US20050271745A1 - Magnetic nanoparticle compositions, and methods related thereto - Google Patents

Magnetic nanoparticle compositions, and methods related thereto Download PDF

Info

Publication number
US20050271745A1
US20050271745A1 US10/888,189 US88818904A US2005271745A1 US 20050271745 A1 US20050271745 A1 US 20050271745A1 US 88818904 A US88818904 A US 88818904A US 2005271745 A1 US2005271745 A1 US 2005271745A1
Authority
US
United States
Prior art keywords
magnetic nanoparticle
magnetic
nanoparticle composition
metal
combination
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/888,189
Inventor
Cordula Gruettner
Joachim Teller
Fritz Westphal
Robert Ivkov
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.)
NANOTX Inc
micromod Partikeltechnologie GmbH
Aspen Medisys LLC
Original Assignee
Individual
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
Priority claimed from US10/360,561 external-priority patent/US20040156852A1/en
Application filed by Individual filed Critical Individual
Priority to US10/888,189 priority Critical patent/US20050271745A1/en
Assigned to MICROMOD PARTIKELTECHNOLOGIE GMBH reassignment MICROMOD PARTIKELTECHNOLOGIE GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GRUETTNER, CORDULA, TELLER, JOACHIM, WESTPHAL, FRITZ
Assigned to TRITON BIOSYSTEMS, INC. reassignment TRITON BIOSYSTEMS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IVKOV, ROBERT
Assigned to TRITON BIOSYSTEMS, INC. reassignment TRITON BIOSYSTEMS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MICROMOD PARTIKELTECHNOLOGIE GMBH
Publication of US20050271745A1 publication Critical patent/US20050271745A1/en
Assigned to NANOTX, INC. reassignment NANOTX, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: ONCOLOGIC, INC., TRITON BIOSYSTEMS, INC.
Assigned to ADURO BIOTECH reassignment ADURO BIOTECH FICTITIOUS BUSINESS NAME STATEMENT Assignors: NANOTX CORP.
Assigned to ASPEN MEDISYS, LLC reassignment ASPEN MEDISYS, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ADURO BIOTECH
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/26Iron; Compounds thereof
    • 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
    • A61K47/60Medicinal 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 the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • 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
    • A61K47/61Medicinal 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 the organic macromolecular compound being a polysaccharide or a derivative thereof
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • 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
    • A61K47/6923Medicinal 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 an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1851Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule
    • A61K49/1857Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule the organic macromolecular compound being obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. PLGA
    • A61K49/186Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule the organic macromolecular compound being obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. PLGA the organic macromolecular compound being polyethyleneglycol [PEG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1851Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule
    • A61K49/1863Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an organic macromolecular compound, i.e. oligomeric, polymeric, dendrimeric organic molecule the organic macromolecular compound being a polysaccharide or derivative thereof, e.g. chitosan, chitin, cellulose, pectin, starch
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1875Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle coated or functionalised with an antibody
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1241Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins
    • A61K51/1255Granulates, agglomerates, microspheres
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y25/00Nanomagnetism, e.g. magnetoimpedance, anisotropic magnetoresistance, giant magnetoresistance or tunneling magnetoresistance
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01FMAGNETS; INDUCTANCES; TRANSFORMERS; SELECTION OF MATERIALS FOR THEIR MAGNETIC PROPERTIES
    • H01F1/00Magnets or magnetic bodies characterised by the magnetic materials therefor; Selection of materials for their magnetic properties
    • H01F1/0036Magnets or magnetic bodies characterised by the magnetic materials therefor; Selection of materials for their magnetic properties showing low dimensional magnetism, i.e. spin rearrangements due to a restriction of dimensions, e.g. showing giant magnetoresistivity
    • H01F1/0045Zero dimensional, e.g. nanoparticles, soft nanoparticles for medical/biological use
    • H01F1/0054Coated nanoparticles, e.g. nanoparticles coated with organic surfactant
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01FMAGNETS; INDUCTANCES; TRANSFORMERS; SELECTION OF MATERIALS FOR THEIR MAGNETIC PROPERTIES
    • H01F1/00Magnets or magnetic bodies characterised by the magnetic materials therefor; Selection of materials for their magnetic properties
    • H01F1/44Magnets or magnetic bodies characterised by the magnetic materials therefor; Selection of materials for their magnetic properties of magnetic liquids, e.g. ferrofluids
    • H01F1/445Magnets or magnetic bodies characterised by the magnetic materials therefor; Selection of materials for their magnetic properties of magnetic liquids, e.g. ferrofluids the magnetic component being a compound, e.g. Fe3O4
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/811Of specified metal oxide composition, e.g. conducting or semiconducting compositions such as ITO, ZnOx
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/827Nanostructure formed from hybrid organic/inorganic semiconductor compositions
    • Y10S977/83Inorganic core or cluster coated with organic or biological shell
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/832Nanostructure having specified property, e.g. lattice-constant, thermal expansion coefficient
    • Y10S977/838Magnetic property of nanomaterial

Definitions

  • the magnetic nanoparticle composition is prepared via a two-step process wherein a preformed metal-containing magnetic material is processed through a turbulent flow zone in a first step, and the resulting improved magnetic material of the first step is utilized in combination with a biocompatible coating material to generate a magnetic nanoparticle composition, via a turbulent flow zone in a second step.
  • the magnetic nanoparticle composition is prepared via a two-step process wherein a metal-containing magnetic material is generated from a metal-containing solution, via a turbulent flow zone, in a first step, and the resulting magnetic material of the first step is utilized in combination with a biocompatible coating material to generate a magnetic nanoparticle composition, via a turbulent flow zone, in a second step.
  • FIG. 2 b Size Plot of Lot-No. 152-W1, of dextran-coated iron oxide, first fraction (example 2);
  • FIG. 2 c Size Plot of Lot-No. 152-W2, of dextran-coated iron oxide, second fraction (example 2);
  • AMF an abbreviation for alternating magnetic field
  • alternating magnetic field refers to a magnetic field that changes the direction of its field vector periodically, for example in a manner that is sinusoidal, triangular, or rectangular.
  • the AMF may also be added to a static magnetic field, such that only the AMF component of the resulting magnetic field vector changes direction. It will be appreciated that an alternating magnetic field is accompanied by an alternating electric field and is electromagnetic in nature.
  • One aspect of the present invention relates to magnetic nanoparticle compositions that comprise one or more metal-containing magnetic nanoparticles and a suitable medium for suspending the nanoparticles.
  • the magnetic nanoparticles comprise a susceptor that comprises a suitable biocompatible coating material.
  • the magnetic nanoparticles possess a low-field magnetization when an external magnetic field is applied to the magnetic nanoparticles.
  • the magnetic material of the nanoparticles exhibits a low-field magnetization greater than that of the material from which it was derived.
  • the magnetic nanoparticle compositions and a sub-structure, preferably a ligand form the basis for the bioprobe compositions useful in the treatment of the body, body part, tissue, cell, or body fluid of a subject.
  • Another aspect of the present invention relates to methods for producing biocompatible magnetic nanoparticles that may be used in the treatment of various indications and in various biological applications. These methods for synthesizing the nanoparticles utilize the “core-shell method” or the “one-pot method”.
  • the core-shell methods disclosed herein employ a two-step process; in the first step, 1) for example, a magnetic metal oxide is formed (core) from either a magnetic preformed material or a suitable metal-containing precursor material, which is then 2) coated with a biocompatible polymer—the coating (shell).
  • Suitable proteins include an extracellular matrix protein, proteoglycan, glycoprotein, albumin, peptide, and gelatin.
  • the polymer comprises a hydrogel polymer, a histidine-containing polymer, or a combination of a hydrogel polymer and a histidine-containing polymer.
  • the polymer for use herein is dextran or dextran that contains functional groups.
  • the preferred functional groups are hydroxyl, oxyalkyl, and carboxyl functional groups.
  • the resulting magnetic nanoparticles have at least an average 50 mass percent of metal and an average hydrodynamic diameter of less than 200 nm.
  • the targeting ligand is the ING-1 antibody.
  • ING-1 (heMAb) is a high-affinity, human-engineeredTM monoclonal antibody that recognizes a 40 kDa epithelial cell adhesion molecule (Ep-CAM—also known as 17-1A, EPG40, GA733-2, KSA and EGP).
  • Ep-CAM epithelial cell adhesion molecule
  • ING-1 was developed using human engineering, in which individual residues in the variable region of a murine antibody are replaced with the amino acid found in the human framework, such that immunogenicity of the variable region is reduced.
  • ING-1 is produced from Chinese hamster ovary (CHO) cells containing synthetic heavy and light chain genes encoding the modified variable regions linked to human IgG1 and kappa constant region cDNA, respectively.
  • Magnetite will display a Verwey transition near 100° K. on low-temperature remanence curves as the domains transition from blocked to unblocked behavior. On the other hand, maghemite has no Verwey transition. If magnetite particles show superparamagnetic behavior, then it is usually difficult to see the magnetic effects of the Verwey transition over the thermal unblocking effect of SP in this type of experiment. In this case, no Verwey transition was observed.
  • the PEG-COOH modification leads to higher negative zetapotentials of the nanoparticles in the basic pH range and higher positive zetapotentials in the acidic pH range.
  • the range of saturation of the particle surface with functional groups can be derived by achieving constant zetapotentials at theoretical PEG-COOH densities greater than 125 nanomol (nmol)/mg.
  • the nanoparticles of lots 202-G3 and 202-G4 show only low to medium coverage with PEG-COOH groups (as shown in FIG. 5 ).
  • the average hydrodynamic diameter of the ChL6 coated nanoparticles (Lot 206-G) was determined by PCS to be in the range of 45 nm-55 nm.
  • the iron content in the particles was determined to be in the range of 56% to 60% (w/w).
  • the examples 6-8 provide a detailed description of the synthetic procedures for the production of iron oxide nanoparticles by high pressure homogenization (HPH) (example 6), for the production of dextran coated nanoparticles from HPH-produced iron oxide (example 7), for the functionalization of nanoparticles, and methods used for conjugating ING-1 antibody to produce the final product for in vivo application (example 8).
  • HPH high pressure homogenization
  • dextran 6 g was dissolved in 20 ml of water. 22 ml of dextran coated nanoparticles of Lot 178-W (example 7) having a particle concentration of 46 mg/ml was added to the dextran solution. The particle suspension was heated to 100° C., with a rotation speed of 400 rpm for 1 hour. After cooling to room temperature, the magnetic particles were separated using a permanent magnet to remove the excess dextran from the supernatant. The particles were suspended in 20 ml of water to yield a particle concentration of 58 mg/ml.

Abstract

Disclosed are biocompatible magnetic nanoparticle compositions for various therapeutic or biological applications, and methods related thereto. Specifically, the present invention pertains to magnetic nanoparticle compositions prepared via high-pressure homogenization processes that include a turbulent flow zone. The methods of production may involve a two-step or a one-step process. The disclosed magnetic nanoparticle compositions may be useful in the treatment of the body, body part, tissue, cell, or body fluid of a subject for a variety of indications. The disclosed magnetic nanoparticle compositions may also be useful in the fixation, separation, transportation, marking or coding of targets, or energy transformation processes.

Description

    CROSS REFERENCE TO RELATED APPLICATION
  • This is a continuation-in-part application claiming the benefit of and priority to U.S. patent application Ser. No. 10/360,561 filed on Feb. 6, 2003, and German patent application number 103 33 631 filed on Jul. 10, 2003, which are incorporated by reference.
  • TECHNICAL FIELD
  • The present invention relates generally to biocompatible magnetic nanoparticle compositions for various biological and therapeutic applications, and methods related thereto. Specifically, the present invention pertains to magnetic nanoparticle compositions prepared via high-pressure homogenization processes that include a turbulent flow zone.
  • BACKGROUND
  • A number of commercial applications for magnetic nanoparticles exist today. Examples include the isolation, fixation and cleaning of cells, parts of cells, nucleic acids, enzymes, antibodies, proteins or peptides; phagocyte diagnostics in cell biology; parts of diagnostic assays or therapeutic drug forms in clinical chemistry; contrast media, radionuclide or drug carrier in clinical diagnostics; and in bio- and technical chemistry as solid phase for the analysis of molecular recognition phenomena and hetero catalytic processes.
  • Various polymer coated metal oxide particles for biological applications have been disclosed since the mid-1980's. Examples include magnetizable nanoparticles below 200 nanometers (nm) that open new possibilities for transport and separation of cells, cell parts, bioactive molecules and radionuclides (e.g., U.S. 2003/0099954, Miltenyi; WO 01/17662 Zborowski; WO 02/43708, Alexiou); markers for contrast enhanced nuclear magnetic imaging or diagnostic methods (e.g., U.S. 2003/0092029A1, Josephson; WO 01/74245, Johansson; and U.S. Pat. No. 5,427,767, Kresse); and mechanical and thermal modifications of living cells (e.g., DE 10020376A1, Koch; and U.S. Pat. No. 6,541,039, Lesniak).
  • All such applications involve magnetizable metal oxide particles coated with biocompatible polymer layers and combined to form composite particles of 5 nm to 500 nm in diameter in a colloidal stable water-based suspension. The coating material is selected to eliminate any undesired interaction between the particles and the biological material, i.e. to make the particles biocompatible, so as to guarantee a sufficient tolerability of the coated particle with vital cells, influence the mechanism of metabolism of the particles in the living environment, or enable a selective binding at the cell surfaces, or enable the controlled release of enclosed substances. Due to their magnetic properties, the particles may align with applied magnetic fields and react to changes of said fields.
  • Various methods to produce these metal oxide, particularly as iron oxide, particles have been reported. Examples of these methods include sintering at high temperatures followed by mechanical grinding, clustering under vacuum conditions, and wet chemical synthesis from solution. The precipitation of iron oxide can be initiated under non-aqueous conditions (e.g., U.S. Pat. No. 4,677,027, Porath) and be continued under aqueous conditions (e.g., U.S. Pat. No. 5,160,725, Pilgrim), or it can be accomplished completely under aqueous conditions (e.g., U.S. Pat. No. 4,329,241, Massart). Due to toxicity concerns, an aqueous formulation is generally used for biological applications (e.g., U.S. Pat. No. 4,101,435, Hasegawa). Wet chemical synthesis can be preceded by a coating with polymer components (Core-shell-Method), or it can be performed in the presence of the polymer (One-pot-Method). For the core-shell-method, it is necessary to add stabilizing substances to the iron oxide, because these tend to aggregate in aqueous solutions, thereby limiting aggregation of the oxide particles. Amphiphilic substances (e.g., WO 01/56546, Babincova) or additional nanoparticles with electrically charged surfaces can be selected as stabilizers (e.g., U.S. Pat. No. 4,280,918, Homola). Surface-active substances used as stabilizers may influence and limit the functionality of said surface. Iron-containing magnetizable nanoparticle composites produced by the One-pot method are accepted for medical applications due to their physical and chemical properties as well as to their pharmacological stability. The One-pot method involves the coating of the polymer directly during the formation of the iron oxide to stabilize the formation process and the growth of the crystals from the solution. The most commonly used coating material is dextran in its various forms. Also used are other biocompatible carbohydrates, such as arabinogalactan, starch, glycosaminoglycan and proteins (e.g., U.S. Pat. No. 6,576,221, Kresse). One such method is the precipitation of Fe(II)- and Fe(III)-salts in the presence of dextran (e.g., U.S. Pat. No. 4,452,773, Molday). A modified method involves the use of ultrasound treatment followed by thermal treatment in the same apparatus (e.g., U.S. Pat. No. 4,827,945, Groman). The quality of this method is enhanced by magnetic classification (e.g., WO 9007380, Miltenyi). Further encapsulation/coating may improve the biocompatibility of the nanoparticles due to the use of amphiphilic substances for stabilization. (e.g., U.S. Pat. No. 5,545,395 Tournier; and EP 0272091, Eley).
  • Suspensions of nanoparticles produced using the techniques described hereinabove typically contain particles with varying characteristics such as size. The non-homogeneous nature of the particle suspension degrades the performance of the suspension for many biological applications. Methods to improve the homogeneity of the particles are utilized for the production of dispersed aqueous systems that are intended for use as injectable fluids. Such homogenization methods include the rotor-stator- and high-pressure-methods. The use of liquid-jet- or liquid-slot-nozzle-high-pressure homogenization machines (e.g., available from Microfluidics, a division of MFIC, Corp., Newton, Mass.) enables high mechanical energy deposition, which is especially useful for the production of liposomes (e.g., U.S. Pat. No. 5,635,206, Ganter) or injectable active substances (e.g., U.S. Pat. No. 5,595,687, Raynolds).
  • High-pressure homogenization machines for the production of metal oxide nanoparticle compositions using controlled coalescence followed by drying in emulsion, in which the non-aqueous component contains an oxide as sol, have been reported in conjunction with industrial production of catalytic materials (e.g., U.S. Pat. No. 5,304,364, Costa) as well as electrographic pigment particles, ceramic powders, felt materials, spray layers, active substances carriers, and ion exchange resins (e.g., U.S. Pat. No. 5,580,692, Lofftus). An emulsion is the dispersion of multi-phase systems of two or more insoluble liquids. Emulsions consist of at least one continuous (outer) phase (e.g. water) and one isolated (dispersed or inner) phase (e.g., oil). Emulsions are thermodynamically unstable. High-pressure homogenization (HPH) is often used for the preparation or stabilization of emulsions and suspensions in pharmaceutical, cosmetic, chemical and food industries. It is also known that nano-scale metal oxides can be prepared using high shear forces in a fluidizing apparatus but it concerns only plain oxides, i.e., those without any coating (e.g., U.S. Pat. No. 5,417,956, Moser). For some applications, pressures up to 200 Mega Pa (MPa) or higher are used.
  • SUMMARY OF THE INVENTION
  • Various methods for producing magnetic nanoparticle compositions exist. However, a major problem has been the inability to produce biocompatible magnetic nanoparticle compositions having enhanced homogeneity, especially those having high metallic content and high magnetic mobility.
  • In view of the above, there is a need for biocompatible magnetic nanoparticle compositions that have enhanced homogeneity and comprise high metallic content nanoparticles possessing high magnetic mobility. There is also a need for methods for producing such compositions.
  • It is, therefore, an aspect of the present invention to provide biocompatible magnetic nanoparticle compositions having enhanced homogeneity.
  • It is also an aspect of the present invention to provide magnetic nanoparticle compositions comprising high metallic content nanoparticles that possess high magnetic mobility.
  • It is another aspect of the present invention to provide methods for producing magnetic nanoparticle compositions, particularly with the use of high-pressure homogenization processes that include a turbulent flow zone.
  • It is another aspect of the present invention to provide applications for biocompatible magnetic nanoparticle compositions comprising high metallic content nanoparticles that possess high magnetic mobility.
  • The present invention pertains to biocompatible magnetic nanoparticle compositions that possess high metallic content and exhibit enhanced homogeneity and high magnetic mobility, which contributes to higher heating rates. Higher heating rates are desirable for various therapeutic applications. These compositions are preferably prepared via high-pressure homogenization processes, and are useful for various therapeutic and biological applications.
  • In one embodiment, the magnetic nanoparticle composition comprises one or more metal-containing magnetic nanoparticles and a suitable medium for suspending the nanoparticles. The nanoparticles comprise a suitable biocompatible coating material. The magnetic nanoparticles possess a low-field magnetization when an external magnetic field is applied to the magnetic nanoparticles. The magnetic material of the nanoparticles exhibits a low-field magnetization greater than that of the material from which it was derived. The nanoparticles have at least an average 50 mass percent of metal and an average hydrodynamic diameter of less than 200 nm.
  • In another embodiment, the magnetic nanoparticle composition is prepared via a two-step process wherein a preformed metal-containing magnetic material is processed through a turbulent flow zone in a first step, and the resulting improved magnetic material of the first step is utilized in combination with a biocompatible coating material to generate a magnetic nanoparticle composition, via a turbulent flow zone in a second step. In another embodiment, the magnetic nanoparticle composition is prepared via a two-step process wherein a metal-containing magnetic material is generated from a metal-containing solution, via a turbulent flow zone, in a first step, and the resulting magnetic material of the first step is utilized in combination with a biocompatible coating material to generate a magnetic nanoparticle composition, via a turbulent flow zone, in a second step. In another embodiment, the magnetic nanoparticle composition is prepared via a one-step process wherein a metal-containing magnetic material is generated from a metal-containing solution, and processed with a biocompatible coating material, via a turbulent flow zone, to form a magnetic nanoparticle composition. The resulting magnetic nanoparticles may be separated from the carrier medium by an external magnetic field, for example, via permanent magnets.
  • The biocompatible coating material may comprise a polymer, metal compound, transfection agent, or any combination thereof. The polymer may comprise a naturally occurring, synthetic, or semi-synthetic polymer. Preferably, the polymer is dextran or dextran that contains functional groups, e.g., sulfoalkyl, aminoalkyl, epoxyalkyl and carboxyalkyl whereby the alkyl chain may be substituted with heteroatoms, e.g., oxygen. The metal-containing magnetic material comprises a metal, metal oxide, metal oxide-hydrate, metal hydroxide, metallic alloy of two or more metals, or any combination thereof. The metal-containing magnetic material has ferro-, antiferro-, ferri-, antiferri- or superparamagnetic properties. Preferably, the metal-containing magnetic material comprises an oxide of iron, more preferably, the iron oxide is magnetite, hematite, maghemite, or any combination thereof. The magnetic nanoparticle composition may also comprise sub-structures that may comprise ligands or chelators, or that may comprise one or more bioactive substances.
  • The present invention also pertains to applications for biocompatible magnetic nanoparticle compositions, namely therapeutic and biological applications. The therapeutic applications pertain to the treatment of the body, body part, tissue, cell, or body fluid of a subject for a variety of indications, including but not limited to, cancer of any type, such as bone marrow, lung, vascular, neuro, colon, ovarian, breast and prostate cancer, epitheleoid sarcomas, adverse angiogenesis, restenosis, amyloidosis, tuberculosis, multiple sclerosis, cardiovascular plaque, vascular plaque, obesity, malaria, and illnesses due to viruses, such as HIV and AIDS. The therapeutic compositions may be administered via injection, topical application, transdermal application, oral ingestion, rectal insertion, inhalation through the mouth or nose, or any combination thereof. The biological applications include fixation, separation, transportation, marking or coding of targets, or energy transformation processes. The magnetic nanoparticle compositions may also be used in the separation, purification, or any combination thereof of nucleic acids, nucleic acid derivatives, nucleic acid fragments, proteins, protein derivatives, protein fragments, or any combination thereof.
  • The above summary of the present invention is not intended to describe each illustrated embodiment or every implementation of the present invention. The figures and the detailed description that follow particularly exemplify these embodiments.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The invention may be more completely understood in consideration of the following detailed description of various embodiments of the invention in connection with the accompanying drawings, in which:
  • FIG. 1 schematically illustrates a bioprobe configuration, according to an embodiment of the present invention;
  • FIG. 2 graphically illustrates size plots of various samples, according to an embodiment of the present invention;
  • FIG. 2 a: Size Plot of Lot-No. 146-T (iron oxide, example 1);
  • FIG. 2 b: Size Plot of Lot-No. 152-W1, of dextran-coated iron oxide, first fraction (example 2);
  • FIG. 2 c: Size Plot of Lot-No. 152-W2, of dextran-coated iron oxide, second fraction (example 2);
  • FIG. 3 graphically illustrates the zetapotential results for iron oxide of Lot-No. 146-T (example 1) and dextran-coated iron oxide of Lot-No. 152-W (example 2), according to an embodiment of the present invention;
  • FIG. 4 illustrates a process flow chart for samples 152-W1, 152-W2 and 152-W3 (examples 1 and 2), according to an embodiment of the present invention;
  • FIG. 5 graphically illustrates zetapotential-pH function for nanoparticles of type 152-W with different PEG-COOH densities of Lot-No. 202-G1-202-G4 (example 3), according to an embodiment of the present invention; and
  • FIG. 6 graphically illustrates isoelectric point (IEP) of nanoparticles of type 152-W with different PEG-COOH densities, according to an embodiment of the present invention.
  • While the invention is amenable to various modifications and alternative forms, specifics thereof have been shown by way of example in the drawings and will be described in detail. It should be understood, however, that the intention is not to limit the invention to the particular embodiments described. On the contrary, the intention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the invention as defined by the appended claims.
  • DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
  • The present invention pertains to biocompatible magnetic nanoparticle compositions that possess high metallic content and exhibit enhanced homogeneity and high magnetic mobility, which contributes to higher heating rates. Higher heating rates are desirable for various therapeutic applications. These compositions are preferably prepared via high-pressure homogenization processes, and are useful for various therapeutic and biological applications.
  • 1. Definitions
  • The terms “targeted therapy system”, “targeted nanotherapeutics”, and “targeted therapy”, as used herein, refer to the methods and devices that involve the targeted delivery of bioprobe compositions for the treatment of an indication, including those disclosed in U.S. patent applications U.S. 2003/0032995, U.S. 2003/0028071, Ser. Nos. 10/360,578, and 10/360,561.
  • The term “hyperthermia”, as used herein, refers to heating of tissue to temperatures between 40° C. and 46° C.
  • The term “susceptor”, as used herein, refers to a magnetic energy susceptive particle that, when exposed to an energy source, either heats or physically moves.
  • The term “magnetic nanoparticle composition”, as used herein, refers to a composition comprising a susceptor that comprises a biocompatible coating and a suitable medium.
  • The term “ligand”, as used herein, refers to a molecule or compound that attaches to a susceptor (or a coating on the susceptor) and targets and attaches to a biological marker. A monoclonal antibody specific for Her-2 (an epidermal growth factor receptor protein) is an exemplary ligand.
  • The term “marker”, as used herein, refers to an antigen or other substance to which the ligand is specific. Her-2 protein is an exemplary marker.
  • The term “target”, as used herein, refers to the matter for which deactivation, rupture, disruption or destruction is desired, such as a diseased cell, a pathogen, or other undesirable matter. A marker may be attached to the target. Breast cancer cells are exemplary targets.
  • The term “bioprobe”, as used herein, refers to a susceptor comprising a biocompatible coating and at least one sub-structure. The term “bioprobe compositions”, as used herein, refers to a composition comprising a magnetic nanoparticle composition and at least one 1 sub-structure. The sub-structure, preferably a ligand, acts to guide the bioprobe to a target. The term “bioprobe system”, as used herein, refers to a bioprobe specific to a target that is optionally identified via a marker.
  • The term “linker molecule”, as used herein, refers to an agent that targets particular functional groups on a ligand and on a susceptor or a coating, and thus forms a covalent link between the ligand and the susceptor or the coating.
  • The term “indication”, as used herein, refers to a medical condition, such as a disease. Breast cancer is an exemplary indication.
  • The term “energy source”, as used herein, refers to a device that is capable of delivering AMF energy to the bioprobe's susceptor.
  • The term “AMF” (an abbreviation for alternating magnetic field), as used herein, refers to a magnetic field that changes the direction of its field vector periodically, for example in a manner that is sinusoidal, triangular, or rectangular. The AMF may also be added to a static magnetic field, such that only the AMF component of the resulting magnetic field vector changes direction. It will be appreciated that an alternating magnetic field is accompanied by an alternating electric field and is electromagnetic in nature.
  • It is to be understood that the singular forms of “a”, “an”, and “the”, as used herein and in the appended claims, include plural reference unless the context clearly dictates otherwise.
  • 2. Nanoparticle Compositions
  • The present invention pertains to magnetic nanoparticle compositions and methods for producing these compositions.
  • 2.1. Magnetic Nanoparticle Compositions
  • One aspect of the present invention relates to magnetic nanoparticle compositions that comprise one or more metal-containing magnetic nanoparticles and a suitable medium for suspending the nanoparticles. The magnetic nanoparticles comprise a susceptor that comprises a suitable biocompatible coating material. The magnetic nanoparticles possess a low-field magnetization when an external magnetic field is applied to the magnetic nanoparticles. The magnetic material of the nanoparticles exhibits a low-field magnetization greater than that of the material from which it was derived. The magnetic nanoparticle compositions and a sub-structure, preferably a ligand, form the basis for the bioprobe compositions useful in the treatment of the body, body part, tissue, cell, or body fluid of a subject.
  • 2.2. High Pressure Homogenization (“HPH”) Process for Producing Magnetic Nanoparticle Compositions
  • Another aspect of the present invention relates to methods for producing biocompatible magnetic nanoparticles that may be used in the treatment of various indications and in various biological applications. These methods for synthesizing the nanoparticles utilize the “core-shell method” or the “one-pot method”. The core-shell methods disclosed herein employ a two-step process; in the first step, 1) for example, a magnetic metal oxide is formed (core) from either a magnetic preformed material or a suitable metal-containing precursor material, which is then 2) coated with a biocompatible polymer—the coating (shell).
  • The two-step process may be accomplished by first processing a preformed metal-containing magnetic material to yield a metal-containing magnetic material having improved magnetic properties, and then generating a magnetic nanoparticle composition using the material resulting from the first step and a biocompatible coating material wherein both steps are processed via a turbulent flow zone. The two-step process may also be accomplished by first generating a metal-containing magnetic material from a metal-containing solution, and then generating a magnetic nanoparticle composition using the material resulting from the first step and a biocompatible coating material, wherein both steps are processed via a turbulent flow zone.
  • The one-pot method disclosed herein employs a one-step process, in which a metal-containing magnetic material is generated from a metal-containing solution, and it is processed with a biocompatible coating material, via a turbulent flow zone, to yield a magnetic nanoparticle composition.
  • The preformed metal-containing magnetic material for use herein comprises a metal, metal oxide, metal oxide-hydrate, metal hydroxide, metal alkoxide, metallic alloy with another metal, or any combination thereof. The magnetic preformed material has ferro-, antiferro-, ferri-, antiferri-, or superparamagnetic properties. Preferably, the magnetic preformed material comprises an oxide of iron, more preferably, the iron oxide is magnetite, hematite, maghemite, or any combination thereof. The iron oxide may be doped with bi- or tri-valent metal ions. Preferably, the magnetic preformed material comprises a combination of Fe-(II) and Fe-(III), more preferably Fe-(II) and Fe-(III) in a molar ratio of 1:1 to 1:2.
  • The metal-containing solution suitable for use herein can be prepared from one or a combination of two or more different metals, different metal compounds, or different valences of a metal, alone or in combination. Preferably, the solution comprises a combination of Fe-(II) and Fe-(III) salts, more preferably Fe-(II) and Fe-(III) salts in a molar ratio of 1:1 to 1:2.
  • To produce the core from a metal-containing solution, for example, an iron oxide suspension is produced by precipitation of ferric and ferrous chlorides in the presence of ammonia. After precipitation, the particles are separated from the liquid and re-suspended. In the second step, these suspended iron oxide precipitates, or cores, are coated with a biocompatible polymer under high-pressure homogenization conditions. Preferably, homogenization occurs above 100 bar, more preferably above 1000 bar. High-pressure homogenization takes advantage of high shear forces that are generated when a large quantity of fluid is forced through a nozzle with a high pressure to produce a polymer-coated particle. The fluid that is forced through the nozzle contains the precipitated particles, to which a liquid containing dissolved biocompatible coating material, preferably a polymer, is added. The resulting mixture is then further processed in the homogenizer by repeated cycling through the nozzle. The resulting magnetic nanoparticles may be separated from the carrier medium by an external magnetic field, for example, via permanent magnets.
  • Preferably, the resulting magnetic nanoparticles form a stable aqueous colloid or a stable colloid in physiological solution.
  • The first and the second steps are preferably processed in a liquid carrier medium at a temperature in the range from about 40° C. and the boiling point of the medium, more preferably in the range from about 75° C. to about 95° C. Preferably, the liquid medium comprises water or an aqueous alkaline solution having an ammonia, sodium or potassium hydroxide basis. Preferably, the components are processed at a flow rate in the range from about 20 ml/min to about 200 ml/min through a turbulent flow zone in each step.
  • The biocompatible coating material suitable for use herein comprises a polymer, metal compound, or any combination thereof. The polymeric coating materials suitable for use herein may be naturally occurring, synthetic, semi-synthetic, or any combination thereof. The synthetic polymer may comprise polyamine, polyimine, polyvinyl, polyol, polyether, polycarboxylic acid, polysilicic acid, polyacrylate, polysiloxane, polyalkylene glycol, parylene, polylactic acid, polyglycolic acid, or any derivative thereof, or any combination thereof.
  • In one embodiment, the polymer comprises a functional group, preferably, a reactive or an ionic group, or any combination thereof. In another embodiment, the polymer comprises a homo-polymer, a co-polymer, or a polymer-blend. In another embodiment, the polymer comprises a biological material, such as polysaccharides, preferably dextran, polyamino acids, proteins, lipids, fatty acids, heparin, heparin sulfate, chondroitin sulfate, chitin, chitosan, alginate, glycosaminoglycan, cellulose, starch, any derivative thereof, and any combination thereof. Suitable proteins include an extracellular matrix protein, proteoglycan, glycoprotein, albumin, peptide, and gelatin. In another embodiment, the polymer comprises a hydrogel polymer, a histidine-containing polymer, or a combination of a hydrogel polymer and a histidine-containing polymer. Preferably, the polymer for use herein is dextran or dextran that contains functional groups. The preferred functional groups are hydroxyl, oxyalkyl, and carboxyl functional groups.
  • The metal compounds (of the biocompatible coating material) suitable for use herein include hydroxyapatite, metal carboxylate, sulfonate, phosphate, ferrite, phosphonate, and oxides of Group IV elements of the Periodic Table of Elements. These materials may form a composite coating with the biological or synthetic polymers. Where the magnetic particle is formed from a biocompatible magnetic material, the surface of the particle itself may operate as the biocompatible coating.
  • The nanoparticles of the resulting magnetic nanoparticle composition may also comprise sub-structures that may comprise ligands, chelators or a combination thereof, or a bioactive substance. The sub-structures may be bound covalently or by physical interaction to uncoated portion of susceptor 142, to coating 144, or to an uncoated portion of susceptor 142 and partially covered by coating 144, or they may be intercalated with coating 144. The ligands or chelators may comprise a peptide, protein, nucleic acid, enzyme, antibody, antibody fragment, or any combination thereof. The ligands suitable for use herein are presented in Table I. The preferred antibody for use herein is ING-1. The bioactive substance may comprise a pharmaceutical agent, including chemotherapeutic and hormone, peptide, lipid, biochemical factor, or any combination thereof.
  • The resulting magnetic nanoparticles may be subjected to an external magnetic field. In such a case, the initial susceptibility (the ratio of the nanoparticle magnetic moment to applied magnetic field near zero field, especially in the range between −20 and +20 Oersted) of the resulting magnetic nanoparticles is larger than that of the magnetic material from which it was derived.
  • The resulting magnetic nanoparticles have at least an average 50 mass percent of metal and an average hydrodynamic diameter of less than 200 nm.
  • 3. Applications
  • Another aspect of the present invention relates to applications for the magnetic nanoparticle compositions, which include targeted therapies and biological applications. The magnetic nanoparticles of the compositions of the present invention possess high metallic content and exhibit enhanced homogeneity and high magnetic mobility, which contribute to higher heating rates. Higher heating rates are desirable for various therapeutic applications.
  • 3.1. Targeted Nanotherapeutics
  • The bioprobe compositions of the present invention are useful in the treatment of the body, body part, tissue, cell, or body fluid of a subject. The bioprobe compositions, as a component of targeted nanotherapeutic systems, may be used in the treatment of a variety of indications, including but not limited to, cancer of any type, such as bone marrow, lung, vascular, neuro, colon, ovarian, breast and prostate cancer, epitheleoid sarcomas, AIDS, adverse angiogenesis, restenosis, amyloidosis, tuberculosis, cardiovascular plaque, vascular plaque, obesity, malaria, and illnesses due to viruses, such as HIV.
  • The targeted therapy system, based on the principles of hyperthermia, comprises a bioprobe system in conjunction with an energy source to treat an indication. FIG. 1 discloses a bioprobe configuration according to an embodiment of the present invention. A bioprobe 190 comprises a magnetic energy susceptive particle 142, also referred to as a susceptor, and at least one targeting ligand 140, such as, but not limited to, an antibody, which may be located on an exterior portion of susceptor 142. The susceptor 142 may comprise a biocompatible coating 144. Coating material 144 may fully or partially coat susceptor 142. Heat is generated in susceptor 142 when susceptor 142 is exposed to an energy source, such as AMF. Coating 144 may enhance the heating properties of bioprobe 190, particularly if coating 144 has a high viscosity, for example, coating is a polymeric material. The coated susceptor is produced in accordance to the HPH processes and examples described herein. The targeting ligand 140 may be selected to seek out and attach to a target, such as a particular type of cell or disease matter. The targeting ligand 140 may be specific to a substance, such as an antigen, on the target, referred to as a marker.
  • The temperature to which susceptor 142 heats is dependent upon, inter alia, the magnetic properties of the material, characteristics of the magnetic field, and the cooling capacity of the target site. If bioprobe 190 contains a magnetite (Fe3O4) particle 142, then a diameter of susceptor 142 may be in the range of about 8 nm to about 80 nm, more specifically in the range of 10 nm and 40 nm. In this case, bioprobes 190 may be sufficiently small to evade the liver, and yet the magnetic particle 142 still retains a sufficient magnetic moment for heating in an applied AMF. Magnetite particles larger than about 8 nm generally tend to be ferrimagnetic and thus appropriate for disease treatment. If other elements, such as cobalt, are added to the magnetite, this size range can be smaller. This results directly from the fact that cobalt generally possesses a larger magnetic moment than magnetite, which contributes to the overall magnetic moment of cobalt-containing susceptor 142. In general, the size of bioprobe 190 may be about 0.1 nm to about 200 nm, depending upon the disease indication and materials from which the bioprobe is composed.
  • The presence of coating 144 and the composition of the coating material may form an integral part of the energy loss, and thus the heat produced, by bioprobe 190. In addition, coating 144 may serve additional purposes. Coating 144 may provide a biocompatible layer separating the magnetic material from proteins and enzymes as well as the immunologic defenses in a patient, thereby controlling the residence time of the nanoparticles in the blood or tissue fluids. This control of residence time allows one to select targeting ligands 140 that are best suited for a particular tissue type. In addition, coating 144 may serve to protect the patient from potentially toxic elements in susceptor 142. The coating 144 may also serve to prevent particle aggregation, as bioprobes 190 may be suspended in a fluid. The coating 144 may preserve the integrity of contained metal oxide to prevent further oxidation or chemical change, and thus may preserve the structural and magnetic properties of the oxide. It may also be advantageous to coat bioprobe 190 with a biocompatible coating that is biodegradable or resorbable. In such an application, both the coating 144 and the susceptor 142 may be digested and absorbed by the body.
  • The coating 144 may also serve to facilitate transport of bioprobe 190 into a cell, a process known as transfection. Such coating materials, known as transfection agents, may include vectors, prions, polyaminoacids, cationic liposomes, amphiphiles, non-liposomal lipids, or any combination thereof. A suitable vector may be a plasmid, a virus, a phage, a viron, or a viral coat. The bioprobe coating may comprise a combination of transfection agents with organic and/or inorganic materials, such that the particular combination may be tailored for a particular type of a diseased cell and a specific location within a patient's body.
  • To ensure that bioprobe 190 selectively attaches to, or otherwise associates with, the target, an appropriate ligand 140 may be combined with bioprobe 190. The association of a ligand or ligands with bioprobes 190 allows for targeting of cancer or disease markers on cells. It also allows for targeting biological matter in the patient The term ligand relates to compounds which may target molecules including, for example, proteins, peptides, glycans, antibodies, antibody fragments, saccharides, carbohydrates, cytokines, chemokines, lipids, nucleotides, lectins, receptors, steroids, neurotransmitters, imprinted polymers, Cluster Designation/Differentiation (CD) markers, and the like. Examples of protein ligands include cell surface proteins, membrane proteins, proteoglycans, glycoproteins, peptides, and the like. Example nucleotide ligands include complete nucleotides, complimentary nucleotides, and nucleotide fragments. Example lipid ligands include phospholipids, glycolipids, and the like. Ligand 140 may be covalently bonded to or physically interacted with susceptor 142 or coating 144. Ligand 140 may be bound covalently or by physical interaction to an uncoated portion of susceptor 142. Ligand 140 may be bound covalently or by physical interaction directly to an uncoated portion of susceptor 142 and partially covered by coating 144. Ligand 140 may be bound covalently or by physical interaction to a coated portion of bioprobe 190. Ligand 140 may be intercalated to the coated portion of bioprobe 190. Covalent bonding may be achieved with a linker molecule. Examples of functional groups used in linking reactions include amines, sulfhydryls, carbohydrates, carboxyls, hydroxyls, and the like. The linking agent may be a homobifunctional or heterobifunctional crosslinking reagent, such as carbodiimides, sulfo-N-hydroxy succinamide (NHS) ester linkers, and the like. The linking agent may also be an aldehyde crosslinking reagent, such as glutaraldehyde. The linking agent may be selected to link ligand 140 to susceptor 142 or coating 144 in a preferable orientation, specifically with the active region of the ligand 140 available for targeting. Physical interaction does not require that the linking molecule and ligand 140 be bound directly to susceptor 142 or to coating 144 by non-covalent means such as, for example, absorption, adsorption, or intercalation.
  • The targeting ligands, and their corresponding targets and markers for given indications are presented in Table I.
    TABLE I
    BIOPROBE SYSTEMS AND INDICATIONS
    BIOPROBE SYSTEM
    TARGET MARKER LIGAND INDICATION
    Endothelial cells of Integrin vβ3 Ber EP4 antibody Metastatic breast cancer, metastatic
    growing blood LM609 antibody colon carcinoma
    vessels of metastatic Integrin antagonist
    cancer cells
    Cancer cells Unglycosylated DF3 Anti-DF3 antibody Breast cancer
    antigen
    Cancer cells Kallikreins Anti-kallikrein Ovarian and prostate cancer
    antibody
    Cancer cells ErbB2 (HER-2/neu) Anti-ErbB2 antibody, Breast and ovarian cancers
    and scFv (F5), IDM-1
    (aka MDX-210)
    variants
    Cancer cells Prostate specific MDX-070 and 7E11- Prostate cancer
    membrane antigen C5.3 antibodies
    (PSMA)
    MCF-7 breast 43 Kd membrane 323/A3 antibody Breast cancer
    cancer cells associated
    glycoprotein
    Receptor tyrosine Vascular endothelial Anti-FLT1 antibody Tumour angiogenesis
    kinases- growth factor Anti-FLK1 antibody, Tumour angiogenesis
    FLT1 (VEGF) and VEGFB 2C3 antibody
    FLK1 and placental growth
    factor receptors
    (PGFR)
    Metastatic cancer CAR (coxsackie Anti-CAR antibody Metastatic prostate cancer
    cells adenovirus cell-
    surface receptor)
    Vascular smooth Urokinase type Urokinase type Cancer
    muscle cells of plasminogen plasminogen activator
    cancer cells activator receptor (uPA)
    (uPAR)
    Blood vessels of Plasminogen Anti-PAI-1 antibody Breast cancer
    cancer cells activator inhibitor
    1(PAI-1)
    Epithelial ovarian Matrix Anti-MMP-9 antibody Ovarian carcinomas with lymph node
    tumour cells metaloproteinase 9 metastasis.
    (MMP-9)
    Cancer cells Cyclin A Anti-cyclin A antibody Squamous cell carcinoma of the tongue
    Cancer cells Cyclin D Anti-cyclin D(1, 2, 3) Malignant breast cancer, head and neck
    antibody squamous cell carcinomas, mantle cell
    carcinomas, laryngeal squamous cell
    carcinomas
    Kidney cortex tissue Cyclin E Anti-cyclin E antibody Human renal cell carcinoma
    Tumorigenic human Cyclin E Anti-cyclin E antibody Breast cancer
    breast epithelial
    cells
    Malignant epithelial Cyclin E Anti-cyclin E antibody Transitional cell carcinoma of the
    bladder tissue urinary bladder
    Cancer cells Cdc 2 Anti-cdc 2 antibody Breast cancer
    Malignant epithelial P27 Anti-phospho p27 Transitional cell carcinoma of the
    bladder tissue antibody urinary bladder
    Cancer cells P73 Anti-p73 antibody Lung carcinogenesis, bladder
    carcinogenesis, neuroblastoma, breast
    cancer
    Cancer cells Ras Anti-ras antibody Breast cancer
    Cancer cells c-myc Anti C-myc antibody Breast cancer
    Cancer cells c-fms Anti-c-fms antibody Breast cancer
    Cancer cells Hepatocyte growth Anti-HGFR antibody Colorectal cancer
    factor receptor
    (HGFR)
    Cancer cells c-met Anti-c-met antibody Gastric and colon cancers, hepatomas,
    ovarian cancer, skin cancer
    Large granular Apoptosis related Anti-CD95 (Fas) Leukaemia, prostate cancer
    lymphocyte (LGL) factors: antibody
    leukaemia cells Fas
    FasL
    Cancer cells Non-receptor protein Anti c-src-polyclonal Metastatic colorectal cancer, and late
    tyrosine kinase V- antibody stage breast cancer
    Src and C-Src
    Cancer cell CAR (coxsackie Onyx-015 adenovirus Lung, ovarian, other cancers
    adenovirus cell-
    surface receptor)
    Cancer cell Epidermal growth Molecule 225 antibody Cancer
    factor receptor
    (EGFR)
    Cancer cells D6 antigen Anti-D6 antibody Vascular tumours including Kaposi's
    sarcoma
    Cancer cells 2C4 antigen Anti-2C4 antibody Breast, prostate, other cancers
    Cancer cells Cytokeratin S5A10-2 antibody Non-small cell lung cancer
    epithelial marker
    and/or telomerase
    reverse transcriptase
    Cancer cells Carcinoembryonic MFE-23 scFv of anti- Colorectal cancer
    antigen (CEA) CEA antibody
    Cancer cells Proliferating cell Anti-PCNA antibody Breast cancer
    nuclear antigen
    (PCNA)
    Cancer cells Neu 3, a membrane Anti-neu 3 sialidase Colon cancer
    associated sialidase antibody
    Cancer cells P13KC2 beta (cancer Anti-P13KC2beta Lung cancer
    cell signal mediator) antibody
    Cancer cells Guanylyl cyclase-C Anti-GC-C antibody Esophageal or gastric cancer
    (GC-C) receptor
    Cancer cells Transforming Anti-TGFB antibody Breast cancer
    growth factor beta
    (TGFB) receptor
    Cancer cells Platelet derived Anti-PDGF-A Lung cancer
    growth factor antibody Bone cancer
    receptor (PDGFR) Anti-PDGF-B antibody
    PDGFR-A (alpha)
    PDGFR-B (beta)
    Cancer cells and Vascular endothelial Tie1 Cancer
    blood vessels growth factors Tie2 Cancer
    VEGER
    Angiopoietin
    Cancer cells Mucin family of Anti-MUC-1 antibody, Colorectal and ovarian carcinomas
    receptors 12E antibody
    3D antibody
    A5 antibody
    Cancer cells TAG-72 B72.3 antibody Breast and lung cancers
    Cancer cells Human milk fat NCL-HMFG1 and Breast, lung, colon, and prostate
    globule receptor NCL-HMFG2 cancers
    antibodies
    Methionine synthase Cobalamin receptor B12 (riboflavin, and Breast, lung, colon, sarcomatous
    and L- variants) cobalamin thyroid or central nervous system
    methylmalonyl-CoA and variants such as malignancies cancer
    mutase adenosylcobalamin
    transcobalamin
    Cancer cells Glioma chloride Scorpion toxin- Gliomas
    channel chlorotoxin and
    chlorotoxin-like
    molecules
    Cancer cells 40 kD glycoprotein NR-LU-10 antibody Small cell lung cancer
    antigen
    CNS cells and tissue Brain-specific Anti-BEHAB antibody Gliomas
    chondroitin sulphate
    proteoglycan
    Brain enriched
    hyaluronan binding
    protein (BEHAB-
    aka brevican
    Cancer cells Catenins Anti-alpha catenin Colorectal carcinoma, non-small cell
    Alpha catenin antibody lung cancer
    Beta catenin Anti-beta catenin Breast cancer
    Gamma catenin antibody Thyroid cancer
    Anti-gamma catenin
    antibody
    Cancer cells Interleukin (IL) IL13-PE38 antibody Kidney, brain, breast, and head and
    receptors neck cancers, and Kaposi's sarcoma
    IL13 receptor
    Cancer cells Mesothelin receptor Anti-mesothelin Mesotheliomas
    antibody, and Ovarian cancer and mesotheliomas
    SS1(dsFv) variant
    Cancer cells CD44 surface Anti-CD44 antibody Prostate cancer
    adhesion molecule
    Cancer cells EGFRvIII Ua30:2 antibody Brain, colorectal, pancreatic, billary,
    L8A4 antibody liver cancers and soft tissue sarcomas.
    DH8.3 antibody
    81C6 antibody
    Receptor tyrosine Vascular endothelial Anti-FLT1 antibody Atherosclerotic plaques
    kinases FLT1 growth factor
    (VEGF) and VEGFB
    Smooth muscle cells Basic fibroblast Anit-bFGF antibody Restenosis
    in the lumen of growth factor
    blood vessels receptor (bFGFR)
    Vulnerable plaque Oxidized low density Oxidation-specific Atherosclerosis and vascular disease
    lipoprotein (OxLDL) antibodies (Ox-AB)
    MDA-2 antibody
    Vulnerable plaque Malondialdehyde- IK17 antibody Atherosclerosis and vascular disease
    modified LDL
    (MDA-LDL)
    M. Tuberculosis APA-antigen Anti-APA antibody Tuberculosis
    bacilli
    Retrovirus infected TGFA (alpha) Anti-TGFA antibody HIV
    cells
    Leukocytes Alpha4 subunit of Antegren Multiple sclerosis
    alpha4beta1-integrin
    (VLA-4) and
    alpha4beta7-integrin
    Receptor tyrosine Vascular endothelial Anti-FLT1 antibody Autoimmune joint destruction (arthritis,
    kinases FLT1 growth factor lupus, etc)
    (VEGF) and VEGFB
    Plasmodium Apical membrane Anti-AMA-1 antibody Malaria
    falciparum antigen-1 (AMA-1)
    Cells of the immune CD30 AC10, HeFi1, and Immunological disorders other than
    system derivatives of AC10 cancer
    and HeFi1
    Hepatitis C virus Hepatitis C virus 19D9D6 Monoclonal Hepatitis C infection
    core protein Antibody
    Tumor vascular cells Vascular endothelial MV833 and HuMV833 Cancer
    growth factor antibodies
    (VEGF)
    Tumor cells Cytokeratin Anti-cytokeratin Epitheleoid sarcomas
    AE1/3 and anti-
    CAM5.2 antibodies
    Tumor cells Thomsen M170, chimeric M170, Breast, Prostate, Ovarian, and Lung
    Friedenreich (TF) MaB 170H.82R1808 cancers
    antigen
    Tumor cells CEA HumaSpect ™, Colon and Ovarian cancers
    Votumumab,
    Mab 88BV59
    Tumor cells EFG-r ABX-EGF Colon, NSCLC, Prostate, and Renal
    cancers
    Tumor cells EGF-r HuMax-EGFr Head, Neck, Breast, Colon, Prostate,
    Lung, and Ovarian cancers
    Tumor cells EGF-r TheraCIM ™, h-R3 Head and Neck cancers
    Tumor cells CEA KSB309 ™ Oral cavity, and Pharngial cancers
    Tumor cells CEA 4B5-H Melanoma
    Tumor cells GD2 ganglioside ABX-MA1 Melanoma, Neuroblastoma, NSCLC
    Tumor cells CTLA4; CD152 MDX-010 Melanoma
    Tumor cells GD2 ganglioside TriGem, Mab-1A7 Melanoma
    Tumor cells CA125; MUC-16 ACA-125 Ovarian cancer
    Tumor cells Polymorphic R1549, Pemtumomab, Ovarian, Stomach, Breast, Lung, and
    epithelial mucin MuHMFg1, HuHMFg1 Prostate cancers
    Tumor cells CA125 OvaRex ™, Mab- Ovarian cancer
    B42.13, Ov
    Tumor cells VB2-011, H-11 ScFv, Breast, Ovarian, and Colorectal cancers
    Novo Mab-G2ScFv
    Tumor cells CEA CEA-Cide, Breast, Colon, and Lung cancers
    Labetuzumab
    Tumor cells VEGF Avastin ™, Breast, Colorectal, NSCLC, and Renal
    Bevacizumab, cancers
    rhuMAb-VEGF
    Tumor cells LewisY Ag SGN-15, cBR96 Breast, NSCLC, and Ovarian cancers
    Tumor cells HER2 OmniTag ™, Breast, Ovarian, Lung, and Prostate
    Pertuzumab, cancers
    rhuMAb 2C4
    Tumor cells MUC1 BrevaRex ™, Mab Breast, Ovarian, and Multiple Myeloma
    AR20.5 cancer
    Tumor cells MUC1 Therex ™, R1550, Breast, Ovarian, Pancreatic, and Gastric
    HuHMFG1 cancers
    Tumor cells Ep-CAM ING-1 Breast, Lung, Prostate, and Pancreatic
    cancers
    Tumor cells αvβ3 integrin Vitaxin ™, huLM609 Solid tumors
    Tumor cells αvβ3 integrin Mab-MEDI-522, Advanced solid tumors
    huLM609
  • Antibodies can also be attached on the surface of magnetic nanoparticles, e.g., by direct covalent binding of amino groups of the antibody molecule to carbodiimide activated carboxylic acid groups on the particle surface, by binding of biotinylated antibodies on streptavidin coated nanoparticles, and by binding of the antibodies on protein A or protein G coated nanoparticles. Nucleic acids can be immobilized on the surface of nanoparticles with covalently attached oligonucliotides.
  • In one embodiment, the targeting ligand is the ING-1 antibody. ING-1 (heMAb) is a high-affinity, human-engineered™ monoclonal antibody that recognizes a 40 kDa epithelial cell adhesion molecule (Ep-CAM—also known as 17-1A, EPG40, GA733-2, KSA and EGP). ING-1 was developed using human engineering, in which individual residues in the variable region of a murine antibody are replaced with the amino acid found in the human framework, such that immunogenicity of the variable region is reduced. ING-1 is produced from Chinese hamster ovary (CHO) cells containing synthetic heavy and light chain genes encoding the modified variable regions linked to human IgG1 and kappa constant region cDNA, respectively.
  • In vitro studies have demonstrated that ING-1(heMAb) is active against a number of human tumor cell lines, and that ING-1(heMAb) has high affinity for adenocarcinomas of the breast, prostate, colon, NSC lung and pancreas.
  • The treatment of a patient with the bioprobe compositions of the present invention involves the administration of the composition to a subject, and administration of energy to the bioprobes in the composition, after a prescribed period of time for the bioprobe to locate and attach to a markered target, so as to destroy or inactivate the target or inhibit or destroy the vascularity of a tumor. The energy may be administered directly into a subject's body, body part, tissue, or body fluid (such as blood, blood plasma, blood serum, or bone marrow), or extracorporeally to a subject's body, organ or body fluid. The bioprobe composition may be administered via injection, topical application, transdermal application, oral ingestion, rectal insertion, inhalation through the mouth or nose, or any combination thereof. The targeted thermotherapy may be combined with at least one other treatment.
  • Exemplary energy forms useful herein include AMF, radio frequency, microwave, acoustic, or a combination thereof, and may be created using a variety of mechanisms. The preferred energy form is AMF.
  • 3.2. Biological Applications
  • The magnetic nanoparticle compositions of the present invention are also useful for various biological applications, including, but not limited to, the fixation, separation, transportation, marking or coding of targets, or energy transformation processes. In one embodiment, the magnetic nanoparticles recognize, selectively fixate and controllably release metal ions, via ligands and chelators. In another embodiment, the nanoparticles are used as radionuclide carriers, wherein the nanoparticles are doped with radioactive isotopes or the nanoparticles are processed to render them radioactive, e.g., for use in radio-nuclide therapy. In another embodiment, the magnetic nanoparticles are used in the separation or purification of biomolecules, or any combination thereof. In such applications, the magnetic nanoparticle compositions may be used in the separation, purification, or any combination thereof of nucleic acids, nucleic acid fragments, nucleic acid derivatives, proteins, protein derivatives, protein fragments, or any combination thereof. In one embodiment, the magnetic nanoparticles bind to cells, and are used for the sorting and purification of cells. In another embodiment, the magnetic nanoparticles are used in analytical processes or diagnostic assays, for example, as contrast media.
  • EXAMPLES
  • Having generally described the invention, a more complete understanding thereof may be obtained by reference to the following examples that are provided for purposes of illustration only and do not limit the invention.
  • The Examples Described Herein Below Utilize the Following Analysis Techniques:
  • Photon correlation spectroscopy (PCS) is a technique used for measuring the random diffusion of particles to determine their size. PCS measurements lead to the determination of the average hydrodynamic diameter of the nanoparticles in suspension. The weighing of data analysis is carried out according to the signal intensity, the volume, and the number of the nanoparticles. There are three types of data analysis. The first, a monomodal analysis performs an analysis of only the cumulants. In the second, an analysis of the multimodal distribution is performed, resulting in 24 size classes on the X-axis. The Y scale of the plot yields the relative intensity of light scattered by particles in each size class. This is therefore known as an intensity plot. The third analysis method is known in the industry as CONTIN mode. This method provides the distributions and detects traces of aggregates. Therefore, the CONTIN analysis has been selected as the standard analytical method herein for determination of the size distribution from PCS data.
  • The quality of the size distribution is characterized by the polydispersity index (PI). This value is calculated as the deviation between the measured autocorrelation function and the mathematically adapted correlation function. This value is below 0.2 for nanoparticles with a small size distribution, and between 0.2 and 0.5 for broad size distributions. PI values higher than 0.5 indicate that the measurement is not reliable and that the data cannot be analyzed.
  • The instrument utilized for conducting PCS is Zetasizer 3000 (Malvern Instruments Ltd., Malvern, Worcestershire, U.K.), which is calibrated for the size range of 10 nm -1500 nm.
  • Neutron Powder Diffractometry (NPD) is a method to validate the magnetic form and crystalline structure of the particles and is a diffraction technique that is analogous to X-ray powder diffraction, with notable differences. Because the incident radiation is composed of neutrons, which interact with the atomic nuclei of the sample, the result is a nuclear diffraction pattern that provides average distances between atomic nuclei within a crystal. Because the neutrons possess a magnetic moment, this technique yields information about the magnetic properties of materials not available from its electromagnetic counterparts.
  • Laser Doppler Velocimetry (LDV) is used for measuring the movement of particles in an electric field to determine the zetapotential of particles. Zetapotential measurement of iron oxide particles provides information about the charge load of the particles.
  • The instrument utilized for conducting zetapotential measurements is Zetasizer 3000 (Malvern Instruments Ltd., Malvern, Worcestershire, U.K.), which is calibrated with the zetapotential standard DTS0050 (Malvern Instruments Ltd., Malvern, Worcestershire, U.K.).
  • Examples 1-4 and 8 herein below provide a detailed description of the synthetic procedures for producing nanoparticles, methods used for characterizing the nanoparticles, methods for functionalizing the nanoparticles, and methods used for conjugating ChL6 and ING-1 antibodies (which are used only as an exemplary system) to produce the final product for in vivo use. The general approach for the synthesis of the nanoparticle compositions in these examples is the core-shell method, as follows:
  • First, the initial iron oxide suspension was produced from precipitation of ferric and ferrous chlorides in the presence of ammonia (example 1). Second, the iron oxide cores were coated with dextran under high-pressure homogenization conditions (example 2). Third, the nanoparticles were functionalized with carboxylic acid groups via a polyethylene glycol spacer (example 3). And, fourth, the ChL6 antibody (example 4) or ING-1 antibody (example 8) was covalently bound to the particle surface.
  • Example 1 Precipitation and Analysis of Iron Oxide (Lot 146-T)
  • 54.1 g (0.2 mol) of FeCl3×6H2O and 31.4 g (0.158 mol) of FeCl2×4H2O were dissolved in 300 milliliter (ml) of de-ionized water. The solution of the iron salts was stirred with a mechanical stirrer at 500 revolutions per minute (rpm) in a 1 L two neck flask at room temperature. 220 ml of 25% ammonium hydroxide was added to the solution with a peristaltic pump over a period of 30 minutes (min). After addition of the ammonium hydroxide, the stirring process was continued for 10 min. The iron oxide suspension was purified via centrifugation in two 500 ml centrifuge beakers at 1100 rpm for 10 min. After removal of the supernatant, the iron oxide pellet was re-suspended two times in 200 ml of de-ionized water. The re-suspension was centrifuged three more times for successive neutralization of the iron oxide suspension. The iron oxide suspension was centrifuged again in two 500 ml beakers at 1100 rpm for 10 min. After removal of the supernatant, the iron oxide was re-suspended two times in 25 ml of de-ionized water and combined in one 500 ml beaker, followed by another centrifugation at 1100 rounds per minute (rpm) for 10 min.
  • The pellet was again re-suspended in 50 ml de-ionized water and centrifuged at 1100 rpm for 10 min. The supernatant was added to the product container. The process of re-suspension of the pellet in 50 ml of de-ionized water aliquot, centrifugation at 1100 rpm at 10 min, and addition of the obtained supernatants was repeated two more times to produce an overall yield of 200 ml iron oxide suspension. For final removal of larger aggregates, the iron oxide suspension was centrifuged at 700 rpm for 10 min. The supernatant was filtered through a glass-fiber filter (MN 85/90, Macherey-Nagel, Germany; size exclusion: greater than 600 nm). Finally, 200 ml iron oxide suspension of Lot 146-T, with a solids concentration of 45 milligrams per milliliter (mg/ml) was obtained (with a solid iron oxide yield of 9 g).
  • A representative plot of the size distribution obtained for Lot 146-T particles is shown in FIG. 2 a. The peak analysis by particle number is consistent with size determinations by transmission electron microscopy.
  • The PCS size measurement of the iron oxide suspension of Lot 146-T leads to a mean hydrodynamic diameter of 400 nm-450 nm. The polydispersity index of 0.227 shows a monomodal size distribution. Thus, the individual iron oxide crystals have aggregated in suspension. This effect has implications for further processing, namely coating with dextran. Before the precipitates can be coated with dextran to yield the nominal nanoparticle that forms the bioprobe, the precipitated particles must be re-suspended and stabilized. Other characterization techniques must be employed to determine the nominal crystal size. For example, zetapotential measurement of iron oxide particles provides information about the charge loaded to the particles. In order to make these particles biocompatible, it is essential that the charge of the particles is almost, and ideally, zero. The Zetasizer 3000 was combined with an autotitrator (available from Mettler-Toledo, Inc., Columbus, Ohio) to determine the zetapotential dependence on the solution pH. The nanoparticle suspension was first titrated with 0.1 M of sodium-hydroxide (NaOH) to increase the pH value to about 10. Then, the zetapotential-pH profile was measured during a titration with 0.1 M of hydrogen chloride (HCl) from pH=9 to pH=3.
  • The zetapotential-pH function of the iron oxide nanoparticles of Lot 146-T is shown in FIG. 3. We observe a significant influence of the pH value on the zetapotential. In the basic to neutral pH range, the nanoparticles have strongly negative surface potentials in the range of −43 millivolt (mV) to −29 mV. After passing the isoelectric point at pH=6.1, the nanoparticles show high positive surface potentials of +27 mV to +35 mV in the acidic pH range from 5 to 3.
  • The magnetic properties of Lot 146-T iron oxide nanoparticles were analyzed using a variety of standard magnetic techniques. Saturation magnetization (Ms), magnetic coercivity (Hc), and remanence ratio (ratio of magnetic remanence to saturation magnetization (Mr,Ms)) were measured. These magnetic measurements are indicators of the dominant magnetic properties of the particles, and can be used to determine the relative concentrations of magnetite (Fe3O4) and maghemite (γ-Fe2O3). These magnetic measurements and the determined magnetite content of the particles are presented in Table II.
    TABLE II
    PROPERTIES OF MAGNETIC NANOPARTICLES
    Category of
    Sample/ Ms (300° K) Hc % Mag- Tb magnetic
    Lot ID (emu/mg-Fe) (Oe) Mr/Ms netite (Kelvin) properties
    146-T 0.1085 <5 0.0261 85.4 48 super-
    paramagnetic
  • The standard value of saturation magnetization for pure magnetite is 0.1271 emu/mg-Fe, and for pure maghemite is 0.1087 electromagnetic unit (emu)/mg-Fe. The remanence ratio provides a measure of magnetic domain character to the particles, on average, i.e., single or multi-domain. The theoretical value for a magnetic nanoparticle that exhibits stable single-domain properties is Mr/Ms=0.5. Superparamagnetic (SP) behavior is observed when the low-temperature remanence curves have a blocking temperature (Tb) distribution with “magnetic” particle sizes less than 30 nm.
  • Magnetite will display a Verwey transition near 100° K. on low-temperature remanence curves as the domains transition from blocked to unblocked behavior. On the other hand, maghemite has no Verwey transition. If magnetite particles show superparamagnetic behavior, then it is usually difficult to see the magnetic effects of the Verwey transition over the thermal unblocking effect of SP in this type of experiment. In this case, no Verwey transition was observed.
  • In preparation for Neutron Powder Diffractometry, the Lot 146-T nanoparticle suspensions were dried in a vacuum oven for several days. This is necessary to remove all traces of water, which, because of the scattering cross section of hydrogen, produces a high background to the data. For this study, measurements were conducted using the neutron powder diffractometer located at the Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Md. The Si(531) monochromator was used because it provides the best resolution at high scattering angles, although longer data collection times are needed, often from 4 hour (h) to 24 h. This monochromator has a takeoff angle of 1200 and produces neutrons with wavelength 1.590 angstrom (Å). Diffraction intensities are approximately 30% of that obtained with the Cu(311) monochromator. With 7′ collimation, perfectly Gaussian line shapes with widths full width at half maximum (FWHM) as small as 10′ (delta d/d=8×10−4, where d is the line width) are observed, but for most samples the intrinsic sample broadening exceeds the instrument resolution. Thus, use of the Si(531) monochromator is restricted to those applications where highest resolution is clearly needed and where samples are available in sufficient quality and quantity.
  • Based on the measurements, we conclude that the mean crystal size of the nanoparticles is about 12 nm and that the sample consists mostly of magnetite, and that the mean magnetic moment of the octahedrally coordinated Fe atoms in the crystalline lattice is 4.4 Bohr magnetons, while that of the tetrahedrally coordinated atoms is 3.9 Bohr magnetons. This latter result is consistent with the conclusions arrived from the magnetic measurements, i.e., that about 20% maghemite exists in the crystals.
  • The mean crystalline size obtained from NPD measurements also helps explain the lack of an observed Verwey transition in the magnetic properties, the low coercivity, and low remanence ratio. Together, these results demonstrate that the dominant SP behavior of the nanoparticles is due to their small size (<<30 nm) and not to the composition consisting of a significant maghemite component.
  • Example 2 Synthesis and Characterization of Dextran-Coated Nanoparticles (Lot 152-W)
  • 21 g of dextran (molecular weight (MW)=40.000 Daltons, available from Carl Roth, GmbH, Karlsruhe, Germany) was dissolved in 100 ml of de-ionized water. 75 ml of iron oxide suspension from Lot 146-T (concentration=45 mg/ml) was homogenized in a high-pressure-homogenizer (Fluidizer Y110, Microfluidics, Inc., Newton, Mass.) at 500 bar for 7 min. This is a pressured, air driven plunger pump that drives the fluid through a 100 micrometer (μm) diamond gap in order to force a high shear stress. The pre-heated dextran solution (about 45° C.) was added to the suspension, warmed in the homogenizer to 90° C. at 1000 bar and processed at 1000 bar for 30 min at a temperature between 87° C.-92° C. After cooling to room temperature, the nanoparticle suspension was transferred into a crystallizing dish (diameter: 12 centimeter (cm)) and placed on a permanent magnet plate for 1 hour. Then, the supernatant was transferred to a second crystallizing dish (diameter: 12 cm) and placed on a permanent magnet plate for 16 hours. The pellet from the first crystallizing dish was re-suspended in 20 ml of isotonic saline to yield nanoparticle fraction of Lot 152-W1. After 16 hours of magnetic separation, the supernatant from the second crystallizing dish was removed and stored. The pellet was re-suspended in 20 ml of isotonic saline to yield nanoparticle fraction of Lot 152-W2.
  • The remaining supernatant was utilized to produce the nanoparticle fraction of Lot 152-W3 by separation of the nanoparticles in a high gradient magnetic field. Columns filled with coated iron grains were introduced into a permanent magnetic field. The supernatant was filled into the columns. The magnetic particles were retained in the column. The non-magnetic material, especially the excess dextran, was passed through the column. After washing the particles with isotonic saline, the column was removed from the magnetic field, releasing the nanoparticles from the column, and the nanoparticles were re-suspended in isotonic saline. The process flow chart for lots 152-W1, 152-W2 and 152-W3 is shown in FIG. 4. All particle fractions were filtered through sterile filters (Rotilabo®, 0.22 μm, PES, P668.1; available from Carl Roth GmbH). The particle yield of the single fractions was determined by gravimetry. The iron concentration of all fractions was measured spectrophotometrically using the Spectroquant®-Kit (Merck & Co., Inc., Whitehouse Station, N.J.; VWR International GmbH, Darmstadt, Germany). The properties of the magnetic nanoparticles are presented in Table III.
    TABLE III
    PROPERTIES OF MAGNETIC NANOPARTICLES
    Particle Iron Weight % Particle
    Volume concentration concentration (w/w) yield
    Lot ID. [ml] [mg/ml] [mg/ml] iron oxide [g]
    152-W1 35 45 22 67 1.57
    152-W2 20 34 17 69 0.68
    152-W3 110 16 5 43 1.76
  • Size characterization: The hydrodynamic diameters of the nanoparticle fractions of lot 152-W were measured via PCS (see FIG. 2 b and FIG. 2 c). The resulting data is presented in Table IV.
    TABLE IV
    DIAMETER OF MAGNETIC NANOPARTICLES
    Mean diameter [nm] Mean diameter [nm] Mean diameter [nm]
    (Analysis by (Analysis by (Analysis by
    Lot ID. intensity) volume) number)
    152-W1 109.1 79.3 63.8
    152-W2 82.0 56.8 48.5
    152-W3 50.4 22.2 16.6
  • The zetapotential measurement of dextran-coated iron oxide nanoparticles of Lot 152-W is shown in FIG. 3. The comparison of the zetapotential-pH functions of the initial iron oxide particles of Lot 146-T and the corresponding dextran-coated particles shows a significant decrease over the entire pH range of 3-9. The isoelectric point of the iron oxide suspension at pH=6.1 was shifted to 5.4 due to the dextran coating under HPH conditions.
  • Example 3 PEG-Carboxylation of Dextran-Coated Nanoparticles (Lot 202-G)
  • 6 ml of a suspension of dextran-coated nanoparticles of 152-W-type (example 2) with a particle concentration of 25 mg/ml in 0.1 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3) was treated with increasing amounts of carbodiimide activated 3,6-dioxaoctanedioic acid to yield the lots of nanoparticles 202-G1-202-G4 with various PEG-COOH densities, as presented in Table V.
    TABLE V
    THEORETICAL PEG-COOH DENSITY OF MAGNETIC
    NANOPARTICLES
    Lot ID. of Amounts of EDC, and 3,6-
    PEG-COOH dioxaoctanedioic acid [mg Theoretical PEG-COOH
    particles per 150 mg particle] density [nmol/mg]
    202-G1 18 625
    202-G2 3.6 125
    202-G3 0.72 25
    202-G4 0.144 5
  • Equal amounts (as shown in Table V) of 1-(3-dimethyl-aminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) and 3,6-dioxaoctanedioic acid were dissolved in 1.5 ml of 0.5 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3), incubated at 50° C. for 10 min, and added to the particle suspension. After shaking the particle suspension for 2 hours at room temperature, the particles were washed twice with water after separation from the solution using a permanent magnet, resulting in a 5 ml of suspension of PEG-carboxylated nanoparticles with a concentration of about 20 mg/ml. The zetapotential-pH profile was determined for all lots of PEG-COOH modified nanoparticles by titration with 0.1 M NaOH to pH=10, and then with 0.1 M HCl to pH=3.
  • In general, the PEG-COOH modification leads to higher negative zetapotentials of the nanoparticles in the basic pH range and higher positive zetapotentials in the acidic pH range. From the zetapotential-pH functions of particles with increasing PEG-COOH densities, the range of saturation of the particle surface with functional groups can be derived by achieving constant zetapotentials at theoretical PEG-COOH densities greater than 125 nanomol (nmol)/mg. Thus, the nanoparticles of lots 202-G3 and 202-G4 show only low to medium coverage with PEG-COOH groups (as shown in FIG. 5). The isoelectric point of the nanoparticles changed from pH=5.9 for the particles 202-G4 with the lowest density of PEG-COOH on the surface to the acidic direction to pH=4.8 for corresponding particles 202-G1 with the maximum saturation of the particle surface with PEG-COOH groups (as shown in FIG. 6).
  • Example 4 Covalent Binding of ChL6 Antibody on the Surface of Dextran-Coated Nanoparticles (Lot 206-G)
  • 2.4 mg (12.6 mmol) of 1-(3-dimethyl-aminopropyl)-3-ethylcarbodiimide hydrochloride and 4.8 mg (42 mmol) of N-hydroxysuccinimide were dissolved in 1.25 ml of 0.5 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3) and added to 5 ml of particle suspension containing PEG-COOH functionalized of lot 202-G2 (example 3) with a concentration of 20 mg/ml. After shaking the particle suspension for 2 hours, the particles were washed with 5 ml of phosphate buffered saline (pH=7.4) after separation from the solution using a permanent magnet, and suspended in 5 ml of phosphate buffered saline (pH=7.4). 1 mg of ChL6 antibody (obtained from UC-Davis) was dissolved in 1 ml of phosphate buffered saline (pH=7.4) and added to the particle suspension. After shaking the particle suspension for 4 hours at room temperature, the remaining active sites on the particle surface were blocked by addition of 2 ml of 0.3 M glycine in phosphate buffered saline (pH=7.4). After shaking the particle suspension for further 30 min at room temperature, the particles were washed three times, each time with 5 ml of phosphate buffered saline (pH=7.4) after separation from the solution with a permanent magnet, resulting in a 5 ml suspension having a particle concentration of 7 mg/ml. The average hydrodynamic diameter of the ChL6 coated nanoparticles (Lot 206-G) was determined by PCS to be in the range of 45 nm-55 nm. The iron content in the particles was determined to be in the range of 56% to 60% (w/w).
  • Example 5 Synthesis of Carboxy-Dextran-Coated Nanoparticles (Lot 200-W)
  • Analogous to example 2, the iron oxide described in example 1 was coated with carboxydextran (available from Innovent e.V., Jena, Germany) instead of dextran to study the influence of carboxylic acid groups, which are already present in the initial dextran structure, on the integrity and density of the dextran shell. The resulting nanoparticles of Lot 200-W have a COOH density of 180 nmol/mg (determined via polyelectrolyte titration) and can be directly coated with antibodies according to the procedure described in example 4.
  • The examples 6-8 provide a detailed description of the synthetic procedures for the production of iron oxide nanoparticles by high pressure homogenization (HPH) (example 6), for the production of dextran coated nanoparticles from HPH-produced iron oxide (example 7), for the functionalization of nanoparticles, and methods used for conjugating ING-1 antibody to produce the final product for in vivo application (example 8).
  • Example 6 Synthesis of Iron Oxide Nanoparticles by High Pressure Homogenization (Lot 176-W)
  • 13.5 g of FeCl3*6H2O and 7.85 g of FeCl2*4H2O were dissolved in 250 ml of water. The temperature of this solution was increased to 80° C. The iron salt solution was filled into the reservoir of the Microfluidizer M-110Y and homogenized at a pressure of 1000 bar. 250 ml of 2 N sodium hydroxide solution was heated to 80° C. and added to the iron salt solution while stirring. The homogenization was carried out at 90° C. for 30 min. The resultant suspension was then cooled to room temperature. The iron oxide suspension was neutralized by successive centrifugation and washed with water to yield a stable aqueous iron oxide colloid. The average hydrodynamic diameter of the resulting nanoparticles was determined (via PCS) to be in the range of 200 nm-500 nm.
  • Example 7 Synthesis and Characterization of Dextran-Coated Nanoparticles from HPH-Produced Iron Oxide (Lot178-W)
  • 21 g of dextran (MW=40,000 D, Fluka) was dissolved in 100 ml of water and heated to 50° C. 200 ml of iron oxide suspension from Example 6 containing 2.6 g of iron oxide was placed in the Microfluidizer M-110Y and processed at 700 bar for 10 min. While stirring, the pre-heated dextran solution was added to the suspension, and the homogenization process was continued for an additional 40 min at a pressure of 1000 bar. The temperature during this process was maintained the range of 85° C.-90° C. The resultant suspension was cooled to room temperature. The excess dextran was removed by magnetic separation of the nanoparticles, and the nanoparticles were re-suspended in water. The average hydrodynamic diameter of the resulting nanoparticles was determined (via PCS) to be in the range of 20 nm-80 nm. The iron content of the nanoparticles was determined to be in the range of 45% to 55% (w/w).
  • Example 8 ING-1 Antibody Binding on Dextran Coated Nanoparticles (Lot 07304-G)
  • 6 g of dextran was dissolved in 20 ml of water. 22 ml of dextran coated nanoparticles of Lot 178-W (example 7) having a particle concentration of 46 mg/ml was added to the dextran solution. The particle suspension was heated to 100° C., with a rotation speed of 400 rpm for 1 hour. After cooling to room temperature, the magnetic particles were separated using a permanent magnet to remove the excess dextran from the supernatant. The particles were suspended in 20 ml of water to yield a particle concentration of 58 mg/ml.
  • 2.6 ml of this particle suspension was diluted with 2.4 ml of water to a particle concentration of 30 mg/ml. 3.6 mg (18.8 mmol) of 1-(3-dimethyl-aminopropyl)-3-ethylcarbodiimide hydrochloride and 3.6 mg (20.2 μmol) of 3,6-dioxaoctanedioic acid were dissolved in 1.25 ml of 0.5 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3), incubated at 50° C. for 10 min, and added to the particle suspension. After shaking the particle suspension for 2 hours at room temperature, the particles were washed twice with 5 ml of 0.1 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3) after separation from the solution using a permanent magnet, resulting in a 5 ml suspension of PEG-carboxylated nanoparticles with a concentration of 10 mg/ml.
  • 6 mg (31.4 μmol) of 1-(3-dimethyl-aminopropyl)-3-ethylcarbodiimide hydrochloride and 12 mg (104 μmol) of N-hydroxysuccinimide were dissolved in 1.25 ml of 0.5 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3), and added to the particle suspension. After shaking the particle suspension for 2 hours, the particles were washed with 5 ml of phosphate buffered saline (pH=7.4) after separation from the solution using a permanent magnet, and suspended in 4 ml of phosphate buffered saline (pH=7.4). 500 μg of ING-1 antibody (obtained from XOMA Inc., Oakland, Calif.) was dissolved in 200 μl of phosphate buffered saline (pH=7.4), and added to the particle suspension. After shaking the particle suspension for 4 hours at room temperature, remaining active sites on the particle surface were blocked by addition of 2 ml of 0.3 M glycine in phosphate buffered saline (pH=7.4). After shaking the particle suspension for further 30 min at room temperature, the particles were washed three times, each time with 5 ml of phosphate buffered saline (pH=7.4) after separation from the solution using a permanent magnet, resulting in a 5 ml suspension having a particle concentration of 8 mg/ml. The average hydrodynamic diameter of the ING-1 coated nanoparticles (Lot 07204-G) was determined by PCS to be in the range of 60 nm-70 nm. The iron content in the nanoparticles was determined to be in the range of 58% to 60% (w/w).
  • Example 9 Synthesis of Oligo-dT20 Modified Dextran-Coated Nanoparticles (Lot 208-G)
  • 6 mg (31.4 μmol) of 1-(3-dimethyl-aminopropyl)-3-ethylcarbodiimide hydrochloride and 12 mg (104 μmol) of N-hydroxysuccinimide were dissolved in 1.25 ml of 0.5 M β-morpholino ethanesulfonic acid hydrate buffer (pH=6.3) and added to 5 ml of a suspension of PEG-carboxylated nanoparticles having a concentration of 10 mg/ml (from example 8). After shaking the particle suspension for 2 hours at room temperature, the particles were washed with 5 ml of imidazol buffer (pH=7.0) after separation from the solution using a permanent magnet, and suspended in 4 ml of imidazol buffer (pH=7.0). 200 nmol of oligo-dT20 C6 amino link (5′) (available from Thermo Electron Corporation, Ulm, Germany) was dissolved in 200 μl of imidazol buffer (pH=7.0) and added to the particle suspension. After shaking the particle suspension for 4 hours at room temperature, the particles were washed three times, each time with 5 ml of 10 mM Tris buffer (pH=7.5) and 500 mM of KCl for further binding of nucleic acids. The average hydrodynamic diameter of the oligo-dT20 coated nanoparticles (Lot 208-G) was determined by PCS to be in the range of 55 nm-65 nm. The iron content in the nanoparticles was determined to be in the range of 58% to 60% (w/w).
  • Example 10 Calorimetric Measurements of Nanoparticle SAR Values
  • Equipment utilized:
      • TIG 10/300 RF power supply with tank circuit (Huttinger Electronic Inc., Farmington, Conn.)
      • RF coil (solenoid) having 8 turns, 0.75″ ID, and 2.2″ length (Triton BioSystems, Inc., Chelmsford, Mass.)
      • UMI-4 temperature probe controller with FOT-M/2M temperature probe and charting software (Fiso, Inc., Quebec, Canada)
      • TDS-3014 Oscilloscope (Tektronix, Inc., Beaverton, Oreg.)
      • Litz wire magnetic probe (Centre for Induction Technology, Inc., Auburn Hills, Mich.)
  • The apparatus was calibrated by setting the power supply at 20, 40, 60, 80, 90 and 100% of output voltage. At each setting, the peak magnetic field strength was measured at the center of the coil using the litz wire probe and oscilloscope. The field was also measured at small axial and radial offsets and it was determined the field was uniform within 10% throughout the planned 1 ml sample volume. The peak field strengths were charted versus the power supply's percent voltage output, and this curve allowed the presetting of field strength via the power supply's voltage readout. A calorimetric calibration was performed as well. This was accomplished by running de-ionized (DI) water samples immediately before SAR testing at each planned field strength, and following the same heating procedure. The temperature curves from the DI samples were subtracted from the SAR samples, thereby taking into account the heat losses (or additions) through the insulation.
  • After thorough vortexing, 1 ml samples were dispensed into 8 mm glass test tubes. In some cases they were diluted with de-ionized water to keep the heating rate within easily measurable limits. (In all cases the sample size was maintained at 1 ml.) The test tube was vortexed, wrapped in ceramic felt insulation and placed in the solenoid coil. It was placed on a glass pedestal such that the liquid sample was axially and radially centered within the coil. The temperature probe was immersed in the sample and the charting software started. At 10 seconds the RF was applied and at 60 seconds the RF was curtailed. After subtracting the DI calibration curve from the raw warm-up data, a characteristic heating curve resulted. The slope of this curve and the particle and iron concentrations were entered into the following formulae;
    SAR particle(W/gm)=4180*slope (° C./sec)/particle concentration (mg/ml)
    SAR iron(W/gm)=4180*slope (° C./sec)/iron concentration (mg/ml)
  • These SAR values were used to optimize the heating characteristics of the iron oxide nanoparticles. The SARiron values for Lots 146-T, 152-W1, 152-W2, and 178-W are presented in Table VI.
    TABLE VI
    SARiron VALUES FOR PARTICLES
    PRODUCED BY HPH PROCESS
    SARiron of A. SARiron of SARiron of SARiron of
    Lot 146-T B. Lot 152-W1 Lot 152-W2 Lot 178-W
    Field (W/gmiron) (W/gmiron) (W/gmiron) (W/gmiron)
    370 188 216 252 179
    730 234 273 314 602
    1080 211 287 321 1311
    1300 151 301 358 2090
    1670 271 NA NA NA
  • The HPH process (as illustrated via Examples 1-7) was used for enhancing the heating rates of coated nanoparticles, particularly the dextran-coated iron oxide nanoparticles, which exhibit heating when exposed to alternating magnetic field (AMF) amplitude of about 370 to about 500 Oe. First, a magnetic material having an Fe(II)/Fe(III)-ratio=1.58:2.0 is selected. This material exhibits constant heating rates when exposed to an AMF amplitude of about 370 to about 1600 Oe (when produced via iron oxide precipitation without HPH process). The iron oxide is cleaned, and HPH process is utilized to coat the nanoparticles with dextran. Due to this process, the heating rates of the nanoparticles are enhanced and occur when exposed to AMF amplitudes of about 370 to 500 W/g Fe with SAR values of 240 W/g Fe or greater.
  • The compositions of the present invention are applicable to human subjects (patient), as well as mammals, organ donors, cadavers and the like.
  • As noted above, the present invention pertains to biocompatible magnetic nanoparticle compositions prepared via high-pressure homogenization processes for various therapeutic and biological applications. The present invention should not be considered limited to the particular embodiments described above, but rather should be understood to cover all aspects of the invention as fairly set out in the appended claims. Various modifications, equivalent processes, as well as numerous structures to which the present invention may be applicable will be readily apparent to those skilled in the art to which the present invention is directed upon review of the present specification. The claims are intended to cover such modifications and devices.

Claims (66)

1. A method for preparing a magnetic nanoparticle composition, comprising:
a. generating a metal-containing magnetic material by processing a preformed metal-containing magnetic material through a turbulent flow zone in a first step, and generating a magnetic nanoparticle composition using the resulting improved magnetic material of the first step and a biocompatible coating material, via a turbulent flow zone, in a second step; or
b. generating a metal-containing magnetic material from a metal-containing solution via a turbulent flow zone in a first step, and generating a magnetic nanoparticle composition using the resulting magnetic material of the first step and a biocompatible coating material, via a turbulent flow zone, in a second step; or
c. forming a magnetic nanoparticle composition by generating a metal-containing magnetic material from a metal-containing solution and processing it with a biocompatible coating material, via a turbulent flow zone, in a single step.
2. A method according to claim 1, wherein in the first and the second steps are carried out in a liquid medium comprising water, or an aqueous alkaline solution having an ammonia sodium or potassium hydroxide basis.
3. A method according to claim 1, wherein the preformed metal-containing magnetic material comprises one or a combination of two or more of a) different metals, b) different metal compounds, c) metal with different valences, or d) any combination thereof.
4. A method according to claim 3, wherein the preformed metal-containing magnetic material comprises a combination of Fe-(II) and Fe-(III) in a molar ratio of 1:1 to 1:2.
5. A method according to claim 1, wherein the metal-containing solution is prepared from a metal, metal salt, metal alkoxyde, metal hydroxide, metal oxide, metal oxide-hydrate, metallic alloy of two or more metals, or any combination thereof.
6. A method according to claim 5, wherein the metal-containing solution comprises a combination of Fe-(II) and Fe-(III) salts in a molar ratio of 1:1 to 1:2.
7. A method according to claim 1, wherein the generated metal-containing magnetic material comprises a metal, metal salt, metal alkoxyde, metal hydroxide, metal oxide, metal oxide-hydrate, metallic alloy of two or more metals, or any combination thereof.
8. A method according to claim 7, wherein the metal oxide is iron oxide.
9. A method according to claim 8, wherein the iron oxide is magnetite, hematite, maghemite, or any combination thereof.
10. A method according to claim 8, wherein the iron oxide is doped with bi- or tri-valent metal ions.
11. A method according to claim 1, wherein the biocompatible coating material comprises a polymer, metal compound, transfection agent, or any combination thereof.
12. A method according to claim 11, wherein the polymer is naturally occurring, synthetic, or semi-synthetic.
13. A method according to claim 11, wherein the polymer comprises at least one reactive or ionic group, or any combination thereof.
14. A method according to claim 11, wherein the polymer comprises a homo-polymer, a co-polymer, or a polymer-blend.
15. A method according to claim 11, wherein the polymer comprises a biological material.
16. A method according to claim 15, wherein the polymer comprises a polysaccharide, polyamino acid, protein, lipid, fatty acid, heparin, heparin sulfate, chondroitin sulfate, chitin, chitosan, alginate, glycosaminoglycan, cellulose, starch, histidine-containing polymer, hydrogel polymer, any derivative thereof, or any combination thereof.
17. A method according to claim 16, wherein the polysaccharide is dextran.
18. A method according to claim 17, wherein the dextran comprises one or more functional groups.
19. A method according to claim 12, wherein the synthetic polymer comprises a polyvinyl compound, polyamine, polyimine, polyol, polyether, polycarboxylic acid, polysilicic acid, polyacrylate, polysiloxane, polyalkylene glycol, parylene, polylactic acid, polyglycolic acid, or any derivative thereof, or any combination thereof.
20. A method according to claim 2, wherein the first and the second steps are processed in a liquid carrier medium at a pressure above 100 bar.
21. A method according to claim 2, wherein the first and the second steps are processed in a liquid carrier medium at a pressure above 1000 bar.
22. A method according to claim 1, wherein the first and the second steps are processed in a liquid carrier medium at a temperature in the range from about 40° C. and the boiling point of the medium.
23. A method according to claim 1, wherein the first and the second steps are processed in a liquid carrier medium at a temperature in the range from about 75° C. to about 95° C.
24. A method according to claim 1, wherein the components are processed at a flow rate in the range from about 20 ml/min to about 200 ml/min through a turbulent flow zone in each step.
25. A method according to claim 2, wherein the resulting magnetic nanoparticles form a stable aqueous colloid.
26. A method according to claim 2, wherein the resulting magnetic nanoparticles form a stable colloid in physiological solution.
27. A method according to claim 2, wherein the resulting magnetic nanoparticles are separated from the carrier medium via an external magnetic field.
28. A method according to claim 27, wherein the resulting magnetic nanoparticles are separated with permanent magnets.
29. A magnetic nanoparticle composition, comprising:
a. at least one metal-containing magnetic nanoparticle possessing a low-field magnetization when an external magnetic field is applied to the at least one magnetic nanoparticle; and
b. a suitable medium for suspending the at least one nanoparticle,
wherein the at least one nanoparticle has a hydrodynamic diameter less than 200 nm, and contains at least 50 mass percent of metal, and
wherein the at least one metal-containing magnetic nanoparticle comprises a biocompatible coating material.
30. A magnetic nanoparticle composition according to claim 29, wherein the external magnetic field has an amplitude in the range from about 0 to about 400 Oersted.
31. A magnetic nanoparticle composition according to claim 29, wherein the nanoparticle composition has a PEG density in the range from about 2 μmol/g to about 250 μmol/g.
32. A magnetic nanoparticle composition according to claim 29, wherein the at least one magnetic nanoparticle comprises a) a metal, b) a metal oxide, c) a metal oxide-hydrate, d) a metal hydroxide, e) a metallic two or more metals, or f) any combination thereof.
33. A magnetic nanoparticle composition according to claim 32, wherein the at least one magnetic nanoparticle comprises an oxide of iron.
34. A magnetic nanoparticle composition according to claim 33, wherein the iron oxide is magnetite, hematite, maghemite, or any combination thereof.
35. A magnetic nanoparticle composition according to claim 33, wherein the iron oxide is doped with bi- or tri-valent metal ions.
36. A magnetic nanoparticle composition according to claim 29, wherein the at least one magnetic nanoparticle has ferro-, antiferro-, ferri-, antiferri- or superparamagnetic properties.
37. A magnetic nanoparticle composition according to claim 29, wherein the biocompatible coating material comprises a polymer.
38. A magnetic nanoparticle composition according to claim 37, wherein the polymer is naturally occurring, synthetic, or semi-synthetic.
39. A magnetic nanoparticle composition according to claim 37, wherein the polymer comprises a homo-polymer, a co-polymer, or a polymer-blend.
40. A magnetic nanoparticle composition according to claim 37, wherein the polymer comprises at least one reactive or ionic group, or any combination thereof.
41. A magnetic nanoparticle composition according to claim 37, wherein the polymer comprises a biological material.
42. A magnetic nanoparticle composition according to claim 37, wherein the polymer comprises a polysaccharide, polyamino acid, protein, lipid, fatty acid, heparin, heparin sulfate, chondroitin sulfate, chitin, chitosan, alginate, glycosaminoglycan, cellulose, starch, histidine-containing polymer, hydrogel polymer, any derivative thereof, or any combination thereof.
43. A magnetic nanoparticle composition according to claim 42, wherein the polysaccharide is dextran.
44. A magnetic nanoparticle composition according to claim 43, wherein the dextran comprises one or more functional groups.
45. A magnetic nanoparticle composition according to claim 38, wherein the synthetic polymer comprises a polyvinyl compound, polyamine, polyimine, polyol, polyether, polycarboxylic acid, polysilicic acid, polyacrylate, polysiloxane, polyalkylene glycol, parylene, polylactic acid, polyglycolic acid, or any derivative thereof, or any combination thereof.
46. A magnetic nanoparticle composition according to claim 29, wherein the magnetic nanoparticle composition comprises one or more sub-structures.
47. A magnetic nanoparticle composition according to claim 46, wherein the sub-structures comprise at least one ligand, chelator, or a combination thereof.
48. A magnetic nanoparticle composition according to claim 47, wherein the at least one ligand or chelator comprises a peptide, protein, nucleic acid, enzyme, antibody, antibody fragment, or any combination thereof.
49. A magnetic nanoparticle composition according to claim 48, wherein the antibody is ING-1.
50. A magnetic nanoparticle composition according to claim 46, wherein the sub-structures comprise a bioactive substance.
51. A magnetic nanoparticle composition according to claim 50, wherein the bioactive substance comprises a pharmaceutical agent, peptide, lipid, biochemical factor, or any combination thereof.
52. A magnetic nanoparticle composition produced according to the method of claim 1.
53. A magnetic nanoparticle composition according to claim 52, wherein the magnetic nanoparticle composition comprises one or more sub-structures.
54. A magnetic nanoparticle composition according to claim 29, wherein the composition is used in the fixation, separation, transportation, marking or coding of targets, or energy transformation processes.
55. A magnetic nanoparticle composition according to claim 54, wherein the composition is used in the separation or purification of biomolecules, or any combination thereof.
56. A therapeutic method according to claim 54, wherein the composition is used in the separation, purification, or any combination thereof of nucleic acids, nucleic acid derivatives, nucleic acid fragments, proteins, protein derivatives, protein fragments, or any combination thereof.
57. A magnetic nanoparticle composition according to claim 54, wherein the composition is used for the sorting and purification of cells.
58. A magnetic nanoparticle composition according to claim 54, wherein the composition is used in radionuclide therapy.
59. A magnetic nanoparticle composition according to claim 54, wherein the composition is used in analytical processes or diagnostic assays.
60. A magnetic nanoparticle composition according to claim 59, wherein the composition is used as contrast media.
61. A therapeutic method, comprising:
a. administering a magnetic nanoparticle composition according to claim 46 to at least a portion of the body, body part, tissue, cell, or body fluid of a subject comprising a target, and
b. administering energy to the magnetic nanoparticle composition combined with the target.
62. A therapeutic method according to claim 61, further comprising the step of applying an alternating magnetic field (AMF).
63. A therapeutic method according to claim 61, wherein the composition is administered via injection, topical application, transdermal application, orally ingestion, rectal insertion, inhalation through the mouth or nose, or any combination thereof.
64. A therapeutic method according to claim 61, wherein the method is utilized for the treatment of a cancer, AIDS, adverse angiogenesis, restenosis, amyloidosis, tuberculosis, multiple sclerosis, cardiovascular plaque, vascular plaque, obesity, malaria, illnesses due to viruses, or any combination thereof.
65. A therapeutic method according to claim 61, wherein the magnetic nanoparticles transport and release bioactive substances.
66. A therapeutic method, comprising:
a. administering a magnetic nanoparticle composition according to claim 53 to at least a portion of the body, body part, tissue, cell, or body fluid of a subject comprising a target, and
b. administering energy to the magnetic nanoparticle composition combined with the target.
US10/888,189 2003-02-06 2004-07-09 Magnetic nanoparticle compositions, and methods related thereto Abandoned US20050271745A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/888,189 US20050271745A1 (en) 2003-02-06 2004-07-09 Magnetic nanoparticle compositions, and methods related thereto

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10/360,561 US20040156852A1 (en) 2003-02-06 2003-02-06 Therapy via targeted delivery of nanoscale particles
DE10331439A DE10331439B3 (en) 2003-07-10 2003-07-10 Magnetic nanoparticles with improved magnetic properties
DE10331439.3 2003-07-10
US10/888,189 US20050271745A1 (en) 2003-02-06 2004-07-09 Magnetic nanoparticle compositions, and methods related thereto

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/360,561 Continuation-In-Part US20040156852A1 (en) 2001-07-25 2003-02-06 Therapy via targeted delivery of nanoscale particles

Publications (1)

Publication Number Publication Date
US20050271745A1 true US20050271745A1 (en) 2005-12-08

Family

ID=33560046

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/888,189 Abandoned US20050271745A1 (en) 2003-02-06 2004-07-09 Magnetic nanoparticle compositions, and methods related thereto
US10/564,228 Expired - Fee Related US7691285B2 (en) 2003-07-10 2004-07-09 Magnetic nanoparticles having improved magnetic properties

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/564,228 Expired - Fee Related US7691285B2 (en) 2003-07-10 2004-07-09 Magnetic nanoparticles having improved magnetic properties

Country Status (7)

Country Link
US (2) US20050271745A1 (en)
EP (2) EP1648381A4 (en)
JP (2) JP2007530422A (en)
AT (1) ATE535001T1 (en)
CA (1) CA2534687A1 (en)
DE (1) DE10331439B3 (en)
WO (2) WO2005006356A1 (en)

Cited By (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050090732A1 (en) * 2003-10-28 2005-04-28 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles
US20070259133A1 (en) * 2004-12-27 2007-11-08 Kyoto University Ordered Alloy Phase Nanoparticle, Method of Manufacturing the Same Ultra-High-Density Magnetic Recording Medium, and Method of Manufacturing the Same
US20070281034A1 (en) * 2006-05-15 2007-12-06 Kirpotin Dmitri B Magnetic microparticles comprising organic substances
US20080213382A1 (en) * 2007-01-19 2008-09-04 Triton Biosystems, Inc. Thermotherapy susceptors and methods of using same
US20090082611A1 (en) * 2004-02-20 2009-03-26 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
WO2009075774A2 (en) * 2007-12-05 2009-06-18 Massachusetts Institute Of Technology Glycosaminoglycan-coated particles and uses thereof
US20090216320A1 (en) * 2006-04-21 2009-08-27 The Children's Hospital Of Philadelphia Magnetic Gradient Targeting And Sequestering Of Therapeutic Formulations And Therapeutic Systems Thereof
US20100260780A1 (en) * 2004-02-20 2010-10-14 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US20100330704A1 (en) * 2008-06-12 2010-12-30 Canon Kabushiki Kaisha Composite particle, method for producing the same, dispersion solution, magnetic biosensing apparatus and magnetic biosensing method
US20110076767A1 (en) * 2004-02-20 2011-03-31 The Children's Hospital Of Philadelphia Magnetically-driven biodegradable gene delivery nanoparticles formulated with surface-attached polycationic complex
US20110105825A1 (en) * 2009-10-31 2011-05-05 Qteris Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
WO2011075516A2 (en) 2009-12-18 2011-06-23 President And Fellows Of Harvard College Active scaffolds for on-demand drug and cell delivery
US20110301452A1 (en) * 2010-06-07 2011-12-08 Michael Maschke Microcapsule for local treatment of a tumor and method for positioning a magnetic gradient field guiding magnetic nanoparticles to a target location as well as apparatus for positioning a magnetic gradient field
US20120021010A1 (en) * 2010-04-29 2012-01-26 University Of Calcutta Antiplatelet agent and methods of using the same
CN102344117A (en) * 2010-08-06 2012-02-08 同济大学附属上海市肺科医院 Method for preparing composite nano microsphere for enriching lung cancer cells
US20120064140A1 (en) * 2009-05-12 2012-03-15 Wuxi Now Materials Corp. Composite nanogranules from polymer/inorganic nanoparticles, preparation method thereof and use of the same
CN102513046A (en) * 2011-12-21 2012-06-27 上海大学 Magnetic polyelectrolyte microcapsule and its preparation method
US20120283503A1 (en) * 2011-04-29 2012-11-08 The Johns Hopkins University Nanoparticle loaded stem cells and their use in mri guided hyperthermia
WO2013012924A2 (en) 2011-07-18 2013-01-24 President And Fellows Of Harvard College Engineered microbe-targeting molecules and uses thereof
US20130277600A1 (en) * 2008-07-16 2013-10-24 International Business Machines Corporation Protective coating of magnetic nanoparticles
US20140054162A1 (en) * 2012-08-24 2014-02-27 The University Of Akron Magnetic macroinitiators and magnetically induced chain reactions
US8772030B2 (en) 2003-07-31 2014-07-08 Universita Degli Studi Di Roma “La Sapienza” Cardiac stem cells and methods for isolation of same
WO2014144325A1 (en) 2013-03-15 2014-09-18 President And Fellows Of Harvard College Methods and compositions for improving detection and/or capture of a target entity
US8895597B2 (en) 2010-06-17 2014-11-25 Violette Renard Recinos Combination of local temozolomide with local BCNU
WO2014190040A1 (en) 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
EP2835644A1 (en) * 2013-08-06 2015-02-11 Yerzhan Ussembayev Nanocomposites for encapsulation of cells and method for treating diseases
US9138293B1 (en) * 2012-07-27 2015-09-22 Brent Weisman Intravascular treatment of lesions using magnetic nanoparticles
US20150352231A1 (en) * 2014-06-04 2015-12-10 National Taiwan University Magnetic nanoparticle composition and manufacturing method and use thereof
WO2015192078A1 (en) 2014-06-12 2015-12-17 Cedars-Sinai Medical Center Compositions and methods for treating cancers
US20150367140A1 (en) * 2011-08-26 2015-12-24 Michael Edward Susedik Apparatus for the generation of an energy field for the treatment of cancer in body cavities and parts that are cavity-like
US9233163B2 (en) 2010-12-28 2016-01-12 The Children's Hospital Of Philadelphia Hydrolytically releasable prodrugs for sustained release nanoparticle formulations
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
US9330821B2 (en) 2008-12-19 2016-05-03 Boutiq Science Limited Magnetic nanoparticles
WO2009076673A3 (en) * 2007-12-13 2016-06-09 Aduro Biotech Ligand conjugated thermotherapy susceptors and methods for preparing same
EP2416220A4 (en) * 2009-03-31 2016-07-20 Toda Kogyo Corp Magnetic composite particles, magnetic carrier, and developer
US20160228548A1 (en) * 2014-01-06 2016-08-11 University Of Wyoming Nanoparticle delivery system for targeted anti-obesity treatment
WO2017024114A1 (en) 2015-08-06 2017-02-09 President And Fellows Of Harvard College Improved microbe-binding molecules and uses thereof
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
US10124186B2 (en) 2011-01-24 2018-11-13 Endomagnetics Limited System for automatically amending energy field characteristics in the application of an energy field to a living organism for treatment of invasive agents
US20190038748A1 (en) * 2009-11-02 2019-02-07 Pulse Therapeutics, Inc. Devices for controlling magnetic nanoparticles to treat fluid obstructions
CN109420177A (en) * 2017-08-28 2019-03-05 香港中文大学 Material and method for effective DNA delivery nanostructure in vivo to atherosclerotic plaque
CN109516503A (en) * 2019-01-11 2019-03-26 福州大学 A kind of preparation method of high stability water-base nano magnetic fluid
US10359678B2 (en) 2014-04-07 2019-07-23 The Regents Of The University Of California Highly tunable magnetic liquid crystals
EP3546474A2 (en) 2013-12-18 2019-10-02 President and Fellows of Harvard College Crp capture/detection of gram positive bacteria
CN110828094A (en) * 2019-11-13 2020-02-21 黑龙江省科学院高技术研究院 Preparation method of magnetic fluid for high-magnetic-sedimentation-resistance sealing under strong magnetic field condition
WO2020148206A1 (en) 2019-01-14 2020-07-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and kits for generating and selecting a variant of a binding protein with increased binding affinity and/or specificity
CN112970076A (en) * 2018-07-19 2021-06-15 贝克曼库尔特有限公司 Magnetic particles
WO2021153823A1 (en) * 2020-01-30 2021-08-05 (주)카이바이오텍 Method for preparing chitosan hydrogel-chelator for cancer therapy
CN113913805A (en) * 2021-10-20 2022-01-11 中南大学湘雅医院 Cold spraying modified layer and application thereof
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
US11464858B2 (en) 2018-06-23 2022-10-11 University Of Wyoming Magnetic nanoparticle delivery system for pain therapy
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
WO2023081650A1 (en) * 2021-11-02 2023-05-11 Lodestone Biomedical Llc Implantable biosensor containing a magnetic nanoparticle assay for in vivo analyte detection
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US11918315B2 (en) 2018-05-03 2024-03-05 Pulse Therapeutics, Inc. Determination of structure and traversal of occlusions using magnetic particles

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7731648B2 (en) * 2001-07-25 2010-06-08 Aduro Biotech Magnetic nanoscale particle compositions, and therapeutic methods related thereto
DE10331439B3 (en) * 2003-07-10 2005-02-03 Micromod Partikeltechnologie Gmbh Magnetic nanoparticles with improved magnetic properties
KR100604976B1 (en) * 2004-09-03 2006-07-28 학교법인연세대학교 Water-Soluble Nanoparticles Stabilized with Multi-Functional Group Ligands
DE102005007374A1 (en) * 2005-02-17 2006-08-24 Universität Ulm Nano-particle, useful e.g. as carriers for transport of active substances, comprises hydrophobic and biocompatible functionalized polymer, active material and/or marker
US7598199B2 (en) * 2005-05-10 2009-10-06 Massachusetts Institute Of Technology Catalytic nanoparticles for nerve-agent destruction
JP2007085929A (en) * 2005-09-22 2007-04-05 Jsr Corp Magnetic particles for specific capturing and its manufacturing method
US20070020699A1 (en) * 2005-07-19 2007-01-25 Idexx Laboratories, Inc. Lateral flow assay and device using magnetic particles
CN101242870B (en) * 2005-08-19 2013-01-09 季诺维斯公司 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
DE102005042768A1 (en) 2005-09-08 2007-03-15 Ludwig-Maximilian-Universität Magnetic field-controlled drug transfer for aerosol therapy
CN1772303A (en) * 2005-10-25 2006-05-17 朱宏 Nanometer magnetic powder-antibody targeting medicine for magneto thermical threrapy of malignant tumor
KR100713745B1 (en) * 2006-02-27 2007-05-07 연세대학교 산학협력단 Water-soluble magnetic or metal oxide nanoparticles coated with ligands and preparation method thereof
EP1852107A1 (en) * 2006-04-19 2007-11-07 Nanobiotix Magnetic nanoparticles compositions and uses thereof
TWI321133B (en) * 2006-08-01 2010-03-01 Univ Kaohsiung Medical Folate-receptor-targeting iron oxide nanoparticles coated with poly(ethylene glycol)
CN101553889A (en) * 2006-09-05 2009-10-07 哥伦布纳米制品公司 Magnetic particles and methods of making and using the same
DE102006059418B4 (en) 2006-12-15 2011-06-30 Airbus Operations GmbH, 21129 Redundant aircraft cooling system for redundant aircraft components
ES2324003B1 (en) * 2007-07-23 2010-05-31 Universidad De Granada NANOPARTICULES CONSTITUTED BY A MAGNETIC NUCLEUS AND A POLYMER COATING.
KR20100123674A (en) * 2007-09-21 2010-11-24 싸이티뮨 사이언스, 인크. Nanotherapeutic colloidal metal compositions and methods
US20090226376A1 (en) * 2008-03-05 2009-09-10 General Electric Company Novel Mixed Ligand Core/Shell Iron Oxide Nanoparticles for Inflammation Imaging
BRPI0900815A2 (en) * 2009-04-23 2010-12-28 Sociedade Benef Israelita Bras Hospital Albert Einstein method for isolating exosomes from biological solutions using iron oxide nanoparticles
WO2010149150A2 (en) 2009-06-22 2010-12-29 Deklatec Gmbh Colorless, magnetic polymer particles for highly sensitive detection of biological substances and pathogens within the context of bioanalytics and diagnostics
CN102695473B (en) * 2009-11-06 2015-09-23 纳米医学科学公司 Use the cell of targeted nano-particle and magnetic characteristic thereof as the detection of cancerous cell and other biological material, measurement and imaging
US8101680B1 (en) 2010-10-12 2012-01-24 Sabic Innovative Plastics Ip B.V. Methods of preparing polymer nanocomposites
WO2012121528A2 (en) * 2011-03-04 2012-09-13 Samsung Life Public Welfare Foundation Magnetic nanocomposite specific for thyroid cancer and use thereof
RU2622025C2 (en) 2011-08-10 2017-06-08 Магфорс Аг Agglomerating of magnetic nanoparticles coated by alkoxysilane
US20130331590A1 (en) * 2012-06-08 2013-12-12 William A. Farone Apparatus and Method for Manufacturing Permanently Confined Micelle Array Nanoparticles
CN103438221A (en) * 2013-09-25 2013-12-11 北京交通大学 Optimization method for improving pressure endurance capability of magnetic fluid sealing device
DE102014006519A1 (en) 2014-05-03 2015-11-05 Smart Material Printing B.V. Use of magnetic and / or magnetizable, polymeric micro- and / or nanocomposites for the production of complex, magnetic and / or magnetizable molded parts using additive manufacturers
KR101480169B1 (en) * 2014-05-30 2015-01-08 (주)일신오토클레이브 The Method for Preparation of Monodisperse Iron Oxide Nanoparticles Using High Pressure Homogenizer and Monodisperse Iron Oxide Nanoparticels Thereof
CN104141796B (en) * 2014-07-04 2016-02-10 北京交通大学 The method of magnetic fluid sealing voltage endurance capability is improved with magnetic conductivity nanowire magnetic liquid
CN105203578B (en) * 2015-09-29 2018-03-09 北京市理化分析测试中心 A kind of method for detecting microorganism
WO2018023033A1 (en) 2016-07-29 2018-02-01 Western Michigan University Research Foundation Magnetic nanoparticle-based gyroscopic sensor
WO2019103103A1 (en) * 2017-11-24 2019-05-31 Jsr株式会社 Method for separating cells, and particles and kit for cell separation or concentration
TWI690330B (en) * 2018-10-11 2020-04-11 近鎰生技股份有限公司 Drug carrier, drug structure, use thereof, preparation method thereof and method for inhibiting Helicobacter pylori
WO2020180999A1 (en) * 2019-03-04 2020-09-10 Arizona Board Of Regents On Behalf Of The University Of Arizona High verdet constant nanoparticles and methods for producing and using the same
CN114755337B (en) * 2022-04-18 2023-01-31 河南大学 Disulfide bond mediated photo-crosslinking magnetic silica affinity probe and preparation method and application thereof

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4101435A (en) * 1975-06-19 1978-07-18 Meito Sangyo Kabushiki Kaisha Magnetic iron oxide-dextran complex and process for its production
US4209412A (en) * 1978-05-22 1980-06-24 Hercules Incorporated Process for producing nonstoichiometric ferroso-ferric oxides
US4280918A (en) * 1980-03-10 1981-07-28 International Business Machines Corporation Magnetic particle dispersions
US4329241A (en) * 1979-07-20 1982-05-11 Agence Nationale De Valorisation De La Recherche (Anvar) Magnetic fluids and process for obtaining them
US4452773A (en) * 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
US4501726A (en) * 1981-11-12 1985-02-26 Schroeder Ulf Intravascularly administrable, magnetically responsive nanosphere or nanoparticle, a process for the production thereof, and the use thereof
US4677027A (en) * 1984-10-19 1987-06-30 Exploaterings Ab T.B.F. Polymer coated particles having immobilized metal ions on the surfaces thereof
US4783389A (en) * 1987-03-27 1988-11-08 E. I. Du Pont De Nemours And Company Process for preparation of liquid electrostatic developers
US4824587A (en) * 1985-03-18 1989-04-25 The Dow Chemical Company Composites of coercive particles and superparamagnetic particles
US4827945A (en) * 1986-07-03 1989-05-09 Advanced Magnetics, Incorporated Biologically degradable superparamagnetic materials for use in clinical applications
US5160725A (en) * 1987-03-24 1992-11-03 Silica Gel Gesellschaft Mbh Adsorptions-Technik, Apparatebau Magnetic liquid compositions
US5304364A (en) * 1991-10-17 1994-04-19 Istituto Guido Donegani S.P.A. Method for preparing spherical inorganic oxide-based materials having monomodal particle size distribution
US5417956A (en) * 1992-08-18 1995-05-23 Worcester Polytechnic Institute Preparation of nanophase solid state materials
US5427767A (en) * 1991-05-28 1995-06-27 Institut Fur Diagnostikforschung Gmbh An Der Freien Universitat Berlin Nanocrystalline magnetic iron oxide particles-method for preparation and use in medical diagnostics and therapy
US5545395A (en) * 1992-08-13 1996-08-13 Bracco Research, S.A. Method of imaging using encapsulated magnetite particles
US5580692A (en) * 1992-05-26 1996-12-03 Eastman Kodak Company Solvent extraction in limited coalescence processes
US5595687A (en) * 1992-10-30 1997-01-21 Thomas Jefferson University Emulsion stability
US5635206A (en) * 1994-01-20 1997-06-03 Hoffmann-La Roche Inc. Process for liposomes or proliposomes
US5814687A (en) * 1996-01-31 1998-09-29 Jsr Corporation Magnetic polymer particle and process for manufacturing the same
US6048920A (en) * 1994-08-15 2000-04-11 Xerox Corporation Magnetic nanocomposite compositions and processes for the preparation and use thereof
US6136428A (en) * 1992-01-10 2000-10-24 Imation Corp. Magnetic recording media prepared from magnetic particles having an extremely thin, continuous, amorphous, aluminum hydrous oxide coating
US6541039B1 (en) * 1997-06-20 2003-04-01 Institut Für Neue Materialien Gem. Gmbh Nanoscale particles having an iron oxide-containing core enveloped by at least two shells
US20030092029A1 (en) * 2001-06-06 2003-05-15 Lee Josephson Magneitc-nanoparticle conjugates and methods of use
US6567221B2 (en) * 2001-02-09 2003-05-20 Tamron Co., Ltd. High zoom ratio lens device having focus and zoom cams
US20030099954A1 (en) * 2001-11-26 2003-05-29 Stefan Miltenyi Apparatus and method for modification of magnetically immobilized biomolecules
US6767635B1 (en) * 1999-09-14 2004-07-27 Biomedical Apherese Systeme Gmbh Magnetic nanoparticles having biochemical activity, method for the production thereof and their use

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3545395A (en) * 1968-06-12 1970-12-08 Vladislav Vladimirovich Rastor Ship with ice-breaking attachment
FR2480764B1 (en) * 1980-04-18 1985-10-04 Rhone Poulenc Spec Chim LATEX OF MAGNETIC POLYMERS AND PREPARATION METHOD
JPS6055265A (en) * 1983-09-06 1985-03-30 Fujirebio Inc Measuring method of antigen and antibody by using magnetic particle
DK175531B1 (en) * 1986-12-15 2004-11-22 Nexstar Pharmaceuticals Inc Delivery vehicle with amphiphil-associated active ingredient
FR2618084B1 (en) * 1987-07-15 1989-11-10 Rhone Poulenc Chimie MAGNETISABLE MICROSPHERES BASED ON POLYSILOXANE, THEIR PREPARATION PROCESS AND THEIR APPLICATION IN BIOLOGY
WO1990007380A2 (en) * 1988-12-28 1990-07-12 Stefan Miltenyi Methods and materials for high gradient magnetic separation of biological materials
WO1995027437A1 (en) 1991-10-22 1995-10-19 Mallinckrodt Medical, Inc. Microfluidization of calcium/oxyanion-containing particles
US5358659A (en) * 1992-07-09 1994-10-25 Xerox Corporation Magnetic materials with single-domain and multidomain crystallites and a method of preparation
JPH06148189A (en) * 1992-11-05 1994-05-27 Nippon Telegr & Teleph Corp <Ntt> Magnetic particulate for laser magnetic immunological measurement and its manufacture
JP3404787B2 (en) * 1993-03-12 2003-05-12 三菱ウェルファーマ株式会社 Novel diethylenetriaminepentaacetic acid derivative, complex compound of the derivative with a metal atom, and diagnostic agent containing the complex compound
DE4428851C2 (en) * 1994-08-04 2000-05-04 Diagnostikforschung Inst Nanoparticles containing iron, their production and application in diagnostics and therapy
US5852076A (en) * 1994-11-13 1998-12-22 Minnesota Mining And Manufacturing Company Process for preparing a dispersion of hard particles in solvent
JPH08176212A (en) * 1994-12-21 1996-07-09 Japan Synthetic Rubber Co Ltd Method of surface-modifying magnetic particle
US5667716A (en) * 1996-07-01 1997-09-16 Xerox Corporation High magnetization aqueous ferrofluids and processes for preparation and use thereof
US6467630B1 (en) 1999-09-03 2002-10-22 The Cleveland Clinic Foundation Continuous particle and molecule separation with an annular flow channel
AU2001234831A1 (en) * 2000-02-05 2001-08-14 Florida State University Research Foundation Magnetoliposome composition for targeted treatment of biological tissue and associated methods
WO2001074245A1 (en) * 2000-03-31 2001-10-11 Amersham Health As Method of magnetic resonance imaging
DE10020376A1 (en) * 2000-04-26 2001-11-08 Inst Zelltechnologie E V Dynamic markers
DE10059151C2 (en) 2000-11-29 2003-10-16 Christoph Alexiou Magnetic particles for targeted regional therapy and use of the same
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
DE10154016B4 (en) * 2001-10-26 2004-02-12 Berlin Heart Ag Magnetic fluid and process for its manufacture
DE10331439B3 (en) * 2003-07-10 2005-02-03 Micromod Partikeltechnologie Gmbh Magnetic nanoparticles with improved magnetic properties

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4101435A (en) * 1975-06-19 1978-07-18 Meito Sangyo Kabushiki Kaisha Magnetic iron oxide-dextran complex and process for its production
US4209412A (en) * 1978-05-22 1980-06-24 Hercules Incorporated Process for producing nonstoichiometric ferroso-ferric oxides
US4329241A (en) * 1979-07-20 1982-05-11 Agence Nationale De Valorisation De La Recherche (Anvar) Magnetic fluids and process for obtaining them
US4280918A (en) * 1980-03-10 1981-07-28 International Business Machines Corporation Magnetic particle dispersions
US4501726A (en) * 1981-11-12 1985-02-26 Schroeder Ulf Intravascularly administrable, magnetically responsive nanosphere or nanoparticle, a process for the production thereof, and the use thereof
US4452773A (en) * 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
US4677027A (en) * 1984-10-19 1987-06-30 Exploaterings Ab T.B.F. Polymer coated particles having immobilized metal ions on the surfaces thereof
US4824587A (en) * 1985-03-18 1989-04-25 The Dow Chemical Company Composites of coercive particles and superparamagnetic particles
US4827945A (en) * 1986-07-03 1989-05-09 Advanced Magnetics, Incorporated Biologically degradable superparamagnetic materials for use in clinical applications
US5160725A (en) * 1987-03-24 1992-11-03 Silica Gel Gesellschaft Mbh Adsorptions-Technik, Apparatebau Magnetic liquid compositions
US4783389A (en) * 1987-03-27 1988-11-08 E. I. Du Pont De Nemours And Company Process for preparation of liquid electrostatic developers
US5427767A (en) * 1991-05-28 1995-06-27 Institut Fur Diagnostikforschung Gmbh An Der Freien Universitat Berlin Nanocrystalline magnetic iron oxide particles-method for preparation and use in medical diagnostics and therapy
US5304364A (en) * 1991-10-17 1994-04-19 Istituto Guido Donegani S.P.A. Method for preparing spherical inorganic oxide-based materials having monomodal particle size distribution
US6136428A (en) * 1992-01-10 2000-10-24 Imation Corp. Magnetic recording media prepared from magnetic particles having an extremely thin, continuous, amorphous, aluminum hydrous oxide coating
US5580692A (en) * 1992-05-26 1996-12-03 Eastman Kodak Company Solvent extraction in limited coalescence processes
US5545395A (en) * 1992-08-13 1996-08-13 Bracco Research, S.A. Method of imaging using encapsulated magnetite particles
US5417956A (en) * 1992-08-18 1995-05-23 Worcester Polytechnic Institute Preparation of nanophase solid state materials
US5595687A (en) * 1992-10-30 1997-01-21 Thomas Jefferson University Emulsion stability
US5635206A (en) * 1994-01-20 1997-06-03 Hoffmann-La Roche Inc. Process for liposomes or proliposomes
US6048920A (en) * 1994-08-15 2000-04-11 Xerox Corporation Magnetic nanocomposite compositions and processes for the preparation and use thereof
US5814687A (en) * 1996-01-31 1998-09-29 Jsr Corporation Magnetic polymer particle and process for manufacturing the same
US6541039B1 (en) * 1997-06-20 2003-04-01 Institut Für Neue Materialien Gem. Gmbh Nanoscale particles having an iron oxide-containing core enveloped by at least two shells
US6767635B1 (en) * 1999-09-14 2004-07-27 Biomedical Apherese Systeme Gmbh Magnetic nanoparticles having biochemical activity, method for the production thereof and their use
US6567221B2 (en) * 2001-02-09 2003-05-20 Tamron Co., Ltd. High zoom ratio lens device having focus and zoom cams
US20030092029A1 (en) * 2001-06-06 2003-05-15 Lee Josephson Magneitc-nanoparticle conjugates and methods of use
US20030099954A1 (en) * 2001-11-26 2003-05-29 Stefan Miltenyi Apparatus and method for modification of magnetically immobilized biomolecules

Cited By (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8772030B2 (en) 2003-07-31 2014-07-08 Universita Degli Studi Di Roma “La Sapienza” Cardiac stem cells and methods for isolation of same
US8846396B2 (en) 2003-07-31 2014-09-30 Universita Degli Studi Di Roma “La Sapienza” Methods for the isolation of cardiac stem cells
US20050090732A1 (en) * 2003-10-28 2005-04-28 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles
US9095610B2 (en) 2004-02-20 2015-08-04 Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US20100260780A1 (en) * 2004-02-20 2010-10-14 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US20090082611A1 (en) * 2004-02-20 2009-03-26 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US8562505B2 (en) 2004-02-20 2013-10-22 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US20110076767A1 (en) * 2004-02-20 2011-03-31 The Children's Hospital Of Philadelphia Magnetically-driven biodegradable gene delivery nanoparticles formulated with surface-attached polycationic complex
US9028829B2 (en) 2004-02-20 2015-05-12 The Children's Hospital Of Philadelphia Uniform field magnetization and targeting of therapeutic formulations
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US20070259133A1 (en) * 2004-12-27 2007-11-08 Kyoto University Ordered Alloy Phase Nanoparticle, Method of Manufacturing the Same Ultra-High-Density Magnetic Recording Medium, and Method of Manufacturing the Same
US20090216320A1 (en) * 2006-04-21 2009-08-27 The Children's Hospital Of Philadelphia Magnetic Gradient Targeting And Sequestering Of Therapeutic Formulations And Therapeutic Systems Thereof
US20070281034A1 (en) * 2006-05-15 2007-12-06 Kirpotin Dmitri B Magnetic microparticles comprising organic substances
US20080213382A1 (en) * 2007-01-19 2008-09-04 Triton Biosystems, Inc. Thermotherapy susceptors and methods of using same
WO2009075774A3 (en) * 2007-12-05 2009-12-30 Massachusetts Institute Of Technology Glycosaminoglycan-coated particles and uses thereof
WO2009075774A2 (en) * 2007-12-05 2009-06-18 Massachusetts Institute Of Technology Glycosaminoglycan-coated particles and uses thereof
WO2009076673A3 (en) * 2007-12-13 2016-06-09 Aduro Biotech Ligand conjugated thermotherapy susceptors and methods for preparing same
US20100330704A1 (en) * 2008-06-12 2010-12-30 Canon Kabushiki Kaisha Composite particle, method for producing the same, dispersion solution, magnetic biosensing apparatus and magnetic biosensing method
US20130277600A1 (en) * 2008-07-16 2013-10-24 International Business Machines Corporation Protective coating of magnetic nanoparticles
US9330821B2 (en) 2008-12-19 2016-05-03 Boutiq Science Limited Magnetic nanoparticles
EP2416220A4 (en) * 2009-03-31 2016-07-20 Toda Kogyo Corp Magnetic composite particles, magnetic carrier, and developer
US20120064140A1 (en) * 2009-05-12 2012-03-15 Wuxi Now Materials Corp. Composite nanogranules from polymer/inorganic nanoparticles, preparation method thereof and use of the same
US9139430B2 (en) * 2009-05-12 2015-09-22 Wuxi Now Materials Corp. Composite nanogranules from polymer/inorganic nanoparticles, preparation method thereof and use of the same
US8565892B2 (en) 2009-10-31 2013-10-22 Qteris, Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
US20110105825A1 (en) * 2009-10-31 2011-05-05 Qteris Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
US9844679B2 (en) 2009-10-31 2017-12-19 Qteris, Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
US11612655B2 (en) 2009-11-02 2023-03-28 Pulse Therapeutics, Inc. Magnetic particle control and visualization
US11000589B2 (en) 2009-11-02 2021-05-11 Pulse Therapeutics, Inc. Magnetic particle control and visualization
US20190038748A1 (en) * 2009-11-02 2019-02-07 Pulse Therapeutics, Inc. Devices for controlling magnetic nanoparticles to treat fluid obstructions
US10813997B2 (en) * 2009-11-02 2020-10-27 Pulse Therapeutics, Inc. Devices for controlling magnetic nanoparticles to treat fluid obstructions
WO2011075516A2 (en) 2009-12-18 2011-06-23 President And Fellows Of Harvard College Active scaffolds for on-demand drug and cell delivery
US20120021010A1 (en) * 2010-04-29 2012-01-26 University Of Calcutta Antiplatelet agent and methods of using the same
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US20110301452A1 (en) * 2010-06-07 2011-12-08 Michael Maschke Microcapsule for local treatment of a tumor and method for positioning a magnetic gradient field guiding magnetic nanoparticles to a target location as well as apparatus for positioning a magnetic gradient field
US8895597B2 (en) 2010-06-17 2014-11-25 Violette Renard Recinos Combination of local temozolomide with local BCNU
CN102344117A (en) * 2010-08-06 2012-02-08 同济大学附属上海市肺科医院 Method for preparing composite nano microsphere for enriching lung cancer cells
US9233163B2 (en) 2010-12-28 2016-01-12 The Children's Hospital Of Philadelphia Hydrolytically releasable prodrugs for sustained release nanoparticle formulations
US10124186B2 (en) 2011-01-24 2018-11-13 Endomagnetics Limited System for automatically amending energy field characteristics in the application of an energy field to a living organism for treatment of invasive agents
US20120283503A1 (en) * 2011-04-29 2012-11-08 The Johns Hopkins University Nanoparticle loaded stem cells and their use in mri guided hyperthermia
EP3081937A1 (en) 2011-07-18 2016-10-19 President and Fellows of Harvard College Engineered microbe-targeting molecules and uses thereof
WO2013012924A2 (en) 2011-07-18 2013-01-24 President And Fellows Of Harvard College Engineered microbe-targeting molecules and uses thereof
US9682247B2 (en) * 2011-08-26 2017-06-20 Endomagnetics Limited Apparatus for the generation of an energy field for the treatment of cancer in body cavities and parts that are cavity-like
RU2635653C2 (en) * 2011-08-26 2017-11-14 Эндомагнетикс Лтд Device for energy field generation for treatment of body cavities cancer and cavity organs cancer
US9687668B2 (en) 2011-08-26 2017-06-27 Endomagnetics Limited Treatment of cancer in body cavities and parts that are cavity-like
US20150367140A1 (en) * 2011-08-26 2015-12-24 Michael Edward Susedik Apparatus for the generation of an energy field for the treatment of cancer in body cavities and parts that are cavity-like
CN102513046A (en) * 2011-12-21 2012-06-27 上海大学 Magnetic polyelectrolyte microcapsule and its preparation method
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
US9138293B1 (en) * 2012-07-27 2015-09-22 Brent Weisman Intravascular treatment of lesions using magnetic nanoparticles
US11220687B2 (en) 2012-08-13 2022-01-11 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US10457942B2 (en) 2012-08-13 2019-10-29 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9545611B2 (en) * 2012-08-24 2017-01-17 The University Of Akron Magnetic macroinitiators and magnetically induced chain reactions
US20140054162A1 (en) * 2012-08-24 2014-02-27 The University Of Akron Magnetic macroinitiators and magnetically induced chain reactions
WO2014144325A1 (en) 2013-03-15 2014-09-18 President And Fellows Of Harvard College Methods and compositions for improving detection and/or capture of a target entity
EP3848044A1 (en) 2013-05-21 2021-07-14 President and Fellows of Harvard College Engineered heme-binding compositions and uses thereof
EP3848045A1 (en) 2013-05-21 2021-07-14 President and Fellows of Harvard College Engineered heme-binding compositions and uses thereof
WO2014190040A1 (en) 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
EP2835644A1 (en) * 2013-08-06 2015-02-11 Yerzhan Ussembayev Nanocomposites for encapsulation of cells and method for treating diseases
EP3912986A2 (en) 2013-12-18 2021-11-24 President and Fellows of Harvard College Crp capture/detection of bacteria
EP3546474A2 (en) 2013-12-18 2019-10-02 President and Fellows of Harvard College Crp capture/detection of gram positive bacteria
US9782481B2 (en) * 2014-01-06 2017-10-10 University Of Wyoming Nanoparticle delivery system for targeted anti-obesity treatment
US10456467B2 (en) 2014-01-06 2019-10-29 University Of Wyoming Nanoparticle delivery system for targeted anti-obesity treatment
US11077190B2 (en) * 2014-01-06 2021-08-03 University Of Wyoming Nanoparticle delivery system for targeted anti-obesity treatment
US20160228548A1 (en) * 2014-01-06 2016-08-11 University Of Wyoming Nanoparticle delivery system for targeted anti-obesity treatment
US10359678B2 (en) 2014-04-07 2019-07-23 The Regents Of The University Of California Highly tunable magnetic liquid crystals
US20150352231A1 (en) * 2014-06-04 2015-12-10 National Taiwan University Magnetic nanoparticle composition and manufacturing method and use thereof
US9775824B2 (en) * 2014-06-04 2017-10-03 National Taiwan University Magnetic nanoparticle composition and manufacturing method and use thereof
WO2015192078A1 (en) 2014-06-12 2015-12-17 Cedars-Sinai Medical Center Compositions and methods for treating cancers
EP4306173A2 (en) 2014-06-12 2024-01-17 Cedars-Sinai Medical Center Compositions and methods for treating cancers
US11357799B2 (en) 2014-10-03 2022-06-14 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
EP3763378A1 (en) 2015-08-06 2021-01-13 President and Fellows of Harvard College Improved microbe-binding molecules and uses thereof
WO2017024114A1 (en) 2015-08-06 2017-02-09 President And Fellows Of Harvard College Improved microbe-binding molecules and uses thereof
US11872251B2 (en) 2016-01-11 2024-01-16 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
US11351200B2 (en) 2016-06-03 2022-06-07 Cedars-Sinai Medical Center CDC-derived exosomes for treatment of ventricular tachyarrythmias
US11541078B2 (en) 2016-09-20 2023-01-03 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
US11759482B2 (en) 2017-04-19 2023-09-19 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US10973927B2 (en) * 2017-08-28 2021-04-13 The Chinese University Of Hong Kong Materials and methods for effective in vivo delivery of DNA nanostructures to atherosclerotic plaques
CN109420177A (en) * 2017-08-28 2019-03-05 香港中文大学 Material and method for effective DNA delivery nanostructure in vivo to atherosclerotic plaque
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11918315B2 (en) 2018-05-03 2024-03-05 Pulse Therapeutics, Inc. Determination of structure and traversal of occlusions using magnetic particles
US11464858B2 (en) 2018-06-23 2022-10-11 University Of Wyoming Magnetic nanoparticle delivery system for pain therapy
CN112970076A (en) * 2018-07-19 2021-06-15 贝克曼库尔特有限公司 Magnetic particles
CN109516503A (en) * 2019-01-11 2019-03-26 福州大学 A kind of preparation method of high stability water-base nano magnetic fluid
WO2020148206A1 (en) 2019-01-14 2020-07-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and kits for generating and selecting a variant of a binding protein with increased binding affinity and/or specificity
CN110828094A (en) * 2019-11-13 2020-02-21 黑龙江省科学院高技术研究院 Preparation method of magnetic fluid for high-magnetic-sedimentation-resistance sealing under strong magnetic field condition
WO2021153823A1 (en) * 2020-01-30 2021-08-05 (주)카이바이오텍 Method for preparing chitosan hydrogel-chelator for cancer therapy
CN113913805A (en) * 2021-10-20 2022-01-11 中南大学湘雅医院 Cold spraying modified layer and application thereof
WO2023081650A1 (en) * 2021-11-02 2023-05-11 Lodestone Biomedical Llc Implantable biosensor containing a magnetic nanoparticle assay for in vivo analyte detection

Also Published As

Publication number Publication date
WO2005006356A1 (en) 2005-01-20
US20060163526A1 (en) 2006-07-27
JP2007530422A (en) 2007-11-01
WO2005013897A2 (en) 2005-02-17
US7691285B2 (en) 2010-04-06
DE10331439B3 (en) 2005-02-03
EP1648381A2 (en) 2006-04-26
ATE535001T1 (en) 2011-12-15
EP1644941A1 (en) 2006-04-12
JP4649406B2 (en) 2011-03-09
JP2007538380A (en) 2007-12-27
WO2005013897A3 (en) 2006-02-16
EP1648381A4 (en) 2011-03-09
EP1644941B1 (en) 2011-11-23
CA2534687A1 (en) 2005-02-17

Similar Documents

Publication Publication Date Title
US20050271745A1 (en) Magnetic nanoparticle compositions, and methods related thereto
El-Boubbou Magnetic iron oxide nanoparticles as drug carriers: clinical relevance
Laurent et al. Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, physicochemical characterizations, and biological applications
Schütt et al. Applications of magnetic targeting in diagnosis and therapy—possibilities and limitations: a mini-review
Laurent et al. Superparamagnetic iron oxide nanoparticles: promises for diagnosis and treatment of cancer
Sun et al. Folic acid‐PEG conjugated superparamagnetic nanoparticles for targeted cellular uptake and detection by MRI
Yang et al. Hyaluronic acid conjugated magnetic prussian blue@ quantum dot nanoparticles for cancer theranostics
Arruebo et al. Magnetic nanoparticles for drug delivery
Mahmoudi et al. Superparamagnetic iron oxide nanoparticles (SPIONs): development, surface modification and applications in chemotherapy
Weissig et al. Nanopharmaceuticals (part 2): products in the pipeline
Reddy et al. Magnetic nanoparticles: design and characterization, toxicity and biocompatibility, pharmaceutical and biomedical applications
Lin et al. Development of superparamagnetic iron oxide nanoparticles (SPIONS) for translation to clinical applications
US20160271274A1 (en) Synthesis and use of targeted radiation enhancing iron oxide-silica-gold nanoshells for imaging and treatment of cancer
US7731648B2 (en) Magnetic nanoscale particle compositions, and therapeutic methods related thereto
Sun et al. Bacterial magnetosome: a novel biogenetic magnetic targeted drug carrier with potential multifunctions
Singh et al. Formulation design facilitates magnetic nanoparticle delivery to diseased cells and tissues
US20060140867A1 (en) Coated stent assembly and coating materials
US20110177153A1 (en) targeted nanoparticle drug for magnetic hyperthermia treatment on malignant tumors
Bárcena et al. Applications of magnetic nanoparticles in biomedicine
US20200155682A1 (en) Process for making iron oxide nanoparticle preparations for cancer hyperthermia
Cormode et al. Inorganic nanocrystals as contrast agents in MRI: synthesis, coating and introduction of multifunctionality
Joshi Multifunctional metal ferrite nanoparticles for MR imaging applications
Hanini et al. Ferrite nanoparticles for cancer hyperthermia therapy
Zamay et al. Magnetic Nanoparticles in Theranostics
Shetty et al. Magnetic nano-systems in drug delivery and biomedical applications

Legal Events

Date Code Title Description
AS Assignment

Owner name: MICROMOD PARTIKELTECHNOLOGIE GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRUETTNER, CORDULA;TELLER, JOACHIM;WESTPHAL, FRITZ;REEL/FRAME:015573/0953

Effective date: 20040707

Owner name: TRITON BIOSYSTEMS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:IVKOV, ROBERT;REEL/FRAME:015584/0361

Effective date: 20040707

Owner name: TRITON BIOSYSTEMS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MICROMOD PARTIKELTECHNOLOGIE GMBH;REEL/FRAME:015598/0961

Effective date: 20040707

AS Assignment

Owner name: NANOTX, INC., CALIFORNIA

Free format text: MERGER;ASSIGNORS:TRITON BIOSYSTEMS, INC.;ONCOLOGIC, INC.;REEL/FRAME:021450/0525

Effective date: 20080327

Owner name: NANOTX, INC.,CALIFORNIA

Free format text: MERGER;ASSIGNORS:TRITON BIOSYSTEMS, INC.;ONCOLOGIC, INC.;REEL/FRAME:021450/0525

Effective date: 20080327

AS Assignment

Owner name: ADURO BIOTECH, CALIFORNIA

Free format text: FICTITIOUS BUSINESS NAME STATEMENT;ASSIGNOR:NANOTX CORP.;REEL/FRAME:021450/0900

Effective date: 20080422

Owner name: ADURO BIOTECH,CALIFORNIA

Free format text: FICTITIOUS BUSINESS NAME STATEMENT;ASSIGNOR:NANOTX CORP.;REEL/FRAME:021450/0900

Effective date: 20080422

AS Assignment

Owner name: ASPEN MEDISYS, LLC, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ADURO BIOTECH;REEL/FRAME:028353/0447

Effective date: 20120605

STCB Information on status: application discontinuation

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