acaaggcgtc gaacgacgtc cctccagggt gtccctctcc aaacgtggct 1560 cctggggtgg gcgcggtgga gcagaccccg cgcaaacgtc tgagatgagg atccagtgtg 1620 gtggaattct gcagatatcc agcacagtgg cggccgctcg agtctagata actgatcata 1680 atcagccata ccacatttgt agaggtttta cttgctttaa aaaacctccc acacctcccc 1740 ctgaacctga aacataaaat gaatgcaatt gttgttgtta acttgtttat tgcagcttat 1800 aatggttaca aataaagcaa tagcatcaca aatttcacaa ataaagcatt tttttcactg 1860 cattctagtt gtggtttgtc caaactcatc aatgtatctt aacgcgtaaa ttgtaagcgt 1920 taatattttg ttaaaattcg cgttaaattt ttgttaaatc agctcatttt ttaaccaata 1980 ggccgaaatc ggcaaaatcc cttataaatc aaaagaatag accgagatag ggttgagtgt 2040 tgttccagtt tggaacaaga gtccactatt aaagaacgtg gactccaacg tcaaagggcg 2100 aaaaaccgtc tatcagggcg atggcccact acgtgaacca tcaccctaat caagtttttt 2160 ggggtcgagg tgccgtaaag cactaaatcg gaaccctaaa gggagccccc gatttagagc 2220 ttgacgggga aagccggcga acgtggcgag aaaggaaggg aagaaagcga aaggagcggg 2280 cgctagggcg ctggcaagtg tagcggtcac gctgcgcgta accaccacac ccgccgcgct 2340 taatgcgccg ctacagggcg cgtcaggtgg cacttttcgg ggaaatgtgc gcggaacccc 2400 tatttgttta tttttctaaa tacattcaaa tatgtatccg ctcatgagac aataaccctg 2460 ataaatgctt caataatatt gaaaaaggaa gagtcctgag gcggaaagaa ccagctgtgg 2520 aatgtgtgtc agttagggtg tggaaagtcc ccaggctccc cagcaggcag aagtatgcaa 2580 agcatgcatc tcaattagtc agcaaccagg tgtggaaagt ccccaggctc cccagcaggc 2640 agaagtatgc aaagcatgca tctcaattag tcagcaacca tagtcccgcc cctaactccg 2700 cccatcccgc ccctaactcc gcccagttcc gcccattctc cgccccatgg ctgactaatt 2760 ttttttattt atgcagaggc cgaggccgcc tcggcctctg agctattcca gaagtagtga 2820 ggaggctttt ttggaggcct aggcttttgc aaagatcgat caagagacag gatgaggatc 2880 gtttcgcatg attgaacaag atggattgca cgcaggttct ccggccgctt gggtggagag 2940 gctattcggc tatgactggg cacaacagac aatcggctgc tctgatgccg ccgtgttccg 3000 gctgtcagcg caggggcgcc cggttctttt tgtcaagacc gacctgtccg gtgccctgaa 3060 tgaactgcaa gacgaggcag cgcggctatc gtggctggcc acgacgggcg ttccttgcgc 3120 agctgtgctc gacgttgtca ctgaagcggg aagggactgg ctgctattgg gcgaagtgcc 3180 ggggcaggat ctcctgtcat ctcaccttgc tcctgccgag aaagtatcca tcatggctga 3240 tgcaatgcgg cggctgcata cgcttgatcc ggctacctgc ccattcgacc accaagcgaa 3300 acatcgcatc gagcgagcac gtactcggat ggaagccggt cttgtcgatc aggatgatct 3360 ggacgaagag catcaggggc tcgcgccagc cgaactgttc gccaggctca aggcgagcat 3420 gcccgacggc gaggatctcg tcgtgaccca tggcgatgcc tgcttgccga atatcatggt 3480 ggaaaatggc cgcttttctg gattcatcga ctgtggccgg ctgggtgtgg cggaccgcta 3540 tcaggacata gcgttggcta cccgtgatat tgctgaagag cttggcggcg aatgggctga 3600 ccgcttcctc gtgctttacg gtatcgccgc tcccgattcg cagcgcatcg ccttctatcg 3660 ccttcttgac gagttcttct gagcgggact ctggggttcg aaatgaccga ccaagcgacg 3720 cccaacctgc catcacgaga tttcgattcc accgccgcct tctatgaaag gttgggcttc 3780 ggaatcgttt tccgggacgc cggctggatg atcctccagc gcggggatct catgctggag 3840 ttcttcgccc accctagggg gaggctaact gaaacacgga aggagacaat accggaagga 3900 acccgcgcta tgacggcaat aaaaagacag aataaaacgc acggtgttgg gtcgtttgtt 3960 cataaacgcg gggttcggtc ccagggctgg cactctgtcg ataccccacc gagaccccat 4020 tggggccaat acgcccgcgt ttcttccttt tccccacccc accccccaag ttcgggtgaa 4080 ggcccagggc tcgcagccaa cgtcggggcg gcaggccctg ccatagcctc aggttactca 4140 tatatacttt agattgattt aaaacttcat ttttaattta aaaggatcta ggtgaagatc 4200 ctttttgata atctcatgac caaaatccct taacgtgagt tttcgttcca ctgagcgtca 4260 gaccccgtag aaaagatcaa aggatcttct tgagatcctt tttttctgcg cgtaatctgc 4320 tgcttgcaaa caaaaaaacc accgctacca gcggtggttt gtttgccgga tcaagagcta 4380 ccaactcttt ttccgaaggt aactggcttc agcagagcgc agataccaaa tactgtcctt 4440 ctagtgtagc cgtagttagg ccaccacttc aagaactctg tagcaccgcc tacatacctc 4500 gctctgctaa tcctgttacc agtggctgct gccagtggcg ataagtcgtg tcttaccggg 4560 ttggactcaa gacgatagtt accggataag gcgcagcggt cgggctgaac ggggggttcg 4620 tgcacacagc ccagcttgga gcgaacgacc tacaccgaac tgagatacct acagcgtgag 4680 ctatgagaaa gcgccacgct tcccgaaggg agaaaggcgg acaggtatcc ggtaagcggc 4740 agggtcggaa caggagagcg cacgagggag cttccagggg gaaacgcctg gtatctttat 4800 agtcctgtcg ggtttcgcca cctctgactt gagcgtcgat ttttgtgatg ctcgtcaggg 4860 gggcggagcc tatggaaaaa cgccagcaac gcggcctttt tacggttcct ggccttttgc 4920 tggccttttg ctcacatgtt ctttcctgcg ttatcccctg attctgtgga taaccgtatt 4980 accgccatgc at 4992

Claims (65)

1. A composition comprising:
a plurality of particles comprising a surfactant having an HLB value of less than about 6.0 units associated with a functional composition and a polymer, with the particles having an average diameter of less than about 100 nanometers as measured by atomic force microscopy of the particles following drying of the particles, wherein the functional composition is a member of the group consisting of a bioactive component and a diagnostic agent.
2. The composition of claim 1 wherein the surfactant is a non-ionic surfactant.
3. The composition of claim 1 wherein the surfactant has an HLB value of less than about 5.0 units.
4. The composition of claim 3 wherein the average diameter of the particles is less than about 50 nm.
5. The composition of claim 1 wherein the functional composition is a member of the group consisting of peptides, proteins, and carbohydrates.
6. The composition of claim 5 wherein the average diameter is less than about 50 nm.
7. The composition of claim 1 wherein the functional composition is cisplatin.
8. The composition of claim 1 wherein the functional composition is inorganic.
9. The composition of claim 1 wherein the functional composition is a diagnostic agent that is a fluorescent molecule.
10. The composition of claim 1 wherein the functional composition is a bioactive component that comprises a hydrophilic component.
11. The composition of claim 10 wherein the bioactive component is condensed.
12. The composition of claim 1 wherein the functional composition is a bioactive component that is a member of the group consisting of aptamers, mini-chromosomes, steroids, adrenergic, adrenocotical steroid, adrenocortical suppressant, aldosterone antagonist, and anabolic agents; analeptic, analgesic, anesthetic, anorectic, anti-acne agents; anti-adrenergic, anti-allergic, anti-amebic, anti-anemic, and anti-anginal agents; anti-arthritic, anti-asthmatic, anti-atherosclerotic, antibacterial, and anticholinergic agents; anticoagulant, anticonvulsant, antidepressant, antidiabetic, and antidiarrheal agents; antidiuretic, anti-emetic, anti-epileptic, antifibrinolytic, and antifungal agent; antihemorrhagic, inflammatory, antimicrobial, antimigraine, and antimiotic agents; antimycotic, antinauseant, antineoplastic, antineutropenic, and antiparasitic agents; antiproliferative, antipsychotic, antirheumatic, antiseborrheic, and antisecretory agents; antipasmodic, antihrombotic, anti-ulcerative, antiviral and appetite suppressant agents.
13. The composition of claim 1 wherein the functional composition is a bioactive component that is a member of the group consisting of blood glucose regulator, bone resorption inhibitor, bronchodilator, cardiovascular, and cholinergic agents; fluorescent, free oxygen radical scavenger, gastrointestinal motility effector, glucocorticoid, and hair growth stimulant agent; hemostatic, histamine H2 receptor antagonists; hormone; hypocholesterolemic, and hypoglycemic agents; hypolipidemic, hypotensive, and imaging agents, immunizing and agonist agents; mood regulators, mucolytic, mydriatic, nasal decongestant; neuromuscular blocking agents; neuroprotective, NMDA antagonist, non-hormonal sterol derivative, plasminogen activator, and platelet activating factor antagonist agent.
14. The composition of claim 1 wherein the functional composition is a bioactive component that is a member of the group consisting of platelet aggregation inhibitor, psychotropic, radioactive, scabicide, and sclerosing agents; sedative, sedative-hypnotic, selective adenosine A1 antagonist, serotonin antagonist, and serotonin inhibitor agent; serotonin receptor antagonist, steroid, thyroid hormone, thyroid hormone, thyroid inhibitor agent; thyromimetic, tranquilizer, amyotrophic lateral sclerosis, cerebral ischemia, Pagel's disease agent; unstable angina, vasoconstrictor, vasodilator, wound healing, and xanthine oxidase inhibitor agent; and immunological agents.
15. The composition of claim 1 wherein the functional composition is a bioactive component that comprises a member of the group consisting of antigens isolated from pathogens, viral antigens, fungal antigens, parasitic antigens, and inactivated pathogenic organisms.
16. The composition of claim 1 wherein the surfactant has a critical micelle concentration of less than about 200 micromolar.
17. The composition of claim 1 further comprising a biocompatible oil.
18. The composition of claim 1 wherein the functional composition is a bioactive component that comprises a polynucleic acid, oligonucleotide, antisense molecule, or a polypeptide.
19. The composition of claim 18 wherein the bioactive component is condensed.
20. The composition of claim 19 wherein the composition further comprises a water-miscible solvent.
21. The composition of claim 1 wherein the surfactant is selected from the group consisting of 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol, molecules containing an acetylenic diol portion, and blends of 2, 4, 7, 9-tetramethyl-5-decyn-4, 7-diol.
22. The composition of claim 1 wherein the particles further comprise a cell recognition component.
23. The composition of claim 22 wherein the cell recognition component is a ligand, peptide hormone, or an antibody.
24. The composition of claim 23 wherein the cell recognition component comprises tenascin, hyaluronan, or polyvinylpyrrolidone, or a fragment thereof.
25. The composition of claim 24 wherein the average diameter of the particles is less than about 50 nm.
26. The composition of claim 24 wherein the cell recognition component comprises tenascin or a fragment thereof.
27. The composition of claim 1 wherein the average diameter of the particles is less than about 50 nm.
28. The composition of claim 1 wherein the polymer is an iontophoretic polymer.
29. The composition of claim 1 wherein the polymer is a hydrophobic polymer.
30. The composition of claim 1 wherein the polymer is a hydrophilic polymer.
31. The composition of claim 1 wherein the polymer is chosen from the group consisting of polyamides, polycarbonates, polyalkylenes, polyalkylene glycols, polyalkylene oxides, polyalkylene terepthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkyl cellulose, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, nitro celluloses, polymers of acrylic and methacrylic esters, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxy-propyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxylethyl cellulose, cellulose triacetate, and cellulose sulphate sodium salt.
32. The composition of claim 1 wherein the polymer is chosen from the group consisting of poly(methyl methacrylate), poly(ethylmethacrylate), poly(butylmethacrylate), poly(isobutylmethacrylate), poly(hexlmethacrylate), poly(isodecylmethacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), polyethylene, polypropylene poly(ethylene glycol), poly(ethylene oxide), and poly(ethylene terephthalate).
33. The composition of claim 1 wherein the polymer is chosen from the group consisting of poly(vinyl alcohols), poly(vinyl acetate, poly vinyl chloride polystyrene, polyvinylpryrrolidone, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutylmethacrylate), poly(hexlmethacrylate), poly(isodecl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and poly(octadecl acrylate).
34. The composition of claim 1 wherein the hydrophilic polymer is a member of the group consisting of proteinaceous materials , peptides, carbohydrates.
35. A method of delivering a functional composition across keratinized barrier epithelia to a cell, the method comprising introducing the composition of claim 1 at a position that is separated from the cell by a keratinized barrier epithelium, wherein at least a portion of the plurality of particles passes through the keratinized barrier epithelium to the cell.
36. The method of claim 35 wherein the functional composition is delivered transcutaneously.
37. The method of claim 35 wherein the composition of claim 1 is prepared as a medicament, and the medicament is administered to a patient.
38. A solution comprising the composition of claim 1, the solution comprising a concentration of cations between 20 and 2000 millimolar.
39. The composition of claim 1, further comprising a cation chosen from the group consisting of Ni2+, Mn2+, Mg2+, Ca2+, Al3+, Be2+, Li+, Ba2+, and Gd3+.
40. A medicament comprising the composition of claim 1.
41. The medicament of claim 39 further comprising a form selected from the group consisting of granules, tablets, pellets, films, oral, intravenous, subcutaneous, intraperitoneal, intrathecal, intramuscular, inhalation, topical, transdermal, suppository, pessary, intra urethral, intraportal, intraocular, transtympanic, intrahepatic, intra-arterial, intrathecal, transmucosal, coatings, buccal, and combinations thereof.
42. A method of delivering a functional composition to a patient, the method comprising administering a medicament to the patient that comprises the composition of claim 1.
43. A cell comprising the composition of claim 1, wherein the plurality of particles is associated with the cell.
44. A cell comprising the composition of claim 18, wherein the plurality of particles is associated with the cell.
45. A method of transfecting a cell, the method comprising exposing the cell to the composition of claim 18.
46. A matrix for binding bioactive or diagnostic particles, the matrix comprising the particles of claim 1 and a binder.
47. A method of delivering a medical agent to a cell having caveolae, the method comprising: associating the medical agent with an organic functional composition in vitro to make an association of the medical agent and the organic functional composition, wherein the assoication is passable through cellular caveolae for delivery of the medical agent.
48. The method of claim 47 wherein the association of the agent and the functional composition comprises a particle.
49. The method of claim 48 wherein the particle has a diameter of less than about 100 nanometers as measured by atomic force microscopy of the particles following drying of the particles.
50. The method of claim 48 wherein the particle has a diameter of less than about 50 nm.
51. The method of claim 48 wherein the particle further comprises a surfactant having an HLB value of less than about 6.0 units.
52. The method of claim 51 wherein the particle has an average diameter of less than about 50 nanometers as measured by atomic force microscopy of the particles following drying of the particles.
53. The method of claim 51 further comprising exposing the particle to the cell.
54. The method of claim 47 wherein the association of the agent and the functional composition has a maximum dimension of no more than about 50 nm nanometers as measured by atomic force microscopy following drying of the association of the agent and the functional composition.
55. The method of claim 47 wherein the functional composition comprises a surfactant.
56. The method of claim 47 further comprising exposing the association of the agent and the functional composition to the cell.
57. The method of claim 47 further comprising administering a medicament to a patient, the medicament comprising the association of the medical agent and the organic functional composition.
58. The method of claim 47 wherein the functional composition comprises a surfactant and a hydrophilic polymer.
59. The method of claim 47 wherein the agent comprises a bioactive component that a member of the group consisting of peptides, proteins, and carbohydrates.
60. The method of claim 47 wherein the agent comprises a member of the group consisting of a bioactive component and a diagnostic agent.
61. The method of claim 47 wherein the agent comprises a fragment of a nucleic acid that comprises a nucleic acid sequence.
62. The method of claim 61 wherein the functional composition comprises a surfactant.
63. The method of claim 47 wherein the association is introduced at a position that is separated from the cell by keratinized barrier epithelia, and the association passes through the keratinized barrier epithelia to the cell.
64. The method of claim 63 further comprising exposing the cell to the association of the agent and the functional composition
65. The method of claim 47 wherein the functional composition comprises carbon and hydrogen.
US10/652,814 2000-02-28 2003-08-29 Nanocapsule encapsulation system and method Abandoned US20040137071A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/652,814 US20040137071A1 (en) 2000-02-28 2003-08-29 Nanocapsule encapsulation system and method

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US18528200P 2000-02-28 2000-02-28
US09/796,575 US6632671B2 (en) 2000-02-28 2001-02-28 Nanoparticle encapsulation system and method
US10/652,814 US20040137071A1 (en) 2000-02-28 2003-08-29 Nanocapsule encapsulation system and method

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/796,575 Continuation US6632671B2 (en) 2000-02-28 2001-02-28 Nanoparticle encapsulation system and method

Publications (1)

Publication Number Publication Date
US20040137071A1 true US20040137071A1 (en) 2004-07-15

Family

ID=22680346

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/796,575 Expired - Lifetime US6632671B2 (en) 2000-02-28 2001-02-28 Nanoparticle encapsulation system and method
US10/652,814 Abandoned US20040137071A1 (en) 2000-02-28 2003-08-29 Nanocapsule encapsulation system and method

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/796,575 Expired - Lifetime US6632671B2 (en) 2000-02-28 2001-02-28 Nanoparticle encapsulation system and method

Country Status (9)

Country Link
US (2) US6632671B2 (en)
EP (1) EP1267946A4 (en)
JP (1) JP4751556B2 (en)
CN (1) CN1406140A (en)
AU (2) AU4724401A (en)
CA (1) CA2400172C (en)
HK (1) HK1048772A1 (en)
MX (1) MXPA02008361A (en)
WO (1) WO2001064164A2 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030206958A1 (en) * 2000-12-22 2003-11-06 Cattaneo Maurizio V. Chitosan biopolymer for the topical delivery of active agents
US20050281886A1 (en) * 2004-05-06 2005-12-22 Ivrea Pharmaceuticals, Inc. Particles for the delivery of active agents
US20060222692A1 (en) * 2005-03-31 2006-10-05 Fairfield Clinical Trials Llc Method and compositions for transdermal administration of antimicrobial medications
US20060258569A1 (en) * 2003-10-21 2006-11-16 Mctavish Hugh Compounds and methods for treating cancer
US20060258589A1 (en) * 2004-01-24 2006-11-16 Igf Oncology, Llc Methods for enhancing radiation therapy
US20070098713A1 (en) * 2002-04-08 2007-05-03 Genesegues, Inc. Nanoparticle delivery systems and methods of use thereof
US20070224119A1 (en) * 2004-10-21 2007-09-27 Igf Oncology Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US20080045583A1 (en) * 2006-08-18 2008-02-21 David Delmarre Stable levetiracetam compositions and methods
US20090143339A1 (en) * 2007-11-30 2009-06-04 Martin Purpura Methods using phosphatidylserine, lysophosphatidylserine, and/or salts thereof to increase testosterone levels
US20090297596A1 (en) * 2005-05-23 2009-12-03 Elan Pharma International Limited Nanoparticulate and Controlled Release Compositions Comprising a Platelet Aggregation Inhibitor
US20100119501A1 (en) * 2007-04-30 2010-05-13 Yi-Yan Yang Method of delivering a protein into a cell
US20100173001A1 (en) * 2007-06-14 2010-07-08 Genesegues, Inc. Metal Ion-Treated Biocompatible Polymers Useful for Nanoparticles
US20100216696A1 (en) * 2003-10-21 2010-08-26 Mctavish Hugh Compounds and methods for treating cancer
US20110038987A1 (en) * 2008-02-11 2011-02-17 Dganit Danino Beta-casein assemblies for enrichment of food and beverages and methods of preparation thereof
US20110045092A1 (en) * 2008-02-11 2011-02-24 Livney Yoav D Casein particles encapsulating therapeutically active agents and uses thereof
US20110052703A1 (en) * 2008-02-11 2011-03-03 Yechezkel Barenholz Beta-casein assemblies for mucosal delivery of therapeutic bioactive agents
US8685458B2 (en) 2009-03-05 2014-04-01 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
US8815294B2 (en) 2010-09-03 2014-08-26 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives and a carrier material
US8871276B2 (en) 2008-02-11 2014-10-28 Technion Research And Development Foundation Ltd. Beta-casein assemblies for mucosal delivery of therapeutic bioactive agents
US9084727B2 (en) 2011-05-10 2015-07-21 Bend Research, Inc. Methods and compositions for maintaining active agents in intra-articular spaces
US9675671B2 (en) 2014-01-12 2017-06-13 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof
US10987308B2 (en) 2014-09-03 2021-04-27 Genesegues, Inc. Therapeutic nanoparticles and related compositions, methods and systems
US11324834B2 (en) 2017-05-21 2022-05-10 Igf Oncology, Llc Insulin-like growth factor-chemotherapeputic conjugate for treating myelodysplastic syndrome
WO2022261148A1 (en) * 2021-06-09 2022-12-15 Scribe Therapeutics Inc. Compositions of glycoprotein particles

Families Citing this family (129)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6433154B1 (en) * 1997-06-12 2002-08-13 Bristol-Myers Squibb Company Functional receptor/kinase chimera in yeast cells
EP1267946A4 (en) * 2000-02-28 2008-07-02 Genesegues Inc Nanocapsule encapsulation system and method
AU2001261625B2 (en) 2000-05-16 2006-04-06 Regents Of The University Of Minnesota High mass throughput particle generation using multiple nozzle spraying
US8067032B2 (en) 2000-12-22 2011-11-29 Baxter International Inc. Method for preparing submicron particles of antineoplastic agents
US20050048126A1 (en) 2000-12-22 2005-03-03 Barrett Rabinow Formulation to render an antimicrobial drug potent against organisms normally considered to be resistant to the drug
US20030096013A1 (en) * 2000-12-22 2003-05-22 Jane Werling Preparation of submicron sized particles with polymorph control
US9700866B2 (en) 2000-12-22 2017-07-11 Baxter International Inc. Surfactant systems for delivery of organic compounds
US7247338B2 (en) * 2001-05-16 2007-07-24 Regents Of The University Of Minnesota Coating medical devices
AU2002309172A1 (en) * 2001-06-22 2003-01-08 Pfizer Products Inc. Pharmaceutical compositions containing polymer and drug assemblies
US7081489B2 (en) * 2001-08-09 2006-07-25 Florida State University Research Foundation Polymeric encapsulation of nanoparticles
CN1558755A (en) 2001-09-26 2004-12-29 ���ع��ʹ�˾ Preparation of submicron sized nanoparticles via dispersion and solvent or liquid phase removal
US20060003012A9 (en) 2001-09-26 2006-01-05 Sean Brynjelsen Preparation of submicron solid particle suspensions by sonication of multiphase systems
US7615676B2 (en) * 2001-11-30 2009-11-10 U.S. Department Of Veterans Affairs Transgenic screen and method for screening modulators of brain-derived neurotrophic factor (BDNF) production
US7491810B2 (en) * 2001-11-30 2009-02-17 U.S. Department Of Veterans Affairs Transgenic screen and method for screening modulators of brain-derived neurotrophic factor (BDNF) production
US7053034B2 (en) * 2002-04-10 2006-05-30 Salvona, Llc Targeted controlled delivery compositions activated by changes in pH or salt concentration
DE10236064A1 (en) * 2002-08-07 2004-02-19 Beiersdorf Ag Foaming sunscreen preparations
US6989196B2 (en) * 2002-10-02 2006-01-24 Florida State University Research Foundation Microencapsulation of magnetic material using heat stabilization
US7829694B2 (en) 2002-11-26 2010-11-09 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US7618948B2 (en) 2002-11-26 2009-11-17 Medtronic, Inc. Devices, systems and methods for improving and/or cognitive function through brain delivery of siRNA
US7605249B2 (en) * 2002-11-26 2009-10-20 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US20060018882A1 (en) * 2004-06-21 2006-01-26 Kaemmerer William F Medical devices and methods for delivering compositions to cells
US7732591B2 (en) * 2003-11-25 2010-06-08 Medtronic, Inc. Compositions, devices and methods for treatment of huntington's disease through intracranial delivery of sirna
US7994149B2 (en) 2003-02-03 2011-08-09 Medtronic, Inc. Method for treatment of Huntington's disease through intracranial delivery of sirna
AU2004249172A1 (en) * 2003-06-24 2004-12-29 Baxter International Inc. Specific delivery of drugs to the brain
US8986736B2 (en) * 2003-06-24 2015-03-24 Baxter International Inc. Method for delivering particulate drugs to tissues
WO2005011702A1 (en) * 2003-07-17 2005-02-10 Tibotec Pharmaceuticals Ltd. Process for preparing particles containing an antiviral
US20050202094A1 (en) * 2004-01-29 2005-09-15 Werling Jane O. Nanosuspensions of anti-retroviral agents for increased central nervous system delivery
KR20050081092A (en) * 2004-02-12 2005-08-18 한국과학기술연구원 Composition and formulation of colloidal system comprising biocompatible aqueous-soluble polymer, and preparation method thereof
US7354980B1 (en) 2004-03-12 2008-04-08 Key Medical Technologies, Inc. High refractive index polymers for ophthalmic applications
US20050208090A1 (en) * 2004-03-18 2005-09-22 Medtronic, Inc. Methods and systems for treatment of neurological diseases of the central nervous system
US7842281B2 (en) * 2004-05-10 2010-11-30 The Florida State University Research Foundation Magnetic particle composition for therapeutic hyperthermia
JP2008502706A (en) * 2004-06-15 2008-01-31 バクスター・インターナショナル・インコーポレイテッド Ex vivo application of solid particulate therapeutic agents
US7446157B2 (en) * 2004-12-07 2008-11-04 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications
WO2006065724A2 (en) * 2004-12-14 2006-06-22 Regents Of The University Of Minnesota Casein kinase 2 antisense therapy
CA2598558C (en) * 2005-02-14 2013-04-02 Australian Nuclear Science & Technology Organisation Layered nanoparticles
US7902352B2 (en) * 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
WO2006121960A2 (en) * 2005-05-06 2006-11-16 Medtronic, Inc. Methods and sequences to suppress primate huntington gene expression
US20080280843A1 (en) * 2006-05-24 2008-11-13 Van Bilsen Paul Methods and kits for linking polymorphic sequences to expanded repeat mutations
US9133517B2 (en) * 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
CA2625880C (en) 2005-10-14 2016-10-11 Cynthia M. Goh Composite nanoparticles, nanoparticles and methods for producing same
CN1962155A (en) * 2005-11-10 2007-05-16 鸿富锦精密工业(深圳)有限公司 CO2 laser welding apparatus
US7741396B2 (en) * 2005-11-23 2010-06-22 General Electric Company Composites having tunable dielectric constants, methods of manufacture thereof, and articles comprising the same
JP2009518641A (en) * 2005-12-08 2009-05-07 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ System and method for monitoring in-vivo drug release using overhauser improved NMR
US9108217B2 (en) 2006-01-31 2015-08-18 Nanocopoeia, Inc. Nanoparticle coating of surfaces
WO2007089881A2 (en) 2006-01-31 2007-08-09 Regents Of The University Of Minnesota Electrospray coating of objects
CA2641117C (en) 2006-01-31 2018-01-02 Nanocopoeia, Inc. Nanoparticle coating of surfaces
WO2007127799A2 (en) * 2006-04-25 2007-11-08 Research Foundation Of The City University Of New York Polymer submicron particle preparation by surfactant- mediated precipitation
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
WO2007149868A2 (en) * 2006-06-20 2007-12-27 The Regents Of The University Of California Controlled release encapsulated anti-bacterial and anti-inflammatory nanoparticles
US20080039415A1 (en) * 2006-08-11 2008-02-14 Gregory Robert Stewart Retrograde transport of sirna and therapeutic uses to treat neurologic disorders
US9375440B2 (en) * 2006-11-03 2016-06-28 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US8324367B2 (en) 2006-11-03 2012-12-04 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US7988668B2 (en) * 2006-11-21 2011-08-02 Medtronic, Inc. Microsyringe for pre-packaged delivery of pharmaceuticals
US7819842B2 (en) * 2006-11-21 2010-10-26 Medtronic, Inc. Chronically implantable guide tube for repeated intermittent delivery of materials or fluids to targeted tissue sites
US9040816B2 (en) 2006-12-08 2015-05-26 Nanocopoeia, Inc. Methods and apparatus for forming photovoltaic cells using electrospray
US20080171906A1 (en) * 2007-01-16 2008-07-17 Everaerts Frank J L Tissue performance via hydrolysis and cross-linking
EP1955695A1 (en) * 2007-02-06 2008-08-13 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nanocapsules of lipophilic complexes of nucleic acids
US20080193766A1 (en) * 2007-02-13 2008-08-14 Northern Nanotechnologies Control of Transport to and from Nanoparticle Surfaces
WO2008135828A2 (en) 2007-05-03 2008-11-13 Pfizer Products Inc. Nanoparticles comprising a drug, ethylcellulose, and a bile salt
US8703204B2 (en) 2007-05-03 2014-04-22 Bend Research, Inc. Nanoparticles comprising a cholesteryl ester transfer protein inhibitor and anon-ionizable polymer
EP2162120B1 (en) 2007-06-04 2016-05-04 Bend Research, Inc Nanoparticles comprising a non-ionizable cellulosic polymer and an amphiphilic non-ionizable block copolymer
US9545384B2 (en) 2007-06-04 2017-01-17 Bend Research, Inc. Nanoparticles comprising drug, a non-ionizable cellulosic polymer and tocopheryl polyethylene glocol succinate
US20090042825A1 (en) * 2007-08-06 2009-02-12 Majed Matar Composition, method of preparation & application of concentrated formulations of condensed nucleic acids with a cationic lipopolymer
US9144546B2 (en) 2007-08-06 2015-09-29 Clsn Laboratories, Inc. Nucleic acid-lipopolymer compositions
US7973019B1 (en) 2007-10-03 2011-07-05 Alcon Research, Ltd. Transferrin/transferrin receptor-mediated siRNA delivery
TW200930405A (en) 2007-11-15 2009-07-16 Alcon Res Ltd Low density lipoprotein receptor-mediated siRNA delivery
EP2231169B1 (en) 2007-12-06 2016-05-04 Bend Research, Inc. Pharmaceutical compositions comprising nanoparticles and a resuspending material
US9233078B2 (en) 2007-12-06 2016-01-12 Bend Research, Inc. Nanoparticles comprising a non-ionizable polymer and an Amine-functionalized methacrylate copolymer
EP2225604A1 (en) * 2007-12-28 2010-09-08 Dow Global Technologies Inc. Phase compensation film comprising polymer nanoparticles imbibed with liquid crystal material
JP5241853B2 (en) * 2007-12-28 2013-07-17 ダウ グローバル テクノロジーズ エルエルシー Micro functional materials
ATE537817T1 (en) * 2008-02-18 2012-01-15 Csir NANOPARTICLE CARRIER FOR DELIVERY OF MEDICINAL PRODUCTS AND METHOD FOR THE PRODUCTION THEREOF
BRPI0909780B1 (en) 2008-03-05 2022-05-03 Baxter International Inc Suspension containing a surface-modified particle, pharmaceutical composition comprising said suspension, method for enhancing cellular uptake of an active agent, use of surface-modified particles, and method of preparing a surface-modified particle
IE20080211A1 (en) 2008-03-20 2009-11-25 Nat Univ Ireland Biodegradable nanoshells for delivery of therapeutic and/or imaging molecules
WO2009126742A2 (en) * 2008-04-08 2009-10-15 Appian Labs, Llc Enzyme mediated delivery system
US20100010102A1 (en) * 2008-07-09 2010-01-14 Board Of Regents, The University Of Texas System Triggered release of drugs from polymer particles
US10952965B2 (en) * 2009-05-15 2021-03-23 Baxter International Inc. Compositions and methods for drug delivery
CN107033339A (en) 2009-05-27 2017-08-11 西莱克塔生物科技公司 Immunomodulator polymer compound
WO2011017382A2 (en) * 2009-08-03 2011-02-10 The Regents Of The University Of California Nanofibers and morphology shifting micelles
US8470304B2 (en) 2009-08-04 2013-06-25 Avidas Pharmaceuticals Llc Therapeutic vitamin D sun-protecting formulations and methods for their use
CN102574891A (en) 2009-08-26 2012-07-11 西莱克塔生物科技公司 Compositions that induce t cell help
US9650244B2 (en) 2010-08-25 2017-05-16 Genesegues, Inc. Topical vaccination via DNA microparticles
US20120076735A1 (en) * 2010-03-24 2012-03-29 Genesegues, Inc. Nanoparticles for Extravascular Administration
KR20130108988A (en) 2010-05-26 2013-10-07 셀렉타 바이오사이언시즈, 인크. Dose selection of adjuvanted synthetic nanocarriers
US8709487B1 (en) 2011-04-25 2014-04-29 The United States Of America As Represented By The Secretary Of The Army Nanoparticle entrapment of materials
JP6602536B2 (en) 2011-04-29 2019-11-06 セレクタ バイオサイエンシーズ インコーポレーテッド Controlled release of immunosuppressive agents from synthetic nanocarriers
WO2012161677A1 (en) 2011-05-20 2012-11-29 Alcon Research, Ltd. TRANSFERRIN/TRANSFERRIN RECEPTOR-MEDIATED siRNA DELIVERY
US20130028857A1 (en) 2011-07-29 2013-01-31 Selecta Biosciences, Inc. Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents
WO2013032829A1 (en) * 2011-08-26 2013-03-07 Arrowhead Research Corporation Poly(vinyl ester) polymers for in vivo nucleic acid delivery
US9474715B2 (en) * 2011-11-30 2016-10-25 Andreas Voigt Polymeric drug-delivery material, method for manufacturing thereof and method for delivery of a drug-delivery composition
KR20150127206A (en) 2013-03-14 2015-11-16 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Nanoparticle-based compositions
MX2015015221A (en) 2013-05-03 2016-07-06 Selecta Biosciences Inc Tolerogenic synthetic nanocarriers and therapeutic macromolecules for reduced or enhanced pharmacodynamic effects.
CN112933234A (en) 2013-06-04 2021-06-11 西莱克塔生物科技公司 Repeated administration of non-immunosuppressive antigen-specific immunotherapeutics
CN103550155A (en) * 2013-11-08 2014-02-05 江南大学 Method of restraining production of lysophosphatide in lecithin/non-ionic surfactant mixed micelles
AU2015279738A1 (en) 2014-06-25 2016-12-22 Selecta Biosciences, Inc. Methods and compositions for treatment with synthetic nanocarriers and immune checkpoint inhibitors
JP7272746B2 (en) 2014-09-07 2023-05-12 セレクタ バイオサイエンシーズ インコーポレーテッド Methods and compositions for attenuating exon-skipping antiviral transduction vector immune responses
LT3215133T (en) 2014-11-05 2021-02-25 Selecta Biosciences, Inc. Methods and compositions related to the use of low hlb surfactants in the production of synthetic nanocarriers comprising a rapalog
KR101663775B1 (en) * 2014-11-13 2016-10-07 주식회사 엘지생활건강 Composition for stabilizing polyphenolic effective ingredient
ES2908479T3 (en) 2015-08-26 2022-04-29 Achillion Pharmaceuticals Inc Compounds for the treatment of immune and inflammatory disorders
AR106018A1 (en) 2015-08-26 2017-12-06 Achillion Pharmaceuticals Inc ARYL, HETEROARYL AND HETEROCYCLIC COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
WO2017156513A1 (en) 2016-03-11 2017-09-14 Selecta Biosciences, Inc. Formulations and doses of pegylated uricase
KR20190036520A (en) 2016-06-27 2019-04-04 아칠리온 파르마세우티칼스 인코포레이티드 Quinazoline and indole compounds for the treatment of medical disorders
WO2018064215A1 (en) 2016-09-27 2018-04-05 Selecta Biosciences, Inc. Recombinant immunotoxins for use in the treatment of cancer
JP2020502062A (en) 2016-11-17 2020-01-23 ザ ユニバーシティ オブ ノース カロライナ アット チャペル ヒル Alkylpyrrolopyrimidine analogs and methods of making and using same
EP3565572A1 (en) 2017-01-07 2019-11-13 Selecta Biosciences, Inc. Patterned dosing of immunosuppressants coupled to synthetic nanocarriers
JP2020510687A (en) 2017-03-11 2020-04-09 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Methods and compositions relating to combination treatment with synthetic nanocarriers including anti-inflammatory and immunosuppressive agents
CA3078705A1 (en) 2017-10-13 2019-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
US20190151470A1 (en) * 2018-01-21 2019-05-23 RoverMed BioSciences, LLC Nanoparticles comprising protein-polynucleotide complexes and for delivering protein based complexes
CN108414393A (en) * 2018-06-13 2018-08-17 安徽中医药大学 A method of measuring entrapment efficiency of liposome
BR112021000675A2 (en) 2018-07-16 2021-04-13 Selecta Biosciences, Inc. METHODS AND COMPOSITIONS OF MMA CONSTRUCTIONS AND VECTORS
CN112771070A (en) 2018-07-16 2021-05-07 西莱克塔生物科技公司 Methods and compositions of OTC constructs and vectors
JP2022517466A (en) 2018-10-16 2022-03-09 ジョージア ステイト ユニバーシティー リサーチ ファウンデーション インコーポレイテッド Carbon monoxide prodrug for the treatment of medical disorders
BR112021021566A2 (en) 2019-04-28 2022-04-19 Selecta Biosciences Inc Methods for treating individuals with preexisting immunity to viral transfer vectors
BR112021023594A2 (en) 2019-05-28 2022-02-08 Selecta Biosciences Inc Methods and compositions for attenuated antiviral transfer vector immune response
CN114207440A (en) 2019-06-04 2022-03-18 西莱克塔生物科技公司 PEGylated uricase formulations and dosages
JP2022553345A (en) 2019-10-21 2022-12-22 セレクタ バイオサイエンシーズ インコーポレーテッド Methods and compositions for treating liver disease and disorders
IL292770A (en) 2019-11-08 2022-07-01 Selecta Biosciences Inc Formulations and doses of pegylated uricase
MX2022010568A (en) 2020-02-26 2022-11-30 Selecta Biosciences Inc Methods and compositions using synthetic nanocarriers comprising immunosuppressant.
WO2021211100A1 (en) 2020-04-14 2021-10-21 Selecta Biosciences, Inc. Methods and compositions for inducing autophagy
WO2021183781A1 (en) 2020-03-11 2021-09-16 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers
EP4274571A1 (en) 2021-01-05 2023-11-15 Selecta Biosciences, Inc. Viral vector dosing protocols
US20220323607A1 (en) 2021-04-09 2022-10-13 Selecta Biosciences, Inc. Synthetic nanocarriers comprising an immunosuppressant in combination with high affinity il-2 receptor agonists to enhance immune tolerance
WO2023064367A1 (en) 2021-10-12 2023-04-20 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
WO2023064350A1 (en) 2021-10-12 2023-04-20 Selecta Biosciences, Inc. Viral vector dosing protocols
WO2023086615A1 (en) 2021-11-14 2023-05-19 Selecta Biosciences, Inc. Multiple dosing with viral vectors
US20230263906A1 (en) 2022-01-10 2023-08-24 Selecta Biosciences, Inc. High affinity il-2 receptor agonists and synthetic nanocarrier dose sparing
WO2023172628A1 (en) 2022-03-09 2023-09-14 Selecta Biosciences, Inc. Immunosuppressant in combination with high affinity il-2 receptor agonists and related dosing
WO2023172624A1 (en) 2022-03-09 2023-09-14 Selecta Biosciences, Inc. Immunosuppressants in combination with anti-igm agents and related dosing
US20230372535A1 (en) 2022-03-25 2023-11-23 Selecta Biosciences, Inc. Synthetic nanocarriers comprising an immunosuppressant in combination with high affinity il-2 receptor agonists and anti-igm agents
US20230381277A1 (en) 2022-04-08 2023-11-30 Selecta Biosciences, Inc. High affinity il-2 receptor agonists and immunosuppressants to enhance immune tolerance

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5487390A (en) * 1990-10-05 1996-01-30 Massachusetts Institute Of Technology Gas-filled polymeric microbubbles for ultrasound imaging
US5492814A (en) * 1990-07-06 1996-02-20 The General Hospital Corporation Monocrystalline iron oxide particles for studying biological tissues
US5500224A (en) * 1993-01-18 1996-03-19 U C B S.A. Pharmaceutical compositions containing nanocapsules
US5650543A (en) * 1993-11-29 1997-07-22 Air Products And Chemicals, Inc. Ethoxylated acetylenic glycols having low dynamic surface tension
US5707606A (en) * 1993-01-25 1998-01-13 Sonus Pharmaceuticals, Inc. Phase shift colloids as ultrasound contrast agents
US6008192A (en) * 1997-03-12 1999-12-28 Abbott Laboratories Hydrophilic binary systems for the administration of lipophilic compounds
US6030956A (en) * 1996-10-24 2000-02-29 Boulikas; Teni Combination gene therapy for human cancers
US6139819A (en) * 1995-06-07 2000-10-31 Imarx Pharmaceutical Corp. Targeted contrast agents for diagnostic and therapeutic use
US6165988A (en) * 1995-02-10 2000-12-26 Christian Noe Medicament in particulate form
US6177103B1 (en) * 1998-06-19 2001-01-23 Rtp Pharma, Inc. Processes to generate submicron particles of water-insoluble compounds
US6197346B1 (en) * 1992-04-24 2001-03-06 Brown Universtiy Research Foundation Bioadhesive microspheres and their use as drug delivery and imaging systems
US6255457B1 (en) * 1995-09-08 2001-07-03 Beth Israel Deaconess Medical Center Tissue-specific monoclonal antibodies
US20010018054A1 (en) * 1997-09-18 2001-08-30 Idec Pharmaceuticals Corporation Synergistic composition and methods for treating neoplastic or cancerous growths and for restoring or boosting hematopoiesis
US6322805B1 (en) * 1995-09-21 2001-11-27 Samyang Corporation Biodegradable polymeric micelle-type drug composition and method for the preparation thereof
US6372714B1 (en) * 1997-04-07 2002-04-16 Daiichi Pharmaceutical Co., Ltd. Composition for gene introduction into cell
US6383500B1 (en) * 1996-06-27 2002-05-07 Washington University Particles comprising amphiphilic copolymers, having a crosslinked shell domain and an interior core domain, useful for pharmaceutical and other applications
US6489458B2 (en) * 1997-03-11 2002-12-03 Regents Of The University Of Minnesota DNA-based transposon system for the introduction of nucleic acid into DNA of a cell
US20030170893A1 (en) * 2000-02-28 2003-09-11 Genesegues, Inc. Nanocapsule encapsulation system and method
US20040038406A1 (en) * 2002-04-08 2004-02-26 Genesegues, Inc. Nanoparticle delivery systems and methods of use thereof
US20080113932A1 (en) * 2004-12-14 2008-05-15 Slaton Joel W Casein kinase 2 antisense therapy
US20090238883A1 (en) * 2006-04-28 2009-09-24 Kren Betsy T Liver-specific nanocapsules and methods of using

Family Cites Families (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
DK143689C (en) * 1975-03-20 1982-03-15 J Kreuter PROCEDURE FOR THE PREPARATION OF AN ADVERTISED VACCINE
US4177177A (en) 1976-03-26 1979-12-04 El Aasser Mohamed S Polymer emulsification process
FR2504408B1 (en) 1981-04-24 1986-02-14 Couvreur Patrick PROCESS FOR THE PREPARATION OF SUBMICROSCOPIC PARTICLES, PARTICLES THUS OBTAINED AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
WO1988001165A1 (en) 1986-08-11 1988-02-25 Innovata Biomed Limited Pharmaceutical formulations comprising microcapsules
US4920016A (en) * 1986-12-24 1990-04-24 Linear Technology, Inc. Liposomes with enhanced circulation time
FR2634397B2 (en) 1986-12-31 1991-04-19 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF DISPERSIBLE COLLOIDAL SYSTEMS OF A PROTEIN IN THE FORM OF NANOPARTICLES
EP0309527B1 (en) 1987-04-16 1993-01-20 Christian Bindschaedler Process for preparing a powder of water-insoluble polymer which can be redispersed in a liquid phase and process for preparing a dispersion of the powdered polymer
US4937119A (en) * 1988-12-15 1990-06-26 Hoechst Celanese Corp. Textured organic optical data storage media and methods of preparation
PH30995A (en) 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5145684A (en) * 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
GB9116610D0 (en) 1991-08-01 1991-09-18 Danbiosyst Uk Preparation of microparticles
US6113946A (en) 1992-04-03 2000-09-05 The Regents Of The University Of California Self-assembling polynucleotide delivery system comprising dendrimer polycations
DE4223169C1 (en) 1992-07-10 1993-11-25 Ferring Arzneimittel Gmbh Process for the microencapsulation of water-soluble active substances
CN1245156C (en) 1993-02-22 2006-03-15 美国生物科学有限公司 Methods for in vivo delivery of biologics and compositions useful therefor
US5439686A (en) 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
ATE258685T1 (en) 1993-03-09 2004-02-15 Baxter Int MACROMOLECULAR MICROPARTICLES AND METHOD FOR THE PRODUCTION THEREOF
FR2704754B1 (en) * 1993-05-07 1995-06-30 Oreal USE OF AN ALKYL ESTER OF GLUTATHION IN A COSMETIC OR DERMATOLOGICAL COMPOSITION FOR TOPICAL TREATMENT OF CUTANEOUS AGING.
WO1995003789A1 (en) 1993-07-28 1995-02-09 The Johns Hopkins University School Of Medicine Controlled release of pharmaceutically active substances from coacervate microcapsules
DE4341114A1 (en) * 1993-12-02 1995-06-08 Ifac Gmbh Emulsions comprising non-aqueous phase dispersed in oil phase
US5962427A (en) 1994-02-18 1999-10-05 The Regent Of The University Of Michigan In vivo gene transfer methods for wound healing
FR2721510B1 (en) 1994-06-22 1996-07-26 Rhone Poulenc Rorer Sa Nanoparticles filterable under sterile conditions.
US5858398A (en) * 1994-11-03 1999-01-12 Isomed Inc. Microparticular pharmaceutical compositions
US5629021A (en) 1995-01-31 1997-05-13 Novavax, Inc. Micellar nanoparticles
US5736156A (en) * 1995-03-22 1998-04-07 The Ohio State University Liposomal anf micellular stabilization of camptothecin drugs
US6051258A (en) 1995-06-07 2000-04-18 Emisphere Technologies, Inc. Proteinoid emulsions and methods for preparation and use thereof
US6033645A (en) * 1996-06-19 2000-03-07 Unger; Evan C. Methods for diagnostic imaging by regulating the administration rate of a contrast agent
PT752245E (en) 1995-07-05 2002-09-30 Europ Economic Community BIOCOMPATIBLE AND BIODEGRADABLE NANOPARTICLES FOR THE ABSORPTION AND ADMINISTRATION OF PROTEINACEAL DRUGS
US6143211A (en) 1995-07-21 2000-11-07 Brown University Foundation Process for preparing microparticles through phase inversion phenomena
CA2227287A1 (en) 1995-07-21 1997-02-06 Brown University Research Foundation A method for gene therapy using nucleic acid loaded polymeric microparticles
US5648097A (en) 1995-10-04 1997-07-15 Biotek, Inc. Calcium mineral-based microparticles and method for the production thereof
FR2742357B1 (en) 1995-12-19 1998-01-09 Rhone Poulenc Rorer Sa STABILIZED AND FILTRABLE NANOPARTICLES UNDER STERILE CONDITIONS
DE19605279A1 (en) 1996-02-13 1997-08-14 Hoechst Ag Target cell-specific vectors for the introduction of genes into cells, drugs containing such vectors and their use
KR100236771B1 (en) 1997-04-01 2000-02-01 성재갑 Hyaluronate microparticles for sustained release of drug
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US6143037A (en) 1996-06-12 2000-11-07 The Regents Of The University Of Michigan Compositions and methods for coating medical devices
FR2751222B1 (en) * 1996-07-16 1998-10-09 Capsulis COMPOSITIONS CONTAINING AT LEAST ONE NUCLEIC ACID AND THEIR APPLICATIONS IN THE BIOMEDICAL FIELD, PARTICULARLY IN GENE THERAPY
WO1998028423A2 (en) * 1996-12-20 1998-07-02 Board Of Regents, The University Of Texas System Compositions and methods of use for osteoclast inhibitory factors
US5874111A (en) 1997-01-07 1999-02-23 Maitra; Amarnath Process for the preparation of highly monodispersed polymeric hydrophilic nanoparticles
EP0860167A1 (en) 1997-01-30 1998-08-26 Robert Gurny RNA-and DNA- based active agents in nanoparticles
DE19723308A1 (en) * 1997-06-04 1998-12-10 Wolfgang A Prof Dr Wohlrab Microemulsions useful for topical administration of e.g. local anaesthetics
KR100923172B1 (en) 1997-06-27 2009-10-22 아브락시스 바이오사이언스, 엘엘씨 Novel Formulations of Pharmacological Agents
US6165440A (en) 1997-07-09 2000-12-26 Board Of Regents, The University Of Texas System Radiation and nanoparticles for enhancement of drug delivery in solid tumors
AU5860798A (en) 1997-12-29 1999-07-19 Universite De Geneve Method for producing an aqueous colloidal dispersion of nanoparticles
IT1298575B1 (en) * 1998-02-06 2000-01-12 Vectorpharma Int PHARMACEUTICAL COMPOSITIONS IN THE FORM OF NANOPARTICLES INCLUDING LIPID SUBSTANCES AND ANTIPHILIC SUBSTANCES AND RELATED PROCESS OF
IL143196A0 (en) * 1998-11-20 2002-04-21 Rtp Pharma Inc Method of preparing stable suspensions of insoluble microparticles
JP2002536386A (en) 1999-02-12 2002-10-29 コラジェネシス インク. Collagen-based injection system for delivery of cells or therapeutic agents

Patent Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5492814A (en) * 1990-07-06 1996-02-20 The General Hospital Corporation Monocrystalline iron oxide particles for studying biological tissues
US5487390A (en) * 1990-10-05 1996-01-30 Massachusetts Institute Of Technology Gas-filled polymeric microbubbles for ultrasound imaging
US6197346B1 (en) * 1992-04-24 2001-03-06 Brown Universtiy Research Foundation Bioadhesive microspheres and their use as drug delivery and imaging systems
US5500224A (en) * 1993-01-18 1996-03-19 U C B S.A. Pharmaceutical compositions containing nanocapsules
US5707606A (en) * 1993-01-25 1998-01-13 Sonus Pharmaceuticals, Inc. Phase shift colloids as ultrasound contrast agents
US5650543A (en) * 1993-11-29 1997-07-22 Air Products And Chemicals, Inc. Ethoxylated acetylenic glycols having low dynamic surface tension
US6165988A (en) * 1995-02-10 2000-12-26 Christian Noe Medicament in particulate form
US6139819A (en) * 1995-06-07 2000-10-31 Imarx Pharmaceutical Corp. Targeted contrast agents for diagnostic and therapeutic use
US6255457B1 (en) * 1995-09-08 2001-07-03 Beth Israel Deaconess Medical Center Tissue-specific monoclonal antibodies
US6322805B1 (en) * 1995-09-21 2001-11-27 Samyang Corporation Biodegradable polymeric micelle-type drug composition and method for the preparation thereof
US6383500B1 (en) * 1996-06-27 2002-05-07 Washington University Particles comprising amphiphilic copolymers, having a crosslinked shell domain and an interior core domain, useful for pharmaceutical and other applications
US6030956A (en) * 1996-10-24 2000-02-29 Boulikas; Teni Combination gene therapy for human cancers
US6489458B2 (en) * 1997-03-11 2002-12-03 Regents Of The University Of Minnesota DNA-based transposon system for the introduction of nucleic acid into DNA of a cell
US6008192A (en) * 1997-03-12 1999-12-28 Abbott Laboratories Hydrophilic binary systems for the administration of lipophilic compounds
US6372714B1 (en) * 1997-04-07 2002-04-16 Daiichi Pharmaceutical Co., Ltd. Composition for gene introduction into cell
US20010018054A1 (en) * 1997-09-18 2001-08-30 Idec Pharmaceuticals Corporation Synergistic composition and methods for treating neoplastic or cancerous growths and for restoring or boosting hematopoiesis
US20010019715A1 (en) * 1997-09-18 2001-09-06 Idec Pharmaceuticals Corporation Synergistic composition and methods for treating neoplastic or cancerous growths and for restoring or boosting hematopoiesis
US6177103B1 (en) * 1998-06-19 2001-01-23 Rtp Pharma, Inc. Processes to generate submicron particles of water-insoluble compounds
US20030170893A1 (en) * 2000-02-28 2003-09-11 Genesegues, Inc. Nanocapsule encapsulation system and method
US6632671B2 (en) * 2000-02-28 2003-10-14 Genesegues, Inc. Nanoparticle encapsulation system and method
US20040038406A1 (en) * 2002-04-08 2004-02-26 Genesegues, Inc. Nanoparticle delivery systems and methods of use thereof
US20040038303A1 (en) * 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
US20080220072A1 (en) * 2002-04-08 2008-09-11 Unger Gretchen M Biologic modulations with nanoparticles
US20080113932A1 (en) * 2004-12-14 2008-05-15 Slaton Joel W Casein kinase 2 antisense therapy
US20090238883A1 (en) * 2006-04-28 2009-09-24 Kren Betsy T Liver-specific nanocapsules and methods of using

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Debeaufort et al., Int. J. Food Sci. and Technol., 1995, 30: 183-190. *
Wary et al., Cell, 1996, 87: 733-743. *

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030206958A1 (en) * 2000-12-22 2003-11-06 Cattaneo Maurizio V. Chitosan biopolymer for the topical delivery of active agents
US20070098713A1 (en) * 2002-04-08 2007-05-03 Genesegues, Inc. Nanoparticle delivery systems and methods of use thereof
US20100303929A1 (en) * 2003-10-21 2010-12-02 Mctavish Hugh Compounds and methods for treating cancer
US8501906B2 (en) 2003-10-21 2013-08-06 Igf Oncology, Llc Compounds and methods for treating cancer
US7811982B2 (en) 2003-10-21 2010-10-12 Igf Oncology, Llc Compounds and methods for treating cancer
US20060258569A1 (en) * 2003-10-21 2006-11-16 Mctavish Hugh Compounds and methods for treating cancer
US20100216696A1 (en) * 2003-10-21 2010-08-26 Mctavish Hugh Compounds and methods for treating cancer
US9011880B2 (en) 2003-10-21 2015-04-21 Igf Oncology, Llc Compounds and methods for treating cancer
US20060258589A1 (en) * 2004-01-24 2006-11-16 Igf Oncology, Llc Methods for enhancing radiation therapy
US20050281886A1 (en) * 2004-05-06 2005-12-22 Ivrea Pharmaceuticals, Inc. Particles for the delivery of active agents
US8017102B2 (en) 2004-10-21 2011-09-13 Igf Oncology, Llc Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US8920777B2 (en) 2004-10-21 2014-12-30 Igf Oncology, Llc Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US20070224119A1 (en) * 2004-10-21 2007-09-27 Igf Oncology Toxins and radionuclides coupled to IGF-1 receptor ligands for treatment of cancer
US20080085301A1 (en) * 2005-03-31 2008-04-10 Fairfield Clinical Trials, Llc Method for transdermal administration of antimicrobial medications
US20060222692A1 (en) * 2005-03-31 2006-10-05 Fairfield Clinical Trials Llc Method and compositions for transdermal administration of antimicrobial medications
US20090297596A1 (en) * 2005-05-23 2009-12-03 Elan Pharma International Limited Nanoparticulate and Controlled Release Compositions Comprising a Platelet Aggregation Inhibitor
US20080045583A1 (en) * 2006-08-18 2008-02-21 David Delmarre Stable levetiracetam compositions and methods
US20100119501A1 (en) * 2007-04-30 2010-05-13 Yi-Yan Yang Method of delivering a protein into a cell
US8425900B2 (en) * 2007-04-30 2013-04-23 Agency For Science, Technology And Research Method of delivering a protein into a cell
US20100173001A1 (en) * 2007-06-14 2010-07-08 Genesegues, Inc. Metal Ion-Treated Biocompatible Polymers Useful for Nanoparticles
US20090143339A1 (en) * 2007-11-30 2009-06-04 Martin Purpura Methods using phosphatidylserine, lysophosphatidylserine, and/or salts thereof to increase testosterone levels
US8124594B2 (en) 2007-11-30 2012-02-28 Chemi Nutra, Llc Methods using phosphatidylserine, lysophosphatidylserine, and/or salts thereof to increase testosterone levels
US20110052703A1 (en) * 2008-02-11 2011-03-03 Yechezkel Barenholz Beta-casein assemblies for mucosal delivery of therapeutic bioactive agents
US8865223B2 (en) 2008-02-11 2014-10-21 Technion Research And Development Foundation Ltd. Beta-casein assemblies for mucosal delivery of therapeutic bioactive agents
US8865222B2 (en) 2008-02-11 2014-10-21 Technion Research And Development Foundation Ltd. Beta-casein assemblies for enrichment of food and beverages and methods of preparation thereof
US8871276B2 (en) 2008-02-11 2014-10-28 Technion Research And Development Foundation Ltd. Beta-casein assemblies for mucosal delivery of therapeutic bioactive agents
US20110045092A1 (en) * 2008-02-11 2011-02-24 Livney Yoav D Casein particles encapsulating therapeutically active agents and uses thereof
US20110038987A1 (en) * 2008-02-11 2011-02-17 Dganit Danino Beta-casein assemblies for enrichment of food and beverages and methods of preparation thereof
US9757464B2 (en) 2009-03-05 2017-09-12 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
US8685458B2 (en) 2009-03-05 2014-04-01 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives
US8815294B2 (en) 2010-09-03 2014-08-26 Bend Research, Inc. Pharmaceutical compositions of dextran polymer derivatives and a carrier material
US9084727B2 (en) 2011-05-10 2015-07-21 Bend Research, Inc. Methods and compositions for maintaining active agents in intra-articular spaces
US9675671B2 (en) 2014-01-12 2017-06-13 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof
US9801923B2 (en) 2014-01-12 2017-10-31 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof
US10391147B2 (en) 2014-01-12 2019-08-27 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof
US10967049B2 (en) 2014-01-12 2021-04-06 Igf Oncology, Llc Fusion proteins containing insulin-like growth factor-1 and epidermal growth factor and variants thereof and uses thereof
US10987308B2 (en) 2014-09-03 2021-04-27 Genesegues, Inc. Therapeutic nanoparticles and related compositions, methods and systems
US11324834B2 (en) 2017-05-21 2022-05-10 Igf Oncology, Llc Insulin-like growth factor-chemotherapeputic conjugate for treating myelodysplastic syndrome
WO2022261148A1 (en) * 2021-06-09 2022-12-15 Scribe Therapeutics Inc. Compositions of glycoprotein particles

Also Published As

Publication number Publication date
CA2400172C (en) 2010-04-20
AU2001247244B2 (en) 2005-06-02
US6632671B2 (en) 2003-10-14
EP1267946A2 (en) 2003-01-02
HK1048772A1 (en) 2003-04-17
JP2003524654A (en) 2003-08-19
EP1267946A4 (en) 2008-07-02
JP4751556B2 (en) 2011-08-17
CN1406140A (en) 2003-03-26
CA2400172A1 (en) 2001-09-07
WO2001064164A2 (en) 2001-09-07
WO2001064164A3 (en) 2001-12-20
AU4724401A (en) 2001-09-12
MXPA02008361A (en) 2004-05-17
US20030170893A1 (en) 2003-09-11

Similar Documents

Publication Publication Date Title
US6632671B2 (en) Nanoparticle encapsulation system and method
KR102606929B1 (en) An engineered two-part cellular device for discovery and characterization of T-cell receptor interactions with cognate antigens
Chong et al. A paradigm for peptide vaccine delivery using viral epitopes encapsulated in degradable polymer hydrogel capsules
Moon et al. Interbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responses
KR840002117B1 (en) Process for preparing magnetically-localizable,biodegrodable hipid miscospheres
Rajapaksa et al. Claudin 4-targeted protein incorporated into PLGA nanoparticles can mediate M cell targeted delivery
US5952232A (en) Expandible microparticle intracellular delivery system
US20080199531A1 (en) Methods And Products For Delivering Biological Molecules To Cells Using Multicomponent Nanostructures
US20030143743A1 (en) Transfection of eukaryontic cells with linear polynucleotides by electroporation
AU6707194A (en) Encapsulation of nucleic acids with conjugates that facilitate and target cellular uptake and gene expression
CA2451389C (en) Improved transfection of eukaryotic cells with linear polynucleotides by electroporation
Kreuter Possibilities of using nanoparticles as carriers for drugs and vaccines
WO2004026453A2 (en) Microcapsules and methods of use
KR20200015900A (en) Self-inactivating virus vector
JP2023051923A (en) Tumor-targeting bead vectors and use methods thereof
KR20200085812A (en) Compositions and methods for inhibiting viral vector-induced inflammatory response
DE69822542T2 (en) Encapsulation of a subunit of the bovine herpes virus type 1 in aqueous solvents as a vaccine.
Yoshimura et al. Two-dimensional crystallization of P22 virus-like particles
CN103149111A (en) Method for detecting odor substance butanedione based on olfactory receptor sensor
Kawano et al. Technologies that generate and modify virus-like particles for medical diagnostic and therapy purposes
CN111909918B (en) Endolysin mutant and coding gene and application thereof
KR101093806B1 (en) Nano scaffold for gene delivery and a method for preparing the same
CN116113427A (en) SARS-COV-2 receptor binding domain in natural outer membrane vesicles
CN102839188A (en) Homonymous tandem co-expression bi-color fluorescent protein report gene vector
CN107217074B (en) Method for realizing site-specific integration of multiple large-fragment DNAs in mammalian cells by using ZFN (zero-forcing transcription)

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENESEGUES, INC., MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UNGER, GRETCHEN M;REEL/FRAME:018642/0242

Effective date: 20061214

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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