WO2009035438A1 - Polymeric nanoparticles by ion-ion interactions - Google Patents

Polymeric nanoparticles by ion-ion interactions Download PDF

Info

Publication number
WO2009035438A1
WO2009035438A1 PCT/US2007/019954 US2007019954W WO2009035438A1 WO 2009035438 A1 WO2009035438 A1 WO 2009035438A1 US 2007019954 W US2007019954 W US 2007019954W WO 2009035438 A1 WO2009035438 A1 WO 2009035438A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
ion
polycation
polyanion
chitosan
Prior art date
Application number
PCT/US2007/019954
Other languages
French (fr)
Inventor
Janos Borbely
Magdolna Bodnar
Istvan Hajdu
John F. Hartman
Zsolt Keresztessy
Laszlo Nagy
Gyorgy Vamosi
Original Assignee
Janos Borbely
Magdolna Bodnar
Istvan Hajdu
Hartman John F
Zsolt Keresztessy
Laszlo Nagy
Gyorgy Vamosi
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 Janos Borbely, Magdolna Bodnar, Istvan Hajdu, Hartman John F, Zsolt Keresztessy, Laszlo Nagy, Gyorgy Vamosi filed Critical Janos Borbely
Priority to PCT/US2007/019954 priority Critical patent/WO2009035438A1/en
Publication of WO2009035438A1 publication Critical patent/WO2009035438A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5138Organic macromolecular compounds; Dendrimers obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • 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
    • A61K47/55Medicinal 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 the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal 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 the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic 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
    • A61K47/69Medicinal 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
    • A61K47/6921Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • 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
    • A61K47/6921Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6933Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained by reactions only involving carbon to carbon, e.g. poly(meth)acrylate, polystyrene, polyvinylpyrrolidone or polyvinylalcohol
    • 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
    • A61K47/6921Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6939Medicinal 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 particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5161Polysaccharides, e.g. alginate, chitosan, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin

Definitions

  • the present invention relates to the preparation of nanoparticles from biopolymers such as polycations and polyanions via an ionotropic gellation process, for the purpose of encapsulating nucleic acid such as therapeutic DNA, RNA, siRNA, antisense oligonucleotides (AS-ON) etc., to achieve specific and intracellular delivery of such compounds and provide means of gene therapy.
  • biopolymers such as polycations and polyanions via an ionotropic gellation process
  • nucleic acid such as therapeutic DNA, RNA, siRNA, antisense oligonucleotides (AS-ON) etc.
  • nanodevices which are capable of carrying drugs and other therapeutic agents such as nucleic acids to tissue or cells. Recently many drags have been discovered, which show good efficacy in treatment of cancer or other diseases, however, because of their serious side effects, healthy tissues and organs are affected.
  • the targeted delivery of drugs and chemotherapies using nanodevices offers protection to healthy body segments and also allows dosage reduction.
  • the present nanodevices with their sandwich-like structure are able to protect the active ingredient carried and their surface is designed to avoid immune reactions.
  • Gene therapy means the transferring of genetic material into specific cells of a patient to treat genetic diseases such as hemophilia, muscular dystrophy, cystic fibrosis, cardiovascular and neurological conditions, infectious diseases or cancer by replacing the errant genes, altering gene expression, producing cytotoxic proteins/pro-drug activating enzymes to stop cell proliferation, or vaccinating against viruses by the above means.
  • genetic diseases such as hemophilia, muscular dystrophy, cystic fibrosis, cardiovascular and neurological conditions, infectious diseases or cancer by replacing the errant genes, altering gene expression, producing cytotoxic proteins/pro-drug activating enzymes to stop cell proliferation, or vaccinating against viruses by the above means.
  • Non-viral gene delivery vectors are superior to viral vectors (recombinant viruses) by providing improved safety, flexibility and facile manufacturing.
  • Such polymer- or lipid-based vectors electrostatically bind DNA/RNA and condense it into nano-sized particles (polyplexes or lipoplexes), protect the genes from degradation and mediate cellular entry.
  • Viral vectors allow a (i) high transfection rate and a (H) rapid transcription of the foreign material inserted in the viral genome.
  • (i) safety issues have been raised following the death of a patient during a clinical trial, (H) only small sequences of DNA can be inserted into the virus genome and (Hi) large-scale production may be difficult.
  • toxicity, immune and inflammatory responses can occur and insertional mutagenesis and oncogenic effects have also been observed in vivo.
  • Non-viral systems - synthetic and natural polycations offer (i) low immunogenicity, (H) relatively large sequences may be condensed in small particles, (Hi) good protection to DNA, (iv) easy manufacture, and may be modified to target specific cells and/or diseases.
  • H immunogenicity
  • Hi good protection to DNA
  • iv easy manufacture, and may be modified to target specific cells and/or diseases.
  • problems such as toxicity, lack of biodegradability, low yield of gene transfection, biocompatibility and in particular, low transfection efficiency need to be solved prior to practical use features in shuttling genes into cells.
  • non-viral vectors design criteria for non-viral vectors are as follows. Such systems have to protect nucleic acids from degradation, enable packaging of large DNA plasmids, provide for easy administration, support serum stability and targetability to specific cell types. Simplicity of fabrication and inexpensive synthesis and facile purification are also desired. They have to be robust/stable, facilitate internalization into cells, promote endolysosomal escape of the load, achive nuclear transport and efficient unpackaging for the function of the nucleic acids to be manifested. Infection of non-dividing cells is important in tissue therapy.
  • nanodevices which are capable of targeting drugs to tissue or cells.
  • Many drugs have been discovered, which show good efficacy in treatment of cancer or other diseases, however, because of their serious side effects, healthy tissues and organs are affected.
  • the targeted delivery of drugs and chemotherapies using nanodevices offers protection to healthy body segments and also allows dosage reduction.
  • the present invention relates to the preparation of nanoparticles from biopolymers such as polycations and polyanions via an ionotropic gellation process, for the purpose of encapsulating nucleic acid such as therapeutic DNA, RNA, antisense oligonucleotides (AS- ON), small interfering RNA molecules (siRNA), triple helix forming oligonucleotides (TFO) etc., to achieve specific and intracellular delivery of such compounds to provide means of gene therapy.
  • nucleic acid such as therapeutic DNA, RNA, antisense oligonucleotides (AS- ON), small interfering RNA molecules (siRNA), triple helix forming oligonucleotides (TFO) etc.
  • PC polycation
  • PA polyanion
  • Fig. 4 polyanion
  • the polycation (PC) is complexed with DNA then coated with a polyanion (PA), via an ionotropic gellation process.
  • PA polyanion
  • the PA balances the surface charge, protects DNA, improves circulation time and shields the nucleic acid from the immune system.
  • the self-assembling nature of the nanoparticle provides a simple means of (/) particle preparation without resorting to chemical cross-linking, organic solvents, or other toxic additives (U) controlled degradation of the starting polymers facilitates particle size, and (iii) surface charge and functionality of the nanoparticles are conveniently controlled by varying mixing ratios of the components.
  • the main components are natural and renewable polymers, which are biocompatible, biodegradable and likely non-immunogenic.
  • FIG. 1 is a representation of nanoparticles formed by ion-ion interaction of polyelectrolyte macromolecules.
  • A positively charged polyelectrolyte (dark blue) on the surface.
  • B negatively charged polyelectrolyte (light blue) on the surface. The surface charge is determined by the sequence of mixing.
  • FIG. 2 is a schematic representation of nanodevice.
  • the nanoparticle is loaded with DNA or PvNA or siRNA, AS-ON, TFO etc. and targeting molecules.
  • FIG. 3 shows characterization of chitosan-FITC/poly- ⁇ -glutamic acid-folate nanoparticles (CHIT: ⁇ -PGA 1 : 1, 0.3 mg/ml).
  • FIG. 4. depicts the preparation of nanodevice for DNA/RNA/siRNA delivery.
  • Polycation PC is complexed with DNA then coated with a polyanion (PA), via an ionotropic gelation process.
  • PC is labeled with fluorescein isothiocyanate (FITC) and single stranded DNA was labeled with Cy3 fluorescent dye for microscopic imaging.
  • FITC fluorescein isothiocyanate
  • PA was conjugated with folic acid as cancer cell specific targeting moiety.
  • FIG. 5 is an atomic force microscopic image of CHIT-FITC/DNA-Cy3/PGA-FA nanoparticles.
  • FIG. 6 depicts nanoparticle assisted accumulation of the labeled oligonucleotide DNS- Cy3 in HeLa cells during 1,3,6 and lOmin.of incubation. Combined signals of visible, fluorescein (green) and Cy3 (red) from laser scanning confocal microscopy.
  • Macromolecules with ionizable functional groups such as carboxyl, amino, etc., in an aqueous medium form polycations and polyanions. Under specific conditions polycations and polyanions form nanoparticles by ion-ion interactions. The formation of nanoparticles requires specific reaction parameters, otherwise flocculation and precipitation occurs. However, once the nanoparticles were formed at specific pH and salt concentration, the nanosystem is stable. Sequence ofpolyions
  • Ion-ion interaction can be performed between the functional groups of polyions, and the ratio of original polyions and the order of mixing can affect the particle structure and morphology.
  • the linear polyelectrolyte chains can collapse in compact globules or can extend to coil conformations depending on the pH. The conformation of polymers is an important factor.
  • the final formation of globular nanoparticles is dependent upon interactions between polyelectrolytes. Core-shell or sandwich like morphology can be obtained by varying the ratio of original polyions, the pH and the order of mixing.
  • Figure 1 shows a representation of nanoparticles formed by ion-ion interaction of polyelectrolyte macromolecules.
  • A positively charged polyelectrolyte (Dark blue lines) on the surface.
  • B negatively charged polyelectrolyte (Light blue lines) on the surface. The surface charge is determined by the sequence of mixing.
  • Figure 2 is a schematic representation of nanodevice.
  • the nanoparticle is loaded with DNA or RNA or siRNA, AS-ON, TFO etc. and targeting molecules.
  • the size of nanoparticles depends on the pH of the solution.
  • the hydrodynamic diameter of nanoparticles increases by increasing the pH.
  • the stability of the aqueous solution and the size of nanoparticles depend on the original concentration of polyions.
  • the hydrodynamic diameter of nanoparticles increases with increasing the original concentration of polyions.
  • the stability of the aqueous solution decreases with increasing the original concentrations, and precipitation can be observed in some cases of mixing at high concentration of original polyions.
  • Nanoparticles formed from Poly Acrylic Acid (PAA) and Poly ammonium salt (PAMM) are Nanoparticles formed from Poly Acrylic Acid (PAA) and Poly ammonium salt (PAMM)
  • the pH value of solutions was adjusted to pH 3 by 0.10 mol/dm 3 sodium hydroxide.
  • the solution of PAMM was added to the solution of PAA with gentle stirring.. After 1 hour the pH was increased to 7 resulting in a stable nanosystem with particle size of 50 to 350 run measured by laser light scattering method.
  • the size of nanoparticles is variable in a range of 10-1,000 run by using polymers with different molecular weight. Also the particle size increased at higher pH due to the repulsion of negative charges.
  • the concentration was varied in the range 0.1 mg/ml - 2.0 mg/ml.
  • the ratio of polyelectrolyte and the order of mixing were modulated.
  • the pH was increased with 0.1 M sodium hydroxide solution resulting in stable nanosystems.
  • the hydrodynamic diameter of nanoparticles was in the range of 40 - 480 nm at pH 3, and at pH 7 was 470-1300 nm measured by laser light scattering method. There was some precipitation at higher pH caused by flocculation and coagulation.
  • the size of nanoparticles can be varied by using polymers with different molecular weights.
  • the concentration of CHIT was varied in the range 0.1 mg/ml - 1.0 mg/ml, and of HYAL 0.04 - 0.2 mg/ml.
  • the pH value of solutions was adjusted to pH 3 with 0.10 mol/dm 3 hydrochloric acid.
  • the ratio of polyelectrolyte and the order of mixing were modulated.
  • After 1 hour mixing the pH was increased with 0.1 M sodium hydroxide solution resulting in stable nanosystems.
  • the hydrodynamic diameter of nanoparticles was in the range of 130 - 350 nm at pH 3, and was higher than 600 nm at pH 7 as measured by laser light scattering. There was some precipitation at higher pH caused by flocculation and coagulation.
  • the size of nanoparticles can be varied by using polymers with different molecular weights.
  • Nanoparticles formed from CHIT and Alginic Acid (ALGA) are Nanoparticles formed from CHIT and Alginic Acid (ALGA)
  • the concentration of CHIT was varied in the range 0.1 mg/ml - 1 mg/ml, and of ALGA 0.04 - 0.2 mg/ml.
  • the size of nanoparticles can be varied by using polymers with different molecular weights.
  • Chitosan was partially modified by betaine.
  • the modification was performed by using the carbodiimide technique.
  • CHIT was dissolved in hydrochloric acid media.
  • Betaine was dissolved in water and then adjusted to pH 6.5 with 0.1 M sodium hydroxide solution.
  • Water soluble carbodiimide was added to the betaine solution and the reaction was stirred for 30 min and subsequently mixed with the chitosan solution.
  • the concentration was varied in the range 0.1 mg/ml - 2.0 mg/ml.
  • the ratio of polyelectrolyte and the order of mixing were modulated. After 1 hour mixing the pH was increased by 0.1 M sodium hydroxide solution resulting in stable nanosystems. There was some precipitation at higher pH caused by flocculation and coagulation.
  • the size of nanoparticles can be varied by using polymers with different molecular weight.
  • the vitamin folic acid (FA) was chosen, which has a high affinity for folate receptors (FAR) which are overexpressed in a number of epithelial and myeloid cancer cells.
  • FA was conjugated to poly- ⁇ -glutamic acid (MW 1.3 MDa, GPC) using water soluble carbodiimide.
  • EDC l-Ethyl-3-[3- dimethylaminopropyl]carbodiimide hydrochloride, 8 mg in 1 ml dd. Water
  • FA (12 mg in DMSO) was added and stirred at room temperature for 24 h.
  • the ⁇ -PGA- FA conjugate was purified by dialysis and the number of FA molecules per ⁇ -PGA was estimated by UV-VlS absorption spectroscopy ( ⁇ ma ⁇ i 368 nm, ⁇ 9120; ⁇ ma ⁇ 2 283 nm, ⁇ 25100). This showed that an average of 7 FA molecules was attached to one PGA molecules by this method.
  • Low molecular weight chitosan (MW 320,000 Da, as determined by viscosity measurements, and with a degree of deacetylation of 88%) solution (10 ml, 1 mg/ml in water, solubilised with HCl and pH adjusted to 6.5 with NaOH) was mixed with an aliquot of fluorescein isothiocyanate (FITC, 1 mg/ml in DMSO, 250 ⁇ l) and the reaction mixture was stirred at room temperature for 24 h.
  • FITC fluorescein isothiocyanate
  • Fluorescein-labelled chitosan (CHIT-FITC) was purified by dialysis against water (MW cutoff 10 000 Da, 3 days) and characterised by UV-VIS spectrophotometry, which showed that 71 fluorescein molecules were attached per chitosan molecules by this method.
  • Stable self-assembled polyelectrolytes were developed via an ionotropic gelation process between the folated ⁇ -PGA and the fluorescently labelled chitosan linear chains.
  • aqueous ⁇ -PGA-FA 0.3 mg/ml, pH 9.0
  • CHIT-FITC 0.3 mg/ml, pH 4.0
  • an opaque colloidal system was formed (75% transmittance at ⁇ 500 nm, pH 7.4), which remained stable at room temperature for several weeks at physiological pH.
  • the presence of individual nanoparticles was confirmed and their size distribution characterized by transmission electron microscopy (TEM), atomic force microscopy (AFM) and dynamic light scattering (DLS) as described before.
  • TEM transmission electron microscopy
  • AFM atomic force microscopy
  • DLS dynamic light scattering
  • the overall charge ratio of the nanoparticles (number Of -NH 3 + groups of CHIT vs -COO " groups on ⁇ -PGA) was calculated as +0.67 : -1 based on the weight ratio between CHIT and ⁇ -PGA. This results in a negative zeta-potential at physiological pH, which may contribute to stabilisation of the nanosystem via charge repulsion between individual particles (Majoros et al., 2006; Hong et al., 2007; Hajdu et al., 2007; Berger et al., 2004; Hsieh et al, 2005; Lin et al, 2006; Lin et al., 2007).
  • Figure 3 shows the characterisation of chitosan-FITC/poly- ⁇ -glutamic acid-folate nanoparticles (CHIT: ⁇ -PGA 1:1, 0.3 mg/ml).
  • AFM micrograph with color key for the third dimension (a) AFM micrograph with color key for the third dimension, (b) TEM micrograph of particles and particle size distribution, and (c) the size distribution of the particles was determined by DLS.
  • a stable nanosystem was formed.
  • Nanodevice for single stranded oligonucleotides deliver)'.
  • Nanoparticles were formed from CHIT, DNA and PGA by a general method represented in Figure 4.
  • Figure 4 shows the preparation of nanodevice for DNA/RNA/siRNA, antisense oligonucleoties etc delivery.
  • Polycation PC is complexed with DNA then coated with a polyanion (PA), via an ionotropic gelation process.
  • PC is labelled with fluorescein isothiocyanate (FITC) and single stranded DNA was labeled with Cy3 fluorescent dye for microscopic imaging.
  • FITC fluorescein isothiocyanate
  • PA was conjugated with folic acid as cancer cell specific targeting moiety.
  • Single stranded DNA consisting of 20 nucleotides and with a specific sequence was labelled at the 3' end with Cy3 fluorescent dye (DNA-Cy3).
  • CHIT-FITC was dissolved in water at a concentration of 0.3 mg/ml and at pH 4.
  • DNA-Cy3 was dissolved in water at a concentration of 0.6 mg/ml and at pH 7.4.
  • PGA-FA was dissolved in water at a concentration of 0.3 mg/ml and at pH 9.5.
  • Nanoparticles were formed by either mixing 1 ml of CHIT-FITC solution pre- combined with 50 ⁇ l DNA-Cy3 solution and 1 ml of PGA-FA solution or by mixing 1 ml of CHIT-FITC solution and 1 ml of PGA-FA solution pre-combined with 50 ⁇ l DNA-Cy3 solution.
  • the nanosystems were characterised by dynamic light scattering (DLS) or atomic force microscopy (AFM).
  • DLS dynamic light scattering
  • AFM atomic force microscopy
  • Nanoparticles from Example 8 were tested for intracellular delivery of single stranded oligonucleotides into the human cervical cancer cell line, HeLa, by laser scanning confocal microscopy (LSCM).
  • LSCM laser scanning confocal microscopy
  • Cells were cultured in RPMI 1640 medium (Sigma, St. Louis, MO) supplemented with 10% fetal bovine serum (Fisher Chemicals, Fairlawn, NJ). Cells were grown at 37 0 C in a humidified atmosphere of 5% CO 2 (v/v) in air. All experiments were performed on cells in the exponential growth phase. A culture of the cells were incubated with the nanoparticles for up to 30 min and the internalisation of the oligonucleotide (DNA-Cy3) as well as the CHIT-FITC was observed.
  • DNA-Cy3 internalisation of the oligonucleotide
  • Figure 6 shows Nanoparticle assisted accumulation of the labelled oligonucleotide DNS-Cy3 in HeLa cells during 1,3,6 and 10 minutes of incubation. Combined signals of visible, fluorescein (green) and Cy3 (red) from laser scanning confocal microscopy. Accordingly, a chitosan/poly- ⁇ -glutamic acid-based self-assembling nanoparticulate system as a delivery platform for nucleic acids is provided.
  • the two main components of this polycationic-polyanionic gel-type composite nanosystem are renewable as chitosan is derived from chitin of crustacean shell by alkaline deacetylation while ⁇ -PGA is also easily obtained from Bacillus sp. ferments, where it is produced as slime.
  • CS and ⁇ -PGA are known to be fully biocompatible, biodegradable and likely non-immunogenic and also failed to display any toxicity in our cellular and in vivo studies. Also, the two polymers have a wide range of biomedical applications in separate or in combination. Additional advantages of the CS/ ⁇ - PGA nanosystem as nanocarrier are that (i) its self-assembling nature provides simple preparation without resorting to chemical cross-linking, organic solvents, or other toxic additives (ii) the use of degraded polymers facilitate particle size control, and that (iii) surface charge and functionality of NPs are conveniently tunable by varying component mixing ratios.

Abstract

The present invention relates to biocompatible and biodegradable stimuli-sensitive polymeric nanoparticles, which were formed by ion-ion interaction in aqueous media. Synthetic and biological macromolecules with ionizable functional groups are capable of forming nanoparticles whose size and surface properties are sensitive to environmental influences such as pH, temperature and salt concentration. Nanodevices are designed for therapeutic applications as drug and nucleic acid carriers, and/or for MRI diagnosis as contrast agents. These nanodevices are designed for therapeutic applications as targeted drug carriers. Additionally, they can be used as contrast agents for MRI diagnosis. These nanosystems are also potential carriers for delivery of active ingredients as DNA, RNA, short interfering RNA (siRNA), antisense oligonucleotides (AS-ON), and triple helix forming oligonucleotides (TFO) etc. for pharmaceutical applications. Their adjustable size offers yet another advantage.

Description

POLYMERIC NANOPARTICLES BY ION-ION INTERACTIONS
This application claims priority on U. S. Application Serial No. 60/833,672 entitled "POLYMERIC NANOPARTICLES BY ION-ION INTERACTIONS" filed July 27, 2006, the disclosures of which are incorporated herein by reference.
FIELD OF INVENTION
The present invention relates to the preparation of nanoparticles from biopolymers such as polycations and polyanions via an ionotropic gellation process, for the purpose of encapsulating nucleic acid such as therapeutic DNA, RNA, siRNA, antisense oligonucleotides (AS-ON) etc., to achieve specific and intracellular delivery of such compounds and provide means of gene therapy.
BACKGROUND OF THE INVENTION
There is an increasing demand for nanodevices, which are capable of carrying drugs and other therapeutic agents such as nucleic acids to tissue or cells. Recently many drags have been discovered, which show good efficacy in treatment of cancer or other diseases, however, because of their serious side effects, healthy tissues and organs are affected. The targeted delivery of drugs and chemotherapies using nanodevices offers protection to healthy body segments and also allows dosage reduction. The present nanodevices with their sandwich-like structure are able to protect the active ingredient carried and their surface is designed to avoid immune reactions. Gene therapy means the transferring of genetic material into specific cells of a patient to treat genetic diseases such as hemophilia, muscular dystrophy, cystic fibrosis, cardiovascular and neurological conditions, infectious diseases or cancer by replacing the errant genes, altering gene expression, producing cytotoxic proteins/pro-drug activating enzymes to stop cell proliferation, or vaccinating against viruses by the above means.
A critical barrier to clinical gene therapy is that an efficient and safe delivery vehicle remains to be discovered. Non-viral gene delivery vectors are superior to viral vectors (recombinant viruses) by providing improved safety, flexibility and facile manufacturing. Such polymer- or lipid-based vectors (usually polycations) electrostatically bind DNA/RNA and condense it into nano-sized particles (polyplexes or lipoplexes), protect the genes from degradation and mediate cellular entry.
Viral vectors allow a (i) high transfection rate and a (H) rapid transcription of the foreign material inserted in the viral genome. However (i) safety issues have been raised following the death of a patient during a clinical trial, (H) only small sequences of DNA can be inserted into the virus genome and (Hi) large-scale production may be difficult. Finally, toxicity, immune and inflammatory responses can occur and insertional mutagenesis and oncogenic effects have also been observed in vivo.
Non-viral systems - synthetic and natural polycations offer (i) low immunogenicity, (H) relatively large sequences may be condensed in small particles, (Hi) good protection to DNA, (iv) easy manufacture, and may be modified to target specific cells and/or diseases. However several problems such as toxicity, lack of biodegradability, low yield of gene transfection, biocompatibility and in particular, low transfection efficiency need to be solved prior to practical use features in shuttling genes into cells.
Design criteria for non-viral vectors are as follows. Such systems have to protect nucleic acids from degradation, enable packaging of large DNA plasmids, provide for easy administration, support serum stability and targetability to specific cell types. Simplicity of fabrication and inexpensive synthesis and facile purification are also desired. They have to be robust/stable, facilitate internalization into cells, promote endolysosomal escape of the load, achive nuclear transport and efficient unpackaging for the function of the nucleic acids to be manifested. Infection of non-dividing cells is important in tissue therapy. Further important requirements include general safety, non-toxicity, non- or low immunogenicity and non- pathogenicity (Pack et al., 2005; Tiera et al, 2006; Barron-Peppas et al, 2007; Huang, 2005; Hetzline et al., 2004).
SUMMARY OF INVENTION
There is an increasing demand for nanodevices, which are capable of targeting drugs to tissue or cells. Many drugs have been discovered, which show good efficacy in treatment of cancer or other diseases, however, because of their serious side effects, healthy tissues and organs are affected. The targeted delivery of drugs and chemotherapies using nanodevices offers protection to healthy body segments and also allows dosage reduction.
The present invention relates to the preparation of nanoparticles from biopolymers such as polycations and polyanions via an ionotropic gellation process, for the purpose of encapsulating nucleic acid such as therapeutic DNA, RNA, antisense oligonucleotides (AS- ON), small interfering RNA molecules (siRNA), triple helix forming oligonucleotides (TFO) etc., to achieve specific and intracellular delivery of such compounds to provide means of gene therapy. In the preferred embodiment, polycation (PC) is complexed with DNA and coated with polyanion (PA), via ion-ion interactions (Fig. 4). The present nanodevices with their sandwich-like structure are able to protect the active ingredient cargo. The polycation (PC) is complexed with DNA then coated with a polyanion (PA), via an ionotropic gellation process. The PA balances the surface charge, protects DNA, improves circulation time and shields the nucleic acid from the immune system. The self-assembling nature of the nanoparticle provides a simple means of (/) particle preparation without resorting to chemical cross-linking, organic solvents, or other toxic additives (U) controlled degradation of the starting polymers facilitates particle size, and (iii) surface charge and functionality of the nanoparticles are conveniently controlled by varying mixing ratios of the components. The main components are natural and renewable polymers, which are biocompatible, biodegradable and likely non-immunogenic.
BRIEF DESCRIPTION OF THE DRAWINGS
For a fuller understanding of the invention, reference should be made to the following detailed description, taken in connection with the accompanying drawings, in which:
FIG. 1 is a representation of nanoparticles formed by ion-ion interaction of polyelectrolyte macromolecules. A: positively charged polyelectrolyte (dark blue) on the surface. B: negatively charged polyelectrolyte (light blue) on the surface. The surface charge is determined by the sequence of mixing.
FIG. 2 is a schematic representation of nanodevice. The nanoparticle is loaded with DNA or PvNA or siRNA, AS-ON, TFO etc. and targeting molecules. FIG. 3 shows characterization of chitosan-FITC/poly-γ-glutamic acid-folate nanoparticles (CHIT: γ-PGA 1 : 1, 0.3 mg/ml). (a) AFM micrograph with color key for the third dimension, (b) TEM micrograph of particles and particle size distribution, and (c) the size distribution of the particles determined by DLS.
FIG. 4. depicts the preparation of nanodevice for DNA/RNA/siRNA delivery. Polycation (PC) is complexed with DNA then coated with a polyanion (PA), via an ionotropic gelation process. PC is labeled with fluorescein isothiocyanate (FITC) and single stranded DNA was labeled with Cy3 fluorescent dye for microscopic imaging. PA was conjugated with folic acid as cancer cell specific targeting moiety.
FIG. 5 is an atomic force microscopic image of CHIT-FITC/DNA-Cy3/PGA-FA nanoparticles.
FIG. 6 depicts nanoparticle assisted accumulation of the labeled oligonucleotide DNS- Cy3 in HeLa cells during 1,3,6 and lOmin.of incubation. Combined signals of visible, fluorescein (green) and Cy3 (red) from laser scanning confocal microscopy.
DETAILED DESCRIPTION OF THE INVENTION
Macromolecules with ionizable functional groups such as carboxyl, amino, etc., in an aqueous medium form polycations and polyanions. Under specific conditions polycations and polyanions form nanoparticles by ion-ion interactions. The formation of nanoparticles requires specific reaction parameters, otherwise flocculation and precipitation occurs. However, once the nanoparticles were formed at specific pH and salt concentration, the nanosystem is stable. Sequence ofpolyions
Ion-ion interaction can be performed between the functional groups of polyions, and the ratio of original polyions and the order of mixing can affect the particle structure and morphology. The linear polyelectrolyte chains can collapse in compact globules or can extend to coil conformations depending on the pH. The conformation of polymers is an important factor. The final formation of globular nanoparticles is dependent upon interactions between polyelectrolytes. Core-shell or sandwich like morphology can be obtained by varying the ratio of original polyions, the pH and the order of mixing.
Figure 1 shows a representation of nanoparticles formed by ion-ion interaction of polyelectrolyte macromolecules. A: positively charged polyelectrolyte (Dark blue lines) on the surface. B: negatively charged polyelectrolyte (Light blue lines) on the surface. The surface charge is determined by the sequence of mixing.
Figure 2 is a schematic representation of nanodevice. The nanoparticle is loaded with DNA or RNA or siRNA, AS-ON, TFO etc. and targeting molecules.
Adjusting ofpH
The size of nanoparticles depends on the pH of the solution. The hydrodynamic diameter of nanoparticles increases by increasing the pH.
Surface charge
Surface charge of nanoparticles reveals the sequence of polyion addition. At lower pH, positively charged nanoparticles can be found, independently of the ratio of polyions or order of mixing. By increasing the pH, negatively charged nanoparticles are formed. The ratio of charged free functional groups determines the charge extent of nanoparticles, which depends on the pH and the ratio of functional groups.
Salt effect
The hydrodynamic diameter and the stability of nanoparticles were investigated in KCl solution. It was found that the hydrodynamic diameters decreased with increasing salt concentration, but the stability of the aqueous solutions was independent of the salt concentration.
Adjusting the concentration ofpolyions
The stability of the aqueous solution and the size of nanoparticles depend on the original concentration of polyions. The hydrodynamic diameter of nanoparticles increases with increasing the original concentration of polyions. The stability of the aqueous solution decreases with increasing the original concentrations, and precipitation can be observed in some cases of mixing at high concentration of original polyions.
Example 1
Nanoparticles formed from Poly Acrylic Acid (PAA) and Poly ammonium salt (PAMM)
PAA with Mw=200 IcDa and poly(2-methacryloxyethyltrimethylammonium bromide) were dissolved in water at a concentration of 1 mg/ml. The pH value of solutions was adjusted to pH 3 by 0.10 mol/dm3 sodium hydroxide. The solution of PAMM was added to the solution of PAA with gentle stirring.. After 1 hour the pH was increased to 7 resulting in a stable nanosystem with particle size of 50 to 350 run measured by laser light scattering method. The size of nanoparticles is variable in a range of 10-1,000 run by using polymers with different molecular weight. Also the particle size increased at higher pH due to the repulsion of negative charges.
Example 2
Nanoparticles formed from Chitosan (CHIT) and Poly gamma Glutamic Acid (PGA)
CHIT with Mv=320 kDa and PGA with Mw=I .3 MDa were dissolved in distilled water. The concentration was varied in the range 0.1 mg/ml - 2.0 mg/ml. The pH value of the solutions was adjusted to pH=3 with 0.10 mo I/dm3 hydrochloric acid. The ratio of polyelectrolyte and the order of mixing were modulated. After 1 hour mixing, the pH was increased with 0.1 M sodium hydroxide solution resulting in stable nanosystems. The hydrodynamic diameter of nanoparticles was in the range of 40 - 480 nm at pH 3, and at pH 7 was 470-1300 nm measured by laser light scattering method. There was some precipitation at higher pH caused by flocculation and coagulation.
The size of nanoparticles can be varied by using polymers with different molecular weights.
Example 3
Nanoparticles formed from CHIT and Hyaluronic Acid (HYAL)
CHIT with Mv=320 kDa and HYAL with Mw=2.5 MDa were dissolved in water. The concentration of CHIT was varied in the range 0.1 mg/ml - 1.0 mg/ml, and of HYAL 0.04 - 0.2 mg/ml. The pH value of solutions was adjusted to pH 3 with 0.10 mol/dm3 hydrochloric acid. The ratio of polyelectrolyte and the order of mixing were modulated. After 1 hour mixing the pH was increased with 0.1 M sodium hydroxide solution resulting in stable nanosystems. The hydrodynamic diameter of nanoparticles was in the range of 130 - 350 nm at pH 3, and was higher than 600 nm at pH 7 as measured by laser light scattering. There was some precipitation at higher pH caused by flocculation and coagulation.
The size of nanoparticles can be varied by using polymers with different molecular weights.
Example 4
Nanoparticles formed from CHIT and Alginic Acid (ALGA)
CHIT with Mv=320 kDa and ALGA with Mv=30 kDa were dissolved in water. The concentration of CHIT was varied in the range 0.1 mg/ml - 1 mg/ml, and of ALGA 0.04 - 0.2 mg/ml. The pH value of solutions was adjusted to pH=3 by 0.10 mol/dm3 hydrochloric acid. The ratio of polyelectrolyte and the order of mixing were modulated. After 1 hour mixing the pH was increased by 0.1 M sodium hydroxide solution resulting stable nanosystems at a pH=3. There was some precipitation at higher pH caused by flocculation and coagulation.
The size of nanoparticles can be varied by using polymers with different molecular weights.
Example 5
Nanoparticles formed from modified CHIT and PGA
Chitosan was partially modified by betaine. The modification was performed by using the carbodiimide technique. CHIT was dissolved in hydrochloric acid media. Betaine was dissolved in water and then adjusted to pH 6.5 with 0.1 M sodium hydroxide solution. Water soluble carbodiimide was added to the betaine solution and the reaction was stirred for 30 min and subsequently mixed with the chitosan solution.
The modified CHIT and PGA with a Mw=I .3 MDa were dissolved in water. The concentration was varied in the range 0.1 mg/ml - 2.0 mg/ml. The pH values of the solutions was adjusted to pH=3 with 0.10 mol/dm3 hydrochloric acid. The ratio of polyelectrolyte and the order of mixing were modulated. After 1 hour mixing the pH was increased by 0.1 M sodium hydroxide solution resulting in stable nanosystems. There was some precipitation at higher pH caused by flocculation and coagulation.
The size of nanoparticles can be varied by using polymers with different molecular weight.
Example 6
Preparation of FITC-labeled and folic acid (FA) conjugated CHIT-PGA nanoparticles.
To assess the suitability of CHIT/PGA nanosystem for intracellular delivery of bioactive compounds including nucleic acids, first additional components were incorporated to allow e.g. cancer cell specific targeting and detection of cellular uptake. As a targeting moiety, the vitamin folic acid (FA) was chosen, which has a high affinity for folate receptors (FAR) which are overexpressed in a number of epithelial and myeloid cancer cells.
FA was conjugated to poly-γ-glutamic acid (MW 1.3 MDa, GPC) using water soluble carbodiimide. After the dropwise addition of EDC (l-Ethyl-3-[3- dimethylaminopropyl]carbodiimide hydrochloride, 8 mg in 1 ml dd. Water) to the γ-PGA solution (50 ml, 1 mg/ml, pH 6.5), the reaction mixture was stirred at room temperature for 30 min. FA (12 mg in DMSO) was added and stirred at room temperature for 24 h. The γ-PGA- FA conjugate was purified by dialysis and the number of FA molecules per γ-PGA was estimated by UV-VlS absorption spectroscopy (λmaλi 368 nm, ε 9120; λmaλ2283 nm, ε 25100). This showed that an average of 7 FA molecules was attached to one PGA molecules by this method.
Low molecular weight chitosan (MW 320,000 Da, as determined by viscosity measurements, and with a degree of deacetylation of 88%) solution (10 ml, 1 mg/ml in water, solubilised with HCl and pH adjusted to 6.5 with NaOH) was mixed with an aliquot of fluorescein isothiocyanate (FITC, 1 mg/ml in DMSO, 250 μl) and the reaction mixture was stirred at room temperature for 24 h. Fluorescein-labelled chitosan (CHIT-FITC) was purified by dialysis against water (MW cutoff 10 000 Da, 3 days) and characterised by UV-VIS spectrophotometry, which showed that 71 fluorescein molecules were attached per chitosan molecules by this method.
Stable self-assembled polyelectrolytes were developed via an ionotropic gelation process between the folated γ-PGA and the fluorescently labelled chitosan linear chains. When an equal volume of aqueous γ-PGA-FA (0.3 mg/ml, pH 9.0) and CHIT-FITC (0.3 mg/ml, pH 4.0) were mixed under continuous stirring, an opaque colloidal system was formed (75% transmittance at λ 500 nm, pH 7.4), which remained stable at room temperature for several weeks at physiological pH. The presence of individual nanoparticles was confirmed and their size distribution characterized by transmission electron microscopy (TEM), atomic force microscopy (AFM) and dynamic light scattering (DLS) as described before. The analyses demonstrated that the CHIT-FITC/γ-PGA-FA nanosystem consists of spherical particles with a smooth surface both in aqueous environment and in a dried state (Figure 3a, b). TEM micrograph showed a size range of 30 - 160 nm with a mean value of 67.8 nm (Figure 3b), while DLS reported a bimodal distribution for hydrodynamic diameter ranging between 70 - 90 nm and 160 - 200 nm with mean values of 80 and 178 nm, respectively (Figure 3c). This is consistent with the particles swelling in an aqueous environment. The overall charge ratio of the nanoparticles (number Of -NH3 + groups of CHIT vs -COO" groups on γ-PGA) was calculated as +0.67 : -1 based on the weight ratio between CHIT and γ-PGA. This results in a negative zeta-potential at physiological pH, which may contribute to stabilisation of the nanosystem via charge repulsion between individual particles (Majoros et al., 2006; Hong et al., 2007; Hajdu et al., 2007; Berger et al., 2004; Hsieh et al, 2005; Lin et al, 2006; Lin et al., 2007).
Figure 3 shows the characterisation of chitosan-FITC/poly-γ-glutamic acid-folate nanoparticles (CHIT: γ-PGA 1:1, 0.3 mg/ml). (a) AFM micrograph with color key for the third dimension, (b) TEM micrograph of particles and particle size distribution, and (c) the size distribution of the particles was determined by DLS.
Example 7
Nanodevice for DNA delivery.
CHIT with Mv=320 kDa was dissolved in water at pH= 3. An aqueous solution of DNA with Mw=32 kDa and with specific sequence was added. A stable nanosystem was formed. In the second step, PGA with Mw= 1.2 MDa was added to cover the residual surface. The sandwich- like composite nanodevice containing the DNA molecules was stable at pH=7 and the NaCl concentration was 0.1 g/dm3.
Example 8
Nanodevice for single stranded oligonucleotides deliver)'.
Nanoparticles were formed from CHIT, DNA and PGA by a general method represented in Figure 4.
Figure 4 shows the preparation of nanodevice for DNA/RNA/siRNA, antisense oligonucleoties etc delivery. Polycation (PC) is complexed with DNA then coated with a polyanion (PA), via an ionotropic gelation process. PC is labelled with fluorescein isothiocyanate (FITC) and single stranded DNA was labeled with Cy3 fluorescent dye for microscopic imaging. PA was conjugated with folic acid as cancer cell specific targeting moiety.
CHIT with Mv=320 kDa was labelled with FITC as described in Example 6. PGA with Mw= 1.2 MDa was conjugated with folic acid (FA) as described in Example 6. Single stranded DNA consisting of 20 nucleotides and with a specific sequence was labelled at the 3' end with Cy3 fluorescent dye (DNA-Cy3). CHIT-FITC was dissolved in water at a concentration of 0.3 mg/ml and at pH 4. DNA-Cy3 was dissolved in water at a concentration of 0.6 mg/ml and at pH 7.4. PGA-FA was dissolved in water at a concentration of 0.3 mg/ml and at pH 9.5. Nanoparticles were formed by either mixing 1 ml of CHIT-FITC solution pre- combined with 50 μl DNA-Cy3 solution and 1 ml of PGA-FA solution or by mixing 1 ml of CHIT-FITC solution and 1 ml of PGA-FA solution pre-combined with 50 μl DNA-Cy3 solution.
The nanosystems were characterised by dynamic light scattering (DLS) or atomic force microscopy (AFM). The results showed that both types of nanoparticles (which differ in the order in which components were mixed) had an effective diameter of 75-77 nm in aqueous environment (by DLS with a distribution between 46-148 nm) or up to 31 nm in a dried state (by AFM) as shown in Figure 5. Figure 5 shows an atomic force microscopic image of CfflT-FITC/DNA-Cy3/PGA-FA nanoparticles.
Example 9
Intracellular delivery of single stranded oligonucleotides by CHIT/PGA nanopar tides.
Nanoparticles from Example 8 were tested for intracellular delivery of single stranded oligonucleotides into the human cervical cancer cell line, HeLa, by laser scanning confocal microscopy (LSCM). Cells were cultured in RPMI 1640 medium (Sigma, St. Louis, MO) supplemented with 10% fetal bovine serum (Fisher Chemicals, Fairlawn, NJ). Cells were grown at 37 0C in a humidified atmosphere of 5% CO2 (v/v) in air. All experiments were performed on cells in the exponential growth phase. A culture of the cells were incubated with the nanoparticles for up to 30 min and the internalisation of the oligonucleotide (DNA-Cy3) as well as the CHIT-FITC was observed. The experiments clearly showed that the DNA was delivered into the cytoplasm of HeLa cells (Figure 6). The form of entrance appeared to be vesicular giving rise to an uneven distribution of the DNA within the cytoplasm. After longer incubation, the DNA-Cy3 compound showed accumulation within the nucleus of the cells (Figure 6 10 min). In control experiments, performed by exposing the cells to DNA-Cy3 without encapsulation within the nanoparticles showed that the DNA did not accumulate within the cells or nucleus.
Figure 6 shows Nanoparticle assisted accumulation of the labelled oligonucleotide DNS-Cy3 in HeLa cells during 1,3,6 and 10 minutes of incubation. Combined signals of visible, fluorescein (green) and Cy3 (red) from laser scanning confocal microscopy. Accordingly, a chitosan/poly-γ-glutamic acid-based self-assembling nanoparticulate system as a delivery platform for nucleic acids is provided. The two main components of this polycationic-polyanionic gel-type composite nanosystem are renewable as chitosan is derived from chitin of crustacean shell by alkaline deacetylation while γ-PGA is also easily obtained from Bacillus sp. ferments, where it is produced as slime. CS and γ-PGA are known to be fully biocompatible, biodegradable and likely non-immunogenic and also failed to display any toxicity in our cellular and in vivo studies. Also, the two polymers have a wide range of biomedical applications in separate or in combination. Additional advantages of the CS/γ- PGA nanosystem as nanocarrier are that (i) its self-assembling nature provides simple preparation without resorting to chemical cross-linking, organic solvents, or other toxic additives (ii) the use of degraded polymers facilitate particle size control, and that (iii) surface charge and functionality of NPs are conveniently tunable by varying component mixing ratios.
Thus, it will be seen that the advantages set forth above, and those made apparent from the foregoing description, are efficiently attained and since certain changes may be made in the above construction without departing from the scope of the invention, it is intended that all matters contained in the foregoing description or shown in the accompanying drawings shall be interpreted as illustrative and not in a limiting sense.
It is also to be understood that the following claims are intended to cover all of the generic and specific features of the invention herein described, and all statements of the scope of the invention which, as a matter of language, might be said to fall therebetween. Now that the invention has been described, REFERENCES
Barron-Peppas, L. et al. (2007) Encapsulation of nucleic acids and opportunities for cancer treatment. Pharmaceutical Res. 24, 618-627.
Pack, D. W. et al. (2005) Design and development of polymers for gene delivery. Nat. Rev.
Drug Discovery 4, 581-593.
Tiera, M. J. et al. (2006) Synthetic and natural polycations for gene therapy: State of the art and new perspectives. Curr. Gene Therapy 6, 59-71.
Huang, M. (2005) Transfection efficiency of chitosan vectors: Effect of polymer molecular weight and degree of deacetylation. J. Controlled Release 106, 391-406.
Hetzline, S. E. et al. (2004) Progress in the development of nucleic acid therapeutics for cancer. Cancer Biol. Therapy 3, 4-12.
Majoros, I. J., Thomas, T., and Baker, J. R. Jr. (2006) Molecular engineering in nanotechnology: Engineered drug delivery. Handbook of Theoretical and Computational
Nanotechnology, (Reith, M. and Schommers, W. Eds.) VoI 6, pp 673-717.
Hong, S., Leroueil, P. R., Majoros, I. J., Orr, B. G., Baker, J. R. Jr., and Banasak HoIl, M. M.
(2007) The binding avidity of a nanoparticle-based multivalent targeted drug delivery platform. Chem. Biol. 14, 107-115.
Hajdu, L, Bodnar, M., Filipcsei, G., Hartmann, J. F., Daroczi, L., Zrinyi, M., and Borbely, J.
(2007) Nanoparticles prepared by self-assembly of chitosan and poly-γ-glutamic acid. Coll.
Polym. Sci., in press.
Berger, J., Reist, M., Mayer, J. M., Felt, O., and Gurny, R. (2004) Structure and interactions in chitosan hydrogels formed by complexation or aggregation for biomedical applications.
Eur. J. Pharm. Biopharm. 57, 35-52.
Hsieh, C-Y., Tsai, S.-P., Wang, D.-M., Chang, Y.-N., Hsieh, H.-J. (2005) Preparation of γ-
PGA/chitosan composite tissue engineering matrices. Biomaterials 26, 5617-5623. Lin, Y.-H., Chung, C. K., Chen, C-T., Liang, H.-F., Chen, S.-C, and Sung, H.-W. (2006) Preparation of nanoparticles composed of chitosan/poly-g-glutamic acid and evaluation of their permeability through caco-2 cells. Biomacromolecules 6, 1104-1112.
Lin, Y.-H., Mi, F.-L., Chen, C-T., Chang, W.-C, Peng, S.-F., Liang, H.-F., and Sung, H.-W. (2007) Preparation and characterization of nanoparticles shelled with chitosan for oral insulin delivery. Biomacromolecules 8, 146-152.

Claims

Claims We claim
1. A method of preparing core-shell nanoparticles comprising complexing polycations with one or more nucleic acids and coating the complex with polyanion (PA), in an aqueous solution via ion-ion interactions.
2. The method according to claim 1 wherein the nucleic acid is a natural nucleic acid.
3. The method according to claim 1 wherein the nucleic acid is a synthetic nucleic acid.
4. The method according to claim 1 wherein the nucleic acid is a single stranded nucleic acid.
5. The method according to claim 1 wherein the nucleic acid is a double stranded nucleic acid.
6. The method according to claim 1 wherein the nucleic acid is DNA.
7. The method according to claim 1 wherein the nucleic acid is RNA
8. The method according to claim 1 wherein the nucleic acid is an antisense oligonucleotides (AS-ON).
9. The method according to claim 1 wherein the nucleic acid is a small interfering RNA molecule (siRNA).
10. The method according to claim 1 wherein the nucleic acid is a triple helix forming oligonucleotides (TFO).
11. The method according to claim 1 wherein said polycation is a polyammonium salt (PAMM).
12. The method according to claim 1 wherein said polycation is chitosan.
13. The method according to claim 12 wherein the weight ratio of chitosan to nucleic acid is about 10: 1.
14. The method according to claim 13 wherein the chitosan component ranges in molecular weight from about 60 kDa to 320 kDa.
15. The method of claim 1, wherein said polyanion is selected from a group consisting of polyacrylic acid (PAA), poly γ glutamic acid (PGA), hyaluronic acid (HYAL) and alginic acid (ALGA).
16. The method according to claim 12 wherein said chitosan is reacted with betaine.
17. The method according to claim 12 wherein said chitosan is reacted with FITC.
18. The method according to claim 15 wherein said poly γ glutamic acid is reacted with carbodiimide and folic acid.
19. The method according to claim 1 wherein said ion-ion interaction is by means of ionotropic gellation.
20. The method according to claim 1 wherein said polycation is chitosan and said polyanion is poly γ glutamic acid (PGA)
21. The method according to claim 20 wherein the weight ratio of chitosan to poly γ glutamic acid is about 1: 1.
22. The nucleic acid delivery system according to claim 21 wherein PGA component range in molecular weight about 80 kDa to about 1 300 kDa.
23. A method for performing gene therapy comprising encapsulating a nucleic acid in a nanoparticle formed from the reaction of a polycation (PC) and a polyanion (PA), via an ion-ion reaction
24. The method according to claim 23 wherein the polycation (PC) is complex ed with DNA then coated with a polyanion (PA), via ionotropic gellation.
25. A nucleic acid delivery system comprising a nucleic acid encapsulated in a nanoparticle formed from the reaction of a polycation (PC) and a polyanion (PA), via an ion- ion reaction.
26. The system according to claim 25 wherein the polycation (PC) is complexed with DNA then coated with a polyanion (PA), via ionotropic gellation.
27. A method of delivering a nucleic acid to a cell comprising the steps of: providing a polycation; providing a nucleic acid; providing a polyanion; combining the polycation, the nucleic acid and the polyanion under conditions sufficient to form polycation-nucleic acid- polyanion complexes; and contacting a target cell with the polycation-nucleic acid-polyanion complexes.
PCT/US2007/019954 2007-09-13 2007-09-13 Polymeric nanoparticles by ion-ion interactions WO2009035438A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2007/019954 WO2009035438A1 (en) 2007-09-13 2007-09-13 Polymeric nanoparticles by ion-ion interactions

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2007/019954 WO2009035438A1 (en) 2007-09-13 2007-09-13 Polymeric nanoparticles by ion-ion interactions

Publications (1)

Publication Number Publication Date
WO2009035438A1 true WO2009035438A1 (en) 2009-03-19

Family

ID=39323750

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/019954 WO2009035438A1 (en) 2007-09-13 2007-09-13 Polymeric nanoparticles by ion-ion interactions

Country Status (1)

Country Link
WO (1) WO2009035438A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2978421A4 (en) * 2013-03-28 2016-12-14 Bbs Nanotechnology Ltd Stable nanocomposition comprising docetaxel, process for the preparation thereof, its use and pharmaceutical compositions containing it
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994024144A2 (en) * 1993-04-19 1994-10-27 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
DE19810965A1 (en) * 1998-03-13 1999-09-16 Aventis Res & Tech Gmbh & Co Nanoparticles comprising polyelectrolyte complex of polycation, polyanion and biologically active agent, especially useful for controlled drug release on oral administration
EP1354958A1 (en) * 2002-04-18 2003-10-22 Max-Delbrück-Centrum Für Molekulare Medizin Preparation and use of DNA-polyelectrolyte nanoparticles for gene transfer
US20060064331A1 (en) * 2004-08-18 2006-03-23 Cassie Odermott Insurance premium refund incentive program
US20060147539A1 (en) * 2004-10-05 2006-07-06 Hsing-Wen Sung Nanoparticles for protein drug delivery
WO2007031812A1 (en) * 2005-09-16 2007-03-22 Medipol Sa Chitosan-based particles
EP1859792A1 (en) * 2006-05-24 2007-11-28 Advanced in Vitro Cell Technologies, S.L. Nanoparticles of chitosan and hyaluronan for the administration of active molecules
WO2007135164A1 (en) * 2006-05-24 2007-11-29 Advanced In Vitro Cell Technologies, S.A. Nanoparticles of chitosan and hyaluronan for the administration of active molecules

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994024144A2 (en) * 1993-04-19 1994-10-27 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
DE19810965A1 (en) * 1998-03-13 1999-09-16 Aventis Res & Tech Gmbh & Co Nanoparticles comprising polyelectrolyte complex of polycation, polyanion and biologically active agent, especially useful for controlled drug release on oral administration
EP1354958A1 (en) * 2002-04-18 2003-10-22 Max-Delbrück-Centrum Für Molekulare Medizin Preparation and use of DNA-polyelectrolyte nanoparticles for gene transfer
US20060064331A1 (en) * 2004-08-18 2006-03-23 Cassie Odermott Insurance premium refund incentive program
US20060147539A1 (en) * 2004-10-05 2006-07-06 Hsing-Wen Sung Nanoparticles for protein drug delivery
WO2006073950A2 (en) * 2005-01-04 2006-07-13 Nanomega Medical Corporation Nanoparticles for protein drug delivery
WO2007031812A1 (en) * 2005-09-16 2007-03-22 Medipol Sa Chitosan-based particles
EP1859792A1 (en) * 2006-05-24 2007-11-28 Advanced in Vitro Cell Technologies, S.L. Nanoparticles of chitosan and hyaluronan for the administration of active molecules
WO2007135164A1 (en) * 2006-05-24 2007-11-29 Advanced In Vitro Cell Technologies, S.A. Nanoparticles of chitosan and hyaluronan for the administration of active molecules

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BLAGBROUGH I S ET AL: "Polyamines and novel polyamine conjugates interact with DNA in ways that can be exploited in non-viral gene therapy.", BIOCHEMICAL SOCIETY TRANSACTIONS APR 2003, vol. 31, no. 2, April 2003 (2003-04-01), pages 397 - 406, XP002479358, ISSN: 0300-5127 *
DOUGLAS ET AL: "Effects of alginate inclusion on the vector properties of chitosan-based nanoparticles", JOURNAL OF CONTROLLED RELEASE, ELSEVIER, AMSTERDAM, NL, vol. 115, no. 3, 27 October 2006 (2006-10-27), pages 354 - 361, XP005720372, ISSN: 0168-3659 *
TRUBETSKOY V S ET AL: "Recharging cationic DNA complexes with highly charged polyanions for in vitro and in vivo gene delivery", GENE THERAPY, MACMILLAN PRESS LTD., BASINGSTOKE, GB, vol. 10, no. 3, 1 February 2003 (2003-02-01), pages 261 - 271, XP002314302, ISSN: 0969-7128 *
ZAITSEV SERGEY ET AL: "Polyelectrolyte nanoparticles mediate vascular gene delivery", PHARMACEUTICAL RESEARCH (DORDRECHT), vol. 21, no. 9, September 2004 (2004-09-01), pages 1656 - 1661, XP002479357, ISSN: 0724-8741 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3252068A2 (en) 2009-10-12 2017-12-06 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
EP2978421A4 (en) * 2013-03-28 2016-12-14 Bbs Nanotechnology Ltd Stable nanocomposition comprising docetaxel, process for the preparation thereof, its use and pharmaceutical compositions containing it

Similar Documents

Publication Publication Date Title
US20080160096A1 (en) Polymeric nanoparticles by ion-ion interactions
Abedini et al. Overview on natural hydrophilic polysaccharide polymers in drug delivery
Gao et al. Zwitterion-functionalized mesoporous silica nanoparticles for enhancing oral delivery of protein drugs by overcoming multiple gastrointestinal barriers
Csaba et al. Ionically crosslinked chitosan/tripolyphosphate nanoparticles for oligonucleotide and plasmid DNA delivery
Narayanan et al. Versatile carboxymethyl chitin and chitosan nanomaterials: a review
Hu et al. Advances in chitosan-based drug delivery vehicles
Zhou et al. Galactosylated chitosan–polycaprolactone nanoparticles for hepatocyte-targeted delivery of curcumin
Wang et al. PLGA/polymeric liposome for targeted drug and gene co-delivery
Opanasopit et al. Development and characterization of pectinate micro/nanoparticles for gene delivery
Sun et al. Self-assembled biodegradable micellar nanoparticles of amphiphilic and cationic block copolymer for siRNA delivery
Duceppe et al. Factors influencing the transfection efficiency of ultra low molecular weight chitosan/hyaluronic acid nanoparticles
Xue et al. A green and facile method for the preparation of a pH-responsive alginate nanogel for subcellular delivery of doxorubicin
Yang et al. Host–guest interaction-based self-engineering of nano-sized vesicles for co-delivery of genes and anticancer drugs
Vauthier et al. Chitosan-based nanoparticles for in vivo delivery of interfering agents including siRNA
Gonzalez-Gaitano et al. Drug carrier systems based on cyclodextrin supramolecular assemblies and polymers: present and perspectives
Sipoli et al. Scalable production of highly concentrated chitosan/TPP nanoparticles in different pHs and evaluation of the in vitro transfection efficiency
Suh et al. Layer-by-layer nanoparticle platform for cancer active targeting
Li et al. Nanoparticle depots for controlled and sustained gene delivery
Han et al. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery
Rajan et al. Potential drug delivery applications of chitosan based nanomaterials
Lee et al. Prolonged gene silencing by siRNA/chitosan-g-deoxycholic acid polyplexes loaded within biodegradable polymer nanoparticles
CN104623685B (en) A kind of mesenchyma stroma of tumors pH responsive types targeting dendrimers and preparation method thereof
Vora et al. Self-assembled nanocomplexes of anionic pullulan and polyallylamine for DNA and pH-sensitive intracellular drug delivery
Zhang et al. Carboxymethyl chitosan-poly (amidoamine) dendrimer core–shell nanoparticles for intracellular lysozyme delivery
Jang et al. Hyaluronic acid-siRNA conjugate/reducible polyethylenimine complexes for targeted siRNA delivery

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07852402

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07852402

Country of ref document: EP

Kind code of ref document: A1