CA2068632C - Nanocrystalline magnetic iron oxide particles, processes for their preparation and their use in medical diagnostics and therapy - Google Patents

Nanocrystalline magnetic iron oxide particles, processes for their preparation and their use in medical diagnostics and therapy Download PDF

Info

Publication number
CA2068632C
CA2068632C CA002068632A CA2068632A CA2068632C CA 2068632 C CA2068632 C CA 2068632C CA 002068632 A CA002068632 A CA 002068632A CA 2068632 A CA2068632 A CA 2068632A CA 2068632 C CA2068632 C CA 2068632C
Authority
CA
Canada
Prior art keywords
magnetic particles
cross
synthetic
magnetite
proteins
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.)
Expired - Fee Related
Application number
CA002068632A
Other languages
French (fr)
Other versions
CA2068632A1 (en
Inventor
Mayk Kresse
Rudiger Lawaczeck
Detlef Pfefferer
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.)
Bayer Pharma AG
Original Assignee
Institut fuer Diagnostikforschung GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut fuer Diagnostikforschung GmbH filed Critical Institut fuer Diagnostikforschung GmbH
Publication of CA2068632A1 publication Critical patent/CA2068632A1/en
Application granted granted Critical
Publication of CA2068632C publication Critical patent/CA2068632C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0052Thermotherapy; Hyperthermia; Magnetic induction; Induction heating therapy
    • 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
    • A61K49/1833Nuclear 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 a small organic molecule
    • A61K49/1839Nuclear 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 a small organic molecule the small organic molecule being a lipid, a fatty acid having 8 or more carbon atoms in the main chain, or a phospholipid
    • 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
    • A61K49/1866Nuclear 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 the nanoparticle having a (super)(para)magnetic core coated or functionalised with a peptide, e.g. protein, polyamino acid
    • 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
    • A61K49/189Host-guest complexes, e.g. cyclodextrins
    • 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
    • A61K51/1244Preparations 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 microparticles or nanoparticles, e.g. polymeric nanoparticles
    • A61K51/1251Preparations 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 microparticles or nanoparticles, e.g. polymeric nanoparticles micro- or nanospheres, micro- or nanobeads, micro- or nanocapsules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5094Microcapsules containing magnetic carrier material, e.g. ferrite for drug targeting
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2123/00Preparations for testing in vivo
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2400/00Assays, e.g. immunoassays or enzyme assays, involving carbohydrates
    • G01N2400/10Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • G01N2400/38Heteroglycans, i.e. polysaccharides having more than one sugar residue in the main chain in either alternating or less regular sequence, e.g. gluco- or galactomannans, e.g. Konjac gum, Locust bean gum, Guar gum
    • G01N2400/40Glycosaminoglycans, i.e. GAG or mucopolysaccharides, e.g. chondroitin sulfate, dermatan sulfate, hyaluronic acid, heparin, heparan sulfate, and related sulfated polysaccharides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2446/00Magnetic particle immunoreagent carriers
    • G01N2446/20Magnetic particle immunoreagent carriers the magnetic material being present in the particle core
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2446/00Magnetic particle immunoreagent carriers
    • G01N2446/30Magnetic particle immunoreagent carriers the magnetic material being dispersed in the polymer composition before their conversion into particulate form
    • 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
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/24Nuclear magnetic resonance, electron spin resonance or other spin effects or mass spectrometry

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Nanotechnology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Radiology & Medical Imaging (AREA)
  • Dispersion Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Medical Informatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Ceramic Engineering (AREA)
  • Inorganic Chemistry (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Optics & Photonics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Compounds Of Iron (AREA)
  • Hard Magnetic Materials (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Soft Magnetic Materials (AREA)

Abstract

Novel nanocrystalline magnetic particles are provided herein which consist of a magnetic iron oxide core of Fe3 0 4, gamma-Fe2 0 3 or mixtures thereof, and an envelope chemisorbed to the core. The composition of the coating material comprises natural or synthetic glycosaminoglycans and/or their derivatives with molecular weights of 500 Da to 250,000 Da, if necessary, which have been covalently cross-linked with appropriate cross-linking agents and/or modified by specific additives. Processes for preparation of these particles as well as the use thereof in medical diagnostics and/or therapy are also taught.

Description

(a) TITLE OF THE INVENTION
NANOCRYSTALLINE MAGNETIC IRON OXIDE PARTICLES, PROCESSES
FOR THEIR PREPARATION AND THEIR USE IN MEDICAL DIAGNOSTICS
AND THERAPY
(b) TECHNICAL FIELD TO WHICH THE INVENTION RELATES
The invention relates to nanocrystalline magnetic particles, to processes for the preparation of these particles, as well as their use in medical diagnostics and therapy.
(c) BACKGROUND ART
Magnetic substances have found their way into medical use as contrast media for magnetic resonance imaging. It should be noted that the first substance to be approved for use was a Gd-DTPA complex (MAGNEVISTTM). Magnetic iron oxide particles have proven to be especially effective in liver diagnostics. They are now in the pre-clinical and also, to some extent, in the clinical development stage. Such magnetic substances are primarily ferrimagnetic iron oxides (e.g., magnetite), nanoparticles which are surrounded with a coating to form stable aqueous sols.
Besides their use as diagnostic agents, ferrous/ferrimagnetic particles are playing an increasing role in the in vitro separation technique and as "thermoseeds"
in local hyperthermia.
Numerous magnetite preparation processes and uses are described in the scientific and patent law literature.
Hasegawa and Hokkoku (U.S. Patent No. 4,101,435) describe an iron oxide-dextran complex and its preparation.
Rembaum (U.S. Patent No. 4,267,234) refers to magnetic polyglutaraldehyde microparticles which are prepared by suspension n:.~ ~ ~."~ ~ ;~
polymerization in the presence of magnetic particles. Widder and Senyei (US 4,247,406) prepare microparticles from an amino acid-polymer matrix in which magnetic particles are embedded. A similar method with nanoparticles is used by Schroder and Mosbach (WO 83/01738) whereby a crystalline hydrocarbon matrix surrounds the magnetic material. Groman and Josephson (US 4,770,183) use magnetic metal oxide particles which are non-coated and coated with a polysaccharide and/or protein coating. Molday (US 4,452,773) describes the synthesis of ferromagnetic iron oxide cores with a polysaccharide coating. He achieves a Stable sol and can couple proteins to the dextran coating using periodate activation.
Gordon (US 4,731,239) claims the use of ferrous, pare- and diamagnetic particles of iron hydroxide, iron oxide and iron dextran for diagnostic purposes. Additional patents (US
4,767,611: 4,758,429 4,735,796) focus on diagnosis and therapy with the aid of the aforesaid iron dextran or iron transferric dextran particles and alternating electromagnetic fields. The particles can be directed to their targets by antigens, antibodies, enzymes or prosthetic groups. Dries et al. (EP 186616) use magnetic particles of double metal oxide/
hydroxide and a complex agent of proteins or alkaline treated saccharides or polysaccharides.
over the years the complexity of the particles has increased and the target has become more specific. Ranney (EP-361960) uses polyatomic chrome complexes which are bonded to a biocompatible clearable carrier.
The carriers listed are: hydrocarbons, polysaccharides, glycosaminoglycans and structurally analogous synthetic polymers.

These Cr4 S clusters have the disadvantage that the number of unpaired electrons is limited to 12.
Application in diagnostics is complemented by therapeutic aspects (hyperthermia, chemotherapy).
Menz et al. (WO 9001295) aim at a special cellular uptake mechanism (receptor-mediated endocytosis) with their superparamagnetic contrast media. They use magnetic iron oxide particles which are surrounded by an arabinose galactan coating of vegetable extraction the toxicological safety of which has not as yet been confirmed.
Yudelson (WO 8911154) deposits a coating of gelatin and a polymeric acid (preferably gum arabic) on the superparamagnetic particles using coacervation and cross-linking preferably with glutaraldehyde. Pilgrim (European patent application publication number 0284549) uses a different approach by forming a chemical bond between the magnetite surface and synthetic polymer stabilizer substances. The reactive stabilizer substance is chemically bonded to the superparamagnetic particles using phosphate, phosphonate or carboxylate groups. Tissue specific bonding substances can be added to the polyethylene skeleton of the stabilizer substance.
In both cases, preparation and magnetic properties of the superparamagnetic particles are described without furnishing proof of the medical suitability and tolerance of the particles.
Magnetic materials are classified as die-, pare-, or ferromagnetic according to their properties in a magnetic field. Dia- and paramagnetism are atomic/ molecular properties which are based on the magnetic moment of the / v H W
prA ~ e~' ~ i~1 ~n..Y LV
electron. The diamagnetism is based on orbital motion of the electrons and is induced by the applied magnetic field. The magnetization is aligned inversely to the external magnetic field. Paramagnetic substances are characterized by one or more unpaired electrons, magnetization is aligned parallel to the external field. Hence paramagnetic atoms/molecules have a permanent magnetic moment and without an external field these magnetic,moments are not oriented. The susceptibility is ,.
independent of the applied field and is inversely proportional to temperature (Curie°s law).
In solid bodies, strong interaction may develop between adjacent atoms/ions so that spontaneous magnetization results. One generally refers to ferromagnetism with more accurate classification in ferrous, ferric and antiferromagnetism. Examples of ferromagnetic materials are the metals iron, cobalt, nickel, many rare earth elements as well as their alloys. The magnetic moment of adjacent atoms/ions can be aligned anti°parallel in combinations of different types of atoms, of ions with different oxidation numbers or of ions that are constructed on a different lattice position. Complete suspension of spontaneous magnetization is referred to as antiferromagnetism. Partial suspension is referred to as ferrimagnetism. Ferrite with a spinal structure demonstrates ferrimagnetic properties. In all three cases domains are formed in solid bodies with different alignments of spontaneous magnetization. Without an external field the magnetic moments of the domains are randomly distributed and there is no externally directed total magnetic moment. Magnetic moments of the domain are aligned in the external magnetic field. A condition preserved in the permanent magnet even after disconnection of the external magnetic field. This is referred 'to as remanence.
The thermal energy counteracts a spontaneous parallel/antiparallel alignment of the individual magnetic ~a~'s~'~,)e~y, a n P~ ~ i of ._d id ' ~s moments in particles which are smaller than the domain.
Suspensions of such ferrous/ferrimagnetic particles with dimensions smaller than 10 - 50 nm have properties similar to paramagnetic materials. They show no remanence or hysteresis 5 but they show susceptibility values which approach. the corresponding values for the solid body. Therefore, they are often referred to as superparamagnetic particles. Ferrites which have been used as parenteral MR contrast media are included in this category (e. g. Wolf et al., Magnetic Resonance Annual, Neia York, Raven Press 1985, 231-266; Saini et al., Radiology 1987, 162, 217-222).
The MR image and signals of MR spectroscopy are produced due to interaction of three physical parameters: density of the sort of core involved (e. g. protons, 13 carbon, 3lphosphorous) spin-lattice-relaxation time, T1, and spin-spin-relaxation time, T2. T1 and T2 affecting signal intensity in a complicated manner. They are functions of strength of the magnetic field, temperature, reorientation and th:~ type of rhysical interaction among individual molecules. Signal intensity increases proportionally to the density of the observed core. For protons the observed cores can be roughly subdivided between fat and water protons.
Proton density in a physiological environment can only be slightly influenced, e.g. by deuterium oxide substitution.
Relaxation time, on the other hand, can be relatively easily influenced by additional paramagnetic molecules/ions (molecules with unpaired electrons, e.g. spin probes or ions of rare earth elements). The underlying relaxation mechanisms, e.g. by paramagnetic ions, are fairly well understood and are found in relevant monographs and text books (Solomon-Bloembergen equation).
Physical description of the relaxation effect is more difficult in the case of particular aotive substances, da~~~i~:~~~ a s especially when these are not homogeneously distributed.
Influence on T2 time is predominant with ferric/ferromagnetic iron oxide particles. This may result in signal boosting up to complete signal extinction, depending on the given concentration of iron oxide particles and the chosen pulse sequence. Normally, relaxation-time-shortening properties of MR contrast media are expressed by relaxivity. Good -molecularly dissolved - paramagnetic substances have values of apex. 4 1/mmol ~ s (Gd-DTPA) for T2 and T2 relaxivity at 20 0.47 T in water and 40 °C. The dispersions of superparamagnetic magnetite or maghemite are appx. 20 - 40 for the T2 and apex. 200 for the T2 relaxivity. Size, coating, the amount of gamma-Fe2 03 and the degree of the recuperation of crystalline fault sites contribute to the 25 individual value.
Pure magnetic iron oxide particles which could possibly be used as diagnostic agents in nuclear magnetic resonance imaging aggregate in aqueous solution with neutral pH and do 20 not form a stable and parenterally injectable sol. The balance of forces among the particles can be so shifted by coating the magnet core with appropriate substance that the thermal energy (Brownian motion) counteracts the aggregation and sedimentation and a stable gel can be obtained. A number 25 of substances which can be used in this manner are known from the literature and patents.
The coating must provide for a stable suspension. Heat sterilization and/or sterile preparation should be possible 30 in the context of a galenic formulation. In addition, the particles must be of medical diagnostic and/or therapeutic value. Moreover, they must be pharmacologically and toxicologically safe. Most of the coating substances fail to fulfill these criteria.

'; ~_:'~.>,~' °~ °,~
7 ~~, '~~.Y-m ,~
So far dextran encapsulated iron oxides are the furthest developed of all the ferrimagnetic nanoparticles mentioned.
Clinical development has been cancelled in the US due to side effects (e. g. drop in blood pressure) (G. L. Wolf "Current status of I~R imaging contrast agents: special report", Radiology 172, 709-710 (1989)). These dextran magnetites are currently being thoroughly tested by the FDA (Food and Drug Administration) (Diagnostic Imaging, October 1990). Dextran magnetites are instable against mixtures with common solvents. Galenic stability in physialogical NaCl solution cannot be maintained either aver extended periods of time.
Derivatization is limited or only possible under drastic conditions. Physiologically, dextran has an adverse allergic potential: the same being true for protein coated magnetites.
The medical diagnostic usefulness of coated ferric/
ferromagnetic (superparamagnetic) iron oxide particles (parent substance) is based on the fact that, following intravenous injection, they are taken up by phagocytizing monocytes and macrophages of the reticuloendothelial system (RES) of the clinically intact splenic and hepatic tissue but are not taken up by tumors and metastases. With normal spin-echo sequences such locally differentiated uptake leads to signal extinction in the magnetic resonance image of the clinically intact splenic and hepatic tissue shortly after parenteral application. Whereas tumors and metastases in liver and spleen appear bright against dark background. A
similar effect is also found in tumors and metastases in the lymphatic system.
Presently, efforts are being made to avoid singular uptake of liposomes and other particular drug carriers via mononuclear phagocytizing cells (RES). This is important for selective enrichment of tumors/tissues in diagnostics and therapy.

r 9f~:;r :qtr: °?'?~
<. - ' ~_J .. b a..D ,a~
The therapeutic benefit of the aforesaid iron oxide particles is based on the applicability of iron preparations as anti-anemic drugs as well as in magnetic targeting, a possibility of targeted transport of iron oxide particles and adhering substances to the site of action by means of external magnetic fields. Recoil-free sensitization by gamma rays and intracellular H-field coupled hyperthermia are additional applications of the iron oxide particles. Resonant nuclear absorption of gamma quanta with subsequent re-emission or emission of Auger electrons is referred to as the M~ssbauer effect after its discoverer. Utilization of this effect in radiotherapy was described by Mills et a1. (Nature 335, 787 -789, 1988).
In contrast to the iron oxide sots described herein, Mills uses molecularly dissolved substances which in solution show only a negligible recoil-free M~ssbauer effect and consequently a resonance strengthened radiotherapeutic benefit. The advantages of resonant nuclear absorption of gamma-quanta with subsequent re-emission or emission of Auger electrons are obvious. The primary radioactive source is outside of the body and the non-radioactive sensor is introduced to the tumor tissue. Absorption of gamma quanta by the non-radioactive radiation sensors without significant stress on the surrounding tissue is the primary result which is attributable to the large capture cross section of the resonant core absorption. These radiation sensors (antennae) can be placed in the tumor tissue (preferably intracellularly). The radioactive source (transmitter) and the receiver are tuned to each other and must satisfy physical and radiobiological criteria. On the receiving end 57Fe, 99Ru, 119Sn, 121Sb, 127I, 151Eu, and 157Gd are preferred possible substances, all of which are not radioactive. Ferrites and magnetites can be doped with the ~,' ~:: J?~ a.a ,a aforesaid Mossbauer isotopes without significant impact on physical properties.
Further use of ferromagnetic materials is based on the assumption that magnetic materials can be introduced into the tumor in the farm of small particles and externally heated by coupling of electromagnetic alternating fields through eddy current, hysteresis losses in the course of magnetic reorientation. Controllable energy uptake is possible through choice of the Curie point (Curie temperature) based on the composition of the magnetic particles (cf. Lilly et al., Radiology 154, 243-244 (1985)). In either case, particles with dimensions larger than each of the single domains should be used for energy uptake. Here, a compromise will have to be made between the physically necessary and pharmacologically tolerable particle dimension, depending on the mode of application. However, based on electron microscopic images of tissue sections the conclusion may be drawn that nanocrystalline particles also tend to aggregate into larger complexes following intracellular uptake so that these nanocrystalline particles may also be considered for this application. Moreover, initial measurements on phantoms indicate that there are also physical mechanisms which can lead to heating of single domain particles.
Another possible approach to therapy can be taken by insertion of 157Gd into ferrites and accomplishing neutron activation for thermal and epithermal neutrons taking advantage of the large capture cross section of 157Gd. As in resonant nuclear absorption through photons (Mossbauer) described above, tissue containing no 157Gd will scarcely take up neutrons and consequently will not be detrimentally affected. Neutron uptake is primarily concentrated on the areas containing 157Gd. Hence, sufficient enrichment of the tumor provided, radiation damage is inflicted only an the 1~
tumour by secondary radiation (Auger electrons and photons). The particles must be doped with the appropriate isotopes for application of ferrite/magnetite in therapy. The degree of doping of ferrite/magnetite as well as size, charge, hydrophobicity and possibly targeting should be adjusted to the purpose of the therapy. Macrophages are known to take up iron oxide particles and other foreign substances and to undergo concentration on the periphery of tumours. It is also possible to concentrate substances in specific tumours by attaching them to monoclonal antibodies. This enables targeting of radiation in order to inhibit the tumour growth and/or perform therapeutic hyperthermia.
In addition, a positive contribution can be made to synovectomy through extracorporeal and/or intracorporeal radiation sensitization. Moreover, even slight accumulation of macrophages containing iron oxide in the vicinity of a tumour can be used for tumour detection by means of highly sensitive magnetic field probes (SQUIDs).
(d) DESCRIPTION OF THE INVENTION
It is the aim of this invention to minimize or even to overcome the above described disadvantages of the known state of the art. Specifically, it is an object of a main aspect of this invention to provide, with an acceptable input in terms of synthesis technique, multiply modifiable and thus variably applicable, pharmacologically and toxicologically safe magnetic particles which can be prepared under thermally sterilized and aseptic conditions.
Surprisingly, the attempt to meet the catalogue of pharmaceutical demands for medical use has been successful owing to glycosaminoglycan coated magnetites.
The present invention in one broad aspect, now provides nanocrystalline magnetic particles consisting of a magnetic iron oxide core of Fe304, gamma-Fe203 or mixtures thereof and a chemisorbed coating on this core, the composition of such coating material comprising natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights of 500 Da to 250,000 Da combined.
By one variant of this broad aspect of the invention, the coating material is covalently cross-linked by appropriate cross-linking agents and/or is modified by specific additives.

By another variant of this broad aspect of the invention, or of the above-described variant thereof, the particles have core diameters which are smaller than the dimensions of the magnetic singular domains.
By yet another variant of this broad aspect of the invention, or of the above-described variants thereof, the particles are also provided with common pharmaceutical adjuvants for injectable solutions and/or enteral agents as stable aqueous colloid dispersive solutions which are 0.2 ~cm filterable. By one specific variation thereof, the magnetic particles can be heat sterilized.
By yet a further variant of this broad aspect of the invention, or of the above-described variants thereof, the iron oxide core is doped with 6Li, 5'Fe, 6'Ni, Ni, 6'Zn, Zn, Mn, ~Ru, ioiRu m3Cd~ n9sn~ izisb~ iz~I~ isiEu~ issGd~ is6Gd, or ls'Gd.
By yet still another variant of this broad aspect of the invention, or of the above-described variants thereof, the natural or synthetic glycosaminoglycans are chondroitin sulphate, dermatan sulphate, or heparin or synthetic analogues thereof, or other heparinoids.
By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the coating material, after preparation of enveloped magnetic iron oxide particles, is additionally cross-linked by cross-linkers which are common in biochemistry and chemistry of natural products.
By another variant of this broad aspect of the invention, or of the above-described variants thereof, the glycosaminoglycan is heparan sulphate or a synthetic analog thereof;
or the glycosaminoglycan is hyaluronic acid, or a synthetic analog thereof.
By a further variant of this broad aspect of the invention, or of the above-described variants thereof, the mono-, di-, tri-, and oligoamines and/or synthetic and biological oligopeptides/proteins are bonded to the coating material.
By yet a further variant of this broad aspect of the invention, or of the above-described variants thereof, the reduced or oxidized glutathione is bonded, as well as intraparticularly reversibly cross-linked to, the coating material.

By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the surface-active substances are bonded to the coating material.
By another variant of this broad aspect of the invention, or of the above-described variants thereof, the targetable structure substances are bonded to the coating material.
By one specific variation thereof, the targetable structure substances comprise hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, e.g., histones, interleukins, lipoproteins, for example, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements or mixtures thereof. By another specific variation thereof, the magnetic particles include additional chemotherapeutic agents, e.g., cytostatic agents.
By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the cross-linking or bonding of targetable structures and/or surface-active substances are combined.
By yet a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the magnetic particles are surrounded by cage molecules. By a specific variation thereof, the cage molecules comprise subunits of clathrin and/or synthetic analogues.
As described above, the chondroitin magnetites, according to aspects of the present invention, can be classified as physiologically safe. Chondroitin is considered a glycosaminoglycan and is of animal or human origin. It is of ubiquitous presence in the body and has numerous medical applications. The iron oxide core presents no difficulties, as long as it is properly shielded by coating material. After intracellular dissolution of the iron oxide core, the liberated iron is incorporated in the endogenous iron pool. Chondroitin magnetites exhibit extremely good tolerance following heat sterilization with LDso values of 20 mmol/kg (rat and mouse). Well-defined imaging of tumours/metastases has been obtained even from doses as low as 10 ~,mol/kg.
This has provided an almost unprecedented safety margin of 2000. Furthermore, glycosaminoglycans are characterized by high hydrolytic stability of the glycosidic bond.
The entire range of demands can be met by these glycosaminoglycan-coated magnetic iron oxide particles: homogeneous iron oxide core; chemical stability of the coating material with well-known positive activity spectrum; preparation of nanocrysalline magnetic particles under non-invasive conditions; expansion of synthesis according to the modular principle (starting from a basic body, specific task oriented modifications can be added), high efficiency of diagnostic and therapeutic action; good tolerance, no significant side effects, no necessary additives to avoid possible side effects;
higher margin of safety (factor 2000 as MR tumour/metastases diagnostic agent);
continued galenic stability even after adjustment of the isotonic value with salt and after heat sterilization in the final receptacle; and low number of decomposition products after extended storage. On the other hand, dextran magnetite, protein magnetite, gum arabic gelatin magnetite or magnetite with synthetic coating materials can only partially satisfy such catalogue of demands. According to aspects of this invention, if the particles are used in therapy, the magnetic core can be doped with 6Li, s'Fe, 6lNi, Ni, 6'Zn, Zn, Mn, 99Ru~ ioiRu u3Cd~ u9sn~ iaisb~ ia~I~ ~siEu~ issGd~ is6Gd~ or ls~Gd.
Especially preferred variants of the magnetic particles are, according to various aspects of this invention, characterized by the following features:
a) the core consists of magnetic iron oxide with core diameters which are smaller than the dimension of the individual magnetic domains; and/or b) they are available with the common pharmaceutical adjuvants for injectable solutions and/or enteral agents as stable aqueous colloid dispersion solutions which are 0.2 ~,m filterable and can be thermally sterilized; and/or c) the core, if doped, is preferably doped with 6Li, s'Fe, 6'Ni, 'SIEu, 'S'Gd;
and/or d) the natural or synthetic glycosaminoglycans are chondroitin sulphates, dermatan sulphates, heparan sulphates, heparin or their synthetic analogues, or other heparinoids; and/or e) the coating material, after the preparation of the coated magnetic iron oxide particles, is additionally cross-linked by cross-linking agents which are common in biochemistry and the chemistry of natural substances; and/or f) mono- di-, tri-, and oligoamines and/or synthetic and biological oligopeptides and proteins are bonded to the coating material; and/or g) reduced or oxidized glutathione is bonded to the coating material and is cross-linked with intraparticular reversibility; and/or h) surface-active substances are bonded to the coating material; and/or i) targetable structure substances, preferably hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, e.
g. , histones, interleukins, lipoproteins, for example, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acid or their fragments or structural elements or mixtures thereof, if necessary with the addition of chemotherapeutic agents, preferably cytostatic agents, are bonded to the coating material;
and/or j) cross-linking, or bonding of targetable structures and/or surface-active substances can be combined as desired; and/or k) they are surrounded by cage molecules, preferably consisting of subunits of clathrins and/or synthetic analogues.
By another aspect of this invention, a process is provided for the preparation of nanocrystalline magnetic particles consisting of a magnetic iron oxide core of Fe304, gamma-Fez03 or mixtures thereof and an envelope chemisorbed to the core, the process comprising the steps of synthesizing iron oxide cores, and enveloping the iron oxide cores, under biometric conditions, with a composition comprising natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights of 500 Da to 250,000 Da combined.
By one variant of this broad process aspect of this invention, free functional groups of the coating material are activated and intraparticularly cross-linked by adding a cross-linking agent, and modifying the cross-linked coating by the addition of surface-active substances, or targetable structure substances.
By another variant of this broad process aspect of this invention, or of the above variant thereof, the process includes the step of adding chemotherapeutic agents and/or 5 residual low-molecular weight residues.
By another variant of this broad process aspect of this invention, or of the above variants thereof, natural or synthetic glycosaminoglycans or their derivatives which are used as a coating substance, comprises chondroitin sulphate, dermatan sulphate, heparin sulphate or synthetic analogues thereof, hyaluronic acid or synthetic analogues thereof, 10 or heparin or synthetic analogues.
By yet another variant of this broad process aspect of this invention, or of the above variants thereof, the process includes the step of activating functional groups of the coating material of the magnetite stock solution by means of water-soluble carbodiimide derivatives, or in a two-phase system by lipophilic carbodiimides, and 15 subsequently purifying and isolating the activated magnetite. By one specific variation thereof, the process includes the step of adding bifunctional cross-linking agents which are common in biochemistry and chemistry of natural products and/or adding mono-, di-, tri-, and oligoamines, synthetic or biological oligopeptides, or reduced or oxidized glutathione to the activated magnetite solution, then non-invasively separating non-reacted educt by dialysis and finally adjusting magnetite with its hydrophilia altered and/or cross-linked to the desired final concentration. By another specific variation thereof, the process includes the step of adding and admixing surface-active substances to the activated magnetite solution. By a further specific variation thereof, the process includes the step of adding targetable structure substances to the activated magnetite solution. By a variation of that further specific variation, the targetable structure substances comprise hormones, cholesterol, lipids, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, e.g., histones, interleukins, lipoproteins, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements as well as mixtures thereof. By still another variation of that further specific variation, the process includes the step of admixing chemotherapeutic agents to the magnetite solution or to the magnetite. By yet a further variation thereof, the process includes the step of admixing and adding lower molecular weight residues which influence the physiological distribution pattern of the particles to the activated magnetite solution. By a specific further variation thereof, the process includes the step of adding clathrin to the activated magnetite solution.
Especially preferred variants of the process of this aspect of this invention are characterized by the following features:
a) natural or synthetic glycosaminoglycans or derivatives thereof, preferably chondroitin sulphates, dermatan sulphates, heparan sulphates, heparin or synthetic analogues thereof which are used as coating substances; and/or b) the functional groups of the coating material of the magnetite stock solution are activated with water-soluble carbodiimide derivatives or in a two-phase system with lipophilic carbodiimides, and thereafter the activated magnetite is purified and isolated; and/or c) bifunctional cross-linking agents which are common in biochemistry and in the chemistry of natural products and/or mono-, di-, tri-, and oligoamines, synthetic or biological oligopeptides, or reduced or oxidized glutathione are added to the activated magnetite solution, with non-reacted educts being non-invasively separated by dialysis and the magnetite cross-linked and/or with its hydrophilia altered is then adjusted to its desired final concentration; and/or d) surface-active substances are admixed and added to the activated magnetite solution; and/or e) the activated magnetite solution which are added thereto are targetable structure substances, preferably hormones, cholesterol, lipids, tumour lectins, adhesion proteins, fusion proteins, transport proteins, or transport units, alkaline proteins, e.g., histones, interleukins, lipoproteins, for example, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, compounds which contain residual sugar, which plays a role in complement and immune recognition, as well as ribonucleic or deoxyribonucleic acids, as well as fragments or structural elements thereof, or a mixture thereof; and/or f) chemotherapeutic agents are admixed to the activated magnetite solution, or are added to the magnetite; and/or g) lower molecular weight substances affecting the physiological distribution pattern of the particles are admixed and added to the activated magnetite solution, and/or h) clathrin is admixed to the activated magnetite solution.
Furthermore, by another broad aspect, the invention also provides diagnostic and/or therapeutic agents comprising isotope-doped iron oxide cores of Fe304, gamma Fe203 or their mixtures, which are surrounded by a biodegradable coating of natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights between 500 - 250,000 Da.
By one variant of this broad aspect of the invention, the coating material is covalently cross-linked by appropriate cross-linking agents and/or is modified by specific additives.
By another variant of this broad aspect of the invention, or of the above-described variant thereof, the particles have core diameters which are smaller than the dimensions of the magnetic singular domains.
By yet another variant of this broad aspect of the invention, or of the above described variants thereof, the particles are also provided with common pharmaceutical adjuvants for injectable solutions and/or enteral agents as stable aqueous colloid dispersive solutions which are 0.2 ~.m filterable. By one specific variation thereof, the magnetic particles can be heat sterilized.
By yet a further variant of this broad aspect of the invention, or of the above described variants thereof, the iron oxide core is doped with 6Li, 5'Fe, 6'Ni, Ni, 6'Zn, Zn, Mn, ~Ru, ioiRu nsCd~ u9sn~ izisb~ i2~I~ isiEu~ issGd~ is6Gd, or ls'Gd.
By yet still another variant of this broad aspect of the invention, or of the above-described variants thereof, the natural or synthetic glycosaminoglycans are chondroitin sulphate, dermatan sulphate, or heparin or synthetic analogues thereof, or other heparinoids.

Ig By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the coating material after preparation of enveloped magnetic iron oxide particles, is additionally cross-linked by cross-linkers common in biochemistry and chemistry of natural products.
By another variant of this broad aspect of the invention, or of the above-described variants thereof, the glycosaminoglycan is heparan sulphate or a synthetic analog thereof, or the glycosaminoglycan is hyaluronic acid or a synthetic analog thereof.
By a further variant of this broad aspect of the invention, or of the above-described variants thereof, the mono-, di-, tri-, and oligoamines and/or synthetic or biological oligopeptides/proteins are bonded to the coating material.
By yet a further variant of this broad aspect of the invention, or of the above-described variants thereof, the reduced or oxidized glutathione is bonded, as well as intraparticularly reversibly cross-linked to, the coating material.
By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the surface-active substances are bonded to the coating material.
By another variant of this broad aspect of the invention, or of the above-described variants thereof, the targetable structure substances are bonded to the coating material.
By one specific variation thereof, the targetable structure substances comprise of hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins or transport units, alkaline proteins, e.g., histones, interleukins, lipoproteins, for example, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements or mixtures thereof. By another specific variation thereof, the magnetic particles include additional chemotherapeutic agents, e.g., cytostatic agents.
By a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the cross-linking or bonding of targetable structures and/or surface-active substances are combined.

By yet a still further variant of this broad aspect of the invention, or of the above-described variants thereof, the magnetic particles are surrounded by cage molecules. By a specific variation thereof, the cage molecules comprise subunits of clathrin and/or synthetic analogues.
Especially preferred variants of the diagnostic and/or therapeutic agents, according to aspects of this invention, are characterized by the following features:
a) the core, if doped, is preferably doped with 6Li, 5'Fe, 6'Ni, 'S'Eu, 'S'Gd;
and/or b) the natural or synthetic glycosaminoglycans are chondroitin sulphates, dermatan sulphates, heparan sulphates, heparin or their synthetic analogues which are cross linked with bifunctional cross-linking agents which are common in biochemistry and the chemistry of natural products and/or are cross-linked with mono-, di-, tri-, and oligoamines, synthetic and biological oligopeptides, reduced or oxidized glutathione; and/or c) surface-active substances are bonded to the cross-linked coating; and/or d) the cross-linked coating which are bonded thereto are targetable structure substances, preferably hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins, transport units, alkaline proteins, e.g., histones, interleukins, lipoproteins, for example, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, compounds which contain residual sugar which plays a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids as well as fragments or structural elements thereof or mixtures thereof, if necessary, with addition of chemotherapeutic agents, preferably cytostatics; and/or e) subunits of clathrin surround the enveloped particles in cage form; and/or f) the magnetic particles are incorporated in liposomes, chylomicrons, cells, cell organelles bacterial, virus shells and are surrounded by lipids in the form of a double layer.
The invention in another aspect provides the use of the diagnostics and/or therapeutic agents as described generically, sub-generically and specifically hereinabove in the preparation of diagnostic and/or therapeutic agents for radiotherapy, hyperthermia, chemotherapy and MR diagnosis as well as biomagnetic probes.
In contrast to the particles of the present state of the art, the nanocrystalline particles according to aspects of this invention described herein are constructed according 5 to the modular principle. Based on a parent substance which consists of a magnetic iron oxide core and an envelope of, for example, glycosaminoglycan, they can be modified in solution according to the purpose of application.
Such modifications can be achieved by covalent or non-covalent adding, activation and/or cross-linking as well as by combinations thereof. It has proven helpful first to 10 cross-link the coating material in vitro before coupling the targetable structures.
Coupling of targetable structures without prior cross-linkage might cause destabilization of the magnetic particles. The parent substance can thus be supplemented with targetable structures and the addition of chemotherapeutic agents for application to selective tumour diagnostics/therapy. In addition, there are many possibilities to modify the parent 15 substance itself. This diversity is associated with a wide spectrum of applications. One can differentiate between the parent substance which is identified and taken up by the reticuloendothelial system (RES) due to its particular nature, and serves, for example, MRI contrasting of liver and spleen tumours (metastases). Targeting of other organs/tissues can be achieved after parenteral administration by extending retention in 20 the blood stream, acceleration of the extravasation and organ or tissue selection by attachment of targetable structures. Uptake by the RES is perceived as a reaction parallel to the reaction with the target organ and can be influenced by size, charge, surface, hydrophobia and pre-saturation of the RES.
The chemisorbed coating substances used are biodegradable substances of animal/human origin which are capable of stabilizing the iron oxohydroxy complex and of ensuring a synthesis of the coated iron oxide under biomimetic conditions.
Precipitation of the iron oxide at alkaline level can thus be avoided. The stock sots can be prepared from blood isotones, for example, by the addition of mannitol and stable NaCI. The size of the emerging particles can be controlled during synthesis, for example, by proper choice of the chondroitin/iron ratio, and must not be adjusted to a fixed value by fractionation procedures.
No organic solvents are used in the preparation process according to aspects of this invention. In contrast to dextran magnetites, particle stability is guaranteed in several organic solvents. The substances are useful in many diagnostic and therapeutic areas. Chemical bonding of coating molecules to the iron oxide core, as described in the European patent application (publication number 0284549), is avoided in order to guarantee the biological degradability. The animal/human origin of the coating material is reflected in good tolerance. "Pro drug" forms and coating substances with pH
dependent fracture sites can also be produced.
The envelope of glycosaminoglycan with carboxylic acid groups ensures solid chemisorption to the iron oxide core (and with the sulphuric acid and the residual N-acetyl also provides for the sufficient stability of the sol). The iron oxide core in this context is sufficiently shielded so that, in contrast to the majority of the conventional formulations, physiological incompatibilities with parenteral application are adequately compensated.
Possible coating materials are water-soluble glycosaminoglycans, e. g. , chondroitin sulphates, keratin sulphates, dermatan sulphates, heparin, hyaluronic acid, heparan sulphates or synthetic analogues. (Hyaluronic acid is often considered as one of the four primary representatives of glycosaminoglycans [mucopolysaccharide polysulphuric acid esters] as mentioned before). Hyaluronic acid differs from chondroitin sulphate, keratin sulphate, dermatan sulphate and heparan (heparin) sulphate in that it does not contain any residual sulphuric acid and is itself not protein conjugated, but it promotes the proteoglycan aggregation. Glycosaminoglycans together with connecting tissue proteins form the ubiquitously present proteoglycans.
In the context of conventional wet chemical procedures of magnetite preparation, acid Fe(II) : Fe(III) solutions are adjusted at increased temperature to a pH
value at an alkaline level by adding lye (e.g. NaOH,) or NH40H. The forming magnetite precipitates in the presence of the coating agent and entrains it, or the coating agent is added after formation of the magnetites. In some cases, ultrasound is applied during magnetite synthesis and leads to reduction in size of the forming particles (see, Menz et al. WO 90/295). The subsequent steps include peptization, neutralization, purification, addition of stabilizers as well as filling and heat sterilization, if possible, in the final receptacle.
While these processes which differ slightly from each other by initial concentrations, reaction temperatures, rates of adding, choice of the coating agent and the processing steps, the processes described here result in formation of glycosaminoglycan-coated magnetites in a neutral medium (pH7) and in ambient temperatures. Also, ultrasonic treatment is unnecessary. The pH value can be held constantly neutral in a chemostat. The alkaline range in which many coating agents and biological materials are hydrolytically instable can be avoided. This proves to be a significant advantage, since the number of possible decomposition products can thus be kept low.
According to broad aspects of this invention, the glycosaminoglycans of the magnetites provide for almost complete shielding of the surface, so that specific interactions are minimized or avoided. Extensive hydrate coating is under discussion for erythrocytes as a result of the poly-N-acetyl-lactosamines (J. Vitala and J.
Jarnefelt, TIBS, October 1985, 392-395). It appears to be important for macrophage uptake of the parent substance that the coating surrounding the core does not cause any specific reactions, yet can still be activated by the immune system so that the opsonized particles are taken up by the macrophages. Choice and derivation of the coating (charge and hydrophobia) in addition to size, offer a possibility, for other purposes, passively to influence extracelluar interaction of the magnetites. Moreover, direct drug targeting is possible by attachment of specific recognition tags (e.g., monoclonal antibodies, hormones, guide proteins). Similar to the wide range of liposome applications, the interactions of parenterally applied particles so far have only been phenomenologically understood and are a current field of biochemical, pharmacological and medical research.
According to broad aspects of this invention, the described magnetic iron oxide particles which (with or without additional doping) together with a coating of glycosaminoglycan and the derivatives thereof or the synthetic analogous substances provide the advantage that the coating materials are biodegradable products.
Iron is liberated from the iron oxide following the intracellular uptake and is incorporated into the iron pool (haemoglobin, iron storage proteins). The normal iron pool of an adult is increased only insignificantly by one single application of 10-30 ,u mol/kg body weight, a dose which is normally sufficient for diagnostic MR tomography. Extreme pH
values can be avoided with the underlying synthesis, except for dissolution of the salts of iron so that the susceptibility to hydrolysis and/or pH induced chemical modification of the coating material is of minor importance.
Iron oxide particles which are dissolved in water with a coating of glycosaminoglycan represent the stock solution. The physical and pharmacological parameters of the particles can be influenced in situ by attaching lower and higher molecular substances/ligands/guide structures and by cross-linking as well as inclusion in and addition to cells or cell components, as described in the following examples. This actually enables adaptation to specific diagnostic and/or therapeutic problems. The approach has been so chosen that the pre made magnetites (made according to Example 1 or 2 hereinafter), are transformed by activation of reactive groups into a form which allows coupling in a physiologically acceptable form to the appropriate ligands/spacers/substrates/targetable structures.
(e) DESCRIPTION OF THE FIGURES
In the accompanying drawings:
FIG. 1 is an MR image, namely an axial slice of a mouse liver before (baseline) and 1, 10, and 30 minutes after application of 10 ~. mol/kg of chondroitin magnetite (chondroitin-4-sulphate) prepared according to Example 1;
FIG. 2 is a graph of the relative intensity (MRI liver signal/MRI reference signal) in percent of the baseline value following intravenous administration of 20 ~, mol/kg chondroitin magnetite as ordinate as a function of time as abscissa;
FIG. 3 is a graph of the relative intensity (MRI liver signal/MRI reference signal) 24 h after intravenous administration of chondroitin magnetite (prepared according to Example 1) with Sl (liver/reference) as ordinate;

FIG. 4 is an X-ray diffractogram of a lyphophilized transferrin magnetite conjugate prepared by periodate activation of chondroitin-4-sulphate-magnetite and subsequent coupling to transferrin; and FIG. 5 is an electron microscopic representation of the transferrin magnetite conjugate.
(f7 AT LEAST ONE MODE FOR CARRYING OUT THE INVENTION
Example 1: (Conventional Synthesis) 18.24 gm chondroitin-4-sulphate (Sigma Type A) is dissolved under heat in 400 ml of distilled water and gassed with nitrogen. 20.6 gm of Fe(II) chloride are dissolved under nitrogen in 210 ml of 1 M Fe(III) chloride solution. The freshly prepared Fe (II)/Fe(III) solution is slowly added drop-wise ( .: 0.5 ml/min) under nitrogen rinse to the 75°C chondroitin sulphate solution so that the precipitate formed at the point of dripping is immediately dissolved. Then, previously degassed 3N NaOH solution is slowly added. The solution is then titrated to pH 10. Immediately thereafter, it is neutralized and boiled for 3 h on the reflux side and is continuously regulated in the process to pH 7. After cooling to room temperature and centrifuging (10 min., RPM) the supernatant solution is diafiltered with 10 1 distilled water over a 3 kDa hollow fibre cartridge and is then reduced in a rotary evaporator to a volume of 250 ma.
The pH value is adjusted to 7. After 0.2 ~,m filtration the ready-to-use solution is autoclaved at 121°C. The yield amounts to 100% relative to the employed iron.
The data of the chondroitin-4-sulphate prepared according to this example is shown hereinbelow in Table 1:
Table 1: Data of chondroitin magnetite (chondroitin-4-sulphate) prepared according to Example 1.
Size: 52 nm (with laser light diffraction) Relaxivity: Tl 401/mmolOs TZ 1201/mmolOs Fe concentration: 0.33 M

Acute tolerance LDSO (mouse) 25 mmol/kg Half life in blood 4.4 min.
(with a dosage of 100 ~, mol/kg in rat) Example 2: (Biomimetic Synthesis) 5.0 gm chondroitin-4-sulphate are dissolved in 200 ma iron III hydroxide sol, corresponding to 6.6 mM Fe (see Jander/Blasius, Lehrbuch dr analytischen and praparativen anorganischen Chemie, S. Hirzel Verlag, Stuttgart). Possible cloudiness is 10 filtered off. A nearly neutralized solution of 1.282 gm ammonium iron(II) sulphate hexahydrate is added dropwise to the solution heated to 37°C, with the pH slowly drifting towards acidic being readjusted with diluted lye to physiological pH
values. The completed magnetite is purified and autoclaved, which provides for accelerated recuperation of possible fault positions of the magnetite crystals.
Example 3: (Doping The procedure was the same as in Example I, except that 5 % of the employed Fe(II) is replaced by 6'Ni as a Mossbauer active isotope for use in the resonant nuclear absorption therapy. 19.57 gm FeCl2 and 1.05 gm 6lNiClZ are to be used instead of the 20.6 gm FeCl2.
Example 4: (Desulphatizingyto Chan a tg he Hydrophilic) 10 ml chondroitin sulphate magnetite according to Example 2 are eluted over a DOWEXTM cation exchanger and are neutralized with appx. 7.5 ml pyridine. The pyridinium salt of chondroitin sulphate magnetite is obtained following lyophilization.
The salt is dissolved in DMSO/ethanol or DMSO/H20 and is maintained at 80 °C for 5 h. After cooling the solution is diluted with water and is adjusted with NaOH
to pH 9-9.5. This adjusted dispersion of desulphated chondroitin magnetite is dialysed against water, ultrafiltered ( 10 kDa) and filtrated under sterile conditions.

Example 5: (Activation of Free Carboxylic Acids) 1 ml chondroitin-4-sulphate magnetite according to Example 2 is diluted 1:10 in distilled water and the pH value is adjusted to 4.5 with HCI. Water-soluble carbodiimide [1-ethyl-3(3-dimethylaminopropyl)-carbodiimide-HCl (EDC.Pierce)] is added under agitation in 1.5-fold surplus quantity (compared to the functional groups to be activated) and the pH value is held constant at 4.5 (1h at 4°C). On completion of reaction, the non-reacted educt is carefully separated by dialysis from the activated magnetite and the substance retained is lyophilized.
The EDC reaction will have to be modified if consequent reactions are to be initiated with the EDC-activated magnetite (e.g. coupling to biogenic ligands) which are not stable at pH 4-5 or would lose biological activity. N-hydroxysulphosuccinimide (Sulfo-NHS, Pierce) must be added to the chondroitin magnetite or the reaction mixture of chondroitin magnetite and ligand and/or cross-linker for the coupling-cross-linking under physiological conditions or in the course of a biomimetic synthesis in order to enhance the stability of the intermediate product at physiological pH values and thus to increase yield and ensure biological activity. A stable intermediate product is obtained by adding sulfo-NHS (see Staros et al., Analytical Biochemistry 156, 220-222, 1986) in contrast to EDC reaction without sulfo-NHS additive where an O-acyl-urea derivative is intermediately formed which requires acidic reaction conditions and is sensitive to hydrolysis. The additional reagent has no influence on the final product. An amide bond is also produced in response to addition of an aminic ligand, and the lower molecular weight reagent is removed from the colloidal solution by dialysis of the ultrafiltration, following mediated condensation between acid and amine components.
Example 6: (Desulphatized EDC) 10 ml desulphatized chondroitin magnetite, prepared according to Example 4, are converted with EDC according to Example 5. This results in activated magnetites which, due to reaction with the acid group of the coating material, is negatively charged or neutral depending on EDC content.

Example 7: (Condensation with Glucosamine) Magnetite solutions, according to Example 5 are mixed with a 1.5-fold surplus quantity of glucosamine. An amide bond results from the condensation reaction between EDC activated carboxylic acid and glucosamine. The resulting magnetite, which has undergone change in hydrophilia, is dialysed against distilled water and adjusted to the desired concentration.
Example 8: (Cross-linking with Ethylenediamine) The procedure was as in Example 7, but bifunctional ethylenediamine is added instead of glucosamine. In order to avoid interparticular cross-linking, the latter is done at lower magnetite concentrations with these concentrations being increased on completion of cross-linkage.
Example 9: (Transferrin Coupling, 10 ml magnetite solution, according to Example 5, with pH adjusted to 7.4, are mixed with 10 ma (10-20 mg/ml) human transferrin which had been transformed into biologically active Fe(III) transferrin using an appx. 10-fold molar surplus of iron (II) in a phosphate citrate buffer. The mixture is agitated for 6 h at 4°C, with the pH value being maintained at the physiological level. This is followed by ultrafiltration at 4°C in order to separate the unbonded transferrin. The chondroitin magnetite which is not cross-linked or bonded with Fe transferrin and the chondroitin magnetite that is bonded with biologically non-active transferrin are separated by CNBr SEPHAROSETM 4 B
(Pharmacia) which had been previously bonded with anti-H-transferrin (see van Ejik and van Noort, J. Clin. Chem. Clin. Biochem., Vol. 14, 475-478, 1976).
This is followed by an assay of content and after adjustment of desired final concentration, by sterile filtration. Yield of iron: transferrin + 1.1 to 5.1 (w/w).
Storage is at 4 °C, with the stability relative to native transferrin being significantly improved, as is known for immobilized proteins. Other targetable structures, e.g., monoclonal antibodies and/or cytostatic agents, can be coupled to the EDC
activated magnetite (Example 5), according to the same reaction pattern.

Example 10: (Hydrophobic Ma ng etite) 1 gm chondroitin magnetite lyophilized according to Example 1 is mixed with ma DMSO/acetone and acidified. Dicyclohexylcarbodiimide (a 5-fold surplus quantity relative to carboxylic acid) is subsequently added to the solution and is mixed with the lipophilic amine benzylamine in a 2-fold surplus quantity relative to carboxylic acids.
The result is hydrophobic magnetite. The magnetites are 50 nm in size in ethanol (70nm in water) .
Example 11: (Lipid Double Layer Magnetite) The procedure is the same as in Example 7; however phosphatidylethanolamine (cephalin from egg yolk) is stoichiometrically (relative to EDC group) added as the amine component under N2 and maintained at 4°C for 2 h. The cephalin displaces the EDC
with the ethanolamine head group and bonds to the chondroitin magnetite. A
complete double layer is formed by addition of phosphatidylcholine (lecithin) in a 1.5-fold surplus quantity, relative to the cephalin or by a mixture of lecithin and cholesterol. Change of the solvent (EtOH water mixture) proves to be advantageous during the cephalin formation. The ethanol must be removed to enable formation of the double layer.
Example 12: (Clathrin Ca,.
Clathrin is isolated in monomeric form by well known procedures [see Review B. M. F. Pearse & R. A. Crother, "Structure and Assembly of Coated Vesicles", An.
Rev. Biophys. Biophys. Chem. 16, 49-68 (1987)]. Addition of pre-made magnetites (see Examples 1, 2) in Fe : protein = 1:l to 5:1 ratio (w/w) is followed by condensation of the clathrin monomers to typical clathrin cages by reduction of pH to 6.2 and addition to MgCl2. Magnetites which were not incorporated in the process of condensation and empty clathrin cages are chromatographically separated from bonded cages.
Example 13: (Small Ma neg tites) Preparation of chondroitin magnetite according to Example 2 is so modified that the reaction solution is repumped through a filter cartridge with some of the magnetite particles appearing in the filtrate. In this manner, especially small magnetites are produced which are highly suitable for intravenous lymphography and have a longer half life in blood.
Example 14: (Erythrocyte Inclusion) Erythrocytes are separated from the other blood components in the customary fashion and are mixed with physiological NaCI solution. The premade magnetites are added (0.56 mg/ma Fe to 1 mg/ma protein). The solution is subsequently pressurized with 50 bar nitrous oxide at 37 °C according to Disclosure DE 3812816 Al. Following pressure relief, non-included magnetite is separated by centrifugation. The result is envelopes which have taken up magnetite in a non-erythrocyte process.
As indicated hereinbefore, FIG. 1 is an MR image of an axial slice of a mouse liver before (baseline) and 1, 10, and 30 minutes after application of 10 ~, mol/kg of chondroitin magnetite (chondroitin-4-sulphate), prepared according to Example 1. CSI
GE 2T. Magnetites are taken up by the RES in clinically-intact liver tissue which takes up the magnetites so that the signal is extinguished. Tumour or tumour metastases have no RES and consequently cannot take up magnetite and do not exhibit any variation in signal intensity. The metastases are not visible in the baseline image, but are clearly distinguishable from the intact liver tissue in response to a dosage of as little as 10 ~, mol/kg.
FIG. 2, shows the time dependence in the form of a graph of the kinetics of magnetite degradation in rat liver for chondroitin magnetite prepared according to Example 1 (chondroitin-4-sulphate). Represented is the relative intensity (MRI
liver signal/MRI reference signal) in percent of the baseline value following intravenous administration of 20 ~c mol/kg chondroitin magnetite as a function of time.
FIG. 3, shows the dosage dependence after application in the form of a graph.
Represented is the relative intensity (MRI liver signal/MRI reference signal) 24 h after intravenous administration of chondroitin magnetite (prepared according to Example 1) as a function of the applied dosage.

FIG. 4 shows a powder diffraction system in the form of an X-ray diffractogram of a lyphophilized transferrin magnetite conjugate prepared by periodate activation of chondroitin-4-sulphate-magnetite and subsequent coupling to transferrin. The lines can be labelled cuboid in the manner of a spinet structure. The X-ray diffractogram shows 5 the magnetite structure remains intact even after periodate activation.
FIG. 5 is an electron microscopic representation of the transferrin magnetite conjugate as described for FIG. 4. The magnification is 1 : 300,000. The electron edited magnetite core is clearly visible. No aggregates appear.

Claims (45)

1. Nanocrystalline magnetic particles consisting of a magnetic iron oxide core of Fe3 0 4, gamma-Fe2 0 3 or mixtures thereof, and a chemisorbed coating on said core, wherein the composition of said coating material comprises natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights of 500 Da to 250,000 Da combined.
2. The nanocrystalline magnetic particles according to claim 1, wherein said coating material is covalently cross-linked by cross-linking agents and/or is modified by specific additives.
3. Magnetic particles according to claim 1 or claim 2, wherein said particles have core diameters which are smaller than the dimensions of the magnetic singular domains.
4. Magnetic particles according to claims 1 to 3, wherein said particles are provided with common pharmaceutical adjuvants for injectable solutions and/or enteral agents as stable aqueous colloid dispersive solutions which are 0.2 µm filterable.
5. Magnetic particles according to claim 4, which can be heat sterilized.
6. Magnetic particles according to claims 1 to 5, wherein said iron oxide core is doped with 6Li, 57Fe, 61Ni, Ni, 67Zn, Zn, Mn, 99Ru, 101Ru 113Cd, 119Sn, 121Sb, 127I, 151Eu, 155Gd, 156Gd, or 157Gd.
7. Magnetic particles according to claims 1 to 6, wherein said natural or synthetic glycosaminoglycans are chondroitin sulphate, dermatan sulphate, heparin or synthetic analogues thereof, or other heparinoids.
8. Magnetic particles according to claims 1 to 7, wherein said coating material, after preparation of enveloped magnetic iron oxide particles, is additionally cross-linked by cross-linkers which are common in biochemistry and chemistry of natural products.
9. Magnetic particles according to claims 1 to 7, wherein said glycosaminoglycan is heparan sulphate or a synthetic analog thereof.
10. Magnetic particles according to claims 1 to 7, wherein said glycosaminoglycan is hyaluronic acid, or a synthetic analog thereof.
11. Magnetic particles according to claims 1 to 10, wherein mono-, di-, tri-, and oligoamines and/or synthetic and biological oligopeptides/proteins are bonded to said coating material.
12. Magnetic particles according to claims 1 to 11, wherein reduced or oxidized glutathione is bonded, as well as intraparticularly reversibly cross-linked to, said coating material.
13. Magnetic particles according to claims 1 to 12, wherein surface-active substances are bonded to said coating material.
14. Magnetic particles according to claims 1 to 13, wherein targetable structure substances are bonded to said coating material.
15. Magnetic particles according to claim 14, wherein said targetable structure substances comprise hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, histones, interleukins, lipoproteins, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements, or mixtures thereof.
16. Magnetic particles according to claim 15, including the addition of chemotherapeutic agents.
17. Magnetic particles according to claim 16, wherein said chemotherapeutic agents comprise cytostatic agents.
18. Magnetic particles according to claims 1 to 17, wherein said cross-linking or bonding of targetable structures and/or surface-active substances are combined.
19. Magnetic particles according to claims 1 to 18, wherein said magnetic particles are surrounded by cage molecules.
20. Magnetic particles according to claim 19, wherein said cage molecules comprise subunits of clathrin and/or synthetic analogues.
21. Diagnostic and/or therapeutic agents comprising isotope-doped iron oxide cores of Fe3 0 4, gamma-Fe2 0 3 or their mixtures, which are surrounded by a biodegradable coating of natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights between 500 - 250,000 Da.
22. Diagnostic and/or therapeutic agents according to claim 21, wherein said coating molecules are cross-linked by cross-linking agents, and are modified by surface-active substances, targetable structure substances or lower molecular weight residues.
23. Diagnostic and/or therapeutic agents according to claim 21 or claim 22, wherein said iron oxide core is doped with 6Li, 57Fe, 61Ni, Ni, 67Zn, Zn, Mn, 99Ru, 101Ru, 113Cd, 119Sn, 121Sb, 127I, 151Eu, 155Gd, 156Gd, or 157Gd.
24. Diagnostic and/or therapeutic agents according to claims 21 to 23, wherein said natural or synthetic glycosaminoglycans are chondroitin sulphate, dermatan sulphate, heparan sulphate, heparin or its synthetic analogues which are cross-linked by bifunctional cross-linking agents which are common in biochemistry and chemistry of natural products and/or are cross-linked with mono-, di-, tri-, and oligoamines, synthetic or biological oligopeptides, or reduced or oxidized glutathione.
25. Diagnostic and/or therapeutic agents according to claims 24, including surface-active substances which are bonded to said cross-linked coating.
26. Diagnostic and/or therapeutic agents according to claims 24, including targetable structure substances which are bonded to said cross-linked coating material.
27. Diagnostic and/or therapeutic agents according to claim 26, wherein said targetable structure substances comprise hormones, cholesterol, lipids, ether lipids, proteins, monoclonal antibodies, lectins, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, histones, interleukins, lipoproteins, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements or mixtures thereof.
28. Diagnostic and/or therapeutic agents according to claim 27, including the addition of chemotherapeutic agents.
29. Diagnostic and/or therapeutic agents according to claim 28, wherein said chemotherapeutic agents comprise cytostatic agents.
30. Diagnostic and/or therapeutic agents according to claims 21 to 29, including chronologically-variable combination of cross-linking substances, bonding of targetable structures, surface-active substances and chemotherapeutic agents.
31. Diagnostic and/or therapeutic agents according to claims 21 to 30, wherein said coated particles are surrounded in cage form by subunits of clathrin and/or analogues thereof.
32. Diagnostic and/or therapeutic agents according to claims 21 to 31, wherein said magnetic particles are incorporated in liposomes, chylomicrons, cells, cell organelles, bacteria and virus coatings and are surrounded by lipids in the form of a double layer.
33. Process for the preparation of nanocrystalline magnetic particles consisting of a magnetic iron oxide core of Fe3 0 4, gamma-Fe2 0 3 or mixtures thereof and an envelope chemisorbed to the core, comprising the steps of: (a) synthesizing iron oxide cores; and (b) enveloping said iron oxide cores under biometric conditions with a composition comprising natural or synthetic glycosaminoglycans and/or derivatives thereof with molecular weights of 500 Da to 250,000 Da combined.
34. Process according to claim 33, wherein said free functional groups of said coating material are activated and intraparticularly cross-linked by adding a cross-linking agent, and modifying said cross-linked coating by the addition of surface-active substances, or targetable structure substances.
35. Process according to claim 33 or claim 34, including the steps of adding chemotherapeutic agents and/or residual low-molecular weight residues.
36. Process according to claims 33 to 35, wherein said natural or synthetic glycosaminoglycans or their derivatives, comprise chondroitin sulphate, dermatan sulphate, heparin sulphate or a synthetic analogue thereof, hyaluronic acid or a synthetic analogue thereof, or heparin or synthetic analogues thereof as coating substance.
37. Process according to claims 33 to 36, including the step of activating functional groups of said coating material of said magnetite stock solution by means of water-soluble carbodiimide derivatives, or in a two-phase system by lipophilic carbodiimides, and subsequently purifying and isolating the activated magnetite.
38. Process according to claim 37, including the steps of: adding bifunctional cross-linking agents which are common in biochemistry and chemistry of natural products and/or mono-, di-, tri-, and oligoamines, synthetic or biological oligopeptides, or reduced or oxidized glutathione to said activated magnetite solution; non-invasively separating non-reacted educt by dialysis; and adjusting magnetite with its hydrophilic altered and/or cross-linked to the desired final concentration.
39. Process according to claim 37 or claim 38, including the step of adding and admixing surface-active substances to said activated magnetite solution.
40. Process according to claims 37, claim 38 or claim 39, including the step of adding targetable structure substances to said activated magnetite solution.
41. Process according to claim 40, wherein said targetable structure substances comprise hormones, cholesterol, lipids, tumour lectins, adhesion proteins, fusion proteins, transport proteins and transport units, alkaline proteins, histones, interleukins, lipoproteins, LDL, glycolipids, interferons, tumour-necrosis factors, protein A or adjuvants, residual glycosyl or general sugar residues which play a role in complement and immune recognition as well as ribonucleic or deoxyribonucleic acids or their fragments or structural elements or mixtures thereof.
42. Process according to claim 41, including the step of admixing chemotherapeutic agents to said magnetite solution or to said magnetite.
43. Process according to claims 37 to 42, including the step of admixing and adding lower molecular weight residues which influence the physiological distribution pattern of said particles to said activated magnetite solution.
44. Process according to claims 37 to 43, including the step of adding clathrin to the activated magnetite solution.
45. Use of the diagnostic and/or therapeutic agents according to claims 21 to 30, for the preparation of diagnostic and/or therapeutic materials for radiotherapy, hyperthermia, chemotherapy and MR diagnostics and as biomagnetic probes.
CA002068632A 1991-05-28 1992-05-13 Nanocrystalline magnetic iron oxide particles, processes for their preparation and their use in medical diagnostics and therapy Expired - Fee Related CA2068632C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE4117782A DE4117782C2 (en) 1991-05-28 1991-05-28 Nanocrystalline magnetic iron oxide particles, processes for their production and diagnostic and / or therapeutic agents
DEP4117782.7 1991-05-28

Publications (2)

Publication Number Publication Date
CA2068632A1 CA2068632A1 (en) 1992-11-29
CA2068632C true CA2068632C (en) 2002-10-01

Family

ID=6432830

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002068632A Expired - Fee Related CA2068632C (en) 1991-05-28 1992-05-13 Nanocrystalline magnetic iron oxide particles, processes for their preparation and their use in medical diagnostics and therapy

Country Status (15)

Country Link
US (1) US5427767A (en)
EP (1) EP0516252B1 (en)
JP (1) JP3306810B2 (en)
AT (1) ATE156021T1 (en)
AU (1) AU653220B2 (en)
CA (1) CA2068632C (en)
DE (2) DE4117782C2 (en)
DK (1) DK0516252T3 (en)
ES (1) ES2106134T3 (en)
GR (1) GR3024898T3 (en)
IE (1) IE921595A1 (en)
IL (1) IL101929A (en)
NO (1) NO305105B1 (en)
NZ (1) NZ242669A (en)
ZA (1) ZA923299B (en)

Families Citing this family (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU611458B2 (en) * 1985-03-19 1991-06-13 Randell L. Mills Apparatus providing diagnosis and selective tissue necrosis
US5672334A (en) * 1991-01-16 1997-09-30 Access Pharmaceuticals, Inc. Invivo agents comprising cationic metal chelators with acidic saccharides and glycosaminoglycans
US5707604A (en) * 1986-11-18 1998-01-13 Access Pharmaceuticals, Inc. Vivo agents comprising metal-ion chelates with acidic saccharides and glycosaminoglycans, giving improved site-selective localization, uptake mechanism, sensitivity and kinetic-spatial profiles
EP0580878B1 (en) * 1992-06-01 1996-01-10 BASF Aktiengesellschaft The use of dispersions of magneto-ionic particles as MRI contrast media
JPH08508721A (en) * 1993-03-17 1996-09-17 シリカゲル ゲス.エム.ビー.エイチ Superparamagnetic particles, their manufacturing method and their use
US5411730A (en) * 1993-07-20 1995-05-02 Research Corporation Technologies, Inc. Magnetic microparticles
US5616568A (en) * 1993-11-30 1997-04-01 The Research Foundation Of State University Of New York Functionalized derivatives of hyaluronic acid
AU2419695A (en) * 1994-05-12 1995-12-05 Otsuka Pharmaceutical Co., Ltd. Contrast medium for magnetic resonance imaging
DE4419256A1 (en) * 1994-06-01 1995-12-07 Rau Freiherr Von Nagell Helmut Intravenous admin of iron
DE4421159C1 (en) * 1994-06-20 1995-08-24 Thomas Bruns Use of ferric oxide for treating immune deficiency
DE4428851C2 (en) * 1994-08-04 2000-05-04 Diagnostikforschung Inst Nanoparticles containing iron, their production and application in diagnostics and therapy
EP0783325B2 (en) 1994-09-27 2004-03-31 Amersham Health AS Contrast agent
DE19500665A1 (en) * 1995-01-12 1996-07-18 Axel Prof Dr Haase Process for the spatially resolving imaging of an area of a biological object with the help of electromagnetic rays using contrast media
DE19508772C2 (en) * 1995-03-01 1998-01-29 Schering Ag Methods and connections for the detection of analytes by means of remanence measurement and their use
US6106866A (en) * 1995-07-31 2000-08-22 Access Pharmaceuticals, Inc. In vivo agents comprising cationic drugs, peptides and metal chelators with acidic saccharides and glycosaminoglycans, giving improved site-selective localization, uptake mechanism, sensitivity and kinetic-spatial profiles, including tumor sites
US5695901A (en) * 1995-12-21 1997-12-09 Colorado School Of Mines Nano-size magnetic particles for reprographic processes and method of manufacturing the same
GB9600427D0 (en) * 1996-01-10 1996-03-13 Nycomed Imaging As Contrast media
AU716667B2 (en) 1996-01-10 2000-03-02 Nycomed Imaging As Contrast media
DE19600744A1 (en) * 1996-01-11 1997-07-17 Werner Alois Prof Dipl Kaiser Magnetic substance for local hyperthermic treatment of mainly small tumors
US6368586B1 (en) 1996-01-26 2002-04-09 Brown University Research Foundation Methods and compositions for enhancing the bioadhesive properties of polymers
US5985312A (en) * 1996-01-26 1999-11-16 Brown University Research Foundation Methods and compositions for enhancing the bioadhesive properties of polymers
GB2311138A (en) * 1996-03-15 1997-09-17 Nycomed Imaging As ESR-enhanced MRI using magnetic particles as contrast agents
US6986942B1 (en) 1996-11-16 2006-01-17 Nanomagnetics Limited Microwave absorbing structure
US6713173B2 (en) * 1996-11-16 2004-03-30 Nanomagnetics Limited Magnetizable device
US20060003163A1 (en) * 1996-11-16 2006-01-05 Nanomagnetics Limited Magnetic fluid
AU9661598A (en) * 1997-09-10 1999-03-29 Yale University Radiation dosimetry with magnetic resonance detectable compounds
US6607829B1 (en) 1997-11-13 2003-08-19 Massachusetts Institute Of Technology Tellurium-containing nanocrystalline materials
US6375634B1 (en) 1997-11-19 2002-04-23 Oncology Innovations, Inc. Apparatus and method to encapsulate, kill and remove malignancies, including selectively increasing absorption of x-rays and increasing free-radical damage to residual tumors targeted by ionizing and non-ionizing radiation therapy
US6207392B1 (en) * 1997-11-25 2001-03-27 The Regents Of The University Of California Semiconductor nanocrystal probes for biological applications and process for making and using such probes
US6428811B1 (en) 1998-03-11 2002-08-06 Wm. Marsh Rice University Temperature-sensitive polymer/nanoshell composites for photothermally modulated drug delivery
US6699724B1 (en) 1998-03-11 2004-03-02 Wm. Marsh Rice University Metal nanoshells for biosensing applications
US7101575B2 (en) 1998-03-19 2006-09-05 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Production of nanocapsules and microcapsules by layer-wise polyelectrolyte self-assembly
EP1159951A1 (en) * 2000-06-02 2001-12-05 Societe Des Produits Nestle S.A. Use of exogenous lactic bacteria strain against Actinomyces naeslundii-related diseases
AU773799B2 (en) * 1998-08-12 2004-06-10 Societe Des Produits Nestle S.A. Incorporation of exogenous lactic bacteria into the oral microflora
EP0988864A1 (en) * 1998-09-23 2000-03-29 Luboldt, Wolfgang, Dr.med. Dipl.-Phys. Contrast agent for magnetic resonance tomography
US6470220B1 (en) * 1999-03-29 2002-10-22 The Regents Of The University Of California Diagnosis and treatment of cancers using in vivo magnetic domains
US7871597B2 (en) 1999-04-09 2011-01-18 Amag Pharmaceuticals, Inc. Polyol and polyether iron oxide complexes as pharmacological and/or MRI contrast agents
JP4970678B2 (en) 1999-06-10 2012-07-11 マックス−プランク−ゲゼルシャフト・ツア・フェルデルング・デア・ヴィッセンシャフテン・エー・ファオ Encapsulation of crystals by multilayer coating
WO2001037721A2 (en) * 1999-11-22 2001-05-31 The Research Foundation Of State University Of New York Magnetic nanoparticles for selective therapy
US6530944B2 (en) 2000-02-08 2003-03-11 Rice University Optically-active nanoparticles for use in therapeutic and diagnostic methods
JP5074652B2 (en) * 2000-02-08 2012-11-14 ライスユニバーシティ Optically active nanoparticles used in therapeutic and diagnostic methods
WO2001067105A1 (en) * 2000-03-06 2001-09-13 Dade Behring Marburg Gmbh Carriers coated with polysaccharides, their preparation and use
US20050013775A1 (en) * 2000-06-01 2005-01-20 Kotov Nicholas A. Bioconjugates of nanoparticles as radiopharmaceuticals
US6689338B2 (en) 2000-06-01 2004-02-10 The Board Of Regents For Oklahoma State University Bioconjugates of nanoparticles as radiopharmaceuticals
DE10042023C2 (en) * 2000-08-08 2003-04-10 Biognostic Ag Capsules that encapsulate solid particles of signal-generating substances and their use in bioassays for the detection of target molecules in a sample
US7077859B2 (en) * 2000-12-22 2006-07-18 Avantec Vascular Corporation Apparatus and methods for variably controlled substance delivery from implanted prostheses
US7153703B2 (en) * 2001-05-14 2006-12-26 Board Of Trustees Of The University Of Arkansas N. A. Synthesis of stable colloidal nanocrystals using organic dendrons
ITTO20011191A1 (en) * 2001-12-18 2003-06-18 Itw Ind Components Srl SERVICE DEVICE FOR A REFRIGERATOR AND REFRIGERATOR PROVIDED WITH A DEVICE.
IL162734A0 (en) 2002-02-01 2005-11-20 Ariad Gene Therapeutics Inc Phosphorus-containing compounds & uses thereof
US20030185757A1 (en) * 2002-03-27 2003-10-02 Mayk Kresse Iron-containing nanoparticles with double coating and their use in diagnosis and therapy
US7249604B1 (en) * 2002-05-10 2007-07-31 Vasmo, Inc. Medical devices for occlusion of blood flow
US7670623B2 (en) * 2002-05-31 2010-03-02 Materials Modification, Inc. Hemostatic composition
AU2006203585B2 (en) * 2002-05-31 2008-11-06 Materials Modification, Inc. A hemostatic composition
US7273904B2 (en) 2002-10-03 2007-09-25 The Board Of Trustees Of The University Of Arkansas Nanocrystals in ligand boxes exhibiting enhanced chemical, photochemical, and thermal stability, and methods of making the same
GB2393728A (en) * 2002-10-04 2004-04-07 Nanomagnetics Ltd Magnetic nanoparticles
WO2004040593A1 (en) * 2002-10-30 2004-05-13 Council Of Scientific And Industrial Research Process for preparing nanosized acicular magnetic maghemite phase iron oxide particles
US7560160B2 (en) * 2002-11-25 2009-07-14 Materials Modification, Inc. Multifunctional particulate material, fluid, and composition
US20040109824A1 (en) * 2002-12-06 2004-06-10 Hinds Kathleen Allison Particles for imaging cells
AU2003900335A0 (en) * 2003-01-22 2003-02-13 Sirtex Medical Limited Microparticles for selectively targeted hyperthermia
US7229600B2 (en) * 2003-01-31 2007-06-12 Nanoproducts Corporation Nanoparticles of rare earth oxides
DE10331439B3 (en) * 2003-07-10 2005-02-03 Micromod Partikeltechnologie Gmbh Magnetic nanoparticles with improved magnetic properties
US7007972B1 (en) 2003-03-10 2006-03-07 Materials Modification, Inc. Method and airbag inflation apparatus employing magnetic fluid
US6982501B1 (en) 2003-05-19 2006-01-03 Materials Modification, Inc. Magnetic fluid power generator device and method for generating power
US7118727B2 (en) * 2003-06-16 2006-10-10 General Electric Company Method of making oxide particles
US8651113B2 (en) * 2003-06-18 2014-02-18 Swr&D Inc. Magnetically responsive nanoparticle therapeutic constructs and methods of making and using
US7723311B2 (en) * 2003-06-18 2010-05-25 Nanobiomagnetics, Inc. Delivery of bioactive substances to target cells
US7344491B1 (en) 2003-11-26 2008-03-18 Nanobiomagnetics, Inc. Method and apparatus for improving hearing
GB0316912D0 (en) 2003-07-18 2003-08-20 Oxford Instr Superconductivity Therapeutic treatment
US7200956B1 (en) 2003-07-23 2007-04-10 Materials Modification, Inc. Magnetic fluid cushioning device for a footwear or shoe
TW593158B (en) * 2003-07-31 2004-06-21 Ind Tech Res Inst Magnetic nanoparticle
US7504082B2 (en) * 2003-07-31 2009-03-17 Industrial Technology Research Institute Magnetic nanoparticles comprising Gadolinium and method of fabrication
US7396589B2 (en) * 2003-07-31 2008-07-08 Industrial Technology Research Institute Core-shell magnetic nanoparticles comprising an inner-transition element
US7448389B1 (en) 2003-10-10 2008-11-11 Materials Modification, Inc. Method and kit for inducing hypoxia in tumors through the use of a magnetic fluid
CN100502948C (en) * 2003-10-22 2009-06-24 复旦大学附属中山医院 Magnetic high polymer nanometer micro-balls type medicine carrying system, and its preparation method
US7393924B2 (en) * 2004-01-06 2008-07-01 Franco Vitaliano Smart bio-nanoparticle elements
US20070003482A1 (en) * 2004-01-15 2007-01-04 Koninklijke Philips Electronic, N.V. Ultrasound contrast agents for molecular imaging
EP1718341A2 (en) * 2004-01-20 2006-11-08 Alnis Biosciences, Inc. Articles comprising magnetic material and bioactive agents
FR2867180B1 (en) * 2004-03-02 2006-06-16 Univ Claude Bernard Lyon HYBRID NANOPARTICLES COMPRISING A HEART OF LN203 CARRIERS OF BIOLOGICAL LIGANDS AND PROCESS FOR THEIR PREPARATION
DE102004013637A1 (en) 2004-03-19 2005-10-13 Capsulution Nanoscience Ag Process for the preparation of CS particles and microcapsules using porous templates as well as CS particles and microcapsules
US20070281007A1 (en) * 2004-08-27 2007-12-06 Jacob Jules S Mucoadhesive Oral Formulations of High Permeability, High Solubility Drugs
EP1642648A1 (en) * 2004-09-30 2006-04-05 Roche Diagnostics GmbH Apparatus and method for regulating the temperature of a liquid
EP1827506A2 (en) * 2004-12-17 2007-09-05 Koninklijke Philips Electronics N.V. Targeting contrast agents or targeting therapeutic agents for molecular imaging and therapy
US20070150051A1 (en) * 2005-01-10 2007-06-28 Duke Fiduciary, Llc Vascular implants and methods of fabricating the same
US8287583B2 (en) * 2005-01-10 2012-10-16 Taheri Laduca Llc Apparatus and method for deploying an implantable device within the body
WO2006106507A2 (en) * 2005-04-04 2006-10-12 Given Imaging Ltd. Device and method for pathology detection
US20100041983A9 (en) * 2005-04-04 2010-02-18 Elisha Rabinovitz Device and method for pathology detection
DE102005016873A1 (en) 2005-04-12 2006-10-19 Magforce Nanotechnologies Ag New nano-particle useful for production of composition to treatment and/or prophylaxis of proliferative illnesses, cancer and bacterial infections, where nano-particle is bonded therapeutic substance
CZ301067B6 (en) * 2006-02-24 2009-10-29 Ústav makromolekulární chemie AV CR Iron oxide-based superparamagnetic nanoparticles with modified surface, process of their preparation and use
JP5282031B2 (en) * 2006-05-15 2013-09-04 ドミトリィ ビー カーポティン Magnetic fine particles containing organic matter
US8501159B2 (en) * 2006-12-18 2013-08-06 Colorobbia Italia S.P.A. Magnetic nanoparticles for the application in hyperthermia, preparation thereof and use in constructs having a pharmacological application
EP2128095A4 (en) * 2007-01-05 2011-12-21 Tokyo Inst Tech Spherical ferrite nanoparticle and method for production thereof
WO2008084477A2 (en) * 2007-01-08 2008-07-17 Yossi Gross In-situ filter
ES2409759T3 (en) 2007-01-21 2013-06-27 Hemoteq Ag Medical product for the treatment of stenosis of the channels of the body and for the prevention of threatening stenosis
US9526642B2 (en) * 2007-02-09 2016-12-27 Taheri Laduca Llc Vascular implants and methods of fabricating the same
US8512697B2 (en) * 2007-05-16 2013-08-20 The University Of North Carolina At Chapel Hill Delivery of micro- and nanoparticles with blood platelets
DE102007028777A1 (en) * 2007-06-22 2008-12-24 Carl Zeiss Meditec Ag transport device
US9192697B2 (en) 2007-07-03 2015-11-24 Hemoteq Ag Balloon catheter for treating stenosis of body passages and for preventing threatening restenosis
FR2922106B1 (en) * 2007-10-16 2011-07-01 Univ Claude Bernard Lyon USE OF NANOPARTICLES BASED ON LANTHANIDES AS RADIOSENSITIZING AGENTS.
US9943490B2 (en) * 2007-11-05 2018-04-17 The Trustees Of Princeton University Composite flash-precipitated nanoparticles
WO2009075774A2 (en) * 2007-12-05 2009-06-18 Massachusetts Institute Of Technology Glycosaminoglycan-coated particles and uses thereof
US8246932B2 (en) * 2008-03-28 2012-08-21 General Electric Company Non-radioactive traceable metal isotope-enriched nanoparticles and method of their use for determining biodistribution
JP5663472B2 (en) * 2008-05-16 2015-02-04 ザ・ユニバーシティ・オブ・シドニー Administrable composition
CN102105554A (en) * 2008-06-10 2011-06-22 阿肯色大学托管委员会 Indium arsenide nanocrystals and methods of making the same
US11235062B2 (en) * 2009-03-06 2022-02-01 Metaqor Llc Dynamic bio-nanoparticle elements
US11096901B2 (en) 2009-03-06 2021-08-24 Metaqor Llc Dynamic bio-nanoparticle platforms
WO2010120905A2 (en) * 2009-04-15 2010-10-21 Board Of Trustees Of Michigan State University Novel nano-probes for molecular imaging and targeted therapy of diseases
EP2451496B1 (en) 2009-07-10 2015-07-22 Boston Scientific Scimed, Inc. Use of nanocrystals for a drug delivery balloon
EP2962707B1 (en) * 2009-07-17 2019-07-24 Boston Scientific Scimed, Inc. Drug delivery balloons with improved crystal size and density
CA2781329A1 (en) 2009-11-20 2011-05-26 Toda Kogyo Corporation Magnetic iron oxide fine particles, and magnetic particle-containing water dispersion and process for producing the same
US20110160645A1 (en) * 2009-12-31 2011-06-30 Boston Scientific Scimed, Inc. Cryo Activated Drug Delivery and Cutting Balloons
US9492480B2 (en) * 2010-04-12 2016-11-15 Ramot At Tel Aviv University Ltd. Iron oxide nanoparticles for use in treating non-infectious inflammatory disorders
US10192660B2 (en) * 2010-07-02 2019-01-29 Sri Lanka Institute of Nanotechnology (Pvt) Ltd. Process for preparation of nanoparticles from magnetite ore
EP2611476B1 (en) 2010-09-02 2016-08-10 Boston Scientific Scimed, Inc. Coating process for drug delivery balloons using heat-induced rewrap memory
US10398668B2 (en) 2010-10-19 2019-09-03 The Board Of Regents Of The University Of Oklahoma Glutamate treatment of cardiovascular disorders
US9744235B2 (en) 2010-10-19 2017-08-29 The Board Of Regents Of The University Of Oklahoma Treatment of cardiovascular disorders with targeted nanoparticles
US8740872B2 (en) 2010-10-19 2014-06-03 The Board Of Regents Of The University Of Oklahoma Magnetically-targeted treatment for cardiac disorders
US8669360B2 (en) 2011-08-05 2014-03-11 Boston Scientific Scimed, Inc. Methods of converting amorphous drug substance into crystalline form
US9056152B2 (en) 2011-08-25 2015-06-16 Boston Scientific Scimed, Inc. Medical device with crystalline drug coating
EP2982652A1 (en) * 2014-08-08 2016-02-10 Universität für Bodenkultur Wien Ultra-dense shell core-shell nanoparticles
EP3193841B1 (en) * 2014-09-17 2019-09-18 Erol, Bülent Development of magnetic-micro particles that provide controlled glycosaminoglycan (gag) release and the intravesical usage of it in interstitial cystitis
KR102147451B1 (en) * 2017-07-21 2020-08-24 네오-나노메딕스.인크 Iron Oxide Nanoparticle doped with alkali metals or alkali earth metals capable of gigantic self-heating in the biocompatible magnetic field and Preparation Method thereof
US20200338218A1 (en) * 2017-11-06 2020-10-29 Yasuhiko Tabata Hydrogel particle and method for producing same, cell or cell structure each enclosing hydrogel particle therein, method for evaluating activity of cell using hydrogel particle, and use of hydrogel particle as sustained release preparation
EP3621089B1 (en) 2018-09-10 2021-08-25 Ivoclar Vivadent AG Dental material with magnetic particles with improved colour shielding

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5913521B2 (en) * 1975-06-19 1984-03-30 メイトウサンギヨウ カブシキガイシヤ Method for producing magnetic iron oxide/dextran complex
US4267234A (en) * 1978-03-17 1981-05-12 California Institute Of Technology Polyglutaraldehyde synthesis and protein bonding substrates
US4247406A (en) * 1979-04-23 1981-01-27 Widder Kenneth J Intravascularly-administrable, magnetically-localizable biodegradable carrier
EP0093757A1 (en) * 1981-11-12 1983-11-16 Ulf SCHRÖDER 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
US4731239A (en) * 1983-01-10 1988-03-15 Gordon Robert T Method for enhancing NMR imaging; and diagnostic use
US4735796A (en) * 1983-12-08 1988-04-05 Gordon Robert T Ferromagnetic, diamagnetic or paramagnetic particles useful in the diagnosis and treatment of disease
US4767611A (en) * 1984-07-03 1988-08-30 Gordon Robert T Method for affecting intracellular and extracellular electric and magnetic dipoles
PT81498B (en) * 1984-11-23 1987-12-30 Schering Ag METHOD FOR PREPARING COMPOSITIONS FOR DIAGNOSTICS CONTAINING MAGNETIC PARTICLES
US4758429A (en) * 1985-11-04 1988-07-19 Gordon Robert T Method for the treatment of arthritis and inflammatory joint diseases
JPH07110815B2 (en) * 1985-11-18 1995-11-29 ボ−ド・オブ・リ−ジェンツ、ザ・ユニバ−シティ−・オブ・テキサス・システム Polychelating agent for image and spectral enhancement (and spectral shift)
US5055288A (en) * 1987-06-26 1991-10-08 Advanced Magnetics, Inc. Vascular magnetic imaging method and agent comprising biodegradeable superparamagnetic metal oxides
US4770183A (en) * 1986-07-03 1988-09-13 Advanced Magnetics Incorporated Biologically degradable superparamagnetic particles for use as nuclear magnetic resonance imaging agents
US5141739A (en) * 1986-07-03 1992-08-25 Advanced Magnetics, Inc. Delivery of x-ray contrast agents using receptor mediated endocytosis
DE3709851A1 (en) * 1987-03-24 1988-10-06 Silica Gel Gmbh Adsorptions Te NMR DIAGNOSTIC LIQUID COMPOSITIONS
DE3812816A1 (en) * 1988-04-16 1989-11-02 Lawaczeck Ruediger Dipl Phys P METHOD FOR SOLUBILIZING LIPOSOMES AND / OR BIOLOGICAL MEMBRANES AND THE USE THEREOF
US4965007A (en) * 1988-05-10 1990-10-23 Eastman Kodak Company Encapsulated superparamagnetic particles
EP0670167A1 (en) * 1988-08-04 1995-09-06 Advanced Magnetics Incorporated Receptor mediated endocytosis type diagnostic agents
WO1990001939A1 (en) * 1988-08-19 1990-03-08 Meito Sangyo Kabushiki Kaisha Agent for thermotherapy
EP0361960A3 (en) * 1988-09-29 1992-01-02 RANNEY, David F. Methods and compositions for magnetic resonance imaging
US5213788A (en) * 1988-09-29 1993-05-25 Ranney David F Physically and chemically stabilized polyatomic clusters for magnetic resonance image and spectral enhancement
FR2652741B1 (en) * 1989-10-10 1994-03-04 Laboratoires Care System ANTIMICROBIAL COMPOSITION FOR APPLICATION TO THE SKIN, APPLICATIONS AS A BODY DEODORANT AND CUTANEOUS BACTERICIDE.
DK0526503T3 (en) * 1990-04-10 1997-10-27 Imarx Pharmaceutical Corp Polymers as contrast media for magnetic resonance imaging
FR2660864A1 (en) * 1990-04-13 1991-10-18 Guerbet Sa CONTRAST COMPOSITION, PROCESS FOR PREPARING THE SAME, AND IMAGING APPLICATION.
US5143716A (en) * 1991-02-01 1992-09-01 Unger Evan C Phosphorylated sugar alcohols, Mono- and Di-Saccharides as contrast agents for use in magnetic resonance imaging of the gastrointestinal region

Also Published As

Publication number Publication date
NO921767D0 (en) 1992-05-05
DE4117782C2 (en) 1997-07-17
DK0516252T3 (en) 1997-08-25
DE59208743D1 (en) 1997-09-04
EP0516252B1 (en) 1997-07-30
CA2068632A1 (en) 1992-11-29
US5427767A (en) 1995-06-27
EP0516252A2 (en) 1992-12-02
IE921595A1 (en) 1992-12-02
IL101929A (en) 1994-08-26
NO305105B1 (en) 1999-04-06
NO921767L (en) 1992-11-30
IL101929A0 (en) 1992-12-30
ATE156021T1 (en) 1997-08-15
GR3024898T3 (en) 1998-01-30
DE4117782A1 (en) 1992-12-03
AU653220B2 (en) 1994-09-22
AU1618492A (en) 1992-12-03
JP3306810B2 (en) 2002-07-24
ZA923299B (en) 1992-12-30
ES2106134T3 (en) 1997-11-01
EP0516252A3 (en) 1993-07-28
NZ242669A (en) 1995-08-28
JPH07122410A (en) 1995-05-12

Similar Documents

Publication Publication Date Title
CA2068632C (en) Nanocrystalline magnetic iron oxide particles, processes for their preparation and their use in medical diagnostics and therapy
EP0275285B1 (en) Biodegradable superparamagnetic materials used in clinical applications
US5069216A (en) Silanized biodegradable super paramagnetic metal oxides as contrast agents for imaging the gastrointestinal tract
US5219554A (en) Hydrated biodegradable superparamagnetic metal oxides
US4770183A (en) Biologically degradable superparamagnetic particles for use as nuclear magnetic resonance imaging agents
US6207134B1 (en) Ultrafine lightly coated superparamagnetic particles for MRI
Arruebo et al. Magnetic nanoparticles for drug delivery
US5055288A (en) Vascular magnetic imaging method and agent comprising biodegradeable superparamagnetic metal oxides
Mornet et al. Magnetic nanoparticle design for medical diagnosis and therapy
Sun et al. Bacterial magnetosome: a novel biogenetic magnetic targeted drug carrier with potential multifunctions
US5948384A (en) Particulate agents
EP0861667A2 (en) Particulate agents
US5547682A (en) Preparation and use of novel injectable RES avoiding inorganic particles for medical application
JP2007530422A (en) Magnetic nanoparticle composition and related methods
KR101244140B1 (en) Positively charged superparamagnetic iron oxide nanoparticles, contrast agent using the same and method for preparation thereof
US20150165070A1 (en) Magnetic nanoparticles dispersion, its preparation and diagnostic and therapeutic use
WO1999055230A1 (en) Hyaluronan-based imaging agents
JPH07505638A (en) Magnetic resonance imaging methods and compositions
IE913240A1 (en) Particulate agents
Antonelli et al. Red blood cells constructs to prolong the life span of iron-based magnetic resonance imaging/magnetic particle imaging contrast agents in vivo
BĂLĂIŢĂ et al. Polymer magnetic particles in biomedical applications
Ghutepatil et al. SURFACE FUNCTIONALIZATION OF MNPS FOR MAGNETIC HYPERTHERMIA THERAPY
CA2201158A1 (en) Contrast agent
Watawe et al. Nanotechnology in Drug Administration and Food

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed