US20160015824A1 - Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same - Google Patents

Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same Download PDF

Info

Publication number
US20160015824A1
US20160015824A1 US14/403,315 US201314403315A US2016015824A1 US 20160015824 A1 US20160015824 A1 US 20160015824A1 US 201314403315 A US201314403315 A US 201314403315A US 2016015824 A1 US2016015824 A1 US 2016015824A1
Authority
US
United States
Prior art keywords
lipid nanoparticle
lipid
hsa
nanoparticle
agents
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
US14/403,315
Other languages
English (en)
Inventor
Robert J. Lee
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.)
Ohio State Innovation Foundation
Original Assignee
Ohio State Innovation Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ohio State Innovation Foundation filed Critical Ohio State Innovation Foundation
Priority to US14/403,315 priority Critical patent/US20160015824A1/en
Assigned to OHIO STATE INNOVATION FOUNDATION reassignment OHIO STATE INNOVATION FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, ROBERT J.
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: OHIO STATE UNIVERSITY
Publication of US20160015824A1 publication Critical patent/US20160015824A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH reassignment NATIONAL INSTITUTES OF HEALTH CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE OHIO UNIVERSITY
Assigned to NATIONAL INSTITUTES OF HEALTH reassignment NATIONAL INSTITUTES OF HEALTH CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: THE OHIO STATE UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48284
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • A61K47/48815
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • A61K47/544Phospholipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6907Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a microemulsion, nanoemulsion or micelle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21064Peptidase K (3.4.21.64)

Definitions

  • LNs lipid nanoparticles
  • NAs nucleic acids
  • a liposome is a vesicle composed of one or more lipid bilayers, capable of carrying hydrophilic molecules within an aqueous core or hydrophobic molecules within its lipid bilayer(s).
  • LNs Lipid nanoparticles
  • Drug delivery by LNs via systemic route requires overcoming several physiological barriers.
  • the reticuloendothelial system (RES) is responsible for clearance of LNs from the circulation. Once escaping the vasculature and reaching the target cell, LNs are typically taken up by endocytosis and must release the drug into the cytoplasm prior to degradation within acidic endosome conditions.
  • NAs nucleic acids
  • siRNA siRNA
  • other therapeutic oligonucleotides are a major technical challenge that has limited their potential for clinical translation.
  • oligonucleotide ON
  • LN formulation should be able to (1) protect the drug from enzymatic degradation; (2) traverse the capillary endothelium; (3) specifically reach the target cell type without causing excessive immunoactivation or off-target cytotoxicity; (4) promote endocytosis and endosomal release; and (5) form a stable formulation with high colloidal stability and long shelf-life.
  • LNs that encapsulate therapeutic oligonucleotides with high efficiency and fulfill physical and biological criteria for efficacious delivery.
  • the LNs comprise hyper-cationized and/or pH-responsive HSA-polymer conjugates.
  • the HSA-polymer conjugate comprises HSA-PEI or HSA-PEHA.
  • APC hyper-cationized albumin-polymer conjugates
  • LNs particles are also described herein as lipid-coated albumin nanoparticles (LCANs)
  • lipid nanoparticle comprising at least one lipid and albumin conjugated to a positively charged polymer.
  • the LN comprises a hyper-cationized albumin-polycation conjugate (APC).
  • the polycation comprises a polyamine selected from the group consisting of spermine, dispermine, trispermine, tetraspermine, oligospermine, thermine, spermidine, dispermidine, trispermidine, oligospermidine, putrescine, polylysine, polyarginine, a polyethylenimine of branched or linear type, and polyallylamine.
  • the positively-charged polymer consists essentially of a polyethylenimine.
  • the polyethylenimine has a molecular weight not greater than 50 kDa, or from about 200 Da to about 2000 Da.
  • the positively-charged polymer comprises pentaethylenehexamine (PEHA) or tetraethylenepentamine (TEPA).
  • the LN comprises a polyethylenimine conjugated to human serum albumin.
  • the conjugation is via one or more cross linking agents.
  • the LN comprises PEHA conjugated to HSA.
  • multiple PEHA molecules are linked to each HSA molecule.
  • between about two (2) and about twenty (2) PEHA molecules can be linked to each HSA molecule.
  • eleven (11) PEHA molecules are linked to each HSA molecule.
  • the LN comprises a mixture of two or more low molecular weight polymers.
  • the at least one lipid comprises a cationic lipid, a neutral lipid, and a PEGylated lipid, with or without cholesterol.
  • At least one lipid comprises 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), L- ⁇ -phosphatidylcholine (SPC), and d-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS).
  • DOTAP 1,2-dioleoyl-3-trimethylammonium-propane
  • SPC L- ⁇ -phosphatidylcholine
  • TPGS d-alpha-tocopheryl polyethylene glycol 1000 succinate
  • the lipids are in a 25:70:5 molar ratio of DOTAP:SPC:TPGS.
  • the LN encapsulates molecules selected from nucleic acids, chemo therapeutic agents, or combinations thereof.
  • the encapsulated molecules comprise a nucleic acid selected from plasmid DNAs, antisense oligonucleotides, miRs, anti-miRs, shRNAs, siRNAs, or combinations thereof.
  • the encapsulation rate of therapeutic agents or nucleotides is 40% or higher.
  • the LN has a diameter under 300 nm, or under 200 nm, or about 98 nm.
  • the polymer is bound only to an external surface of the nanoparticle via direct connection or via a crosslinker.
  • the LN further comprises a polyethylene glycol-conjugated lipid.
  • the polyethylene glycol-conjugated lipid is selected from the group consisting of polysorbate 80, TPGS, and mPEG-DSPE.
  • the polyethylene glycol-conjugated lipid is present at a concentration less than about 15.0 molar percent.
  • the LN further comprises a ligand capable of binding to a target cell or a target molecule.
  • the ligand is an antibody or an antibody fragment.
  • the ligand is selected from cRGD, galatose-containing moieties, transferrin, folate, low density lipoprotein, or epidermal growth factors.
  • a pharmaceutical composition comprising a lipid nanoparticle having at least one lipid and albumin conjugated to a positively charged polymer, and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition is administered perorally, intravenously, intraperitoneally, subcutaneously, or transdermally.
  • the pharmaceutical composition is prepared as an orally administered tablet, a sterile solution, a sterile suspension, a lyophilized powder, or a suppository.
  • a method of making a lipid-coated albumin nanoparticle involves synthesizing a human serum albumin-pentaethylenehexamine (HSA-PEHA) conjugate; adding at least one lipid to the HSA-PEHA conjugate; adding a nucleic acid to the mixture of lipids and the HSA-PEHA conjugate to obtain an LCAN precursor; and subjecting the LCAN precursor to a dialysis or diafiltration step to make a lipid-coated albumin nanoparticle.
  • HSA-PEHA human serum albumin-pentaethylenehexamine
  • the at least one lipid comprises DOTAP, SPC, and TPGS at a 25:70:5 ratio.
  • the nucleic acid is selected from pDNAs, antisense oligonucleotides, miRs, anti-miRs, shRNAs, siRNAs, or combinations thereof. Further provided herein is the product made from the described method.
  • a method of diagnosing or treating a cancer or infectious disease involves administering an effective amount of a pharmaceutical composition comprising at least one lipid, albumin conjugated to a positively-charged polymer, and a pharmaceutically acceptable excipient, to a patient in need thereof.
  • a delivery system comprising at least one lipid and a macromolecule conjugated to a polymer, wherein the macromolecule forms an electrostatic complex with a nucleic acid.
  • a method of using a lipid nanoparticle involves encapsulating a nucleic acid in a lipid nanoparticle, wherein the lipid nanoparticle comprises albumin conjugated to a polymer, incorporating the lipid nanoparticle into a pharmaceutical composition, and administering the pharmaceutical composition to a patient in need thereof.
  • FIG. 1 Gel mobility shift analysis of HSA-PEI(600) (APC)-oligodeoxynucleotide (ODN) complexes at varying ODN-to-HSA-PEI(600) w/w ratios.
  • APC HSA-PEI(600)
  • ODN oligodeoxynucleotide
  • FIG. 2 Zeta potential of LN (LN)-HSA-PEI(600)-LOR-2501 (APC-ODN) complexes.
  • FIGS. 3A-B Downregulation of RNR R1 mRNA expression by LOR-2501 in LCANs.
  • the LCANs were prepared at varying APC concentrations under different media conditions: FIG. 3A displays serum-free media; FIG. 3B displays media containing 10% FBS.
  • RNR R1 mRNA expression relative to actin was determined by RT-PCR where untreated KB cells served as a baseline for mRNA expression.
  • FIG. 4 Cell viability study of KB cells treated with LCAN-HSA-PEI(600)-LOR-2501 (APC) complex. Transfection was performed in serum-free media. Cell viabilities are expressed as a percentage relative to the mean viability of the untreated KB cells.
  • FIG. 5 Gel mobility shift analysis of HSA-PEHA-LOR-2501 (APC-ODN) complexes at varying ODN-to-APC w/w ratios. LOR-2501 was used as the ODN in this study.
  • FIG. 6 Downregulation of RNR R1 mRNA expression by LOR-2501 in LCANs.
  • the LCANs were prepared at varying APC concentrations under serum-free conditions.
  • RNR R1 mRNA expression relative to actin was determined by RT-PCR where untreated KB cells served as a baseline for mRNA expression.
  • FIG. 7 Bcl-2 down regulation in KB cells by lipid-coated albumin nanoparticle (LCAN)-G3139 as compared to LN-G3139.
  • LCAN lipid-coated albumin nanoparticle
  • FIG. 8 An example scheme for synthesizing an APC.
  • FIG. 9 The mechanism of action for hyper-cationized pH-responsive APCs.
  • FIG. 10 Upregulation of p27/kip1 mRNA by LCAN loaded with anti-miR-221 in CAL-51 breast cancer cells.
  • FIG. 11 Upregulation of estrogen mRNA by LCAN loaded with anti-miR-221 in CAL-51 cells.
  • the estrogen receptor is a target of miR-221.
  • NA-based therapies are being developed to promote or inhibit gene expression. As mutations in genes and changes in miRNA profile are believed to be the underlying cause of cancer and other diseases, NA-based agents can directly act upon the underlying etiology, maximizing therapeutic potential.
  • Non-limiting examples of NA-based therapies include: plasmid DNA (pDNA), small interfering RNA (siRNA), small hairpin RNA (shRNA), microRNA (miR), mimic (mimetic), anti-miR/antagomiR/miR inhibitor, and antisense oligonucleotide (ASO).
  • NA-based therapies faced several obstacles in their implementation since transporting NAs to their intracellular target was particularly challenging and since NAs are relatively unstable and subject to degradation by serum and cellular nucleases. Further, the high negative charges of NAs made it impossible for transport across the cell membrane, further limiting utility.
  • LNs described herein provide a useful platform for the delivery of both traditional therapeutic compounds and NA-based therapies.
  • Drugs formulated using LNs provide desirable pharmacokinetic (PK) properties in vivo, such as increased blood circulation time and increased accumulation at the site of solid tumors due to enhanced permeability and retention (EPR) effect.
  • the LNs may be surface-coated with polyethylene glycol to reduce opsonization of LNs by serum proteins and the resulting RES-mediated uptake, and/or coated with cell-specific ligands to provide targeted drug delivery.
  • LNs with a highly positive charge tend to interact non-specifically with non-target cells, tissues, and circulating plasma proteins, and may cause cytotoxicity.
  • LNs with a highly negative charge cannot effectively incorporate NAs, which are themselves negatively charged, and may trigger rapid RES-mediated clearance, reducing therapeutic efficacy.
  • LNs with a neutral to moderate charge are best suited for in vivo drug and gene delivery.
  • the LNs described herein comprise hyper-cationized albumin-polymer conjugates (APCs).
  • APCs hyper-cationized albumin-polymer conjugates
  • the term “hyper-cationized” mean each polycation carries multiple positive charges. In particular embodiments, up to 20 polycations can be linked to each albumin molecule. These factors result in a much higher overall charge content for APCs compared to traditional cationized albumin, which typically comprises an albumin conjugate where carboxyl groups are replaced with single-positive-charge amine functional groups. Because of their high charge density, the APCs are able to very efficiently interact with polyanions such as an oligonucleotide particle, molecule, compound or formulation having multiple cations or positive charges.
  • lipid nanoparticle refers to a vesicle formed by one or more lipid components.
  • the lipid components described herein may include cationic lipids.
  • Cationic lipids are lipids that carry a net positive charge at any physiological pH. In certain embodiments, the positive charge is used for association with negatively charged therapeutics such as ASOs via electrostatic interaction.
  • Suitable cationic lipids include, but are not limited to: 3 ⁇ -[N—(N′,N′-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride (DC-Chol); 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP); 1,2-dioleoyl-3-dimethylammonium-propane (DODAP); dimethyldioctadecylammonium bromide salt (DDAB); 1,2-dilauroyl-sn-glycero-3-ethylphosphocholine chloride (DL-EPC); N-[1-(2,3-dioleyloxy)propyl]-N—N—N-trimethyl ammonium chloride (DOTMA); N-[1-(2,3-dioleyloxy)propyl]-N—N—N-dimethyl ammonium chloride (DODMA); N,N-dioctade
  • cationic lipids in available preparations could be used, such as LIPOFECTIN® (from GIBCO/BRL), LIPOFECTAMINE® (from GIBCO/MRL), siPORT NEOFX® (from Applied Biosystems), T RANSFECTAM® (from Promega), and TRANSFECTIN® (from Bio-Rad Laboratories, Inc.).
  • LIPOFECTIN® from GIBCO/BRL
  • LIPOFECTAMINE® from GIBCO/MRL
  • siPORT NEOFX® from Applied Biosystems
  • T RANSFECTAM® from Promega
  • TRANSFECTIN® from Bio-Rad Laboratories, Inc.
  • the cationic lipids may be present at concentrations up to about 80.0 molar percent of total lipids in the formulation, or from about 5.0 to about 50.0 molar percent of the formulation.
  • the LN formulations presently disclosed may also include anionic lipids.
  • Anionic lipids are lipids that carry a net negative charge at physiological pH. These anionic lipids, when combined with cationic lipids, are useful to reduce the overall surface charge of LNs and introduce pH-dependent disruption of the LN bilayer structure, facilitating nucleotide release by inducing nonlamellar phases at acidic pH or induce fusion with the cellular membrane.
  • anionic lipids include, but are not limited to: fatty acids such as oleic, linoleic, and linolenic acids; cholesteryl hemisuccinate; 1,2-di-O-tetradecyl-sn-glycero-3-phospho-(1′-rac-glycerol) (diether PG); 1,2-dimyristoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (sodium salt); 1,2-dimyristoyl-sn-glycero-3-phospho-L-serine (sodium salt); 1-hexadecanoyl, 2-(9Z,12Z)-octadecadienoyl-sn-glycero-3-phosphate; 1,2-dioleoyl-sn-glycero-3-[phospho-rac-(1-glycerol)] (DOPG); dioleoylphosphatidic acid (DOPG);
  • charged LNs are advantageous for transfection, but off-target effects such as cytotoxicity and RES-mediated uptake may occur.
  • Hydrophilic molecules such as polyethylene glycol (PEG) may be conjugated to a lipid anchor and included in the LNs described herein to prevent LN aggregation or interaction with membranes.
  • Hydrophilic polymers may be covalently bonded to lipid components or conjugated using crosslinking agents to functional groups such as amines.
  • Suitable conjugates of hydrophilic polymers include, but are not limited to: polyvinyl alcohol (PVA); polysorbate 80; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-PEG2000 (DSPE-PEG2000); D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS); dimyristoylphosphatidylethanolamine-PEG2000 (DMPE-PEG2000); and dipalmitoylphosphatidylethanolamine-PEG2000 (DPPE-PEG2000).
  • the hydrophilic polymer may be present at concentrations ranging from about 0 to about 15.0 molar percent of the formulation, or from about 5.0 to about 10.0 molar percent of the formulation.
  • the molecular weight of the PEG used is between about 100 and about 10,000 Da, or from about 100 to about 2,000 Da.
  • the LNs described herein may further comprise neutral and/or cholesterol lipids as helper lipids. These lipids are useful to stabilize the formulation, reduce elimination in vivo, or increase transfection efficiency.
  • the LNs may be formulated in a solution of saccharides such as, but not limited to, glucose, sorbitol, sucrose, maltose, trehalose, lactose, cellubiose, raffinose, maltotriose, dextran, or combinations thereof, to promote lyostability and/or cryostability.
  • Neutral lipids have zero net charge at physiological pH.
  • One or a combination of several neutral lipids may be included in any LN formulation disclosed herein.
  • Suitable neutral lipids include, but are not limited to: phosphatidylcholine (PC, e.g., DSPC, DPPC, DOPC, DMPC, soyPC, eggPC, HSPC), phosphatidylethanolamine (PE, e.g., DOPE, DSPE, DPPE, DSPE, DMPE), ceramide, cerebrosides, sphingomyelin, cephalin, cholesterol, diacylglycerol, glycosylated diacylglycerols, prenols, lysosomal PLA2 substrates, N-acylglycines, and combinations thereof.
  • PC phosphatidylcholine
  • PE e.g., DOPE, DSPE, DPPE, DSPE, DMPE
  • ceramide cerebrosides
  • sphingomyelin cephalin
  • cholesterol diacylglycerol
  • glycosylated diacylglycerols
  • lipids include, but are not limited to: phosphatidic acid, (PG, e.g., DSPG, DMPG, DPPG), and lysophosphatidylethanolamine; sterols such as cholesterol, demosterol, sitosterol, zymosterol, diosgenin, lanostenol, stigmasterol, lathosterol, and dehydroepiandrosterone; and sphingolipids such as sphingosines, ceramides, sphingomyelin, gangliosides, glycosphingolipids, phosphosphingolipids, phytoshingosine; and combinations thereof.
  • PG phosphatidic acid
  • DMPG DMPG
  • DPPG DPPG
  • sterols such as cholesterol, demosterol, sitosterol, zymosterol, diosgenin, lanostenol, stigmasterol, lathosterol, and dehydroepiandrosterone
  • the LN formulations described herein may further comprise fusogenic lipids or fusogenic coatings to promote membrane fusion.
  • fusogenic lipids include, but are not limited to, glyceryl mono-oleate, oleic acid, palmitoleic acid, phosphatidic acid, phosphoinositol 4,5-bisphosphate (PIP 2 ), and combinations thereof.
  • the LN formulations described here may further comprise cationic polymers or conjugates of cationic polymers.
  • Cationic polymers or conjugates thereof may be used alone or in combination with lipid nanocarriers.
  • Suitable cationic polymers include, but are not limited to: polyethylenimine (PEI); pentaethylenehexamine (PEHA); spermine; spermidine; poly(L-lysine); poly(amido amine) (PAMAM) dendrimers; polypropyleneiminie dendrimers; poly(2-dimethylamino ethyl)-methacrylate (pDMAEMA); chitosan; tris(2-aminoethyl)amine and its methylated erivatives; and combinations thereof.
  • PEI polyethylenimine
  • PEHA pentaethylenehexamine
  • spermine spermine
  • spermidine poly(L-lysine)
  • PAMAM poly(amido amine) dend
  • the chain length and branching are important considerations for the implementation of polymeric delivery systems.
  • High molecular weight polymers such as PEI (MW 25,000) are useful as transfection agents, but suffer from cytotoxicity.
  • Low molecular weight PEI (MW 600) does not cause cytotoxicity, but is limited due to its inability to facilitate stable condensation with NAs.
  • conjugation of low molecular weight polymers to a larger molecule such as albumin is a thus a useful method of increasing activity of electrostatic complexation with NA condensation while lowing cytotoxicity of LN formulations.
  • Anionic polymers may be incorporated into the LN formulations presently disclosed as well.
  • Suitable anionic polymers include, but are not limited to: poly(propylacrylic acid) (PPAA); poly(glutamic acid) (PGA); alginates; dextran derivatives; xanthans; derivatized polymers; and combinations thereof.
  • the LN formulation includes conjugates of polymers.
  • the conjugates may be crosslinked to targeting agents, lipophilic moieties, proteins, or other molecules that increase the overall therapeutic efficacy.
  • Suitable crosslinking agents include, but are not limited to: N-succinimidyl 3-[2-pyridyldithio]-propionate (SPDP); dimethyl 3,3′-dithiobispropionimidate (DTBP); dicyclohexylcarbodiimide (DCC); diisopropyl carbodiimide (DIC); 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC); N-hydroxysulfosuccinimide (Sulfo-NHS); N′—N′-carbonyldiimidazole (CDI); N-ethyl-5-phenylisoxazolium-3′sulfonate (Woodward's reagent K); and combinations thereof.
  • SPDP
  • targeting agents to the LN provides increased efficacy over passive targeting approaches.
  • Targeting involves incorporation of specific targeting moieties such as, but not limited to, ligands or antibodies against cell surface receptors, peptides, lipoproteins, glycoproteins, hormones, vitamins, antibodies, antibody fragments, and conjugates or combinations of these moieties.
  • the maximization of targeting efficiency includes the surface coating of the LN with the appropriate targeting moiety rather than pre-mixing of the targeting ligand with other components, which results in partial encapsulation of the targeting agent, rendering it inaccessible to the cellular target. This method optimizes interaction with cell surface receptors.
  • targeting agents may be either directly incorporated into the LN during synthesis or added in a subsequent step.
  • Functional groups on the targeting moiety as well as specifications of the therapeutic application e.g., degradable linkage dictate the appropriate means of incorporation into the LN.
  • targeting moieties that do not have lipophilic regions cannot insert into the lipid bilayer of the LN directly and require prior conjugation to a lipid anchor before insertion or must form an electrostatic complex with the LNs.
  • a targeting ligand cannot directly connect to a lipophilic anchor.
  • a molecular bridge in the form of a crosslinking agent may be utilized to facilitate the interaction.
  • it is advantageous to use a crosslinking agent if steric restrictions of the anchored targeting moiety prevent sufficient interaction with the intended physiological target.
  • the targeting moiety is only functional under certain orientations (e.g., monoclonal antibody), linking to a lipid anchor via crosslinking agent is beneficial.
  • other methods of bioconjugation may be used to link targeting agents to LNs. Reducible or hydrolysable linkages may be applied to prevent accumulation of the formulation in vivo and the related cytotoxicity.
  • LN preparation is suitable to synthesize the LNs of the present disclosure. For example, ethanol dilution, freeze-thaw, thin film hydration, sonication, extrusion, high pressure homogenization, detergent dialysis, microfluidization, tangential flow diafiltration, sterile filtration, and/or lyophilization may be utilized. Additionally, several methods may be employed to decrease the size of the LNs. For example, homogenization may be conducted on any devices suitable for lipid homogenization such as an Avestin Emulsiflex C5®. Extrusion may be conducted on a Lipex Biomembrane extruder using a polycarbonate membrane of appropriate pore size (0.05 to 0.2 ⁇ m). Multiple particle size reduction cycles may be conducted to minimize size variation within the sample. The resultant LNs may then be passed through a size exclusion column such as Sepharose CL4B or processed by tangential flow diafiltration to purify the LNs.
  • a size exclusion column such as Sepharose
  • LNs described herein may further include ethanol in the preparation process.
  • ethanol in the preparation process.
  • the incorporation of about 30-50% ethanol in LN formulations destabilizes the lipid bilayer and promotes electrostatic interactions among charged moieties such as cationic lipids with anionic ASO and siRNA.
  • LNs prepared in high ethanol solution are diluted before administration.
  • ethanol may be removed by dialysis or diafiltration, which also removes non-encapsulated NA.
  • the LNs be sterilized. This may be achieved by passing of the LNs through a 0.2 or 0.22 ⁇ m sterile filter with or without pre-filtration.
  • LNs Physical characterization of the LNs can be carried through many methods. For example, dynamic light scattering (DLS) or atomic force microscopy (AFM) can be used to determine the average diameter and its standard deviation. In certain embodiments, it is especially desirable that the LNs have about a 200 nm diameter, or less. Zeta potential measurement via zeta potentiometer is useful in determining the relative stability of particles. Both dynamic light scattering analysis and zeta potential analysis may be conducted with diluted samples in deionized water or appropriate buffer solution. Cryogenic transmission electron microscopy (Cryo-TEM) and scanning electron microscopy (SEM) may be used to determine the detailed morphology of LNs.
  • DLS dynamic light scattering
  • AFM atomic force microscopy
  • Zeta potential measurement via zeta potentiometer is useful in determining the relative stability of particles. Both dynamic light scattering analysis and zeta potential analysis may be conducted with diluted samples in deionized water or appropriate buffer solution.
  • LNs described herein are stable under refrigeration for several months. LNs requiring extended periods of time between synthesis and administration may be lyophilized using standard procedures. A cryoprotectant such as 10% sucrose may be added to the LN suspension prior to freezing to maintain the integrity of the formulation during lyophilization. Freeze drying of LN formulations is recommended for long term stability.
  • the LCANs described herein have a diameter of less than 300 nm, and, in particular embodiments, between about 50 and about 200 nm in mean diameter.
  • These LNs show enhanced transfection and reduced cytotoxicity, especially under high serum conditions found during systemic administration.
  • the LNs are useful in a wide range of current therapeutic agents and systems, have high serum stability, and can be designed for targeted delivery with high transfection efficiency.
  • PEI high molecular weight polyethylenimine
  • MW ⁇ 600 kDa Low molecular weight PEI
  • APCs hyper-cationized albumin-polymer conjugates
  • APCs may either be used alone to deliver agents such as pDNA or combined with lipid-based formulations to deliver agents such as ASOs and siRNA.
  • Albumin also possesses endosomal lytic activity due to its hydrophobic core, which upon conformational change can be exposed and can induce bilayer disruption or membrane fusion.
  • the APC has an ionization profile that is responsive to pH change. The charge density is increased at endosomal pH, which is acidic.
  • an APC is combined with a cationic lipid combination to assemble a cationic lipid-APC-NA nanoparticle, sometimes herein called LCAN.
  • an APC is combined with an anionic lipid combination to assemble a lipid-APC-NA nanoparticle.
  • the LNs comprise hyper-cationized APCs. These LNs have high transfection efficiency without additional cytotoxicity.
  • An example scheme for synthesizing an APC is shown in FIG. 8 .
  • the mechanism of action for hyper-cationized pH-responsive APCs is shown in FIG. 9 .
  • a low molecular weight pH-sensitive polymer (polyethylenimine MW 600, PEI600) is conjugated to human serum albumin (HSA) via cross linking agents, resulting in a hyper-cationized pH-responsive APC.
  • HSA-PEI600 conjugates to LNs significantly increases downregulation of RRM1 (aka RNR R1) with ASO LOR-2501 (purchased from Alpha DNA) in the presence of serum without substantial cytotoxicity in KB cells (a subline of HeLa).
  • a low molecular weight pentaethylenehexamine is conjugated to HSA via cross linking agents, resulting in a hyper-cationized pH-responsive APC.
  • This particular formulation referred to as a lipid-coated albumin nanoparticle (LCAN)
  • LCAN lipid-coated albumin nanoparticle
  • HSA-PEHA improves the stability and biological activity of the nanoparticles.
  • the lipids in this formulation are DOTAP, SPC, and TPGS, at a ratio of 25:70:5 (mol/mol).
  • the LNs disclosed herein may be designed to favor characteristics such as increased loading of NAs, increased serum stability, reduced RES-mediated uptake, targeted delivery, or pH sensitive release within the endosome. Because of the varied nature of LN formulations, any one of the several methods provided herein may be used to achieve a particular therapeutic aim. Cationic lipids, anionic lipids, polyalkenes, neutral lipids, fusogenic lipids, cationic polymers, anionic polymers, polymer conjugates, peptides, targeting moieties, and combinations thereof may be utilized to meet specific aims.
  • the LNs described herein can be used as platforms for therapeutic delivery of oligonucleotide (ON) therapeutics, such as cDNA, siRNA, shRNA, miRNA, anti-miR, and antisense ODN.
  • ON oligonucleotide
  • These therapeutics are useful to manage a wide variety of diseases such as various types of cancers, leukemias, viral infections, and other diseases.
  • targeting moieties such as cyclic-RGD, folate, transferrin, or antibodies greatly enhance activity by enabling targeted drug delivery.
  • a number of tumors overexpress receptors on their cell surface.
  • suitable targeting moieties include transferrin (Tf), folate, low density lipoprotein (LDL), and epidermal growth factors.
  • LN formulations having particles measuring about 300 nm or less in diameter with a zeta potential of less than 50 mV and an encapsulation efficiency of greater than 20.0% are useful for NA delivery.
  • Non-limiting examples of such therapeutic agents include antineoplastic agents, anti-infective agents, local anesthetics, anti-allergics, antianemics, angiogenesis-inhibitors, beta-adrenergic blockers, calcium channel antagonists, anti-hypertensive agents, anti-depressants, anti-convulsants, anti-bacterial, anti-fungal, anti-viral, anti-rheumatics, anthelminithics, antiparasitic agents, corticosteroids, hormones, hormone antagonists, immunomodulators, neurotransmitter antagonists, anti-diabetic agents, anti-epileptics, anti-hemmorhagics, anti-hypertonics, antiglaucoma agents, immunomodulatory cytokines, sedatives, chemokines, vitamins, toxins, narcotics, imaging agents, and combinations thereof.
  • NA-based therapeutic agents are highly applicable to the LN formulations of the present disclosure.
  • examples of such NA-based therapeutic agents include, but are not limited to: pDNA, siRNA, miRNA, anti-miRNA, ASO, and combinations thereof.
  • modifications to the substituent NA base units and/or phosphodiester linker can be made.
  • Such modifications include, but are not limited to: backbone modifications (e.g., phosphorothioate linkages); 2′ modifications (e.g., 2′-O-methyl substituted bases); zwitterionic modifications (6′-aminohexy modified ODNs); the addition of a terminal lipophilic moiety (e.g., fatty acids, cholesterol, or cholesterol derivatives); and combinations thereof.
  • the modified sequences synergize with the LN formulations disclosed herein. For example, addition of a 3′-cholesterol to an ODN supplies stability to a LN complex by adding lipophilic interaction in a system otherwise primarily held together by electrostatic interaction during synthesis. In addition, this lipophilic attachment promotes cell permeation by localizing the ODN to the outer leaflet of the cell membrane.
  • the LNs described herein may be administered by the following methods: peroral, parenteral, intravenous, intramuscular, subcutaneous, intraperitoneal, transdermal, intratumoral, intraarterial, systemic, or convection-enhanced delivery.
  • the LNs are delivered intravenously, intramuscularly, subcutaneously, or intratumorally. Subsequent dosing with different or similar LNs may use alternative routes of administration.
  • compositions of the present disclosure comprise an effective amount of a LN formulation disclosed herein, and/or additional agents, dissolved or dispersed in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that produce no adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human.
  • the preparation of a pharmaceutical composition that contains at least one compound or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference.
  • LN preparations should meet sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biological Standards.
  • compositions disclosed herein may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection.
  • Compositions disclosed herein can be administered intravenously, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, topically, intramuscularly, subcutaneously, mucosally, in utero, orally, topically, locally, via inhalation (e.g., aerosol inhalation), by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference).
  • the actual dosage amount of a composition disclosed herein administered to an animal or human patient can be determined by physical and physiological factors such as body weight or surface area, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • compositions may comprise, for example, at least about 0.1% of an active compound.
  • the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound.
  • Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight of active pharmaceutical ingredient (API), etc. can be administered, based on the numbers described above.
  • a composition herein and/or additional agents is formulated to be administered via an alimentary route.
  • Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier.
  • a composition described herein may be administered via a parenteral route.
  • parenteral includes routes that bypass the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered, for example but not limited to, intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally (U.S. Pat. Nos. 6,753,514, 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363 are each specifically incorporated herein by reference in their entirety).
  • compositions disclosed herein as free bases or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (i.e., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • a coating such as lecithin
  • surfactants for example
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include agents to achieve isotonicity, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption such as, for example, aluminum monostearate or gelatin.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580).
  • Sterile injectable solutions are prepared by incorporating the compositions in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by sterilization.
  • dispersions are prepared by incorporating the various sterilized compositions into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • some methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • a powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • compositions may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or via inhalation.
  • topical i.e., transdermal
  • mucosal administration intranasal, vaginal, etc.
  • inhalation via inhalation.
  • the compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in their entirety).
  • the delivery of drugs using intranasal microparticle resins (Takenaga et al., 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725,871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts and could be employed to deliver the compositions described herein.
  • transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety), and could be employed to deliver the compositions described herein.
  • compositions disclosed herein may be delivered via an aerosol.
  • aerosol refers to a colloidal system of finely divided solid or liquid particles dispersed in a liquefied or pressurized gas propellant.
  • the typical aerosol for inhalation consists of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent.
  • Suitable propellants include hydrocarbons and hydrocarbon ethers.
  • Suitable containers will vary according to the pressure requirements of the propellant.
  • Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
  • HSA (25%) was purchased from Octapharma.
  • PEHA was purchased from Sigma-Aldrich.
  • a stock solution of PEHA, pH adjusted to 8.0 with M HCl was prepared.
  • HSA was combined with 500 ⁇ of PEHA.
  • 80 ⁇ of the 1-ethyl-3-[3dimethylaminopropyl]carbodiimide hydrochloride (EDC) (from Fisher Scientific) was added to the solution under stiffing. The reaction proceeded at room temperature for >4 h.
  • EDC 1-ethyl-3-[3dimethylaminopropyl]carbodiimide hydrochloride
  • the reaction proceeded at room temperature for >4 h.
  • the produce HSA-PEHA was purified by gel filtration chromatography on a PD-10 desalting column or by dialysis using a MWCO 10,000 membrane to remove unreacted PEHA and byproducts. Protein concentration of the product was determined by a BCA protein assay.
  • the molecular weight of the HSA-PEHA conjugate was determined by matrix-assisted laser desorption-ionization time-of-flight mass spectrometry (MADLI TOF MS). On average, there were 11 PEHA linked to each HSA based on the result showing m/z of 66405.756.
  • the product can be stored at 4° C., frozen, or lyophilized.
  • Lipids 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) (Avanti Polar Lipids), L- ⁇ -phosphatidylcholine derived from soybean (SPC) (Avanti Polar Lipids), and d-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) (Eastman Chemical) were dissolved in ethanol. Lipids were combined at 25:70:5 (mol/mol). Briefly, 3.15, 8.83, and 0.63 ⁇ mol of DOTAP, SPC, and TPGS, respectively, were combined in 4 mL of ethanol.
  • the LCAN-G3139 was stable at 4° C., and was frozen or lyophilized for long term storage.
  • the particle size of LCAN was determined by NICOMP 370 particle size analyzer.
  • the zeta potential was determined on a zetaPALS instrument.
  • Drug loading efficiency was determined by Oligreen ssDNA quantitation reagents.
  • LCAN particles were found to be ⁇ 200 nm in diameter, had a zeta potential of +20 ⁇ +40 mV and a G3139 encapsulation efficiency of greater than >60%.
  • the same process was used to synthesize ligand-conjugated LCANs by incorporating lipid-derivatized ligands into the lipid components during nanoparticle synthesis.
  • Possible ligands include transferring, folate, cRGD, or antibodies.
  • HSA-PEHA conjugate was synthesized as follows. HSA (25%) was purchased from Octapharma. PEHA was dissolved in water and the pH was adjusted to 8.0 with HCl. A 500-fold excess of PEHA was added to the HSA solution, followed by 80-fold excess of EDC under stiffing. The reaction proceeded at room temperature for 4 hr. The product was purified and concentrated by tangential flow diafiltration on a MicroKros cartridge with MW of 10,000 against water. The product protein concentration was determined by BCA protein assay and the PEHA content in the product was determined by TNBS amine content assay using a PEHA-based standard curve. The PEHA-HSA ratio was calculated based on surplus in amine content relative to unmodified HSA and found to be 10.5:1. SDS-PAGE analysis showed that the conjugate migrated as a single band, indicating lack of intermolecular crosslinking in the HSA-PEHA product.
  • G3139 is an 18-mer phosphorothioate ASO against bcl-2.
  • G3139 purchased from AlphaDNA was dissolved in mM HEPES, pH 7.4.
  • the zeta potential (4) was determined on a ZetaPALS (Brookhaven Instruments Corp., Worcestershire, N.Y.). All measurements were carried out in triplicate. The particle size was 98 ⁇ 40 nm and the zeta potential was +23 mV.
  • the product LCAN-G3139 was analyzed for bcl-2 down regulation in KB (human carcinoma) cells. KB cells were plated in 6-well plates at a density of 2 ⁇ 10 4 cells/cm 2 24 h prior to transfection in RPMI 1640 (Life Technologies) medium containing 10% FBS and 1% antibiotics. The medium was removed and replaced with various G3139 ODN formulations in RPMI 1640 culture medium at G3139 concentration of 1 ⁇ M.
  • the control LN-G3139 is a LN formulation with the composition of DOTAP/SPC/TPGS at 25:70:5 (m/m) without the addition of HSA-PEHA and otherwise prepared by the same method as LCAN.
  • forward primer [SEQ ID NO. 1] CCCTGTGGATGACTGAGTACCTG; reverse primer [SEQ ID NO. 2] CCAGCCTCCGTTATCCTGG; and, probe [SEQ ID NO. 3]) CCGGCACCTGCACACCTGGA.
  • Housekeeping gene ABL mRNAs were also amplified concurrently and Bcl-2 mRNA was normalized to ABL mRNA levels.
  • LCAN-G3139 was much more effective in Bcl-2 down regulation than LN-G3139, a typical LN formulation of G3139 that does not contain HSA-PEHA. These data showed that LCAN-G3139 is a superior composition to most LNs and can be used to deliver antisense ASOs and other oligonucleotide drugs, such as siRNA, miR mimics, and anti-miR oligos.
  • Low molecular weight PEI(600) was used in this example.
  • Alternative low molecular weight polymers such as pentaethylenehexamine (PEHA), may also be conjugated using similar techniques.
  • PEHA pentaethylenehexamine
  • HSA-PEI conjugates were produced.
  • HSA-PEI at various w/w ratios (0, 0.5, 1, 3, 6:1, HSA:ODN w/w) were combined with ODN LOR-2501 (0.2 ⁇ M) (purchased from Alpha DNA) to find the optimal retardation ratio using gel mobility shift analysis. Retardation occurred at 3:1 (HSA:ODN w/w) ( FIG. 1 ).
  • DDAB, CHOL, and TPGS lipid stocks dissolved in 100% ethanol were combined at a molar ratio of 60:35:5. 100 ⁇ L lipid mixture in ethanol was added to 900 ⁇ L 1 ⁇ PBS buffer as to form empty LNs in 10% ethanol.
  • the HSA-PEI/ODN complex was then combined with the empty LNs to form LCANs.
  • the formulation was briefly vortexed and allowed to stand for 15 m at room temperature before transfection into KB cells.
  • the concentration of ODN used was 0.2 ⁇ M ( FIG. 2 ). All LCANs containing HSA-PEI exhibited a positive charge ranging between 5 and 25 mV. LCANs without HSA-PEI were neutrally charged.
  • FIG. 3 displays the downregulation of RNR R1 mRNA expression by LOR-2501 in LCANs.
  • KB cells grown in RPMI 1640 medium at 37° C. under 5% CO 2 atmosphere, were plated 24 h prior to transfection at a density of 3.0 ⁇ 10 5 cells per well in a 6-well plate. Cells were grown to approximately 80% confluency and the serum-containing media was removed. Cells were transfected with 1000 ⁇ L transfection media and treated for 4 h. Transfection occurred in the presence of 0% and 10% serum-containing RPMI 1640 media. Experiments were performed with 3 replicates. After treatment was completed, cells were washed with 1 ⁇ PBS and serum-containing RPMI 1640 was restored. At 48 h after treatment was completed, cells were analyzed for RNR R1 expression levels by RT-PCR with actin as a housekeeping gene.
  • Results are shown in FIG. 2 .
  • the 1:3, ODN:HSA LCAN formulation showed the greatest transfection efficacy.
  • the 1:1 ODN:HSA LCAN formulation was the most efficacious.
  • Cell viability 48 h after treatment was assessed by MTT assay ( FIG. 4 ).
  • a similar experiment involving conjugation of PEHA-to-albumin was completed and showed similar transfection activity ( FIGS. 7 and 8 ).
  • LCAN using HSA-PEI based APC were prepared as described above.
  • CAL-51 triple negative breast cancer cells were plated 24 h prior to transfection in a 6-well plate at a density of 2 ⁇ 10 4 cells/cm 2 in DMEM/F12 media supplemented with 1% penicillin/streptomycin and 10% FBS.
  • LCAN was combined with anti-miR-221 (100 nM) to gauge its ability to upregulate the downstream targets of miR-221, p27/Kip1 and the estrogen receptor alpha (ER ⁇ ).
  • RNA from cells was extracted with TRIzol Reagent (Life Technologies) and cDNA was generated by SuperScript® III First-Strand Synthesis System (Life Technologies) per the manufacturer's instructions.
  • RT-PCR was then performed using SYBR green (Life Technologies) and primers for p27/kip1 (Alpha DNA) and ER ⁇ :
  • ⁇ -actin was used as a control.
  • LCAN/anti-miR-221 led to moderate increases in p27/Kip1 expression and slight increases in ER ⁇ expression.
  • HSA-PEHA conjugates were synthesized at a relatively large scale.
  • the HSA:PEHA:EDC molar ratio used during synthesis was 1:1500:200 (mol/mol).
  • 5 g PEHA (MW 232.37, technical grade) was dissolved in 80 mL of ddH 2 O and then adjusted to pH 8.0 using 1 M HCl.
  • 1 g (4 mL) of HSA (25%, Octapharma)) and then 562.5 mg of 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC, dissolved in DMSO) were added into the PEHA solution under stirring. The reaction continued for 3 h at room temperature.
  • the mixture was then dialyzed using MWCO 10,000 Spectrum membrane against ddH 2 O at 4° C.
  • the buffer was replaced every 3-4 h until amines from PEHA became undetectable by the standard ninhydrin or TNBS amine essay in the external buffer at the 3 h time point at the end of the dialysis cycle.
  • the dialysis procedure can be replaced by tangential flow diafiltration, e.g., using a Millipore Pellicon cassette system or a Spectropor hollowfiber system. This method can also be used to concentrate the product to a desirable concentration.
  • the product can be passed through a 0.22 ⁇ m sterile filter into a sterile container and stored at 4° C. For long-term storage, the product can be stored at ⁇ 20° C.
  • the product can also be lyophilized.
  • the product protein concentration was determined using BCA protein assay.
  • the amine content of the HSA-PEHA conjugate was determined by TNBS assay or MALDI-TOF MS based on change in molecular weight relative to HSA.
  • Gel permeation chromatography combined with amine TNBS assay is used to demonstrate the lack of crosslinked HSA and the absence of free PEHA in the product. Due to the modest cost of the reagents used, the yield of the reaction is not critical. The purity of the product is expected to be very high. Exact product specifications can be defined based on PEHA-to-HSA ratio and higher limits of crosslinked HSA and free PEHA in the final product.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Dispersion Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Nanotechnology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US14/403,315 2012-05-23 2013-05-23 Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same Abandoned US20160015824A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/403,315 US20160015824A1 (en) 2012-05-23 2013-05-23 Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261650729P 2012-05-23 2012-05-23
US201361784892P 2013-03-14 2013-03-14
US14/403,315 US20160015824A1 (en) 2012-05-23 2013-05-23 Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same
PCT/US2013/042461 WO2013177421A2 (en) 2012-05-23 2013-05-23 Lipid-coated albumin nanoparticle compositions and methods of making and method of using the same

Publications (1)

Publication Number Publication Date
US20160015824A1 true US20160015824A1 (en) 2016-01-21

Family

ID=48539454

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/900,969 Active US10307490B2 (en) 2012-05-23 2013-05-23 Lipid nanoparticle compositions for antisense oligonucleotides delivery
US14/403,313 Active US9750819B2 (en) 2012-05-23 2013-05-23 Lipid nanoparticle compositions and methods of making and methods of using the same
US14/403,315 Abandoned US20160015824A1 (en) 2012-05-23 2013-05-23 Lipid-Coated Albumin Nanoparticle Compositions and Methods of Making and Method of Using the Same
US15/689,667 Abandoned US20180021447A1 (en) 2012-05-23 2017-08-29 Lipid Nanoparticle Compositions and Methods of Making and Methods of Using the Same

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/900,969 Active US10307490B2 (en) 2012-05-23 2013-05-23 Lipid nanoparticle compositions for antisense oligonucleotides delivery
US14/403,313 Active US9750819B2 (en) 2012-05-23 2013-05-23 Lipid nanoparticle compositions and methods of making and methods of using the same

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/689,667 Abandoned US20180021447A1 (en) 2012-05-23 2017-08-29 Lipid Nanoparticle Compositions and Methods of Making and Methods of Using the Same

Country Status (10)

Country Link
US (4) US10307490B2 (ja)
EP (3) EP2852381B1 (ja)
JP (4) JP6228191B2 (ja)
KR (3) KR20150020180A (ja)
CN (3) CN105163721B (ja)
AU (4) AU2013266232B2 (ja)
BR (2) BR112014029247A2 (ja)
CA (3) CA2874495A1 (ja)
MX (2) MX353567B (ja)
WO (3) WO2013177421A2 (ja)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10987428B2 (en) * 2018-06-01 2021-04-27 City Of Hope Phosphorothioate-conjugated miRNAs and methods of using the same

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8986702B2 (en) 2008-05-16 2015-03-24 Taiga Biotechnologies, Inc. Antibodies and processes for preparing the same
EP2318435B1 (en) 2008-08-28 2015-12-23 Taiga Biotechnologies, Inc. Modulators of myc, methods of using the same, and methods of identifying agents that modulate myc
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
CN104531812A (zh) 2010-10-01 2015-04-22 现代治疗公司 设计核酸及其使用方法
WO2012135805A2 (en) 2011-03-31 2012-10-04 modeRNA Therapeutics Delivery and formulation of engineered nucleic acids
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RU2707251C2 (ru) 2011-10-03 2019-11-25 Модерна Терапьютикс, Инк. Модифицированные нуклеозиды, нуклеотиды и нуклеиновые кислоты и их применение
KR20140102759A (ko) 2011-12-16 2014-08-22 모더나 세라퓨틱스, 인코포레이티드 변형된 뉴클레오사이드, 뉴클레오타이드 및 핵산 조성물
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
EP2833892A4 (en) 2012-04-02 2016-07-20 Moderna Therapeutics Inc MODIFIED POLYNUCLEOTIDES FOR THE PRODUCTION OF PROTEINS AND PEPTIDES ASSOCIATED WITH ONCOLOGY
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
BR112014029247A2 (pt) 2012-05-23 2017-06-27 Univ Ohio State nanopartícula de lipídio, composição farmacêutica, método para fazer uma nanopartícula de lipído, produto, método de tratamento de um distúrbio, e sistema de entrega
EP2877189B1 (en) 2012-07-20 2021-01-06 Taiga Biotechnologies, Inc. Enhanced reconstitution and autoreconstitution of the hematopoietic compartment
JP6144355B2 (ja) 2012-11-26 2017-06-07 モデルナティエックス インコーポレイテッドModernaTX,Inc. 化学修飾mRNA
US10272115B2 (en) 2013-03-11 2019-04-30 Taiga Biotechnologies, Inc. Production and use of red blood cells
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
DK2992874T3 (en) 2013-04-30 2018-11-12 Delta Fly Pharma Inc LIPOSOM FOR TOPIC SUBMISSION AND USE THEREOF
US9802967B2 (en) * 2013-06-17 2017-10-31 The University Of North Carolina At Chapel Hill Polymer coated particles and methods thereof
CN105579582A (zh) 2013-07-25 2016-05-11 埃克西奎雷股份有限公司 用于预防和治疗用途的作为免疫刺激剂的基于球形核酸的构建体
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015130584A2 (en) * 2014-02-25 2015-09-03 Merck Sharp & Dohme Corp. Lipid nanoparticle vaccine adjuvants and antigen delivery systems
WO2015154002A1 (en) * 2014-04-04 2015-10-08 Ohio State Innovation Foundation Oligonucleotide lipid nanoparticle compositions, methods of making and methods of using the same
WO2015175965A1 (en) * 2014-05-15 2015-11-19 The Research Foundation For Suny Compositions targeting the interaction domain between p27kip1 and brk and methods of use thereof
EP3164113B1 (en) 2014-06-04 2019-03-27 Exicure, Inc. Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications
US9855345B2 (en) 2014-10-30 2018-01-02 Delta-Fly Pharma, Inc. Method for producing lipoplex for topical administration and antitumor agent using such lipoplex
JP2017537619A (ja) 2014-11-21 2017-12-21 ノースウェスタン ユニバーシティ 球状核酸ナノ粒子複合体の配列特異的細胞内取込
US10704043B2 (en) 2015-01-14 2020-07-07 Exicure, Inc. Nucleic acid nanostructures with core motifs
US20160287152A1 (en) * 2015-03-30 2016-10-06 Verily Life Sciences Llc Functionalized Nanoparticles, Methods and In Vivo Diagnostic System
EP4353257A2 (en) 2015-04-13 2024-04-17 CureVac Manufacturing GmbH Method for producing rna compositions
RU2718534C2 (ru) * 2015-06-05 2020-04-08 Мираджен Терапьютикс, Инк. Ингибиторы mir-155 для лечения кожной t-клеточной лимфомы (ctcl)
HRP20220156T1 (hr) 2015-09-17 2022-04-15 Modernatx, Inc. Spojevi i pripravci za unutarstaničnu isporuku terapeutskih sredstava
CN105288647B (zh) * 2015-10-10 2018-05-25 中国药科大学 功能化白蛋白及其纳米制剂的制备方法
JP7080172B2 (ja) * 2015-12-10 2022-06-03 モデルナティエックス インコーポレイテッド 治療薬の送達のための組成物及び方法
DK3394030T3 (da) 2015-12-22 2022-03-28 Modernatx Inc Forbindelser og sammensætninger til intracellulær afgivelse af midler
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
CN112587504B (zh) * 2016-10-17 2022-10-18 南京绿叶制药有限公司 一种抑制bcl-2的反义寡聚核酸的脂质纳米粒及其制备方法
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
CA3045017A1 (en) * 2016-12-02 2018-06-07 Taiga Biotechnologies, Inc. Nanoparticle formulations
WO2018107061A1 (en) * 2016-12-09 2018-06-14 Board Of Regents, The University Of Texas System Hybrid exosomal-polymeric (hexpo) nano-platform for delivery of rnai therapeutics
EP3554561B1 (en) 2016-12-14 2023-06-28 Janssen Biotech, Inc. Cd137 binding fibronectin type iii domains
JP7104703B2 (ja) 2016-12-14 2022-07-21 ヤンセン バイオテツク,インコーポレーテツド Cd8a結合フィブロネクチンiii型ドメイン
WO2018111976A1 (en) 2016-12-14 2018-06-21 Janssen Biotech, Inc. Pd-l1 binding fibronectin type iii domains
US20180179577A1 (en) * 2016-12-22 2018-06-28 Jiaming HU Lipid-polymer Hybrid Nanoparticle Biochip and Application Thereof
EP3558407B1 (en) * 2016-12-22 2023-03-15 Biotronik AG Drug releasing coatings for medical devices and methods of making same
US20190328903A1 (en) * 2016-12-30 2019-10-31 Samyang Biopharmaceuticals Corporation Polymer nanoparticle composition for plasmid dna delivery, and preparation method therefor
KR20190132405A (ko) 2017-03-15 2019-11-27 모더나티엑스, 인크. 치료제의 세포내 전달을 위한 화합물 및 조성물
AU2018234828A1 (en) 2017-03-15 2019-09-19 Modernatx, Inc. Lipid nanoparticle formulation
AU2018234814B2 (en) 2017-03-15 2022-06-30 Modernatx, Inc. Crystal forms of amino lipids
WO2018187590A1 (en) 2017-04-05 2018-10-11 Modernatx, Inc. Reduction or elimination of immune responses to non-intravenous, e.g., subcutaneously administered therapeutic proteins
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
WO2018209270A1 (en) 2017-05-11 2018-11-15 Northwestern University Adoptive cell therapy using spherical nucleic acids (snas)
CN107184552B (zh) * 2017-06-07 2021-03-30 东华大学 一种半乳糖化聚乙烯亚胺修饰的载药醇质体的制备方法
MA49421A (fr) 2017-06-15 2020-04-22 Modernatx Inc Formulations d'arn
JP7231147B2 (ja) * 2017-06-29 2023-03-01 国立大学法人東海国立大学機構 Rna導入試薬及びその利用
EP3655776A2 (en) * 2017-07-18 2020-05-27 Avectas Limited Payload delivery across cell membranes using continuous flow fluidic system
US10149898B2 (en) 2017-08-03 2018-12-11 Taiga Biotechnologies, Inc. Methods and compositions for the treatment of melanoma
US20190062788A1 (en) * 2017-08-22 2019-02-28 Rubius Therapeutics, Inc. Lipid nanoparticle methods and compositions for producing engineered erythroid cells
WO2019046809A1 (en) 2017-08-31 2019-03-07 Modernatx, Inc. METHODS OF MANUFACTURING LIPID NANOPARTICLES
CN107982537A (zh) * 2017-11-17 2018-05-04 厦门大学 针对microRNA-155的治疗性药物及其应用
KR102245539B1 (ko) * 2018-02-12 2021-04-29 주식회사 지앤피바이오사이언스 코어-쉘 구조의 마이크로 입자를 유효성분으로 포함하는 성장인자 유전자 발현 증가용 조성물
US20210113641A1 (en) * 2018-03-29 2021-04-22 Institute Of Basic Medical Sciences Chinese Academy Of Medical Sciences Extraction of plant source "medicinal soup" and manual preparation of "herbal medicine" and related products
US11603543B2 (en) * 2018-04-18 2023-03-14 Oisin Biotechnologies, Inc. Fusogenic lipid nanoparticles for target cell-specific production of a therapeutic protein
JP2021523944A (ja) * 2018-05-15 2021-09-09 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー 有害生物防除組成物及びその使用
EP3808344A4 (en) * 2018-06-01 2022-03-30 Sogang University Research Foundation NANOPARTICLE COMPOSITE HAVING IMPROVED ENDOCYTOSIS EFFICIENCY BY SURFACE MODIFICATION USING A LIPID AND METHOD FOR MAKING THE SAME
CN108743962B (zh) * 2018-06-19 2021-07-02 东华大学 一种基于荧光碳点修饰的树状大分子的双载药靶向纳米平台的制备方法
WO2020051223A1 (en) * 2018-09-04 2020-03-12 The Board Of Regents Of The University Of Texas System Compositions and methods for organ specific delivery of nucleic acids
GB2600800B (en) * 2018-09-04 2023-08-16 Univ Texas Compositions and methods for organ specific delivery of nucleic acids
TW202028222A (zh) 2018-11-14 2020-08-01 美商Ionis製藥公司 Foxp3表現之調節劑
KR20210113260A (ko) * 2019-01-04 2021-09-15 온코루스, 인크. 캡슐화된 rna 폴리뉴클레오타이드 및 사용 방법
EP4031524A1 (en) 2019-09-19 2022-07-27 ModernaTX, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
JP2022551204A (ja) 2019-10-14 2022-12-07 アロ・バイオセラピューティクス・カンパニー Cd71結合フィブロネクチンiii型ドメイン
US11781138B2 (en) 2019-10-14 2023-10-10 Aro Biotherapeutics Company FN3 domain-siRNA conjugates and uses thereof
CN110638759A (zh) * 2019-10-29 2020-01-03 珠海丽凡达生物技术有限公司 一种用于体外转染和体内递送mRNA的制剂
KR102198736B1 (ko) * 2020-01-15 2021-01-05 이화여자대학교 산학협력단 생체 내 약물 전달을 위한 지질 나노입자 및 이의 용도
WO2022036170A1 (en) * 2020-08-14 2022-02-17 Arcturus Therapeutics, Inc. Method of lyophilizing lipid nanoparticles
WO2022192674A1 (en) * 2021-03-11 2022-09-15 The Trustees Of The University Of Pennsylvania Targeted therapeutic lipid nanoparticles and methods of use
WO2023070132A1 (en) * 2021-10-22 2023-04-27 Ohio State Innovation Foundation Immunotherapies for the treatment of cancer
CN114306369B (zh) * 2021-12-23 2023-12-26 北京悦康科创医药科技股份有限公司 一种硫代寡核苷酸注射液及其制备方法
WO2023192503A1 (en) * 2022-03-30 2023-10-05 The Johns Hopkins University Compositions of lipid nanoparticles for plasmid dna delivery to the liver and methods for preparing the same
WO2024044370A1 (en) * 2022-08-25 2024-02-29 Ohio State Innovation Foundation Compositions and methods for the delivery of active agents including nucleic acids
WO2024050310A1 (en) * 2022-08-29 2024-03-07 Sorrento Therapeutics, Inc. Lipid-coated nanoparticles

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6248720B1 (en) * 1996-07-03 2001-06-19 Brown University Research Foundation Method for gene therapy using nucleic acid loaded polymeric microparticles
US20020022264A1 (en) * 1995-05-26 2002-02-21 Sullivan Sean M. Delivery vehicles comprising stable lipid/nucleic acid complexes
US20090312402A1 (en) * 2008-05-20 2009-12-17 Contag Christopher H Encapsulated nanoparticles for drug delivery
US20130196434A1 (en) * 2008-07-25 2013-08-01 Alnylam Pharmaceuticals, Inc. ENHANCEMENT OF siRNA SILENCING ACTIVITY USING UNIVERSAL BASES OR MISMATCHES IN THE SENSE STRAND

Family Cites Families (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US590877A (en) * 1897-09-28 Danger-signal for railway-crossings
US2003203A (en) * 1933-05-15 1935-05-28 Kim Young Wo Game
US5725871A (en) 1989-08-18 1998-03-10 Danbiosyst Uk Limited Drug delivery compositions comprising lysophosphoglycerolipid
US5707644A (en) 1989-11-04 1998-01-13 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5466468A (en) 1990-04-03 1995-11-14 Ciba-Geigy Corporation Parenterally administrable liposome formulation comprising synthetic lipids
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
WO1993023011A1 (en) 1992-05-18 1993-11-25 Minnesota Mining And Manufacturing Company Transmucosal drug delivery device
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
IE80468B1 (en) 1995-04-04 1998-07-29 Elan Corp Plc Controlled release biodegradable nanoparticles containing insulin
US5908777A (en) * 1995-06-23 1999-06-01 University Of Pittsburgh Lipidic vector for nucleic acid delivery
US6056973A (en) * 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
TWI225412B (en) 1997-06-23 2004-12-21 Sequus Pharm Inc Liposome-entrapped polynucleotide composition and method
US6395253B2 (en) * 1998-04-23 2002-05-28 The Regents Of The University Of Michigan Microspheres containing condensed polyanionic bioactive agents and methods for their production
JP4638098B2 (ja) * 1999-06-14 2011-02-23 キャンサー・リサーチ・テクノロジー・リミテッド 癌治療
MXPA02002107A (es) * 1999-08-27 2003-08-20 Inex Pharmaceuticals Corp Composiciones para estimular la secrecion de citoquina e inducir una respuesta inmune.
US7060291B1 (en) * 1999-11-24 2006-06-13 Transave, Inc. Modular targeted liposomal delivery system
US6821955B2 (en) * 2000-04-07 2004-11-23 Baylor College Of Medicine Macroaggregated protein conjugates as oral genetic immunization delivery agents
US6613308B2 (en) 2000-09-19 2003-09-02 Advanced Inhalation Research, Inc. Pulmonary delivery in treating disorders of the central nervous system
CA2445947A1 (en) * 2001-04-30 2002-11-07 Targeted Genetics Corporation Lipid-comprising drug delivery complexes and methods for their production
CA2449054C (en) 2001-05-30 2011-01-04 The Scripps Research Institute Integrin targeting liposome for nucleic acid delivery
JP2003088371A (ja) * 2001-09-04 2003-03-25 Mitsubishi Pharma Corp 遺伝子導入用組成物
KR100484371B1 (ko) 2001-10-25 2005-04-20 가부시키가이샤 아텍스 히터선부착 시트체의 제조방법
AU2002340662B2 (en) * 2001-11-07 2008-07-03 Tekmira Pharmaceuticals Corporation Mucosal adjuvants comprising an oligonucleotide and a cationic lipid
US7678386B2 (en) 2002-07-15 2010-03-16 Board Of Regents The University Of Texas Liposomes coated with selected antibodies that bind to aminophospholipids
CN100558740C (zh) * 2002-08-16 2009-11-11 雷克珊公司 反义寡核苷酸用于抑制Akt-1表达的用途
US20040208921A1 (en) 2003-01-14 2004-10-21 Ho Rodney J. Y. Lipid-drug formulations and methods for targeted delivery of lipid-drug complexes to lymphoid tissues
WO2004066949A2 (en) 2003-01-28 2004-08-12 Rexahn Corporation Antisense oligonucleotides that inhibit expression of hif-1
ATE441710T1 (de) * 2003-01-31 2009-09-15 Rexahn Corp Antisense oligonukleotide, die die ausprägungsstufe von hif-1 hemmen
AU2004272646B2 (en) * 2003-09-15 2011-11-24 Arbutus Biopharma Corporation Polyethyleneglycol-modified lipid compounds and uses thereof
KR100638041B1 (ko) 2003-12-24 2006-10-23 주식회사 삼양사 수용성 약물의 경구투여용 나노입자 조성물 및 그의제조방법
JP2006111591A (ja) * 2004-10-15 2006-04-27 Anges Mg Inc 核酸医薬を標的特異的に細胞内送達するための製剤
JP2006298780A (ja) * 2005-04-15 2006-11-02 Tohoku Univ Hgf遺伝子と粗大凝集アルブミン−ポリエチレンイミン(maa−pei)との複合体
US20070087045A1 (en) * 2005-10-14 2007-04-19 Industrial Technology Research Institute Lipid carrier and method of preparing the same
TW200800235A (en) * 2005-10-18 2008-01-01 Otsuka Pharma Co Ltd Carrier composition for nucleic acid transport
WO2007051303A1 (en) * 2005-11-02 2007-05-10 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
US8067380B2 (en) 2005-12-19 2011-11-29 Industrial Technology Research Institute Glutathione-based delivery system
GB2450475A (en) * 2007-06-12 2008-12-31 Univ Nottingham Trent Antimicrobial polymer nanocomposite
US20110177155A1 (en) * 2007-08-21 2011-07-21 Immune Disease Institute, Inc. Methods of delivery of agents to leukocytes and endothelial cells
PL208054B1 (pl) * 2007-09-06 2011-03-31 Akademia Medyczna Im Piastow Śląskich We Wrocławiu Kompozycja lipidowa do wytwarzania lipidowego nośnika dla leków genetycznych i jej zastosowanie
EP2217221B1 (en) * 2007-10-17 2018-06-27 Korea Advanced Institute of Science and Technology Ldl-like cationic nanoparticles for deliverying nucleic acid gene, method for preparing thereof and method for deliverying nucleic acid gene using the same
BRPI0820302A2 (pt) * 2007-11-09 2015-05-19 Univ Northeastern Nanopartículas semelhantes a micelas de automontagem para liberação sistêmica de gene
WO2009067243A2 (en) * 2007-11-20 2009-05-28 Isis Pharmaceuticals Inc. Modulation of cd40 expression
US20110038941A1 (en) 2007-12-27 2011-02-17 The Ohio State University Research Foundation Lipid Nanoparticle Compositions and Methods of Making and Using the Same
WO2009123734A1 (en) 2008-04-04 2009-10-08 The Regents Of The University Of California Functionalized magnetic nanoparticles and methods of use thereof
US8222220B2 (en) 2008-05-13 2012-07-17 George Mason Intellectual Properties, Inc. Nanogenomics for medicine: siRNA engineering
AU2009249403A1 (en) * 2008-05-19 2009-11-26 The University Of North Carolina At Chapel Hill Methods and compositions comprising novel cationic lipids
WO2010014595A2 (en) 2008-07-31 2010-02-04 The Ohio State University Research Foundation Methods and compositions for delivering therapeutic agents in the treatment of b-cell related disorders
WO2010049562A1 (es) * 2008-10-28 2010-05-06 Universidade De Santiago De Compostela Sistemas nanoparticulares elaborados a base de polímeros aniónicos.
WO2010054384A1 (en) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Lipids and compositions for the delivery of therapeutics
EP2601934A1 (en) 2009-01-22 2013-06-12 Ludwig-Maximilians-Universität München Vesicular phospholipid gels comprising proteinaceous substances
ES2351756B1 (es) * 2009-07-28 2011-10-05 Universidad Del Pais Vasco Nanopartículas lipídicas para terapia génica.
CN102803511A (zh) * 2009-11-23 2012-11-28 俄亥俄州立大学 用于影响肿瘤细胞生长、迁移和侵袭的材料和方法
JP2013528665A (ja) * 2010-03-26 2013-07-11 メルサナ セラピューティックス, インコーポレイテッド ポリヌクレオチドの送達のための修飾ポリマー、その製造方法、およびその使用方法
CN103025384B (zh) * 2010-03-26 2016-04-20 俄亥俄州立大学 涉及miR-155对于错配修复和基因组稳定性的调节的材料和方法
US10081542B2 (en) 2010-04-20 2018-09-25 University of Florida Research Foundation, lnc. Nanozymes, methods of making nanozymes, and methods of using nanozymes
US20130260460A1 (en) * 2010-04-22 2013-10-03 Isis Pharmaceuticals Inc Conformationally restricted dinucleotide monomers and oligonucleotides
US20110319473A1 (en) * 2010-06-29 2011-12-29 Surmodics, Inc. Compositions and methods for enhancement of nucleic acid delivery
US8691750B2 (en) * 2011-05-17 2014-04-08 Axolabs Gmbh Lipids and compositions for intracellular delivery of biologically active compounds
CN102552105B (zh) * 2011-10-17 2014-04-02 复旦大学 一种级联脑部靶向药物递送***及其制备方法和用途
BR112014029247A2 (pt) * 2012-05-23 2017-06-27 Univ Ohio State nanopartícula de lipídio, composição farmacêutica, método para fazer uma nanopartícula de lipído, produto, método de tratamento de um distúrbio, e sistema de entrega

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020022264A1 (en) * 1995-05-26 2002-02-21 Sullivan Sean M. Delivery vehicles comprising stable lipid/nucleic acid complexes
US6248720B1 (en) * 1996-07-03 2001-06-19 Brown University Research Foundation Method for gene therapy using nucleic acid loaded polymeric microparticles
US20090312402A1 (en) * 2008-05-20 2009-12-17 Contag Christopher H Encapsulated nanoparticles for drug delivery
US20130196434A1 (en) * 2008-07-25 2013-08-01 Alnylam Pharmaceuticals, Inc. ENHANCEMENT OF siRNA SILENCING ACTIVITY USING UNIVERSAL BASES OR MISMATCHES IN THE SENSE STRAND

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Fayazpour et al., Biomacromolecules, 2006, 7: 2856-2862. *
Fischer et al., Int. J. Pharm., 2001, 225: 97-111. *
Liu et al., Biomaterials, 2010, 31: 330-338. *
Takeuchi et al., FEBS Lett., 1996, 397: 207-209. *
Thöle et al., J. Drug Target., 2002, 10: 337-344. *
Weecharangsan et al., Anticancer Res., 2010, 30: 1-15. *
Zhang et al., ACS Nano, 2008, 2: 1696-1702. *
Zheng et al., Int. J. Pharm., 2010, 390: 1-19. *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10987428B2 (en) * 2018-06-01 2021-04-27 City Of Hope Phosphorothioate-conjugated miRNAs and methods of using the same
US20210386860A1 (en) * 2018-06-01 2021-12-16 City Of Hope PHOSPHOROTHIOATE-CONJUGATED miRNAs AND METHODS OF USING THE SAME
US11857633B2 (en) * 2018-06-01 2024-01-02 City Of Hope Phosphorothioate-conjugated miRNAs and methods of using the same

Also Published As

Publication number Publication date
MX2014014251A (es) 2015-06-17
US20180021447A1 (en) 2018-01-25
BR112014027834A2 (pt) 2017-08-08
JP2015519346A (ja) 2015-07-09
AU2017245294A1 (en) 2017-11-02
AU2013266238A1 (en) 2014-12-18
JP2015525209A (ja) 2015-09-03
AU2013266232B2 (en) 2017-08-03
WO2013177421A3 (en) 2014-02-27
JP2015523969A (ja) 2015-08-20
AU2013266238B2 (en) 2017-07-27
KR20150032945A (ko) 2015-03-31
WO2013177419A2 (en) 2013-11-28
US9750819B2 (en) 2017-09-05
EP2852415A1 (en) 2015-04-01
BR112014029247A2 (pt) 2017-06-27
CN105163721A (zh) 2015-12-16
AU2017245294B2 (en) 2019-09-12
WO2013177421A2 (en) 2013-11-28
EP2852381A2 (en) 2015-04-01
AU2013266232A1 (en) 2014-11-20
CN104582691A (zh) 2015-04-29
US20150118288A1 (en) 2015-04-30
MX353567B (es) 2018-01-18
EP2852380A4 (en) 2016-01-20
JP6220389B2 (ja) 2017-10-25
CN104428005A (zh) 2015-03-18
US20130315937A1 (en) 2013-11-28
CA2874490A1 (en) 2013-11-28
CN105163721B (zh) 2018-05-22
CA2874495A1 (en) 2013-11-28
KR20150020180A (ko) 2015-02-25
CA2874490C (en) 2021-11-16
CA2871477A1 (en) 2013-11-28
EP2852380A2 (en) 2015-04-01
EP2852381B1 (en) 2020-10-07
KR102169891B1 (ko) 2020-10-26
US10307490B2 (en) 2019-06-04
WO2013177415A1 (en) 2013-11-28
MX2014014196A (es) 2015-08-14
JP2018002727A (ja) 2018-01-11
JP6228191B2 (ja) 2017-11-08
WO2013177419A3 (en) 2014-02-20
CN104428005B (zh) 2019-05-10
EP2852381A4 (en) 2016-01-13
AU2013266236A1 (en) 2014-12-18
KR20150032944A (ko) 2015-03-31

Similar Documents

Publication Publication Date Title
AU2017245294B2 (en) Lipid nanoparticle compositions for antisense oligonucleotides delivery
US10555910B2 (en) Oligonucleotide lipid nanoparticle compositions, methods of making and methods of using the same
US11464870B2 (en) Lipid nanoparticles for in-vivo drug delivery, and uses thereof
WO2020051243A1 (en) Lipid nanoparticles and methods of using thereof
RU2799045C1 (ru) Липидные наночастицы для доставки лекарственного средства in vivo и их применение

Legal Events

Date Code Title Description
AS Assignment

Owner name: OHIO STATE INNOVATION FOUNDATION, OHIO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, ROBERT J.;REEL/FRAME:034436/0680

Effective date: 20141208

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:OHIO STATE UNIVERSITY;REEL/FRAME:036243/0813

Effective date: 20150630

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE OHIO UNIVERSITY;REEL/FRAME:053118/0083

Effective date: 20200703

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:THE OHIO STATE UNIVERSITY;REEL/FRAME:053186/0330

Effective date: 20200713