WO2009040811A2 - Polymer nanoparticles coated by magnetic metal oxide and uses thereof - Google Patents

Polymer nanoparticles coated by magnetic metal oxide and uses thereof Download PDF

Info

Publication number
WO2009040811A2
WO2009040811A2 PCT/IL2008/001286 IL2008001286W WO2009040811A2 WO 2009040811 A2 WO2009040811 A2 WO 2009040811A2 IL 2008001286 W IL2008001286 W IL 2008001286W WO 2009040811 A2 WO2009040811 A2 WO 2009040811A2
Authority
WO
WIPO (PCT)
Prior art keywords
ligand
trail
tumor
tnf
metal oxide
Prior art date
Application number
PCT/IL2008/001286
Other languages
English (en)
French (fr)
Other versions
WO2009040811A3 (en
Inventor
Shlomo Margel
Benny Perlstein
Chaya Brodie
Original Assignee
Bar-Ilan University
Henry Ford Hospital
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 Bar-Ilan University, Henry Ford Hospital filed Critical Bar-Ilan University
Priority to JP2010526423A priority Critical patent/JP5624471B2/ja
Priority to EP08808087A priority patent/EP2205282A2/en
Priority to RU2010116167/15A priority patent/RU2472530C2/ru
Publication of WO2009040811A2 publication Critical patent/WO2009040811A2/en
Publication of WO2009040811A3 publication Critical patent/WO2009040811A3/en
Priority to IL204712A priority patent/IL204712A/en
Priority to IL212564A priority patent/IL212564A0/en

Links

Classifications

    • 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
    • A61K49/1878Nuclear 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 the nanoparticle having a magnetically inert core and a (super)(para)magnetic coating
    • 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
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal 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
    • 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
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6935Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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

Definitions

  • the present invention provides nanoparticles consisting of a polymer which is a metal chelating agent coated with a magnetic metal oxide, wherein at least one active agent is covalently bound to the polymer, as well as pharmaceutical compositions and uses thereof.
  • Nanoparticles are spherical particles in sizes ranging from a few nanometers up to 0.1 ⁇ m. Polymeric nano-scaled particles of narrow size distribution are commonly formed by controlled precipitation methods or heterogeneous polymerization techniques, e.g., by optimal emulsion or inverse emulsion polymerization methods. Properties of solid materials undergo drastic changes when their dimensions are reduced to the nanometer size regime. It is important to keep in mind that the smaller the particles are, the larger portion of their constituent atoms is located at the surface. Nanoparticles, particularly in sizes below ca. 20 nm, predominantly exhibit surface and interface phenomena that are not observed in bulk materials, e.g., lower melting and boiling points, lower sintering temperature and reduced flow resistance.
  • nano-scaled particles may provide neat solutions to a variety of problems in materials science, such as composite materials, catalysis, three dimensional structures and photonic uses, and can further be used in biomedical applications such as specific cell labeling and separation, cell growth, affinity chromatography, diagnostics, specific blood purification by hemoper fusion, drug delivery and controlled release (Bockstaller et al., 2003; Hergt et al., 2004; Margel et al., 1999).
  • materials science such as composite materials, catalysis, three dimensional structures and photonic uses
  • biomedical applications such as specific cell labeling and separation, cell growth, affinity chromatography, diagnostics, specific blood purification by hemoper fusion, drug delivery and controlled release (Bockstaller et al., 2003; Hergt et al., 2004; Margel et al., 1999).
  • biomedical applications such as specific cell labeling and separation, cell growth, affinity chromatography, diagnostics, specific blood purification by hemoper fusion, drug delivery and controlled release (
  • nano-scaled particles of different surface chemistry e.g., variety of surface functional groups such as hydroxyl, carboxyl, pyridine, amide, aldehyde and phenyl chloromethyl
  • Such nanoparticles have been designed for various industrial and medical applications, e.g., enzyme immobilization, oligonucleotide and peptide synthesis, drug delivery, specific cell labeling and separation, medical imaging, biological glues and flame retardant polymers (Bunker et al, 1994; Szymonifka and Chapman, 1995; Margel et al, 1999; WO 2004/045494; Galperin et al, 2007).
  • Magnetic iron oxide i.e., magnetite and maghemite
  • nanoparticles are the main particles that have been investigated for biomedical applications, e.g., magnetic hyperthermia, magnetic drug targeting, magnetic cell separation and as MRI contrast agents (Lacoste et al, 1993; Green-Sadan et al, 2005; Leemputten and Horisberger, 1974; Hergt et al, 2004).
  • Magnetic iron oxides nanoparticles are non-toxic and biodegradable, and have already been approved for clinical use as MRI contrast agents. These nanoparticles are usually prepared by adding to an aqueous solution containing stoichiometric concentrations of ferrous and ferric ions, and a polymeric stabilizer such as dextran, wherein a base, e.g., NaOH or ammonia, is added until basic pH (usually above 8.0) is reached. The obtained coated magnetic iron oxide nanoparticles are than washed by different ways, e.g., by magnetic columns or dialysis.
  • a base e.g., NaOH or ammonia
  • WO 99/062079 and corresponding EP 1088315Bl of the same Applicant, herewith incorporated by reference in their entirety as if fully disclosed herein, disclose new uniform magnetic gelatin/iron oxide composite nanoparticles, formed by controlled nucleation of iron oxide onto an iron ion chelating polymer, e.g., gelatin, dissolved in an aqueous solution, followed by stepwise growth of thin layers of iron oxide films onto the gelatin/iron oxide nuclei.
  • These magnetic nanoparticles can be prepared in a very narrow size distribution and in sizes ranging from about 10 nm up to 100 nm.
  • the present invention provides a nanoparticle consisting of a polymer which is a metal chelating agent coated with a magnetic metal oxide, wherein at least one active agent is covalently bound to the polymer.
  • the present invention provides pharmaceutical compositions comprising nanoparticles as defined above and a pharmaceutically acceptable carrier, as well as various methods of use.
  • compositions of the present invention may be used, inter alia, for detection of a tumor; reducing or inhibiting the growth of a tumor or for reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed; reducing or inhibiting the growth of a tumor and monitoring the size thereof; and evaluating responsiveness of tumor cells to treatment with a candidate compound.
  • these compositions may be used for detection of a site of inflammation and treatment of said inflammation, as well as for treatment of type 2 diabetes, obesity and anorexia.
  • the present invention provides a nanoparticle consisting of a polymer which is a metal chelating agent coated with a magnetic metal oxide, wherein at least one agent having an anti-tumor activity selected from a peptide, a peptidomimetic, a polypeptide or a small molecule is bound to the outer surface of the magnetic metal oxide.
  • the present invention further provides pharmaceutical compositions comprising these nanoparticles and a pharmaceutically acceptable carrier, for use in reducing or inhibiting the growth of a tumor, as well as various methods of use.
  • Figs. 1A-1D show transmission electron microscopy (TEM) micrographs of gelatin/iron oxide magnetic composite nanoparticles of increased average diameter, prepared as described in Example 1, by repeating the thin magnetic coating process during the growth step 4, 5, 6 and 7 times (IA, IB, 1C and ID), respectively.
  • TEM transmission electron microscopy
  • Fig. 2 shows a histogram of the diameter of gelatin/iron oxide composite nanoparticles prepared as described in Example 1 and dispersed in water.
  • Figs. 3A-3B show high resolution TEM (HTEM) (3A) and electron diffraction (ED) (3B) picture of gelatin/iron oxide magnetic composite nanoparticles prepared as described in Example 1.
  • HTEM high resolution TEM
  • ED electron diffraction
  • Fig. 4 shows X-ray diffraction (XRD) pattern of gelatin/iron oxide magnetic composite nanoparticles prepared as described in Example 1.
  • Fig. 5 shows mossbauer spectrum of gelatin/iron oxide magnetic composite nanoparticles prepared as described in Example 1.
  • Fig. 6 shows room temperature magnetization (VSM) loop obtained for gelatin/iron oxide magnetic composite nanoparticles prepared as described in Example 1.
  • VSM room temperature magnetization
  • Fig. 7 illustrates the nucleation step of the preparation of fluorescent dye- labeled gelatin/iron oxide magnetic composite nanoparticles, as described in Example 2.
  • Fig. 8 shows the stability of free tumor necrosis factor-related apoptosis- inducing ligand (TRAIL) vs. TRAIL conjugated to gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL), at 1O 0 C during 35 days.
  • TRAIL free tumor necrosis factor-related apoptosis- inducing ligand
  • NP-TRAIL gelatin/iron oxide magnetic composite nanoparticles
  • Figs. 9A-9B show the apoptosis in human A 172 glioma cells (9A) and in glioma spheres established from primary HF2020 tumors (9B) induced by gelatin/iron oxide magnetic composite nanoparticles (NP), free TRAIL (100 ng/ml) and TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-
  • TRAIL 10 ng/ml
  • Fig. 10 shows the effect of free TRAIL and TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) on the apoptosis of glioma spheroids established from the human glioma specimens HF1254, HF1308 and HF2020. Spheroids were plated in 24-well plates and were treated with medium (Control), TRAIL (100 ng/ml), gelatin/iron oxide magnetic composite nanop articles (NP) and TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL, 10 ng/ml). Cell death was determined after 24 h of treatment using LDH assay. The results are the mean ⁇ SE of triplicates in two different experiments. Fig.
  • TRAIL free TRAIL
  • NP-TRAIL TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles
  • NP TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles
  • NP Gelatin/iron oxide magnetic composite nanoparticles
  • PBS PBS
  • Fig. 12 shows the cytotoxic effect of TRAIL-conjugated to non-fluorescent or fluorescent dye-labeled gelatin/iron oxide magnetic composite nanoparticles on Al 72 cells.
  • Al 72 cells were incubated for 5 h with control nanoparticles (NP- Control), TRAIL-conjugated non-fluorescent nanoparticles (NP-TRAIL), control rhodamine-labeled nanoparticles (NPR-Control) or TRAIL-conjugated rhodamine- labeled nanoparticles (NPR-TRAIL).
  • Cell apoptosis was determined using propidium iodide staining and FACS analysis, and the results are the mean ⁇ SE of three different experiments.
  • Fig. 13 shows the specific internalization of TRAIL-conjugated rhodamine- labeled gelatin/iron oxide magnetic composite nanoparticles (NPR-TRAIL) into glioma cells as compared to normal astrocytes.
  • NPR-TRAIL were incubated with A 172 cells and with normal astrocytes for 30 min, and the immunofluorescence of the cells was determined using a confocal microscopy. The results represent one out of three experiments which gave similar results.
  • Fig. 14 shows synergistic effect of ⁇ -irradiation and TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) on U87, A 172, U251 and LN-18 glioma cell lines.
  • Cells were incubated with NP-TRAIL (5 ng TRAIL/ml) or with gelatin/iron oxide magnetic composite nanoparticle (NP) alone for 24 hr, or ⁇ -irradiated (10 Gy for 2 h) and then treated with NP-TRAIL (NP- TRAIL+Rad, 5 ng TRAIL/ml) or with NP alone (NP-Rad) for 24 h.
  • NP-TRAIL ⁇ -irradiation and TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles
  • Fig. 15 shows that both cRGD peptide (cRGD) and cRGD peptide- conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-cRGD) induce autophagy, i.e., an increase of punctuated staining, in glioma U251 cells, compared with control or gelatin/iron oxide magnetic composite nanoparticles (NP) only- treated cells.
  • cRGD cRGD peptide
  • NP-cRGD cRGD peptide- conjugated gelatin/iron oxide magnetic composite nanoparticles
  • Fig. 16 shows the effect of TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) on apoptosis of cervical carcinoma cell line
  • the various cell lines were incubated with TRAIL (100 ng/ml), NP-TRAIL (50 ng TRAIL/ml), nanoparticles (NP) alone or PBS (Control) for 24 h, and data is shown as mean ⁇ SE.
  • Figs. 17A-17B show synergistic effect of ⁇ -irradiation and TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) on glioma stem cells spheroids established from the tumor specimens 2355 (17A) and 2303 (17B).
  • Cells were treated with either TRAIL (100 ng/ml), the nanoparticles (NP) alone (Control) or NP-TRAIL (50 ng/ml) for 24 h, the supernatants were then collected and LDH analysis was performed.
  • IR irradiation
  • TRAIL irradiation-induced TRAIL
  • Ovarian cancer cells were treated with medium alone, IL-12 (50 ng/ml), the nanoparticles (NP, 50 ⁇ g/ml) alone or NP-IL- 12 (50 ng bound IL-12/ml), incubated for 24 h and were then analyzed for cell death using the LDH assay.
  • Fig. 19 shows tumor sections from rats treated with either TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) or the nanoparticles (NP) alone.
  • Human U251 glioma cells were employed as xenografts in rat brains and tumors were allowed to develop for 7 days at which time either NP or NP-TRAIL were intracranially injected at the site of the tumor.
  • TRAIL were examined 7 days after treatment, and cell apoptosis was determined using TUNEL staining (brown staining), which specifically detects apoptotic cells.
  • Fig. 20 shows the effect of TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) on survival of human U251 implanted rats.
  • the nanoparticles (NP) alone, NP-TRAIL or PBS were injected directly into the tumor 7 days post tumor cell implantation, and animals were observed for signs of distress and/or morbidity and were euthanized at that time.
  • Figs. 21A-21C show the effect of TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) (21C) vs. the nanoparticles (NP) alone (21B) or PBS (21A) administration on tumor volume at time of morbidity. Rats were perfused with formalin at time of euthanasia, brains were harvested and H&E staining was performed. Representative tumor volume slices are shown. Number above series of pictures represents the day of euthanasia.
  • Fig. 22 shows that TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) administration leads to a decrease in overall tumor burden as compared with the nanoparticles (NP) alone or PBS administration.
  • Rats were intracranially implanted with human U251 cells and after 7 days, NP-TRAIL, NP or PBS were administered. Animals were euthanized at day 21, and brain tissue was harvested and sectioned for volume determination. Slides were photodoc- umented with identical settings for all slides used in this determination. Volume was determined by measuring greatest width and length of tumor for every 15 th 5 ⁇ m slice. Volume for each slice is determined and multiplied by the number of slices to the next measured slice until edge of tumor is achieved.
  • Fig. 23 shows the ability of gelatin/iron oxide magnetic composite nanoparticles (NP) to track to sites of tumor growth.
  • Rhodamine-labeled NP were implanted in the contralateral hemisphere of rat brains 7 days after U251 tumor cell implantation. Four days later, animals were euthanized, and brains were harvested and snap frozen for sectioning and imaging.
  • Panels A, D and G show images of NPR in the corpus collosum part close to the site of the NPR injection; panels B, E and H shows the site of NPR implantation; and panels C, F and I show images of NPR in the corpus collosum part close to the site of tumor cells.
  • Magnification 4X panels A, B, C); 1OX (panels D, E, F); and 2OX (panels G, H, I).
  • NP-TRAIL TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles
  • Fig. 25 shows increased tumor cell destruction in rats receiving TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) vs. the nanoparticles (NP) or PBS only.
  • Animals were implanted with human U251n cells on day 0 and were administered with PBS, NP or NP-TRAIL on day 7. Animals were euthanized on day 14 and brain tissue processed and hemotoxylin and eosin stained.
  • PBS panels a-b
  • NP panels c-e
  • NP-TRAIL panels f-h.
  • Figs. 26A-26B show that TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL) (26B) induce lower signal intensity both at the margin and inside the tumor, compared with the nanoparticles (NP) alone (26A), as shown by MRI 5 indicating that NP-TRAIL arrive at the borders and inside human glioma xenograft implanted within nude rats.
  • U251n human glioma cells were implanted in nude rats, and NP or NP-TRAIL were intracranially injected in the contra lateral side of the tumor 11 days later.
  • MR images were obtained 8 days later.
  • Each Figure shows 4 consecutive sections of MRI, each section has 4 images with different echo time (TE).
  • Fig. 27 shows the ability of TRAIL-conjugated rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR-TRAIL) to be taken up by tumor cells in vivo.
  • Rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR) alone or NPR-TRAIL were implanted directly within the tumor mass 7 days after GFP-U251 tumor cells implantation. Four days later, animals were euthanized, and brains were harvested and snap frozen for sectioning and imaging (red color and green color indicate the presence of NPR and GFP- U251 tumor cells, respectively).
  • NPR-TRAIL are not only found in areas of tumor mass but also colocalized with tumor cells (panel I). However, NPR alone are found in areas of tumor mass but are not colocalized with tumor cells (panels D, E and particularly F). Control animals were treated with PBS.
  • Fig. 28 shows that cRGD peptide-conjugated rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR-cRGD) migrate towards tumor cells in vivo.
  • NPR-cRGD (10 ⁇ l containing 0.05 mg nanoparticles bound to about 2 ⁇ g cRGD peptide) were injected to the contra-lateral side of the nude rat brains 7 days after GFP-U251 tumor cells implantation.
  • Four days later animals were euthanized, and brains were harvested and snap frozen for sectioning and imaging (red color and green color indicate the presence of rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR) and GFP-U251 tumor cells, respectively).
  • Fig. 29 shows that both cRGD peptide-conjugated rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR-cRGD) and TRAIL- conjugated rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR-TRAIL) migrate towards injury site in vivo.
  • Injury was induced by needle injection of PBS at the left side of the brain, and following 4 days, 5 ⁇ l (25 ⁇ g) of rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR) alone, NPR-TRAIL (50 ng bound TRAIL) and NPR-cRGD were injected to the contra-lateral side of the brain.
  • NPR-TRAIL were localized mainly along the site of injury.
  • NPR-cRGD were distributed all over the side of the injured brain and also along the corpus callosum. Figs.
  • 30A-30B show the effect of TRAIL (10-100 ng/ml), gelatin/iron oxide magnetic composite nanoparticles (NP) and TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL, 10-40 ng bound to TRAIL/ml), both in the absence or presence of proteasome inhibitor (PS, 5 mM), on the bladder carcinoma tumor cells TSU-PRl (30A), as well as on both the breast cancer cells MDA-MB and the normal breast cells MCFlOA (30B). Cell death was determined after 24 h using LDH assay. 100% cell death was determined in Triton X-100- treated cells and data normalized.
  • the present invention provides a nanoparticle consisting of a polymer which is a metal chelating agent coated with a magnetic metal oxide, wherein at least one active agent is covalently bound to the polymer.
  • the magnetic polymer/metal oxide composite nanoparticles of the present invention are based on the magnetic polymer/metal oxide composite nanoparticles disclosed in WO 99/062079, herewith incorporated by reference in their entirety as if fully disclosed herein; however, further comprising at least one active agent that is covalently bound to the polymer inside the nanoparticle.
  • Such nanoparticles may be prepared by any suitable method known in the art, e.g., the process described in detail in Examples 1-3 hereinafter, namely, by controlled nucleation of a magnetic metal oxide, e.g., iron oxide, onto a metal chelating polymer, e.g., gelatin, to which at least one active agent is covalently bound, wherein said polymer is dissolved in an aqueous solution, followed by stepwise growth of thin layers of the magnetic metal oxide films onto the polymer/metal oxide nuclei. As shown in these Examples, the yield of this is almost 100%.
  • a magnetic metal oxide e.g., iron oxide
  • a metal chelating polymer e.g., gelatin
  • the magnetic nanoparticles of the present invention can be prepared in a very narrow size distribution and in sizes ranging from about 10 nm up to about 100 nm.
  • these Examples particularly show the uniformity, atomic order, magnetic properties and crystalline character of the nanoparticles.
  • the nanoparticles of the present invention are superparamagnetic, i.e., they are magnetized in the presence of a magnetic field, but no remanence is observed in the absence of a magnetic field.
  • the size of the nanoparticles of the present invention is less than
  • the metal chelating polymer used for the preparation of the nanoparticles of the present invention may be a polymer having functional groups capable of binding metal ions, particularly iron ions, selected from amino, hydroxyl, carboxylate, -SH, ether, immine, phosphate or sulfide groups.
  • the metal chelating polymer is selected from gelatin, polymethylenimine, chitosan or polylysine, more preferably gelatin.
  • the magnetic metal oxide coating the aforesaid metal chelating polymer is an iron oxide or a ferrite derived from an iron oxide.
  • the iron oxide may be a magnetite, maghemite, or a mixture thereof, and the ferrite is an oxide of the formula (Fe 5 M) 3 O 4 , wherein M represents a transition metal ion, preferably selected from Zn" , Co , Mn or Ni .
  • the magnetic metal oxide used for the preparation of the nanoparticles of the present invention is iron oxide.
  • the at least one active agent being covalently bound to the metal chelating polymer may be selected, without being limited to, from a fluorescent dye, a contrast agent, a peptide, a peptidomimetic, a polypeptide or a small molecule.
  • the active agent covalently bound to the metal chelating polymer is a fluorescent dye.
  • fluorescent dyes include, without being limited to, rhodamine or fluorescein.
  • the active agent covalently bound to the metal chelating polymer is a contrast agent, namely a compound used to improve the visibility of internal bodily structures in either an X-ray imaging or magnetic resonance imaging (MRI).
  • contrast agents for X-ray imaging include, without being limited to, barium sulfate-based contrast agents that are water insoluble, used in the digestive tract only either swallowed or administered as an enema, and iodine-based water soluble contrast agents, which can be used almost anywhere in the body, in particular, intravenously as well as intraarterially, intrathecally (the spine) and intraabdominally.
  • iodinated contrast agents are diatrizoate (Hypaque 50), metrizoate (Isopaque Coronar 370), ioxaglate (Hexabrix), iopamidol (Isovue 370), iohexol (Omnipaque 350), ioxilan (Oxilan), iopromide and iodixanol (Visipaque 320).
  • the active agent covalently bound to the metal chelating polymer is a peptide or a peptidomimetic.
  • arginine-glycine-aspartic acid (Arg-Gly-Asp; RGD) motif of extracellular matrix components such as fibronectin and vitronectin binds to integi ⁇ ns, and integrin-mediated adhesion leads to intracellular signaling events that regulate cell survival, proliferation and migration.
  • RGD-containing peptides Data obtained by phage display methods screening for RGD-containing peptides have shown their selective binding to endothelial lining of tumor blood vessels. RGD peptides also retard signal transmission, affect cell migration and induce tumor cell regression or apoptosis.
  • RGD peptides By binding to integrin of either endothelial or tumor cells, RGD peptides are capable of modulating in vivo cell traffic by inhibition of tumor cell-extracellular matrix and tumor cell-endothelial cell attachments, which are obligatory for metastatic processes.
  • RGD-containing compounds can interfere with tumor cell metastatic processes in vitro and in vivo.
  • Peptides that are specific for individual integrins are of considerable interest and of possible medical significance.
  • the ⁇ v ⁇ 3 integrin was the first integrin shown to be associated with tumor angiogenesis, and RGD peptides that specifically block the ⁇ v ⁇ 3 integrin show promise as inhibitors of tumor and retinal angiogenesis, of osteoporosis and in targeting drugs to tumor vasculature. Consequently, a great amount of work was invested in designing and producing integrin-binding peptides and peptidomimetics.
  • the peptide or peptidomimetic is thus a cyclic RGD (cRGD) peptide or peptidomimetic, or an acyclic RGD-containing peptide or peptidomimetic.
  • the cRGD peptide is the cRGD peptide of the sequence cyclo (Arg-Gly-Asp-D-Phe-Lys (SEQ ID NO: 1).
  • the active agent covalently bound to the metal chelating polymer is a polypeptide.
  • Tumor necrosis factor-related apoptosis-inducing ligand TRAIL, also referred to as Apo-2 ligand, Apo-2L or TRAIL/Apo2L
  • TNF tumor necrosis factor
  • TNF ⁇ also referred to as TNF
  • TNF ⁇ also referred to as TNF
  • TNF ⁇ TNF ⁇
  • TLlA TNF-like ligand
  • lymphotoxix- ⁇ LT ⁇
  • CD30 ligand CD27 ligand, CD40 ligand, OX-40 ligand
  • 4- IBB ligand Apo-1 ligand (also referred as Fas ligand or CD95 ligand)
  • Apo-3 ligand also referred to as RANK ligand, ODF or TRANCE
  • TALL-I also referred to as BIyS, BAFF or THANK
  • TRAIL is a type II transmembrane protein initially identified and cloned based on the sequence homology of its extracellular domain with CD95L (28% identical) and TNF (23% identical).
  • the native sequence of human TRAIL polypeptide is 281 amino acids long; however, some cells can produce a natural soluble form of the polypeptide, through enzymatic cleavage of the polypeptide extracellular region.
  • TRAIL forms homotrimers that bind the receptor molecules, each at the interface between two of its subunits. Indeed, TRAIL like most of the TNF ligand family occurs in both a membrane-bound and a soluble form, which can possess different bioactivity.
  • soluble TRAIL sTRAIL
  • sTRAIL soluble TRAIL
  • TRAIL has a potent ability to induce apoptosis, in vitro, in a variety of tumor cell lines including colon, lung, breast, prostate, bladder, kidney, ovarian and brain tumors, as well as melanoma, leukemia and multiple myeloma, but not in most normal cells, highlighting its potential therapeutic application in cancer treatment (WO 2004/001009; Wang and El-Deiry, 2003; Ashkenazi et al, 1999; Carlo-Stella et al, 2007; Smyth et al, 2003). There are only very few agents that are truly cancer cell- specific in term of efficacy or cell death induction as TRAIL.
  • TRAIL mRNA is constitutively expressed in a wide range of tissues. Although the main biological function of TRAIL seems to be the induction of apoptosis, the complete physiological role of this ligand is not yet fully understood. It appears likely that TRAIL expression on liver natural killer (NK) cells is regulated by IFN ⁇ secreted from NK cells in an autocrine manner, since a large portion of NK cells constitutively produce both TRAIL and IFN ⁇ in wild-type and T-cell-deficient mice.
  • NK liver natural killer
  • TRAIL has an important role in antitumor surveillance by immune cells, and that it mediates thymocyte apoptosis and it is important in the induction of autoimmune diseases.
  • TRAIL induces apoptosis through interacting with its receptors.
  • four homologous human receptors for TRAIL have been identified, including DR4, KILLER/DR5, DcRl (Trail-R3 TRID) and DcR2 (TRAIL-R4), as well as a fifth soluble receptor called osteoprotegerin (OPG), initially identified as a RANKL/OPGL receptor.
  • Both DR4 and DR5 contain a conserved death domain (DD) motif and can signal apoptosis.
  • DD conserved death domain
  • Decoy receptor 1 DcRl
  • DcR2 has close homology to the extra-cellular domains of DR4 and DR5.
  • DcR2 has a truncated, nonfunctional cytoplasmic DD, while DcRl lacks a cytosolic region and is anchored to the plasma membrane though a glycophospholipid moiety.
  • OPG as a receptor for TRAIL is unclear because the affinity for this ligand at physiological temperatures is very low.
  • TRAIL may allow selective killing of tumor cells only (Wei et al, 2006).
  • TRAIL-resistant cancer cells in various tumors. Resistance of cancer cells to TRAIL appears to occur through the modulation of various molecular targets, which may include differential expression of death receptors. Based on molecular analysis of death-receptor signaling pathways several new approaches have been developed to increase the efficacy of TRAIL, including the administration of conventional cancer drugs or irradiation, in combination with TRAIL (Shankar and Srivastava, 2004; Smyth et al., 2003).
  • the polypeptide is a cytokine, for example, tumor necrosis factor (TNF)- ⁇ , TNF- ⁇ ; a TNF-related cytokine or an interleukin (IL).
  • TNF-related cytokines include TNF- related apoptosis-inducing ligand (TRAIL), TALL-I, the TNF-like ligand TLlA 5 lymphotoxin-beta (LT- ⁇ ), CD30 ligands, CD27 ligands, CD40 ligands, OX40 ligands, 4- IBB ligands, Apo-1 ligands and Apo-3 ligands, with TRAIL being preferred.
  • the cytokine is TRAIL.
  • interleukins include any interleukin that has an anti-tumor activity, with IL- 12, IL-23 and IL-27 being preferred.
  • polypeptide is an enzyme
  • the polypeptide is an antibody such as avastin or remicade.
  • Avastin is a monoclonal antibody against vascular endothelial grow factor (VEGF), which is used in the treatment of cancer for inhibiting the growth of tumors by blocking the formation of new blood vessels.
  • VEGF vascular endothelial grow factor
  • Remicade is a monoclonal antibody used for treatment of autoimmune disorders by binding to TNF ⁇ , one of the key cytokines that trigers and sustains the inflammation response, and preventing it from binding to TNF ⁇ receptors.
  • the polypeptide is a hormone, i.e., a polypeptide hormone such as insulin, obestatin or ghrelin.
  • the active agent covalently bound to the metal chelating polymer is a small molecule.
  • the small molecule is an anthracycline chemotherapeutic agent.
  • the anthracycline chemotherapeutic agent may be any chemotherapeutic agent of the anthracycline family including daunombicin (also known as adriamycin), doxorubicin, epirubicin, idarubicin and mitoxantrone.
  • the anthracycline chemotherapeutic agent is doxorubicin, which is a quinine-containing anthracycline and is the most widely prescribed and effective chemotherapeutic agent utilized in oncology.
  • Doxorubicin is indicated in a wide range of human malignancies, including tumors of the bladder, stomach, ovary, lung and thyroid, and is one of the most active agents available for treatment of breast cancer and other indications, including acute lymphoblastic and myelogenous leukemias, Hodgkin's and non-Hodgkin's lymphomas, Ewing's and osteogenic bone tumors, soft tissue sarcomas, and pediatric cancers such as neuroblastoma and Wilms' tumors.
  • the small molecule is an antifolate drug, i.e., a drug which impairs the function of folic acid.
  • an antifolate drug is methotrexate, which is a folic acid analog that inhibits the enzyme dihydrofolate reductase, and thus prevents the formation of tetrahydrofolate that is essential for purine and pyrimidine synthesis, and consequently leads to inhibited production of DNA, RNA and proteins.
  • antifolate agents include trimethoprim, pyrimethamine and pemetrexed. As antifolates interfere with metabolism of nucleotides, their action specifically targets the fast-dividing cells.
  • the small molecule is an antibiotic.
  • the small molecule is an amine- derived hormone, i.e., a derivative of the amino acids tyrosine and tryptophan.
  • amine-derived hormones include catecholamines, e.g., epinephrine, norepinephrin and dopamine, and thyroxine.
  • the small molecule is a lipid- or phospholipid-derived hormone, i.e., a hormone derive from lipids such as linoleic acid and arachidonic acid and phospholipids.
  • lipid- and phospholipid-derived hormones are the steroid hormones derived from cholesterol, e.g., testosterone and Cortisol, and the eicosanoids, i.e., prostaglandins.
  • the small molecule is an antiinflammatory agent.
  • the anti-inflammatory agent may be selected from a corticosteroid, the alkaloid colchicine that is the standard treatment for gout, or non-steroidal antiinflammatory drugs (NSAIDs) such as, but not limited to, aspirin, choline and magnesium salicylates, choline salicylate, celecoxib, diclofenac, diflunisal, etodolac, fenoprofen calcium, flurbiprofen, ibuprofen, indomethacin, ketoprofen, magnesium salicylate, meclofenamate sodium mefenamic acid, meloxicam, nabumetone, naproxen, oxaprozin, piroxicam, rofecoxib, salsalate, sodium salicylate, sulindac, tolmetin sodium and valdecoxib.
  • NSAIDs non-steroidal antiinflammatory drugs
  • the metal chelating polymer is gelatin
  • the metal oxide is iron oxide
  • the agent covalently bound to the gelatin is selected from (i) a fluorescent dye, preferably rhodamine or fluorescein; (ii) a TNF or a TNF-related cytokine, preferably TRAIL; (iii) an anthracycline chemotherapeutic agent, preferably doxorubicin; (iv) an antifolate drug, preferably methotrexate; or (v) a combination thereof.
  • said at least one agent covalently bound to the gelatin is a fuorescent dye or TRAIL.
  • the nanoparticles defined above may further comprise at least one active agent physically or covalently bound to the outer surface of the magnetic metal oxide, wherein said at least one active agent is the same or different from the at least one active agent cobalently bound to the polymer.
  • said active agent is covalently bound to the outer surface of the magnetic metal oxide.
  • the active agent covalently bound to the outer surface of the magnetic metal oxide is bound, in fact, via a molecule containing a functional group attached to the magnetic metal oxide surface.
  • this molecules comprise a polymer selected from a polysaccharide, more preferably chitosan, a protein, more preferably gelatin or albumin, a peptide, or a polyamines.
  • the active agent covalently bound to the outer surface of the magnetic metal oxide is bound, in fact, via an activating ligand attached to the magnetic metal oxide outer surface.
  • the activating ligand is acryloyl chloride, divinyl sulfone (DVS), dicarbonyl immidazole, ethylene glycolbis(sulfosuccinimidylsuccinate) or m- maleimidobenzoic acid N-hydroxysulfosuccinimide ester.
  • the activating ligand is DVS.
  • the activating ligands may further be attached to the polymer extending outside the metal oxide coating.
  • the active agent may also be physically bound to the outer surface of the magnetic metal oxide. This physical binding is based on non- covalent interactions, e.g., hydrophobic bonds, ionic interactions and hydrogen bonds, between the active agent(s) and the outer surface of the magnetic metal oxide.
  • the active agent(s) of (b) is physically bound to the outer surface of the magnetic metal oxide.
  • the at least one active agent being bound to the outer surface of the magnetic metal oxide may be selected, without being limited to, a peptide, a peptidomimetic, a polypeptide or a small molecule, as defined above for the active agent covalently bound to the polymer.
  • said peptide or peptidomimetic is a cyclic RGD (cRGD) peptide or peptidomimetic, preferably the cRGD peptide of SEQ ID NO: 1, or an acyclic RGD- containing peptide or peptidomimetic;
  • said polypeptide is a cytokine, an enzyme, an antibody, preferably avastin or remicade, or a hormone, preferably insulin, obestatin or ghrelin; said cytokine is selected from tumor necrosis factor (TNF)- ⁇ , TNF- ⁇ , a TNF-related cytokine selected from TNF-related apoptosis- inducing ligand (TRAIL), TALL-I, the TNF-like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), a CD30 ligand, a CD27 ligand, a CD40 ligand, an OX40 ligand, a
  • concentrations of the active agent(s) bound, either covalently or physically, to the surface of the magnetic metal oxide may be controlled by changing binding parameters, e.g., active agent(s) concentration in the process.
  • the present invention provides nanoparticles as defined above, wherein said polymer is gelatin, said magnetic metal oxide is iron oxide, said at least one active agent covalently bound to the polymer is selected from (i) a fluorescent dye, preferably rhodamine or fluorescein; (ii) a TNF or a TNF-related cytokine, preferably TRAIL; (iii) an anthracycline chemotherapeutic agent, preferably doxorubicin; (iv) an antifolate drug, preferably methotrexate; or (v) a combination thereof, and said at least one active agent bound to the outer surface of the magnetic metal oxide is selected from: (vi) a TNF or a TNF-related cytokine, preferably TRAIL; (vii) a cRGD peptide, preferably the cRGD peptide of SEQ ID NO: 1; (viii) an interleukin having an anti-tumor activity, preferably IL-
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising magnetic polymer/metal oxide composite nanoparticles as defined above and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition of the present invention may be used for various biological, medical and therapeutical applications.
  • the pharmaceutical composition of the present application is used for diagnostics, drug stabilization, drug delivery and controlled release of drugs.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein the at least one active agent covalently bound to the polymer is a fluorescent dye. This pharmaceutical composition may be used for tumor detection.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein the at least one active agent covalently bound to the polymer is a contrast agent.
  • This pharmaceutical composition may be used for for X-ray imaging or magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • the present invention thus relates to a method for X-ray imaging or magnetic resonance imaging (MRI) comprising administering to an individual in need said pharmaceutical composition.
  • this composition may further comprise a molecule capable of binding a tumor specific cellular marker bound to the outer surface of the magnetic metal oxide.
  • This composition may be used for tumor detection.
  • the present invention thus relates to a method for detection of a tumor comprising administering to an individual in need the pharmaceutical composition defined above.
  • the term "molecule capable of binding a tumor specific cellular marker" as used herein refers to antibodies or fragments thereof directed to tumor associated antigens; receptors or fragments thereof specific for tumor associated ligands; or ligands of tumor associated receptors.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein the at least one active agent covalently bound to the polymer and said at least one active agent bound to the outer surface of the magnetic metal oxide, the same or different, are selected from TNF- ⁇ , TNF- ⁇ , a TNF-related cytokine selected from TNF- related apoptosis-inducing ligand (TRAIL), TALL-I, the TNF-like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), a CD30 ligand, a CD27 ligand, a CD40 ligand, an OX40 ligand, a 4- IBB ligand, an Apo-1 ligand, or an Apo-3 ligand, an interleukin having an anti-tumor activity selected from IL- 12, IL-23 or IL-27, a cRGD
  • said at least one active agent covalently bound to the polymer is selected from TRAIL, doxorubicin, methotrexate or a combination thereof, and said at least one active agent bound to the outer surface of the magnetic metal oxide is selected from TRAIL, the cRGD peptide of SEQ ID NO: 1, IL-12, or a combination thereof.
  • This pharmaceutical composition may be used for reducing or inhibiting the growth of a tumor, or for reducing or inhibiting the growth of tumor cells remaining at a site in a patient from which a tumor has been surgically removed.
  • the present invention thus further relates to a method for reducing or inhibiting the growth of a tumor or for reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed, comprising administering to said patient said pharmaceutical composition.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein the at least one active agent covalently bound to the polymer is a fluorescent dye or a contrast agent, and said at least one active agent bound to the outer surface of the magnetic metal oxide is selected from TNF- ⁇ , TNF- ⁇ , a TNF-related cytokine selected from TNF-related apoptosis-inducing ligand (TRAIL), TALL-I, the TNF- like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), a CD30 ligand, a CD27 ligand, a CD40 ligand, an OX40 ligand, a 4- IBB ligand, an Apo-1 ligand, or an Apo-3 ligand, an interleukin having an anti-tumor activity selected from IL-12, IL-23 or IL-27, a
  • said at least one active agent bound to the outer surface of the magnetic metal oxide is selected from TRAIL, the cRGD peptide of SEQ ID NO: 1, IL- 12, or a combination thereof.
  • This pharmaceutical composition may be used for reducing or inhibiting the growth of a tumor and monitoring the size thereof.
  • the present invention thus further relates to a method for reducing or inhibiting the growth of a tumor and monitoring the size thereof in a patient comprising administering to said patient said pharmaceutical composition.
  • tumor refers to any tumor such as, without being limited to, brain tumors, preferably glioma, colon cancer, lung cancer, breast cancer, prostate cancer, bladder cancer, kidney cancer, ovarioan cancer, melanoma, leukemia or multiple myeloma, preferably glioma, and metastases thereof.
  • the tumor is glioma.
  • Brain tumors in particular, malignant gliomas, belong to the most aggressive human cancers. Patients with malignant glioma have a poor prognosis because these brain tumors respond poorly to radiation or chemotherapy, the conventional treatments of cancer (Wei et al, 2006).
  • Features responsible for the aggressive character of glioma include rapid proliferation, diffuse growth and invasion into distant brain areas in addition to extensive cerebral edema and high levels of angiogenesis. Patients with malignant gliomas posses a median survival of less than one year, wherein there are only occasional long-term survivors.
  • Malignant gliomas including the anaplastic astrocytoma and glioblastoma multiform, are the most common primary brain tumors.
  • Current treatment options include surgery, radiation therapy and chemotherapy; however, prognosis remains extremely poor and the development of alternative therapeutic approaches is thus highly desirable.
  • Gene therapy has been considered as an innovative therapeutic approach for malignant gliomas, and in the last decade there has been a great interest in the development of delivery systems that will allow the expression of exogenous genes in the central nervous system.
  • TRAIL tumor necrosis factor-related apoptosis-inducing ligand
  • HSV replication- deficient herpes simplex virus
  • the tumor is glioma.
  • the pharmaceutical composition of the present invention when aimed for reducing or inhibiting the growth of a tumor, either with or without monitoring the size thereof, or for reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed, may be be used in combination with radiotherapy.
  • the various methods defined above for (i) reducing or inhibiting the growth of a tumor; (ii) reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed; or (iii) reducing or inhibiting the growth of a tumor and monitoring the size thereof may be performed in combination with radiotherapy.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, wherein said magnetic metal oxide is iron oxide, said at least one active agent covalently bound to the polymer is a fluorescent dye or a contrast agent and said at least one active agent bound to the outer surface of the magnetic metal oxide is an anti-inflammatory agent.
  • This pharmaceutical composition may be used for detection of a site of inflammation and treatment of said inflammation in an individual.
  • the present invention thus relates to a method for detection of a site of inflammation and treatment of said inflammation comprising administering to an individual in need said pharmaceutical composition.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein said at least one active agent covalently bound to the polymer and said at least one active agent bound to the outer surface of the magnetic metal oxide, each independently, is insulin.
  • This pharmaceutical composition may be used for treatment of type 2 diabetes.
  • the present invention thus relates to a method for treatment of type 2 diabetes comprising administering to an individual in need said pharmaceutical composition.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein said at least one active agent covalently bound to the polymer and said at least one active agent bound to the outer surface of the magnetic metal oxide, each independently, is obestatin.
  • This pharmaceutical composition may be used for treatment of obesity.
  • the present invention thus relates to a method for treatment of obesity comprising administering to an individual in need said pharmaceutical composition.
  • the pharmaceutical composition of the present invention comprises magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, wherein said at least one active agent covalently bound to the polymer and said at least one active agent bound to the outer surface of the magnetic metal oxide, each independently, is ghrelin.
  • This pharmaceutical composition may be used for treatment of anorexia.
  • the present invention thus relates to a method for treatment of anorexia comprising administering to an individual in need said pharmaceutical composition.
  • the present invention relates to a method for evaluating responsiveness of tumor cells to treatment with a candidate compound, which comprises contacting cells from a biopsy taken from said tumor with magnetic polymer/metal oxide composite nanoparticles as defined above, preferably gelatin/iron oxide composite nanoparticles, and monitoring the viability of the tumor cells, wherein the active agent bound to the outer surface of the magnetic metal oxide in the nanoparticles is the candidate compound to be evaluated and is selected from TNF- ⁇ , TNF- ⁇ , a TNF-related cytokine selected from TNF-related apoptosis-inducing ligand (TRAIL), TALL-I, the TNF-like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), a CD30 ligand, a CD27 ligand, a CD40 ligand, an OX40 ligand, a 4- IBB ligand, an Apo-1 ligand, or an A ⁇ o-3 ligand, an interleukin
  • the present invention provides a nanoparticle consisting of a polymer which is a metal chelating agent coated with a magnetic metal oxide, wherein at least one agent having an anti-tumor activity selected from a peptide, a peptidomimetic, a polypeptide or a small molecule is bound to the outer surface of the magnetic metal oxide.
  • the various definitions with respect to the size of the nanoparticles having an anti-tumor agent bound exclusively to their outer surface, as well as to the metal chelating polymer and the magnetic metal oxide are identical to those defined with respect to the magnetic nanoparticles defined above, in which at least one active agent is covalently bound to the polymer.
  • the agent having an anti-tumor activity may be either covalently or physically bound to the outer surface of the magnetic metal oxide.
  • the agent having anti-tumor activity may be a peptide, a peptidomimetic, a polypeptide or a small molecule.
  • the agent having anti-tumor activity is a peptide or peptidomimetic such as a cRGD peptide or peptidomimetic, or an acyclic RGD- containing peptide or peptidomimetic.
  • the cRGD peptide is the cRGD peptide of SEQ ID NO: 1.
  • the agent having anti-tumor activity is a polypeptide such as a cytokine, for example, TNF- ⁇ , TNF- ⁇ ; a TNF-related cytokine or an interleukin.
  • TNF-related cytokines include TNF-related apoptosis-inducing ligand (TRAIL), TALL-I, the TNF-like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), CD30 ligands, CD27 ligands, CD40 ligands, OX40 ligands, 4- IBB ligands, Apo-1 ligands, and Apo-3 ligands.
  • the cytokine is TRAIL, IL- 12, IL-23 or IL-27, more preferably TRAIL.
  • the agent having anti-tumor activity is a small molecule.
  • small molecules include anthracycline chemotlierapeutic agents and antifolate drugs, as defined above, preferably doxorubicin and methotrexate, respectively.
  • the present invention provides nanoparticles having an anti-tumor agent bound exclusively to their outer surface, wherein said peptide or peptidomimetic is a cRGD peptide or peptidomimetic, preferably the cRGD peptide of SEQ ID NO: 1, or an acyclic RGD-containing peptide or peptidomimetic; said polypeptide is a cytokine selected from TNF- ⁇ , TNF- ⁇ , a TNF-related cytokine selected from TNF-related apoptosis-inducing ligand (TRAIL), TNF- ⁇ , TNF- ⁇ , TALL-I, the TNF-like ligand TLlA, lymphotoxin-beta (LT- ⁇ ), a CD30 ligand, a CD27 ligand, a CD40 ligand, an OX40 ligand, a 4- IBB ligand, an Apo-1 ligand, or an Apo-3 ligand, preferably TNF- ⁇
  • said polymer is gelatin
  • said magnetic metal oxide is iron oxide
  • said at least one agent having an anti-tumor activity is a TNF or a TNF-related cytokine, an anthracycline chemotherapeutic agent, an antifolate drug or a combination thereof.
  • said at least one agent having anti-tumor activity is TRAIL.
  • the present invention provides a pharmaceutical composition comprising nanoparticles having an anti-tumor agent bound exclusively to their outer surface as defined above and a pharmaceutically acceptable carrier, for use in reducing or inhibiting the growth of a tumor.
  • this pharmaceutical composition comprising nanoparticles having an anti-tumor agent bound exclusively to their outer surface, wherein the at least one agent having an anti-tumor activity is TRAIL, the cRGD peptide of SEQ ID NO: 1, IL- 12, doxorubicin, methotrexate or a combination thereof.
  • the nanoparticles having an anti-tumor agent bound exclusively to their outer surface may be used for reducing or inhibiting the growth of a tumor or for reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed.
  • the present invention thus further relates to a method for reducing or inhibiting the growth of a tumor or for reducing or inhibiting the growth of tumor cells left at a site in a patient from which a tumor has been surgically removed, comprising administering to said patient the aforesaid pharmaceutical composition.
  • these nanoparticles may be used in combination with radiotherapy. Therefore, this method may be performed in combination with radiotherapy.
  • the present invention relates to a method for evaluating responsiveness of tumor cells to treatment with a candidate compound, which comprises contacting cells from a biopsy taken from said tumor with nanoparticles having an anti-tumor agent bound exclusively to their outer surface, as defined above, preferably gelatin/iron oxide composite nanoparticles having an anti-tumor agent bound exclusively to their outer surface, and monitoring the viability of the tumor cells, wherein the antitumor agent bound to the outer surface of the magnetic metal oxide of the nanoparticles is the candidate compound to be evaluated.
  • the pharmaceutical composition provided by the present invention may be prepared by conventional techniques, e.g., as described in Remington: The Science and Practice of Pharmacy, 19th Ed., 1995.
  • the composition may be in solid, semisolid or liquid form and may further include pharmaceutically acceptable fillers, carriers or diluents, and other inert ingredients and excipients. Furthermore, the pharmaceutical composition can be designed for a slow release of the nanoparticles.
  • the composition can be administered by any suitable route, e.g. intravenously, orally, parenterally, rectally, transdermally or topically. The dosage will depend on the state of the patient, and will be determined as deemed appropriate by the practitioner.
  • the route of administration may be any route which effectively transports the active compound to the appropriate or desired site of action, the intravenous route being preferred.
  • a solid carrier is used for oral administration, the preparation may be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a lozenge.
  • a liquid carrier is used, the preparation may be in the form of a syrup, emulsion or soft gelatin capsule. Tablets, dragees or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application.
  • Preferable carriers for tablets, dragees or capsules include lactose, corn starch and/or potato starch.
  • malignant gliomas are the most common primary intracranial tumors in patients.
  • the prognosis of these tumors is poor because they poorly respond to radiation or chemotherapy.
  • the difficulty in differentiating tumor and normal brain tissue, and the unusual ability of gliomas to infiltrate the brain pose a serious challenge in glioma therapy and diagnosis.
  • TRAIL is a transmembrane protein initially expressed at the cell membrane and subsequently derived by proteolytic processing. Soluble TRAIL (sTRAIL) can induce apoptosis in tumor cells of diverse origins while sparing most normal cells. However, while most previous studies have been performed in cell culture, the delivery and efficiency of sTRAIL in vivo is significantly less established and successful. The major hindrance for in vivo sTRAIL treatment is due to its short half-life due to proteolytic cleavage, which therefore requires excess amount of sTRAIL. In vivo sTRAIL treatment is particularly inefficient for glioma tumors due to difficulties in the administration of TRAIL to the brain and to the relative insensitivity of gliomas to TRAIL in vivo.
  • the conjugation of TRAIL both to fluorescent and to non-fluorescent gelatin/iron oxide composite nanoparticles significantly stabilized the sTRAIL, minimizing enzymatic degradation of the sTRAIL and thereby decreasing the amount of sTRAIL essential for apoptosis of tumor cells.
  • This approach allows the selective delivery of sTRAIL to tumor cells for efficient apoptosis.
  • the sTRAIL-conjugated nanoparticles selectively tracked infiltrating tumor cells, exerted cytotoxic effects in vivo, and significantly increased the survival of tumor-bearing animals.
  • Tumor cells which were resistant to the cytotoxic effect of the sTRAIL-conjugated nanoparticles were sensitized by using a combined treatment of low-level of ⁇ -irradiation followed by treatment with the sTRAIL-conjugated nanoparticles.
  • these cells were efficiently treated with nanoparticles to which more than one active agent was conjugated.
  • the active agents used for this purpose were sTRAIL, and other cancer drug(s) such as a cRGD peptide, IL- 12, a cRGD peptide and adriamycin. These agents, in different combinations, were bound either to the magnetic metal oxide surface, to the polymer inside the nanoparticle, or to both.
  • these sTRAIL-conjugated nanoparticles served as a marker for tumor imaging by MRI and/or fluorescence.
  • these imaging modalities in animal models implanted with human glioma tumors, it was demonstrated that the sTRAIL-conjugated nanoparticles migrate to the site of the tumors and accumulate around and within the tumors, while the nanoparticles without bound sTRAIL migrate slower and accumulate only at the periphery of the tumor cells.
  • the nanoparticles to which the sTRAIL was conjugated are used as a vehicle for stabilizing the sTRAIL, diagnosis of the tumor and assisting in targeting the tumors for inducing apoptosis.
  • the sTRAIL-conjugated nanoparticles identify and target infiltrating tumor cells.
  • the ability of the sTRAIL-conjugated nanoparticles to specifically target glioma cells can be also employed for determining the border of the tumors in the brain and for distinguishing between recurrent gliomas and radiation-induced necrosis.
  • the efficiency of the sTRAIL-conjugated nanoparticles for targeting and inducing apoptosis of tumor cells other then gliomas, e.g. carcinoma, breast cancer and lung cancer was demonstrated.
  • the invention will now be illustrated by the following non-limiting Examples.
  • Gelatin/iron oxide magnetic composite nanoparticles of sizes ranging from ca. 5 nm up to 100 nm with narrow size distribution were prepared by nucleation followed by controlled growth of magnetic iron oxide thin films onto gelatin/iron oxide nuclei, as described in detail in WO 99/062079.
  • the nucleation step was based on complexation Of Fe +2 ions to chelating sites of the gelatin, followed by partial oxidation (up to approximately 50%) of the chelated Fe +2 to Fe +3 , so that the water soluble gelatin contained both chelated Fe +2 and Fe +3 ions.
  • Gelatin/iron oxide nuclei were than formed by adding NaOH or, alternatively, ammonia aqueous solution up to ca. pH 9.5.
  • the growth of magnetic films onto the gelatin nuclei accomplished by repeating several times the nucleation step.
  • nanoparticles of 15 nm average dry diameter were prepared by adding FeCl 2 solution (10 mmol/5 ml H 2 O) to 80 ml aqueous solution containing 200 mg gelatin (Sigma), followed by NaNO 2 solution (7 mmol/5 ml H 2 O). After a reaction time of 10 min, NaOH aqueous solution (1 N) was added up to pH 9.5. This procedure was repeated four times, or more, if larger particles were required. The formed magnetic nanoparticles were then washed from excess reagents using magnetic gradient columns. Surface analysis demonstrated the present of gelatin both within and on the surface of the nanoparticle.
  • Figs. 1A-1D demonstrate a transmission electron microscopy (TEM) picture of magnetic nanopaticles of increased average diameter formed by repeating the thin magnetic coating process during the growth step, 4 to 7 times, respectively.
  • Fig. 2 shows that magnetic nanoparticles of 15 nm dry average diameter, prepared as described hereinabove and dispersed in water, posses one narrow population with hydrodynamic average diameter of ca. 100 nm.
  • FIG. 3A-3B show high resolution TEM (HTEM) picture (3A), demonstrating crystalline structure with d-spacing of 0.479 nm, and electron diffraction (ED) picture (3B), representing sharp rings indicating the crystalline character of the magnetic nanoparticles.
  • Fig. 4 shows X-ray diffraction (XRD) pattern of the gelatin/iron oxide magnetic composite nanoparticles, indicating that the crystalline cores of these nanoparticles consist nearly completely of maghemite (7-Fe 2 Os). From x-ray line broadening, one deduces a mean diameter of the magnetic cores of 15 nm.
  • XRD X-ray diffraction
  • FIG. 5 shows mossbauer spectrum of the gelatin/iron oxide magnetic composite nanoparticles, further indicating that these nanoparticles consist of maghemite. It is assumed that magnetite (Fe 3 O 4 ) nanoparticles were first produced by this nucleation and growth process, and were then oxidized to the more thermodynamic stable iron oxide, maghemite.
  • Fig. 6 represents hysteresis loop at room temperature of the maghemite nanoparticles of 15 nm dry diameter, indicating that the M(H) curve of these nanoparticles does not exhibit any coercivity and does not saturate at 10,000 Oe, while the magnetic moment obtained at 10,000 Oe is ca. 41 emu g "1 . Both features are typical of superparamagnetic behavior.
  • Uniform gelatin/iron oxide magnetic composite nanoparticles of various sizes and properties were prepared by changing preparation conditions, e.g., oxidizing reagents, iron salt type, pH and temperature, as previously disclosed in WO 99/062079.
  • Uniform nanoparticles of sizes smaller than 15 nm dry diameter down to 5 nm were prepared by gradual surface dissolution of the 15 nm nanoparticles with acids, e.g. HCl at pH ca. 1.0, or iron chelating ligands such as EDTA and oxalic acid. After achieving the desired diameter, the magnetic nanoparticles were wash into distilled water.
  • Fluorescent dye-labeled gelatin/iron oxide magnetic composite nanoparticles in which the fluorescent dye is mainly entrapped within the magnetic composite nanop articles were prepared as described in Example I 5 substituting the gelatin for gelatin covalently bound to a fluorescent dye, as illustrated in Fig. 7.
  • rhodamine-labeled nanoparticles were prepared by adding slowly 0.5 ml dimethyl sulfoxide (DMSO) containing 5 mg of rhodamine isothiocyanate (RITC) to 20 ml aqueous solution containing 200 mg gelatin. The pH of the aqueous solution was raised to 9.5 by adding NaOH aqueous solution (1 N), and the solution was shaken for 1 h at 6O 0 C.
  • DMSO dimethyl sulfoxide
  • RVC rhodamine isothiocyanate
  • This process involves the covalent binding, via thiourea and/or thiourethane bonds, between the part of the hydroxy! and amine groups of the gelatin and the isothiocyanate of the RITC. Excess of RITC was then removed from the gelatin conjugated rhodamine chains by extensive dialysis (cut off: 12-14000) of the former aqueous solution at 60°C against H 2 O, or by washing through a magnetic column. The volume of the solution was adjusted to 80 ml and the synthesis was then continued as described in Example 1.
  • Drug containing gelatin/iron oxide magnetic composite nanoparticles were prepared as described in Example 1, substituting the gelatin for gelatin covalently bound to the drug.
  • adriamycin Aldrich
  • Divinyl sulfone was added to the gelatin/iron oxide magnetic composite nanoparticles prepared as described in Examples 1-3, wherein the initial [DVS]/[nanoparticles] weight ratio was 4/1, and triethylamine was then added to reach a pH of 10.5, followed by incubation at 60 0 C overnight.
  • An albumin coating on the nanoparticles was performed similarly, substituting the gelatin for bovine or human serum albumin.
  • IL 12 was bound onto the gelatin/iron oxide-DVS magnetic composite nanoparticles similarly, substituting the hTRAIL for IL 12.
  • Human TRAIL and/or cRGD peptide were covalently bound to the gelatin/iron oxide composite nanoparticles prepared according to Examples 1-3 and Example 4(iii) via the carbodiimide activation method, as described in Melamed and Margel (2001). Briefly, 123 mg NHS and 82 mg CDC were added to 20 ml MES buffer (0.01 M at pH 5.0, Sigma) containing 200 mg nanoparticles, and the nanoparticles dispersion was then shaken at room temperature for 3 h.
  • the activated nanoparticles were washed extensively by magnetic gradiant columns with PBS, and 5 ml PBS solution containing 8 mg hTRAIL or 50 mg cRGD peptide were then added to 20 ml of the washed activated nanoparticles dispersion.
  • the suspension was shaken at room temperature for about 8 h, and blocking of residual activated groups was then performed with either glycine (200 mg) or cRGD peptide (50 mg) as described in 5(i) or 5(iii), respectively.
  • Human sTRAIL was bound directly to the gelatin/iron oxide nanoparticles via the carbodiimide activation method, as described by Melamed and Margel (2001).
  • 123 mg NHS and 82 mg CDC wereadded to 15 ml MES buffer (0.1M at pH 5.0) containing 10 mg nanoparticles.
  • the nanoparticles mixture was then shaken at room temperature for ca. 3 h.
  • the activated nanoparticles were washed by magnetic gradient columns.
  • PBS solution (2 ml) containing 0.5 mg sTRAIL was then addedto 8 ml of the washed activated nanoparticles PBS dispersion. The dispersion was then shaken at room temperature for ca. 2 h.
  • concentrations of the bound bioactive agents described in 5(i) to 5(vi) were controlled by changing binding parameters, e.g. hTRAIL concentration.
  • Example 6 In vitro studies: targeting, imaging and apoptosis of glioma cells
  • AU cells were cultured in DMEM supplemented with 10% FBS (Hyclone, Logan, UT), 2 mM L-glutamine and 100 ug/ml streptomycin-penicillin (Invitrogen) at 37 0 C under 5% CO 2 .
  • Cell apoptosis was measured using propidium iodide (PI) staining and analyzed by flow cytometry as described by Riccardi and Nicoletti (2006) as well as by ELISA (Cell Death Detection ELISA Kit) using anti-histone antibodies as described by Blass et al. (2002).
  • Cells (10 6 /ml) were plated in six-well plates at 37°C and treated by the indicated treatments (addition of 10 ⁇ l or less phosphate-buffered saline (PBS) or PBS containing the peptide, e.g. TRAIL, or unbound or peptide bound nanoparticles) for 24 h.
  • PBS phosphate-buffered saline
  • TRAIL unbound or peptide bound nanoparticles
  • Detached cells and trypsinized adherent cells were pooled, fixed in 70% ethanol for 1 h on ice, washed with PBS and treated for 15 min with RNase (50 ⁇ M) at room temperature. Cells were then stained with PI (5 ⁇ g/ml) and analyzed on a Becton-Dickinson cell sorter.
  • LDH lactate dehydrogenase
  • TRAIL free and conjugated TRAIL against degradation
  • 1 ml PBS containing either free TRAIL (100 ng) or TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL, 100 ng TRAIL) were incubated at 1O 0 C, and the concentration of TRAIL was measured during 35 days.
  • Fig. 8 shows that the conjugation of TRAIL to the nanoparticles stabilized the TRAIL so that the original concentration of the conjugated TRAIL was maintained up to 35 days in 1O 0 C.
  • the free TRAIL significantly degraded under these conditions.
  • Human glioma cells A 172 were incubated with either TRAIL (100 ng/ml) or
  • NP-TRAIL (10 ng TRAIL/ml) for 5 hours.
  • Cell apoptosis was determined by propidium iodide staining and analyzed by both flow cytometry (FACS) and the morphological appearance of the cells.
  • Fig. 9A shows that free TRAIL induced apoptosis of about 48% of the cells whereas the NP-TRAIL induced apoptosis of about 57% of the cells and the nanoparticles alone did not induce significant cell apoptosis.
  • the apoptosis activity induced by the conjugated TRAIL was at least 10 times higher than that induced by the free one.
  • TRAIL/ml was further tested using glioma spheroids derived from three different tumors, i.e., HF2020, HF1254 and HF1308. These cultures resemble more the original tumors as they maintain their three dimensional structure and cell-cell interaction. As found, glioma spheroids derived from HF2020, shown in Fig. 9B, and HF 1308 underwent cells apoptosis in response to TRAIL or NP-TRAIL, whereas HF 1254 underwent only a low degree of cell apoptosis. In particular, as presented in Fig. 9B and Fig. 10, both TRAIL and NP-TRAIL significantly increased the level of LDH in the HF2020 and the HF 1308 glioma spheroids.
  • NP-Control non-fluorescent nanoparticles
  • NPR rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles
  • NPR- TRAIL TRAIL-conjugated rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles
  • Apoptosis was determined using propidium iodide staining and FACS analysis. As shown in Fig. 12, the apoptosis induced by NPR- TRAIL (51%) was similar to that induced by NP-TRAIL (45%).
  • NP-TRAIL neuropeptide-binding protein
  • human glioma cells A172 and normal human astrocytes were incubated with NPR-TRAIL (10 ng TRAIL/ml) for 30 min and the cells were viewed and photographed using confocal microscopy.
  • NPR-TRAIL entered the A172 cells within 30 min of treatment and accumulated in the ER/golgi region, whereas very weak fluorescent was observed in the normal astrocytes.
  • NPR that were not conjugated to TRAIL did not enter the cells in both cell types (data not shown).
  • Cells were treated with NP alone, NP-TRAIL (5 ng TRAIL/ml), y-radiation (10 Gy), or a combination of NP-TRAIL (5 ng TRAIL/ml) and y-radiation (10 Gy).
  • NP-TRAIL ng TRAIL/ml
  • y-radiation 10 Gy
  • LC3-GFP LC3-GFP plasmid
  • IC3- GFP appeared throughout the cells
  • autophagic cells punctuate staining was observed.
  • the pattern of LC3-GFP in the control cells treated with NP only and in the cells treated with cRGD peptide-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-cRGD) was examined.
  • U251 cells were transfected with LC3-GFP for 24 h, the cells were treated with cRGD peptide (10 ⁇ g/ml), NP only or NP-cRGD (ca. 4 ⁇ g cRGD/ml) for additional 24 h, and the percentage of cells with punctuated GFP staining was then calculated.
  • cRGD peptide 10 ⁇ g/ml
  • NP only or NP-cRGD ca. 4 ⁇ g cRGD/ml
  • the percentage of cells with punctuated GFP staining was then calculated.
  • about 24 h about 30% of the cRGD-treated cells and about 18% of the NP-cRGD-treated cells exhibited an increased punctuated pattern of LC3-GFP. Contrary to that, only 2-3 % of the control or NP-treated cells exhibited punctuated staining.
  • LN- 18 cells that were resistant to TRAIL treatment were incubated for 24 h with NP-TRAIL, TRAIL and cRGD peptide-conjugated nanoparticles (prepared as described in Example 5(iii)) or adriamycin containing TRAIL and cRGD peptide-conjugated nanoparticles (prepared as described in Examples 3 and 5(iii)).
  • Fig. 16 shows the apoptotic effect of
  • TRAIL 100 ng/ml
  • NP-TRAIL 50 ng TRAIL/ml
  • NP alone PBS (control)
  • PI propidium iodide
  • Glioma stem cells were established from the tumor specimens 2355 and 2303, were grown as spheroids and maintained in culture for two months.
  • the glioma stem cells exhibited self-renewal and differentiated on poly-L-Lysine to astrocytes, neurons and oligodendrocytes.
  • sphroids were placed in a 6-well plate and were treated with TRAIL (100 ng/ml), NP alone or NP-TRAIL (50 ng TRAIL/ml) for 24 h. The supernatants were then collected and LDH analysis was performed.
  • ⁇ -radiation and TRAIL were first irradiated with 5 Gy radiation and after 4 hours were treated with either TRAIL or NP-TRAIL for additional 24 h. Cell death was determined using LDH levels in culture supernatants. The apoptotic effect induced by the different treatments was similar in both glioma stem cell lines and illustrated in Figs. 17A- 17B. Insignificant apoptotic effect was observed for NP only, ⁇ -irradiation alone or free TRAIL. NP-TRAIL induced moderate apoptotic effect; however, both NP- TRAIL and free TRAIL, together with ⁇ -irradiation, induced significant apoptosis.
  • Ovarian cancer cells were treated with medium alone, IL- 12 (50 ng/ml), NP
  • IL-12-conjugated gelatin/iron oxide magnetic composite nanoparticles 50 ⁇ g/ml or IL-12-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-IL- 12, 50 ng IL-12/ml), incubated for 24 h and were then analyzed for cell death using the LDH assay. As shown in Fig. 18, both IL- 12 and NP-IL- 12 induced cell death in the cultured cells, as further reflected in the cell morphology.
  • Example 7 In vivo studies: targeting, imaging and apoptosis of glioma cells
  • U251 cells were obtained from ATCC (Manassas,
  • Rats were allowed to acclimate for one week after arrival prior to use. Prior to tumor cell implantation, animals were anesthetized and prepared for sterile surgery. A lcm long incision was made through the scalp and a 26-gauge needle was used to gently puncture, by twisting, a hole through the skull 2 mm to the right and 2 mm to the dorsal of the midline. The animal was then placed into a stereotaxic device (Kopf, Tujunga, CA), equipped with a micro-manipulator and a syringe holder.
  • Kopf Tujunga, CA
  • the needle was lowered 3 mm into the pre-made hole, raised 0.5 mm, and the tumor volume (5 ⁇ l) was slowly injected over a total of 2.5 min. The needle was left in place for 1 min and was then slowly raised over 1 min. The animal was removed from the stereotaxic device, the hole sealed with bone wax, the scalp was sutured and the animal was monitored for recovery. On day 4, 7 or 10 after tumor implantation, PBS (10 ⁇ l) in absence or presence of control nanoparticles (0.05 mg), TRAIL-conjugated nanoparticles (100 ng TRAIL conjugated to 0.05 mg nanoparticles) or free TRAIL (100, 200 or 800 ng) were implanted in the same manner in the ipsi-lateral or the contra-lateral hemisphere.
  • control nanoparticles 0.05 mg
  • TRAIL-conjugated nanoparticles 100 ng TRAIL conjugated to 0.05 mg nanoparticles
  • free TRAIL 100, 200 or 800 ng
  • Gomori's Iron reaction staining (Sheehan and Hrapchak, 1980) for nanoparticle visualization (blue color) was done utilizing 5 ⁇ m sections from formalin-fixed paraffin embedded tissue. The 5 ⁇ m sections were dried in a 6O 0 C oven for 1 h and routinely deparanf ⁇ inized to ddH 2 O. For all remaining steps, acid cleaned glassware was used. Slides were immersed in equal parts of 20% HCl and 10% aqueous potassium ferrocyanide for 10-20 minutes. Slides were then rinsed well in ddH 2 O and counterstained with Nuclear Fast Red for 2 minutes, followed by dehydration, cleared and then cover slipped. Sections of spleen were used as positive controls for each staining.
  • Staining of apoptotic cells was performed using TUNEL staining (brown staining) that specifically detects apoptotic cells (Shah et al., 2003). Briefly, staining of formalin fixed paraffin embedded tissue was performed utilizing the Apoptag Peroxidase In Situ Apoptosis Detection Kit (Chemicon, Temecula, CA) as per manufacturer's instructions. Briefly, 5 ⁇ m sections from formalin-fixed paraffin embedded tissue were dried in a 6O 0 C oven for 1 h and routinely deparafinized. Tissue was then pretreated with proteinase K (20 ⁇ g/ml) for 15 mins at room temp.
  • Slides were washed two times in ddH 2 O and endogenous peroxidase was quenched in 3.0% hydrogen peroxide in PBS for 5 mins at room temperature. Slides were wash twice in PBS and 75 ⁇ l/5cm of equilibration buffer was added to each slide for at least 10 seconds. Excess liquid was tapped off and 55 ⁇ l/5 cm 2 of TdT enzyme was added. Slides were incubated for 1 hour and were then washed in Stop/wash buffer for 15 seconds with agitation, then 10 mins at room temp. Next, the slides were washed in 3 changes of PBS for 1 min each, followed by incubation with 65 ⁇ l/5 cm 2 anti-digoxignenin conjugate for 30 min.
  • Immunohistological and histochemical images were taken at room temperature using a Nikon Eclipse E800M microscope with XlO, X20 and X40 objectives connected to a Nikon DXM1200C digital camera, and digitized using ACT-IC software on Dell Optiplex GX620 computers.
  • Tiff images were imported into Adobe Photoshop for composite production. Insets are magnifications performed using Photoshop. Fluorescent images were taken at room temperature using a Nikon Cl confocal microscope with X4, XlO and X20 objectives connected to a digital camera. Bitmap images were imported into Adobe Photoshop for composite production.
  • Stereotaxic ear bars was used to minimize movement during the imaging procedure. Rectal temperature was maintained at 37 ⁇ 0.5°C using a feedback controlled water bath.
  • a modified fast low angle shot (FLASH) imaging sequence was employed for reproducible positioning of the animal in the magnet at each MRJ session.
  • MR studies were performed using Tl-, T2- and T2*-weighted MRI scans. For detection of iron oxide labeled cells, scans typically employed are T2*W gradient echoes. Average examination times for the Tl-, T2- and T2*-weighted MRI scans were approximately 9, 13 and 13 minutes, respectively, for in vivo studies of brain tumors.
  • T2 weighted images were obtained using standard two-dimensional Fourier transformation (2DTF) multislice (13-15) multiecho (6 echoes) MRI.
  • 2DTF standard two-dimensional Fourier transformation
  • 13-15 multislicecho
  • a series of six sets of images (13-15 slices for each set) were obtained using TEs of 10, 20, 30, 40, 50 and 60 msec and a TR of 3000 msec.
  • T2* weighted images were obtained using standard multislice (13-15 slices) mum " gradient echo (6 echoes) MRI.
  • a series of six sets of images (13-15 slices for each set) were obtained using TEs of 5, 10, 15, 20, 25 and 30 msec and a TR of 3000 msec.
  • Both the T2 and T2* images were used to measure the T2 and T2* maps.
  • the total time for entire sequence was approximately 20 minutes.
  • heating pad was used. Rectal probe were used to monitor the body temperature.
  • TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles In order to examine the in vivo effect of TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles (NP-TRAIL), two glioma animal models were established. In the first model, human U87 or U251 cells were intracranially implanted into nude mice. In the second model, we employed human glioma cells from fresh operative tumor samples were implanted in nude rats, generating tumors that maintain the original properties of the original tumors, thus providing a system that can predict the response of these tumors to different anti-cancer treatments. 7 (U) TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles induce cell apoptosis in glioma
  • NP-TRAIL gelatin/iron oxide magnetic composite nanoparticles
  • NP alone did not induce a significant degree of cell apoptosis; however, NP- TRAIL induced a large degree of cell apoptosis as determined by TUNEL (brown staining)-positive cells. In the PBS-treated rats, no significant cell apoptosis was indicated as well (data not shown).
  • NP-TRAIL 100 ng TRAIL
  • NP-TRAIL In order to determine the effect of NP-TRAIL on tumor burden, PBS, NP alone or NP-TRAIL were delivered intratumoraly into xenogenic human U251 tumors in the brains of nude rats. Animals were euthanized on day 21 post tumor implant and brains were harvested for sectioning and H&E staining. Brains were cut into 2 mm blocks, processed, and blocks with evident tumor were cut in 5 um sections, with every 15 th section kept and stained with H&E, and tumor volume on this slide determined. Max width and height of tumor on each 15 th slide was measured, and total tumor burden determined by multiplying max WxHX(5xl5) for each slide with evident tumor and these numbers added.
  • NP-TRAIL administration significantly decreased tumor burden as compared with NP alone or PBS. No significant difference was seen between NP alone and PBS administration. 7(v) Tumor tracking ability
  • NP ability of NP to travel from the site of implantation to the site of tumor growth is a useful therapeutic potential of NP, particularly for brain tumor therapy.
  • U251 tumor cells were implanted into the left hemisphere of nude rat brains on day 0 and rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles (NPR) were administered parallel to the tumor implantation but in the contralateral hemisphere 7 days later.
  • NPR rhodamine-labeled gelatin/iron oxide magnetic composite nanoparticles
  • NPR-TRAIL rhodamine-labeled TRAIL- conjugated gelatin/iron oxide magnetic composite nanoparticles
  • NP-TRAIL Tumor destruction
  • nude rats were implanted with U251n tumors on day 0.
  • NP-TRAIL, NP alone or PBS were intraneoplastically administered on day 7, and animals were euthanized and tissue harvested on day 14.
  • NP-TRAIL administration led to the development of large areas of tumor destruction in the lower part of the tumor mass, which were not seen in NP alone- or PBS- administered animals.
  • Many necrotic and apoptotic cells were seen within this area (right panels) following NP-TRAIL administration; however were not seen following NP or PBS administration.
  • Fig. 25 demonstrates high magnification of the areas of tumor destruction following therapy.
  • TRAIL-co njugated gelatin/iron oxide magnetic composite nanoparticles arrive at the borders and inside human glioma xenograft implanted within nude rats as shown by MRI
  • NP-TRAIL induced lower signal intensity both at the margin and inside the tumor, compared to NP only (26A). The lower intensity implies the presence of nanoparticles.
  • NPR alone or NPR-TRAIL were implanted directly within the tumor mass 7 days after GFP-U251 tumor cells implantation. Four days later, animals were euthanized, and brains were harvested and snap frozen for sectioning and imaging (red color and green color indicate the presence of NPR and GFP-U251 tumor cells, respectively). As shown in Fig. 27, whereas NPR alone were found in areas around tumor cells but were not colocalized with tumor cells (panels D, E, F), NPR-TRAIL were found in areas of tumor destruction colocalized with tumor cells (panels G, H, I).
  • NPR-cRGD 10 ⁇ l containing 0.05 mg nanoparticles conjugated to about 2 ⁇ g cRGD peptide
  • NPR-cRGD 10 ⁇ l containing 0.05 mg nanoparticles conjugated to about 2 ⁇ g cRGD peptide
  • NPR-TRAIL 50 ng TRAIL
  • NPR-cRGD NPR-cRGD
  • NPR-cRGD have tracking ability but their distribution is not limited to the site of injury and they may bind to inflammatory cells that accumulate there as well.
  • Example 8 The cytotoxic effects of TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles on bladder carcinoma cells, breast cancer cells and normal breast cells
  • NP-TRAIL TRAIL-conjugated gelatin/iron oxide magnetic composite nanoparticles
  • the cancer cell lines particularly used were the bladder carcinoma cells TSU-PRl and the breast cancer cells MDA-MB, and the normal breast cells MCFlOA were used as controls.
  • PS proteasome inhibitor
  • Pharmacologic inhibitors of the proteasome possess in vitro and in vivo antitumor activity. Preclinical studies demonstrate that proteasome inhibition potentiates the activity of other cancer therapeutics such as TRAIL in part by down regulating chemoresistance pathways.
  • Cells (1x10 /well) were treated with different concentrations of TRAIL (10-
  • NP or NP-TRAIL 10-40 ng TRAIL/ml
  • PS 5 niM
  • Cell death was determined after 24 h using LDH assay. 100% cell death was determined in Triton X-100-treated cells and data normalized.
PCT/IL2008/001286 2007-09-24 2008-09-24 Polymer nanoparticles coated by magnetic metal oxide and uses thereof WO2009040811A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2010526423A JP5624471B2 (ja) 2007-09-24 2008-09-24 磁性金属酸化物で被覆されたポリマー性ナノ粒子及びその使用
EP08808087A EP2205282A2 (en) 2007-09-24 2008-09-24 Polymer nanoparticles coated by magnetic metal oxide and uses thereof
RU2010116167/15A RU2472530C2 (ru) 2007-09-24 2008-09-24 Полимерные наночастицы, покрытые оксидом магнитного металла, и их применение
IL204712A IL204712A (en) 2007-09-24 2010-03-24 Trailing gelatin nanoparticles in an oxidized metal casing
IL212564A IL212564A0 (en) 2007-09-24 2011-04-28 Polymer nanoparticles coated by magnetic metal oxide and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US96027007P 2007-09-24 2007-09-24
US60/960,270 2007-09-24

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP12172258.1A Previously-Filed-Application EP2591804A3 (en) 2007-09-24 2008-09-24 Polymer nanoparticles coated by magnetic metal oxide and uses thereof

Publications (2)

Publication Number Publication Date
WO2009040811A2 true WO2009040811A2 (en) 2009-04-02
WO2009040811A3 WO2009040811A3 (en) 2010-03-04

Family

ID=40282352

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2008/001286 WO2009040811A2 (en) 2007-09-24 2008-09-24 Polymer nanoparticles coated by magnetic metal oxide and uses thereof

Country Status (5)

Country Link
US (2) US9295736B2 (und)
EP (3) EP2399610A3 (und)
JP (2) JP5624471B2 (und)
RU (2) RU2472530C2 (und)
WO (1) WO2009040811A2 (und)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101642579A (zh) * 2009-08-14 2010-02-10 江苏大学 壳聚糖修饰顺磁性金属离子磁共振成像对比剂的制备方法
CN101890173A (zh) * 2010-07-16 2010-11-24 江苏大学 磁性水溶性壳聚糖磁共振成像对比剂的制备方法
EP2277544A1 (en) * 2009-07-08 2011-01-26 Nelica Ciobanu Biocompatible magnetic nano-clusters containing iron oxide respectively iron oxide - boron with primary use in magnetic drug targeting and boron neutron capture therapy
WO2011128896A1 (en) * 2010-04-12 2011-10-20 Ramot At Tel-Aviv University Ltd. Iron oxide nanoparticles for use in treating non-infectious inflammatory disorders
EP2460541A1 (en) * 2009-07-30 2012-06-06 FUJIFILM Corporation Kidney imaging agent comprising genetically modified gelatin
WO2012062904A3 (en) * 2010-11-12 2012-08-30 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer
US8354110B2 (en) 2007-03-07 2013-01-15 Uti Limited Partnership Compositions and methods for the prevention and treatment of autoimmune conditions
JPWO2011083845A1 (ja) * 2010-01-08 2013-05-16 富士フイルム株式会社 腫瘍部位に対する標的化剤
WO2014006254A1 (es) 2012-07-03 2014-01-09 Fundación Centro Nacional De Investigaciones Cardiovasculares Carlos Iii Nanopartículas recubiertas de gelatina
US20160000942A1 (en) * 2013-01-04 2016-01-07 Industry-Academic Cooperation Foundation, Yonsei University Mri contrast agent including t1 contrast material coated on surface of nanoparticle support
US9295736B2 (en) 2007-09-24 2016-03-29 Bar Ilan University Polymer nanoparticles coated by magnetic metal oxide and uses thereof
CN105854019A (zh) * 2016-05-06 2016-08-17 暨南大学 Il-27的受体激活剂在制备治疗肥胖症及其并发症产品中的应用
EP2646818A4 (en) * 2010-11-30 2016-09-21 Univ Illinois SILICANANOPARTIKELKONJUGATE
US9603948B2 (en) 2012-10-11 2017-03-28 Uti Limited Partnership Methods and compositions for treating multiple sclerosis and related disorders
US10124045B2 (en) 2013-11-04 2018-11-13 Uti Limited Partnership Methods and compositions for sustained immunotherapy
US10485882B2 (en) 2015-05-06 2019-11-26 Uti Limited Partnership Nanoparticle compositions for sustained therapy
EP3708191A4 (en) * 2017-11-06 2020-11-25 Konica Minolta, Inc. COMPOSITE PARTICLES FOR IMAGING, PROCESS FOR THE PRODUCTION OF COMPOSITE PARTICLES, CELLS, CELLULAR STRUCTURE AND MIXED DISPERSION
US10988516B2 (en) 2012-03-26 2021-04-27 Uti Limited Partnership Methods and compositions for treating inflammation

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9273398B2 (en) * 2010-01-16 2016-03-01 Nanoridge Materials, Inc. Metallized nanotubes
US20110301456A1 (en) * 2010-06-07 2011-12-08 Malignext Targeting Technologies, Inc. Tissue Marking for Lesion Removal
US9572880B2 (en) 2010-08-27 2017-02-21 Sienna Biopharmaceuticals, Inc. Ultrasound delivery of nanoparticles
DK3210591T3 (en) 2010-08-27 2019-04-15 Sienna Biopharmaceuticals Inc COMPOSITIONS AND METHODS OF TARGETED THERMO MODULATION
WO2012033531A1 (en) * 2010-09-10 2012-03-15 Kent State University Nanoparticles based on gadolinium coordination polymers as highly sensitive t1 mri contrast agents
GB201204579D0 (en) 2012-03-15 2012-05-02 Univ Nottingham Trent Coating metal oxide particles
EP2833144B1 (en) * 2012-03-28 2017-11-08 Konica Minolta, Inc. Method for detection biological substance
EP3272388A1 (en) * 2012-10-11 2018-01-24 Nanocomposix, Inc. Silver nanoplate compositions and methods
WO2014093581A1 (en) * 2012-12-14 2014-06-19 Medivation Technologies, Inc. Magnetic retention of regenerative cells for wound repair
EP2958481A4 (en) 2013-02-20 2017-03-08 Sloan-Kettering Institute for Cancer Research Wide field raman imaging apparatus and associated methods
CN105307725B (zh) 2013-04-11 2020-06-16 布里格姆及妇女医院股份有限公司 治疗自身免疫疾病的方法和组合物
EP3003397B1 (en) 2013-05-24 2020-08-19 Danmarks Tekniske Universitet (DTU) Gel formulations for guiding radiotherapy
CA2923817C (en) * 2013-09-09 2021-12-28 Lab-On-A-Bead Ab Manufacture of magnetic particles
US10912947B2 (en) 2014-03-04 2021-02-09 Memorial Sloan Kettering Cancer Center Systems and methods for treatment of disease via application of mechanical force by controlled rotation of nanoparticles inside cells
CA2941635A1 (en) * 2014-03-04 2015-09-11 Memorial Sloan Kettering Cancer Center Applying mechanical force by remote-controlled rotation
JP2017523181A (ja) 2014-07-28 2017-08-17 メモリアル スローン−ケタリング キャンサー センター 医療用同位体のためのユニバーサル結合剤としての金属(メタロイド)カルコゲンナノ粒子
CN107249570B (zh) 2014-11-21 2022-08-09 丹麦技术大学 用于局部药物释放的凝胶制剂
DE102014019388A1 (de) * 2014-12-29 2016-06-30 Susanne Wagner Arzneimittel auf der Basis von Maghämit zur gleichzeitigen Reduzierung der gastrointestinalen Natriumresorption und Phosphatresorption
WO2017004301A1 (en) 2015-07-01 2017-01-05 Memorial Sloan Kettering Cancer Center Anisotropic particles, methods and uses thereof
KR101748120B1 (ko) * 2015-07-13 2017-06-16 서울대학교산학협력단 나노입자-유리체 기반 단백질 복합체를 유효성분으로 포함하는 혈관신생억제용 조성물 및 이의 용도
WO2017027874A1 (en) * 2015-08-13 2017-02-16 Northeastern University Biomaterials for combined radiotherapy and immunotherapy of cancer
CN106902395B (zh) * 2015-12-22 2020-04-07 先健科技(深圳)有限公司 可吸收铁基合金植入医疗器械
KR20170097510A (ko) * 2016-02-18 2017-08-28 연세대학교 산학협력단 T1 mri 조영제로서의 나노입자의 분산 안정도를 증가시키는 방법 및 t1 mri 조영제 나노입자
JP7246930B2 (ja) * 2016-05-18 2023-03-28 モデルナティエックス インコーポレイテッド インターロイキン-12(il12)をコードするポリヌクレオチドおよびその使用
JP2018019686A (ja) * 2016-07-20 2018-02-08 コニカミノルタ株式会社 ゼラチン粒子、ゼラチン粒子の製造方法、ゼラチン粒子内包細胞、ゼラチン粒子内包細胞の製造方法、および細胞構造体
CN106344539B (zh) * 2016-08-25 2019-01-29 湖北大学 一种多功能靶向纳米胶囊抗癌药物的制备方法
US20180207293A1 (en) * 2017-01-25 2018-07-26 2C Tech Corp. Nanoparticles for sustained ophthalmic drug delivery and methods of use
CN108926719B (zh) 2017-05-25 2020-09-01 北京格瑞特森生物医药科技有限公司 用c(RGD-ACP-K)修饰的长循环脂质体
EP3708605A4 (en) * 2017-11-06 2020-12-16 Konica Minolta, Inc. HYDROGEL PARTICLE AND ITS PRODUCTION PROCESS, CELL OR CELLULAR STRUCTURE EACH CONTAINING A HYDROGEL PARTICLE, PROCESS FOR EVALUATING THE ACTIVITY OF A CELL USING A HYDROGEL PARTICLE, AND USE OF A PARTICLE OF HYDROGEL AS A LONG-RELEASE PREPARATION
WO2019108838A1 (en) * 2017-11-29 2019-06-06 The Regents Of The University Of California Cross-linked fluorescent supramolecular nanoparticles and method of making
US20220031605A1 (en) * 2018-09-26 2022-02-03 Ege Universitesi Targeted drug delivery system to be used in treating osteomyelitis
KR102187362B1 (ko) * 2018-12-26 2020-12-07 서울대학교산학협력단 페로토시스를 통한 암세포의 선택적 사멸을 위한 나노 입자, 이의 제조 방법 및 이의 용도
WO2020174476A1 (en) * 2019-02-28 2020-09-03 Hadasit Medical Research Services And Development Ltd. Targeted magnetic vehicles and method of using the same

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999062079A1 (en) * 1998-05-26 1999-12-02 Bar-Ilan University Nucleation and growth of magnetic metal oxide nanoparticles and its use
WO2005017539A2 (en) * 2003-08-14 2005-02-24 The General Hospital Corporation Imaging pathology
WO2007072982A1 (en) * 2005-12-20 2007-06-28 Fujifilm Corporation Protein nanoparticles and the use of the same

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1582956A (en) * 1976-07-30 1981-01-21 Ici Ltd Composite magnetic particles
DK130991D0 (da) 1991-07-04 1991-07-04 Immunodex K S Polymere konjugater
IL104734A0 (en) 1993-02-15 1993-06-10 Univ Bar Ilan Bioactive conjugates of cellulose with amino compounds
US6998116B1 (en) * 1996-01-09 2006-02-14 Genentech, Inc. Apo-2 ligand
US5834121A (en) 1996-01-16 1998-11-10 Solid Phase Sciences Corp. Composite magnetic beads
IL118848A0 (en) 1996-07-14 1996-10-31 Univ Bar Ilan Method for preparing bioactive polymers
EP1311839B1 (en) * 2000-06-21 2006-03-01 Bioarray Solutions Ltd Multianalyte molecular analysis using application-specific random particle arrays
US20020076409A1 (en) * 2000-07-12 2002-06-20 March Carl J. Method for treating cancer
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
DE10144252A1 (de) 2001-08-31 2003-03-27 Fraunhofer Ges Forschung Nanopartikel mit daran immobilisiertem biologisch aktivem TNF
PT1450847E (pt) * 2001-11-13 2011-01-05 Genentech Inc Formulações de ligando de apo2/trail e suas utilizações
US20050129769A1 (en) * 2002-06-03 2005-06-16 Barry Stephen E. Polymeric articles for carrying therapeutic agents
AU2003273272A1 (en) 2002-06-03 2003-12-19 Alinis Biosciences, Inc. Therapeutic agent-containing polymeric nanoarticles
US20060141561A1 (en) 2002-06-24 2006-06-29 Kelley Robert F Apo-2 ligand/trail variants and uses thereof
AU2003282356A1 (en) 2002-11-20 2004-06-15 Bar-Ilan University Biological glue based on thrombin-conjugated nanoparticles
DE60334247D1 (de) 2002-11-27 2010-10-28 Irm Llc Verfahren und zusammensetzungen zur apoptoseinduktion in krebszellen
CN1879065A (zh) * 2003-09-12 2006-12-13 弗尔克斯破产财产公司 用于生物活性剂的定位传递的包含磁性成分和生物相容性聚合物的可磁靶向颗粒
US20050090732A1 (en) * 2003-10-28 2005-04-28 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles
EP1710255A4 (en) * 2003-12-12 2008-09-24 Chugai Pharmaceutical Co Ltd MODIFIED ANTIBODIES RECOGNIZING A TRIMER OR LARGER RECEPTOR
EP1715971A4 (en) 2004-01-28 2010-10-13 Cytimmune Sciences Inc FUNCTIONALIZED COLLOIDAL METAL COMPOSITIONS AND METHODS
KR100617769B1 (ko) * 2004-03-24 2006-08-28 삼성전자주식회사 채널 부호화 장치 및 방법
US20050226911A1 (en) 2004-04-13 2005-10-13 Bringley Joseph F Article for inhibiting microbial growth in physiological fluids
RU2007112952A (ru) * 2004-09-08 2008-10-20 Дженентек, Инк. (Us) Способы применения лигандов рецепторов смерти и cd20-антител
EP1866155A2 (en) 2005-04-05 2007-12-19 Bar-Ilan University New core and core-shell nanoparticles containing iodine for x-ray imaging
EP2399610A3 (en) 2007-09-24 2012-09-05 Bar-Ilan University Polymer nanoparticles coated by magnetic metal oxide and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999062079A1 (en) * 1998-05-26 1999-12-02 Bar-Ilan University Nucleation and growth of magnetic metal oxide nanoparticles and its use
WO2005017539A2 (en) * 2003-08-14 2005-02-24 The General Hospital Corporation Imaging pathology
WO2007072982A1 (en) * 2005-12-20 2007-06-28 Fujifilm Corporation Protein nanoparticles and the use of the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JOSEPHSON LEE ET AL: "Near-infrared fluorescent nanoparticles as combined MR/optical imaging probes" BIOCONJUGATE CHEMISTRY, vol. 13, no. 3, 1 May 2002 (2002-05-01), pages 554-560, XP002267675 ISSN: 1043-1802 *
KIRCHER MORITZ F ET AL: "A dual fluorochrome probe for imaging proteases" BIOCONJUGATE CHEMISTRY, vol. 15, no. 2, 1 March 2004 (2004-03-01), pages 242-248, XP002464245 ISSN: 1043-1802 *
ZIMMER CLAUS ET AL: "MR imaging of phagocytosis in experimental gliomas" RADIOLOGY, vol. 197, no. 2, 1995, pages 533-538, XP002543025 ISSN: 0033-8419 *

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8354110B2 (en) 2007-03-07 2013-01-15 Uti Limited Partnership Compositions and methods for the prevention and treatment of autoimmune conditions
US9295736B2 (en) 2007-09-24 2016-03-29 Bar Ilan University Polymer nanoparticles coated by magnetic metal oxide and uses thereof
EP2277544A1 (en) * 2009-07-08 2011-01-26 Nelica Ciobanu Biocompatible magnetic nano-clusters containing iron oxide respectively iron oxide - boron with primary use in magnetic drug targeting and boron neutron capture therapy
US9259485B2 (en) 2009-07-30 2016-02-16 Fujifilm Corporation Kidney-imaging agent comprising recombinant gelatin
EP2460541A1 (en) * 2009-07-30 2012-06-06 FUJIFILM Corporation Kidney imaging agent comprising genetically modified gelatin
EP2460541A4 (en) * 2009-07-30 2013-02-06 Fujifilm Corp Kidney imaging agent with genetically modified gelatin
CN101642579B (zh) * 2009-08-14 2013-10-23 江苏大学 壳聚糖修饰顺磁性金属离子磁共振成像对比剂的制备方法
CN101642579A (zh) * 2009-08-14 2010-02-10 江苏大学 壳聚糖修饰顺磁性金属离子磁共振成像对比剂的制备方法
JPWO2011083845A1 (ja) * 2010-01-08 2013-05-16 富士フイルム株式会社 腫瘍部位に対する標的化剤
US8961929B2 (en) 2010-01-08 2015-02-24 Fujifilm Corporation Targeting agent for tumor site
US20130129810A1 (en) * 2010-04-12 2013-05-23 Tel Hashomer Medical Research Infrastructure And Services Ltd. Iron oxide nanoparticles for use in treating non-infectious inflammatory disorders
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
WO2011128896A1 (en) * 2010-04-12 2011-10-20 Ramot At Tel-Aviv University Ltd. Iron oxide nanoparticles for use in treating non-infectious inflammatory disorders
CN101890173A (zh) * 2010-07-16 2010-11-24 江苏大学 磁性水溶性壳聚糖磁共振成像对比剂的制备方法
US10172955B2 (en) 2010-11-12 2019-01-08 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer
WO2012062904A3 (en) * 2010-11-12 2012-08-30 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer
US11000596B2 (en) 2010-11-12 2021-05-11 UTI Limited Parttiership Compositions and methods for the prevention and treatment of cancer
US9511151B2 (en) 2010-11-12 2016-12-06 Uti Limited Partnership Compositions and methods for the prevention and treatment of cancer
EP2646818A4 (en) * 2010-11-30 2016-09-21 Univ Illinois SILICANANOPARTIKELKONJUGATE
US10988516B2 (en) 2012-03-26 2021-04-27 Uti Limited Partnership Methods and compositions for treating inflammation
WO2014006254A1 (es) 2012-07-03 2014-01-09 Fundación Centro Nacional De Investigaciones Cardiovasculares Carlos Iii Nanopartículas recubiertas de gelatina
US9603948B2 (en) 2012-10-11 2017-03-28 Uti Limited Partnership Methods and compositions for treating multiple sclerosis and related disorders
US10080808B2 (en) 2012-10-11 2018-09-25 Uti Limited Partnership Methods and compositions for treating multiple sclerosis and related disorders
US10905773B2 (en) 2012-10-11 2021-02-02 Uti Limited Partnership Methods and compositions for treating multiple sclerosis and related disorders
US20160000942A1 (en) * 2013-01-04 2016-01-07 Industry-Academic Cooperation Foundation, Yonsei University Mri contrast agent including t1 contrast material coated on surface of nanoparticle support
US10124045B2 (en) 2013-11-04 2018-11-13 Uti Limited Partnership Methods and compositions for sustained immunotherapy
US11338024B2 (en) 2013-11-04 2022-05-24 Uti Limited Partnership Methods and compositions for sustained immunotherapy
US10485882B2 (en) 2015-05-06 2019-11-26 Uti Limited Partnership Nanoparticle compositions for sustained therapy
CN105854019A (zh) * 2016-05-06 2016-08-17 暨南大学 Il-27的受体激活剂在制备治疗肥胖症及其并发症产品中的应用
EP3708191A4 (en) * 2017-11-06 2020-11-25 Konica Minolta, Inc. COMPOSITE PARTICLES FOR IMAGING, PROCESS FOR THE PRODUCTION OF COMPOSITE PARTICLES, CELLS, CELLULAR STRUCTURE AND MIXED DISPERSION

Also Published As

Publication number Publication date
EP2399610A2 (en) 2011-12-28
US20120141380A1 (en) 2012-06-07
EP2591804A2 (en) 2013-05-15
JP2011207887A (ja) 2011-10-20
US20090110644A1 (en) 2009-04-30
RU2010116167A (ru) 2011-11-10
JP2010540513A (ja) 2010-12-24
US9295736B2 (en) 2016-03-29
WO2009040811A3 (en) 2010-03-04
EP2205282A2 (en) 2010-07-14
RU2472530C2 (ru) 2013-01-20
EP2399610A3 (en) 2012-09-05
EP2591804A3 (en) 2014-04-16
JP5599753B2 (ja) 2014-10-01
RU2011152596A (ru) 2013-06-27
JP5624471B2 (ja) 2014-11-12

Similar Documents

Publication Publication Date Title
US9295736B2 (en) Polymer nanoparticles coated by magnetic metal oxide and uses thereof
Li et al. Folate-decorated anticancer drug and magnetic nanoparticles encapsulated polymeric carrier for liver cancer therapeutics
Prijic et al. Magnetic nanoparticles as targeted delivery systems in oncology
Gupta et al. Recent advances on surface engineering of magnetic iron oxide nanoparticles and their biomedical applications
J Wang et al. Recent advances in superparamagnetic iron oxide nanoparticles for cellular imaging and targeted therapy research
Chen et al. Folic acid-conjugated MnO nanoparticles as a T 1 contrast agent for magnetic resonance imaging of tiny brain gliomas
Yen et al. Multifunctional iron oxide nanoparticles for diagnostics, therapy and macromolecule delivery
Landmark et al. Synthesis, characterization, and in vitro testing of superparamagnetic iron oxide nanoparticles targeted using folic acid-conjugated dendrimers
KR100848931B1 (ko) 양친매성 화합물 및 조직 특이적 결합 성분을 이용한지능형 자성 나노복합체 및 이를 포함하는 조영제
Ke et al. A specific tumor-targeting magnetofluorescent nanoprobe for dual-modality molecular imaging
Lassenberger et al. Individually stabilized, superparamagnetic nanoparticles with controlled shell and size leading to exceptional stealth properties and high relaxivities
US20150093335A1 (en) Cell-targeted magnetic nano-material and biomedical uses thereof
Yang et al. Cooperative dual-activity targeted nanomedicine for specific and effective prostate cancer therapy
Creixell et al. Preparation of epidermal growth factor (EGF) conjugated iron oxide nanoparticles and their internalization into colon cancer cells
Wang et al. Synthesis of tumor-targeted folate conjugated fluorescent magnetic albumin nanoparticles for enhanced intracellular dual-modal imaging into human brain tumor cells
Mauri et al. MnO nanoparticles embedded in functional polymers as T 1 contrast agents for magnetic resonance imaging
Chen et al. Improving sensitivity of magnetic resonance imaging by using a dual-targeted magnetic iron oxide nanoprobe
Zhang et al. Thiol-PEG-carboxyl-stabilized Fe2O3/Au nanoparticles targeted to CD105: Synthesis, characterization and application in MR imaging of tumor angiogenesis
Panahifar et al. Synthesis and in vitro evaluation of bone-seeking superparamagnetic iron oxide nanoparticles as contrast agents for imaging bone metabolic activity
Chen et al. Rotavirus capsid surface protein VP4-coated Fe3O4 nanoparticles as a theranostic platform for cellular imaging and drug delivery
Chiu et al. Effects of PEGylation on capture of dextran-coated magnetic nanoparticles in microcirculation
Wu et al. Modularly assembled magnetite nanoparticles enhance in vivo targeting for magnetic resonance cancer imaging
Tarhan et al. Synthesis and characterization of new branched magnetic nanocomposite for loading and release of topotecan anti-cancer drug
Zhou et al. Mitochondria-localized self-reporting small-molecule-decorated theranostic agents for cancer-organelle transporting and imaging
Bietenbeck et al. Functionalization of clinically approved MRI contrast agents for the delivery of VEGF

Legal Events

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

Ref document number: 08808087

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 204712

Country of ref document: IL

Ref document number: 2010526423

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2821/DELNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2010116167

Country of ref document: RU

Ref document number: 2008808087

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 212564

Country of ref document: IL