WO2014141288A1 - Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament - Google Patents

Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament Download PDF

Info

Publication number
WO2014141288A1
WO2014141288A1 PCT/IN2013/000143 IN2013000143W WO2014141288A1 WO 2014141288 A1 WO2014141288 A1 WO 2014141288A1 IN 2013000143 W IN2013000143 W IN 2013000143W WO 2014141288 A1 WO2014141288 A1 WO 2014141288A1
Authority
WO
WIPO (PCT)
Prior art keywords
imaging
nanocontrast
multifunctional
calcium
nanoparticles
Prior art date
Application number
PCT/IN2013/000143
Other languages
English (en)
Inventor
Manzoor Koyakutty
Anusha ASHOKAN
Shantikumar V. Nair
Original Assignee
Amrita Vishwa Vidyapeetham University
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 Amrita Vishwa Vidyapeetham University filed Critical Amrita Vishwa Vidyapeetham University
Priority to PCT/IN2013/000143 priority Critical patent/WO2014141288A1/fr
Publication of WO2014141288A1 publication Critical patent/WO2014141288A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/662Phosphorus acids or esters thereof having P—C bonds, e.g. foscarnet, trichlorfon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0032Methine dyes, e.g. cyanine dyes
    • A61K49/0034Indocyanine green, i.e. ICG, cardiogreen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0063Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres
    • A61K49/0069Preparation for luminescence or biological staining characterised by a special physical or galenical form, e.g. emulsions, microspheres the agent being in a particular physical galenical form
    • A61K49/0089Particulate, powder, adsorbate, bead, sphere
    • A61K49/0091Microparticle, microcapsule, microbubble, microsphere, microbead, i.e. having a size or diameter higher or equal to 1 micrometer
    • A61K49/0093Nanoparticle, nanocapsule, nanobubble, nanosphere, nanobead, i.e. having a size or diameter smaller than 1 micrometer, e.g. polymeric nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/04X-ray contrast preparations
    • A61K49/0409Physical forms of mixtures of two different X-ray contrast-enhancing agents, containing at least one X-ray contrast-enhancing agent which is not a halogenated organic compound
    • A61K49/0414Particles, beads, capsules or spheres
    • A61K49/0423Nanoparticles, nanobeads, nanospheres, nanocapsules, i.e. having a size or diameter smaller than 1 micrometer
    • 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
    • A61K49/183Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle having a (super)(para)magnetic core coated or functionalised with an inorganic material or being composed of an inorganic material entrapping the MRI-active nucleus, e.g. silica core doped with a MRI-active nucleus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0489Phosphates or phosphonates, e.g. bone-seeking phosphonates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1241Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins
    • A61K51/1244Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins microparticles or nanoparticles, e.g. polymeric nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0002Galenical forms characterised by the drug release technique; Application systems commanded by energy
    • A61K9/0009Galenical forms characterised by the drug release technique; Application systems commanded by energy involving or responsive to electricity, magnetism or acoustic waves; Galenical aspects of sonophoresis, iontophoresis, electroporation or electroosmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y15/00Nanotechnology for interacting, sensing or actuating, e.g. quantum dots as markers in protein assays or molecular motors

Definitions

  • the present invention relates to a nano-sized material that can provide contrast enhancement for multiple imaging methods and also deliver therapeutic molecules such as nucleic acids or chemo drugs to diseased sites such as cancer.
  • the present invention relates to nano-sized synthetic calcium phosphates and calcium apatite nanomaterials showing simultaneous contrast for at least any two of the medical imaging modalities including radio-, raman-, near-infrared fluorescence-, magnetic resonance- and X ray- imaging.
  • This invention relates to the preparation of a nanomaterial based on doped calcium phosphate or calcium apatite contrast agent for multimodal contrast imaging, drug-delivery and therapy applications.
  • Multimodal molecular imaging of disease using a combination of techniques such as single photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), positron emission tomography (PET), X-ray computed tomography (CT) and near- infrared (NIR) fluorescence, raman spectroscopy is an emerging area of research [1,2].
  • SPECT single photon emission computed tomography
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • CT X-ray computed tomography
  • NIR near- infrared fluorescence
  • imaging systems and contrast agents are used independently to derive different anatomical and functional information such as location, size, angiogenesis, metastasis etc. It will be highly useful, if low-cost, but high resolution imaging modalities like optical (fluorescence) techniques can be combined with SPECT, MRI, CT Or raman spectroscopy to derive molecular information about the expression level of specific disease marker or biological process that influence the therapeutic outcome.
  • imaging modalities like optical (fluorescence) techniques can be combined with SPECT, MRI, CT Or raman spectroscopy to derive molecular information about the expression level of specific disease marker or biological process that influence the therapeutic outcome.
  • suitable contrast agents that can show different physical properties compatible with basic principles of imaging technique.
  • different molecular imaging techniques requires separate contrast agents because their working principles are distinctly different.
  • SPECT requires gamma radiation emitting nuclides
  • MRI requires paramagnetic
  • CT computed tomography
  • raman imaging requires raman active metals or dyes
  • in vivo fluorescence imaging requires near-infrared fluorescent contrast agents.
  • radionuclides such as Tc9 m is used for scintigraphy and 3D SPECT imaging
  • paramagnetic gadolinium complexes such as Gd-carboxylates or superparamagnetic iron oxide particles are used as MRI contrast agents
  • iodine or colloidal barium sulphate contrast agents are used in CT
  • Au Au
  • proteins Green fluorescent protein or Red fluorescent proteins
  • QDs quantum dots
  • contrast agents differ significantly.
  • An ideal solution is to provide a single contrast agent that can show all important contrast functionalities at the same time.
  • Luminescent quantum dots, superparamagnetic iron oxide nanoparticles, Ag nanoparticles and Au nanocrystals were studied for optical, magnetic, raman and photo- acoustic/two- photon imaging applications, respectively.
  • bi-functional nanosystems combining two separate agents such as luminescent QDs and magnetic iron oxide or fluorescent dyes and magnetic iron oxide were also reported for combinatorial (MRI and optical) imaging applications.
  • one of the major concerns about nanoparticle based contrast imaging is the efficient incorporation of multiple impurities and toxicity associated with each component within the nanomaterial.
  • luminescent semiconductor nanoparticles were found to be toxic to biological systems due to their heavy- metal composition (cadmium, selenium, tellurium, lead). Further, there exist no biocompatible single nanoparticle that can provide contrast enhancement for multiple imaging options such as SPECT, raman imaging, NI fluorescence, MRI and X-ray imaging.
  • HAp and calcium phosphates are extensively studied and used in clinical practice as implant material for in vivo bone tissue re-generation.
  • the present invention discloses, the preparation of improved calcium apatite and calcium phosphates (tricalcium phosphate, dicalcium phosphate, monocalcium phosphate) nanoparticles doped and labeled with multiple impurities that confer properties such as radioactivity, raman scattering, near-infrared fluorescence, X-ray absorption and super/para-magnetism.
  • the preparation of undoped hydroxyapatite and calcium phosphate nanoparticles is disclosed in several prior arts.
  • stoichiometric hydroxyapatite, Cai 0 (OH) 2 (P04)6 and calcium phosphate nanoparticles are prepared by adding ammonium phosphate or phosphoric acid and ammonium hydroxide mixture solution to a solution of Ca 2 ⁇ precursor.
  • Doped apatite/calcium phosphate nanoparticles is prepared by replacing calcium ions with fluorescent or paramagnetic metal ions.
  • Organic dye doped calcium phosphates can also be prepared by the incorporation of the dye within calcium phosphate matrix during its synthesis, wherein the dye can confer properties such NIR fluorescence or raman scattering.
  • Near infrared fluorescence based imaging is a recent advancement in biomedical research. Generally, in vivo fluorescence imaging using visible light is limited by low penetration of light photons into living tissue due to high absorption and scattering properties of tissue components. However, near- infrared region of the spectrum offers certain advantages because the hemoglobin and water absorb minimally in this spectral window (68o-90onm) so as to allow photons to penetrate for several centimeters deep inside the tissue. This allows mapping of molecular events in intact tissues using near-infrared fluorescence mediated molecular tomography (FMT), a new technique that can three- dimensionally image molecular events like gene expression by resolving fluorescence activation in deep tissues.
  • FMT near-infrared fluorescence mediated molecular tomography
  • an essential component of successful FMT imaging is the development of biocompatible NIR emitting fluorescent probes that can be targeted to the tissue.
  • various paramagnetic and superparamagnetic nanocontrast agents are being developed that has superior magnetic properties that their macro size counterparts.
  • Combining such nanocontrast agents with other imaging modalities such as NIR imaging provides added advantage of combining imaging of anatomic and functional characteristics of the diseased condition.
  • Research in combinatorial imaging approaches are exploring the advantages of combining anatomic imaging modalities such as MRI, 2D/3D CT imaging together with imaging modalities such as NIR, raman and radioactive imaging that give details on molecular characteristics of the pathological condition. It is therefore imperative to develop a biocompatible multimodal contrast agent that can combine the above mentioned modalities in combination to aid better and early stage disease diagnosis and treatment.
  • a novel biocompatible nanoparticle that can provide contrast enhancement for multiple molecular imaging modalities such as radioactive imaging, raman imaging, MRI, X-ray and/or near infrared (NIR) fluorescence together with targeted drug or gene delivery is disclosed.
  • the particle is based on nanoparticles of calcium phosphate (nCP) and calcium hydroxyapatite (nHAp), doped and/or conjugated with more than one impurity ion/molecule.
  • This multifunctional nano-composite particle enable combined molecular imaging using single photon emission computed tomography (SPECT), raman imaging, MRI, CT and/or optical near-infrared fluorescent imaging.
  • Another aspect of this invention relates to a method of making nanomedicine formulations based on the said nCP and nHAp particles, capable of delivering chemodrugs, small molecule inhibitors or nucleic acid drugs such as DNA, RNA, small interfering RNA (siRNA), micro RNA (miRNA) specifically to a targeted disease such as cancer.
  • nucleic acid drugs such as DNA, RNA, small interfering RNA (siRNA), micro RNA (miRNA) specifically to a targeted disease such as cancer.
  • nanoparticle refers to primary inventive nanoparticles formed by calcium phopshate or hydroxyapatite, measuring size about 1-500 nm, preferably l-ioonm, most preferably around 1- 50 nm in size, showing "multifunctional' properties such as radioactivity, raman scattering, NIR fluorescence, magnetism and X-ray absorption in different combinations with at least two properties together.
  • therapeutics refers to chemodrugs, nucleic acids such as DNA or RNA [small interfering RNA (siRNA) or micro RNA (miRNA)], bisphosphonates, antibodies, peptides, proteins, etc that have a therapeutic effect against a disease condition such as cancer, inflammatory disease or autoimmune disease.
  • nucleic acids such as DNA or RNA [small interfering RNA (siRNA) or micro RNA (miRNA)]
  • siRNA small interfering RNA
  • miRNA micro RNA
  • targeting ligand refers to biomolecules that can specifically identify and target another molecule like an antigen or receptor on the surface of cell-membrane of diseased cells such as that of cancer / tumor.
  • Targeting ligand include antibodies, peptides, aptamers, vitamins like folic acid, sugar molecules like mannose ligands, carbohydrates.
  • nanomedicine refers to a composite construct based on the said calcium phosphate or HAp nanoparticle which is connected or alternatively loaded with any of the above said therapeutics which will be delivered to the desired site using the said targeting ligands.
  • nanomedicine construct is formed by either : i) directly connecting the therapeutics and targeting ligands to the surface of said nanoparticles or alternatively, ii) the said nanoparticle will be taken as a core with another biodegradable polymer containing the therapeutics as shell with targeting ligands connected to its surface, or Hi) a biodegradable polymeric nanoparticle which embed nCP or nHAp together with the therapeutic agents and connected with the targeting ligands on its surface
  • the said nanomedicine construct preferably will have a size of 50- 20onm, more preferably 8o-i50nm and most preferably ⁇ ioo-i2onm.
  • the nanomedicine may be produced in the form of dry powders or liquid dispersions.
  • the nanoparticle may also be synthesized via colloidal synthesis or self- assembly of ions in saturated solutions like simulated body fluid (SBF) and may take the form of colloidal crystals with dopant ions or molecules in it.
  • SBF simulated body fluid
  • the term "doped” as used herein, refers to incorporation of small amount of (about 0.001-30 mole % or up to 30 weight %) another substance (inorganic elements, ions or organic molecules or ratioactive isotopes) within the said nanoparticles.
  • the dopant molecules include, elements, ions, dyes or molecular clusters that can provide radioactivity, raman scattering, NIR fluorescence, magnetic contrast by paramagnetic, superparamagnetic, ferromagnetic or ferrimagnetic properties or X-ray absorption.
  • the dopant ions may substitute either Ca or P ions, or may be present as molecular clusters within the interstitial space of nHAp or nCP lattice.
  • labelled refers to attaching the impurity ions, molecules or radioactive isotopes on the surface of the said nanoparticle with the help of a ligand such as bisphosphonates or functional groups such as carboxyl on the surface of the nanoparticle. This may involve electrostatic interactions , covalent-, ionic-, coordinate, or hydrogen bonding or hydrophobic, lipophilic or hydrophilic interactions
  • Figure 1 Schematic showing the method of preparation of multimodal and therapeutic nCP and nHAp
  • Figure 2 (a) XRD of MF-nHAp showing hexagonal HAp crystal structure (b) SEM image of MF-nHAp showing particle size ⁇ 100 nm. (c) DLS data of MF- nHAp giving a size distribution of 100 ⁇ 20nm (d) Digital photograph of MF- nHAp and nHAp solutions ( ⁇ 20 mg/mL) indicating the greenish body color due to ICG doping .
  • Figure 3 (A) Graph showing integrated fluorescence emission intensity at 800 nm with increase in MF-nHAp concentration. Inset: Fluorescence spectra of MF- nHAp showing excitation and emission peaks at 765 nm and 800 nm, respectively.
  • Figure 4 (a) Radioactive emission of 20mg/mL of MF-nCP samples conjugated to Tc-99m of varying concentrations (b) Fluorescence images of MF-nCP samples conjugated to varying concentrations of ICG (c) Graph showing the variation in radioactive emission of MF-nCP samples conjugated to varying concentrations of Tc-99m (d) Graph showing the fluorescence intensity variation of MF-nCP with varying doping % of ICG
  • FIG. 5 (A) Hemolysis data of nHAp and D-nHAp treated whole blood showing non-hemolytic action of the tested samples (B) Digital photograph of the whole blood treated with PBS, D-nHAp and triton showing no leakage of hemoglobin from PBS/D-nHAp treated samples compared to triton treated sample.
  • Figure 6 Scanning electron micrograph of RBC treated with (A) PBS (B) D-nHAp. Enlarged image (C))showing uniform distribution of D- nHAp particles although out the RBC without affecting its morphology (D) EDX spectrum of the D-nHAp that are distributed on the RBC.
  • Figure 7 Cytotoxicity data of D-nHAp treatment on peripheral blood derived mononuclear cells after 24 hours incubation showing no toxicity up to a concentration of 500 ug/mL.
  • Figure 8 Radioactive emission images (a-d) and fluorescence images (e-h) of MF-nCP sample imaged at varying depths in pork tissue.
  • the bottle containing the sample placed in pork tissue was rotated and images taken at varying angles. Depth at which the sample was imaged from, in tissue, is denoted as d and the angle at which the bottle is rotated is denoted as ⁇ .
  • Figure 9 In vivo multimodal imaging of MF-nCP after tail vein injection into mice.
  • Figure (a) shows the fluorescence image
  • (b) shows radioactive emission image
  • (c) shows radioactive emission image overlaid over X-ray images.
  • the method of preparation of said nanoparticle and nanomedicine formulation consists the steps of:
  • the precursor compound, part A is formed from a material selected from the group consisting of sulphate, phosphate, hydroxide, chloride, bromide, iodide, fluoride, nitrate, carbonate or oxide salts of calcium.
  • another precursor compound, Part B is formed from water soluble or miscible salt of phosphates including sodium (Na 3 P0 4 , Na 2 HP0 4 , NaH 2 P0 4 ), potassium (K 3 P0 4 , KsHPC-4, KH 2 P0 4 ) lithium (Li 3 P0 4 , Li 2 HP0 4 , LiH 2 P0 4 ), ammonium ((NH 4 ) 3 P0 4 , (NH 4 ) 2 HP0 4 , NH 4 H 2 P0 4 ) or phosphoric acid
  • the precursor compound containing hydroxyl anions (Part C) is formed by hydroxide salts of sodium, potassium, lithium, ammonium or calcium .
  • the dopant molecules (Part D) in the nanoparticle that gives raman scattering is formed by cyanine dyes (Cy3, Cy5, ICG), rhodamine (rhodamine 6G, tetramethyl rhodamine, rhodamine no), aniline blue, crystal violet, malachite green, basic fuchsin, variamine blue RT salt, triaminopyrimidine sulphate or porphyrins.
  • cyanine dyes Cy3, Cy5, ICG
  • rhodamine rhodamine 6G, tetramethyl rhodamine, rhodamine no
  • aniline blue crystal violet
  • malachite green malachite green
  • basic fuchsin basic fuchsin
  • variamine blue RT salt triaminopyrimidine sulphate or porphyrins.
  • the dopant molecules (Part D) that gives near-infrared fluorescence to the nanoparticle is NIR emitting dyes such as Indocyanine green (ICG).
  • the dopant ions (Part D) in the nanoparticle that gives magnetic contrast includes ions such as chromium(III), manganese(II), iron(II), iron (III), praseodymium (III), neodymium (III), samarium(III), ytterbium(III), gadolinium(lll), terbium(III), dysprosium(III), holmium(III), erbium(III).
  • the dopant ions (Part D) in the nanoparticle that gives X-ray contrast includes ions such as iodine or barium, bismuth, strontium tungsten, tantalum, hafnium, lanthanum, molybdenum, niobium, zirconium.
  • the doped nanoparticle is conjugated with a radioactive element that includes Sm-153, Tc- 99m, 12 3I, 18F, l 3 » I, '"In, l88 Re, l66 Ho, ⁇ ⁇ 0 r 82 Rb through ligands such as bisphOsphonates.
  • a radioactive element that includes Sm-153, Tc- 99m, 12 3I, 18F, l 3 » I, '"In, l88 Re, l66 Ho, ⁇ ⁇ 0 r 82 Rb through ligands such as bisphOsphonates.
  • the nanoparticle with dopant ions / molecules were formed by reacting precursor Part-A, B, C and D at a temperature range of 25-i50°C and pH range of 5 -12.
  • the dopant ions may be mixed preferably with either Part-A or Part B or added separately during the reaction between Part A, B and C.
  • the formed nanoparticles are grown to a preferred size scale, between 1-200 nm by aging of the reactants at time scales of any range between 0-24 Hrs, preferably, 0-6 Hrs and most preferably 1-4 hrs at a temperature range of 5-i50°C.
  • Figure 2b and 2c shows the SEM and DLS images of ICG and Gd3 + co-doped multifunctional hydroxyapatite nanoparticles (MF-nHAp). 90% of the particles synthesized were ⁇ 100 ⁇ 25 nm in size as seen in DLS and SEM data.
  • the XRD data of MF-nHAp gave characteristic peaks of hexagonal HAp crystal lattice as shown in Figure 2a.
  • inset of Figure 3A shows the fluorescence excitation and emission spectra of ICG and Gd3 + co-doped MF-nHAp, peaking at 760 nm and 830 nm proving the capability of the material for non invasive in vivo NIR imaging.
  • the total weight percentage is varied in the range of 0.01 - 1%.
  • the total concentration of ICG in the precursor solution is maintained less than 2xio _ s M to avoid agglomeration of dye molecules.
  • the said nanoparticle can be used for molecular imaging based NIR fluorescence for biomedical applications. This is displayed in phantom experiments as shown in Figure 3B that shows increase in fluorescence with increase in MF-nHAp concentration.
  • the nanoparticles show X-ray contrast properties (radio opacity) together with NIR imaging.
  • X-ray contrast imaging of powder nanoparticle samples of different dopant concentrations are displayed.
  • Undoped and doped samples were made into pellets and imaged using 12.87 KeV X-ray energy (0.4mm filter) for an exposure time of 30 seconds in a digital X-ray imaging station (Kodak in vivo multispectral imaging system, Carestream, USA).
  • the X-Ray density of the material which is a measure of the X-ray absorbed or attenuated, was calculated and plotted in Figure 3C.
  • the said NIR emitting nanoparticles show paramagnetism together with NIR fluorescence and X-ray contrast when co-doping ICG with paramagnetic Gd ions.
  • Figure 3E refers to magnetic studies carried out by keeping the sample under a varying magnetic field, in an instrument called vibrating sample magnetometer. When undoped sample show diamagnetic property the ICG and Gadolinium doped samples show paramagnetic property, which increases with the concentration of dopant ions, gadolinium in this case.
  • FIG. 4a shows the radioactive emission from MF- nCP samples. As seen in the figure tagging of Tc-99m increases with increase in Tc-99m concentration till a saturation value of 250 ⁇ ' (Figure 4c). Figure 4b shows increase in fluorescence with increase in % doping of ICG till a saturation value of 0.16 mole% ( Figure 4d).
  • FIG. 5A shows the hemocompatibility of the doped nHAp (D-nHAp) where D-nHAp and D- nHAp cause hemolytic effects upto a concentration of as shown in Figure 5A .
  • Figure 5B shows the blood samples treated with D-nHAp and triton where hemolysis is clearly visible in triton treated samples and D-nHAp treated sample is comparable to PBS treated blood.
  • Figure 6 shows SEM image of RBC treated with D-nHAp ( Figure 6B) that shows no membrane damage compared to PBS treated RBC ( Figure 6A).
  • Figure 6D shows EDAX data of D-nHAp distributed over RBC.
  • the said nanoparticles are highly bio-compatible to primary human cells, for eg; peripheral blood mononuclear cells, as shown in Figure 7 .
  • Phantom imaging using Tc tagged and ICG doped MF-nCP was done in pork tissue samples to check the efficiency of contrast when imaged in tissue samples.
  • Figure 8 shows that both NIR fluorescence and radioactivity is imageable at varying depths up to 2.5 cm.
  • Later imaging in mice was carried out using the same MF-nCP nanoparticles that proved the ability of the material for multimodal imaging under in vivo conditions.
  • Figure 9 shows the fluorescent and radioactive imaging of mice after tail vein injection of the MF-nCP sample. All these imaging experiment were carried out in an in vivo imaging station (Kodak in vivo multispectral imaging system, Carestream, USA).
  • the high resolution and blood penetration capability of near-infrared light can help doctors, during a surgery, to identify angiogenic blood vessels in the vicinity of a solid tumor, for its effective removal during a surgical procedure.
  • Angiogenesis is a process of formation of larger concentration of new blood vessels around a growing tumor and the removal of such blood vessels is critical in stopping recurrence of tumor.
  • the said nanoparticles conjugated with antibody targeted against angiogenic growth factors may illuminate the cancer related blood vessels together with providing contrast for the tumor.
  • the doped nanoparticles are cationic in nature, the same can be loaded with nucleic acid drugs such as DNA or RNA or its small fractions like siRNA, for therapeutic gene delivery applications.
  • nucleic acid drugs such as DNA or RNA or its small fractions like siRNA, for therapeutic gene delivery applications.
  • mAbs monoclonal antibodies
  • pep peptides
  • similar ligands ligands
  • the said nanoparticles can also be Coated with a biodegradable polymers containing anticancer drugs or nucleic acid drugs.
  • the biodegradable polymers can be formed by polymers such as poly-lactic acid (PLA), poly(lactic-co-glycolic acid) (PLGA), polyvinylpyrrolidon (PVP), polyvinyl alcohol (PVA), polyethyleneimine (PEI), polyethelene glycol (PEG), chitosan, carboxymethyl chitosan, cyclodextrin, thermosensitive polymers such as Poly(JV- isopropylacrylamide) and its derivative or proteins such as bovine or human serum albumin.
  • PLA poly-lactic acid
  • PLGA poly(lactic-co-glycolic acid)
  • PVP polyvinylpyrrolidon
  • PVA polyvinyl alcohol
  • PEI polyethyleneimine
  • PEG polyethelene glycol
  • chitosan carboxymethyl chitosan
  • cyclodextrin thermosensitive polymers
  • thermosensitive polymers such as Poly(JV- isopropylacrylamide) and its derivative or proteins such as
  • the said nanoparticle or drug loaded nanomedicine construct is connected, at the surface with cancer targeting ligands including antibodies, peptides or small molecular ligands such as folic acid or aptamers
  • the present invention provides an injectable composition or composition for oral administration comprising the said nanomedicine according to the present invention as described above, together with a pharmaceutically acceptable medium.
  • the present invention provides a method of image guided delivery of the drug, estimation of drug concentration at diseased site with respect to other regions, estimation of pharmacokinetics and pharmacodynamics, treatment planning and estimation of treatment efficacy after preferred treatment, all the above using any of the said contrast imaging property of the nanoparticle
  • essentially the present invention provides a method of simultaneously detecting and treating the disease like cancer with the help of more than one and upto 05 different molecular imaging techniques using a single nanoparticle system.
  • the inventors have discovered primarily a nanoparticle that provides novel concept for combining a number of important molecular imaging techniques such as radioactivity, raman scattering, near-infrared fluorescence imaging, which can register both spatial and temporal functional properties of a disease at microscopic level such as expression level of a cancer biomarker, with other properties such as super/para-magnetism and X-ray absorbance which gives micro/ macroscopic anatomical information.
  • the inventive step provide a nanomedicine formulation, containing the said nanoparticle as a mairi component together with therapeutic molecules such as chemo drugs or nucleic acids that can treat disease in a targeted manner by conjugation of a ligand specific to a receptor in the diseased tissue /cell.
  • Example 1 Multifunctional contrast agent based on calcium phosphate nanoparticles with 2 dopants for combined NIR and radioactive imaging.
  • the pH during the entire process was maintained at ⁇ 7.4 by the addition of 0.1M NH 4 OH solution.
  • the precipitate was left stirring for another 30 minutes, centrifuged and washed at least 2 times.
  • the entire process was carried out room temperature at ⁇ 25°C. - 3omg/mL of the washed product was treated with lmL of 250 ⁇ Tc-MDP (Tc- 99m conjugated to methylene diphosphate). The mixture is vortexed for 2 minutes and incubated for another 20 minutes.
  • the conjugate is washed at least twice and resuspended in PBS or distilled water for further applications.
  • ICG can be replaced with any NIR emitting dye or raman active dye molecules for the synthesis of other NIR emitting or raman active calcium phosphate nanoparticles.
  • Example 2 Multifunctional contrast agent based on calcium phosphate nanoparticles with more than 2 dopants for combined raman. magnetic. X-ray and radioactive imaging.
  • the entire process was carried out room temperature at ⁇ 25 C. ⁇ 3omg/mL of the washed product was treated with lmL of 250 ⁇ Tc-MDP(Tc-99m conjugated to methylene diphosphate). The mixture was vortexed for 2 minutes and incubated for another 20 minutes. The conjugate was washed at least twice and resuspended in PBS or distilled water for further applications.
  • Aniline blue can be replaced by any other raman active or NIR emitting dye for the synthesis of other raman active or NIR emitting calcium phosphate nanoparticles.
  • Gadolinium can be replaced by any other impurity ion to give magnetic or X-ray contrast properties.
  • Tc-99m can be replaced with any other radioactive element to synthesize other radioactive calcium phosphate nanoparticles.
  • Example 3 Multifunctional contrast agent based on hydroxyapatite nanoparticles with more than 2 dopants for combined raman. magnetic. X-rav and radioactive imaging,
  • Aniline blue can be replaced by any other raman active or NIR emitting dye for the synthesis of other raman active or NIR emitting hydroxyapatite nanoparticles.
  • Gadolinium can be replaced by any other impurity ion to give magnetic or X-ray contrast properties.
  • Tc-99m can be replaced with any other radioactive element to synthesize other radioactive hydroxyapatite nanoparticles.
  • Example 4 Preparation of siRNA incorporated calcium phosphate nanoparticles based nanomedicines capable of image guided delivery.
  • siRNA therapeutic small interfering RNA
  • Antisense strand 3 1 -dTdTCGGCGAGCAACCUUGAGGU— 5 1
  • lmg/ml bare nanoparticles prepared as said in any of the above examples 1-3 is prepared in phosphate buffer saline (PBS, Sigma, USA)) and sonicated for 10 minutes to get a fine dispersion.
  • PBS phosphate buffer saline
  • the nCP-siRNA conjugates were further treated with o.oimg/ml BSA (Bovine Serum Albumin) and o.img/ml EDC (i-ethyl-3-(3- dimethylaminopropyl) carbodiimi.de) for 3omin at 37°C. After 30min, the nanoconjugates are removed by centrifugation and re-suspended in PBS. This forms a protective shell of albumin protein that can be derivatized using targeting ligands.
  • BSA Bovine Serum Albumin
  • EDC i-ethyl-3-(3- dimethylaminopropyl) carbodiimi.de
  • this example provides preparation of a representative nanomedicine formed by loading the said multifunctional calcium phosphate nanoparticles with a representative gene.
  • a gene preferably a marker gene that can be readily detected by simple laboratory tools
  • An appropriate marker gene selected for this example is beta.- galactosidase ( ⁇ -gal) since its expression can be readily detected by addition of X- gal, a substrate which yields a blue color when the active enzyme is present.
  • ⁇ -gal beta.- galactosidase
  • this example is not limited to the said marker gene, but any gene intended for a desired function such as inhibition of tumor growth.
  • Part B Another two solutions containing phosphate group (Part B), ammonium dihydrogen phosphate, (5 mL, 0.3M, 98%, Qualigens, India) and 1 ml NH 4 OH (0.1M, 25% NH 3 , Qualigens, India) are prepared. Both these solutions are added drop wise to the precursor mixture of Part A+D with continuous stirring over a period of 15 minutes. The rate of addition of ammonium hydroxide solution is adjusted to maintain the pH of the reaction medium at ⁇ 7.4 throughout the reaction. After completion of precipitation, the mixture is kept stirring for 1 hour followed by separation of the precipitate by centrifugation at 3000 rpm for lominutes and washing with ice cold water for 3-5 times.
  • the DNA embedded within the doped calcium phosphate matrix is optionally protected from enzymatic degradation using a protective coat of BSA, by treating the nCP-DNA conjugates with o.oimg of BSA and o.img of EDC in 5ml PBS for 30mm at 37°C, followed by washing with water.
  • Example 6 Preparation of nCP-polymer nanomedicine containing chemo drugs capable of image guided delivery
  • a representative nanomedicine formed by the said multifunctional calcium phosphate nanoparticles embedded within polymeric nanoparticle containing chemical drugs, for example a small molecule inhibitor, Sorafenib is presented, l mg/ml bare nanoparticles prepared as said in any of the above examples 1-3 is prepared in DMSO (dimethyl sulfoxide) medium and sonicated for 10 minutes to get a fine dispersion.
  • DMSO dimethyl sulfoxide
  • ⁇ ⁇ Sorafenib in lmg/ml PLGA poly(lactic-co-glycolic acid)
  • DMSO dimethyl sulfoxide
  • ⁇ 500 ⁇ , of H 2 0 is added to the CP-Sorafenib-PLGA mixture to precipitate the polymeric nanoparticles that contain CP and Sorafenib embedded in it.
  • This nanomedicine formulation can be used to deliver drug actively or passively to tumor sites together with imaging using multiple imaging modalities.
  • PLGA other biodegradable polymers such as PEI, PLA, PCL, PVA, PPV etc also can be used for embedding the nCP and chemical drug.
  • Example 7 Bisphosphonate tagged nCP for imaging and therapeutic applications
  • synthesis of bisphosphonate drug, alendronate, tagged Tc99m labeled doped calcium phosphate nanoparticles for imaging and therapy of diseases such as bone metastasis or other bone metabolic disorders, osteoporosis etc is presented.
  • the precipitate was left stirring for another 30 minutes, centrifuged and washed at least 2 times. The entire process was carried out room temperature at ⁇ 25°C. ⁇ 30mg/mL of the washed product was treated with lmL of 250 ⁇ Tc99m-alendronate (Tc-99m conjugated to alendronate drug). The mixture is vortexed for 2 minutes and incubated for another 20 minutes. The conjugate is washed at least twice and resuspended in PBS or distilled water for further applications.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nanotechnology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Organic Chemistry (AREA)
  • Optics & Photonics (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Dispersion Chemistry (AREA)
  • Microbiology (AREA)
  • Inorganic Chemistry (AREA)
  • Immunology (AREA)
  • Medical Informatics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Plant Pathology (AREA)
  • Ceramic Engineering (AREA)
  • Dermatology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne un matériau nanométrique qui peut produire une accentuation de contraste pour de multiples procédés d'imagerie et également délivrer des molécules thérapeutiques telles que des acides nucléiques ou des médicaments chimiothérapeutiques à des sites pathologiques tels qu'un cancer. En particulier, la présente invention concerne des phosphates de calcium synthétique nanométriques et des nanomatériaux d'apatite calcique présentant un contraste simultané pour au moins deux quelconques des modalités d'imagerie médicale comprenant l'imagerie radio, raman, de fluorescence dans l'infrarouge proche, de résonance magnétique et radiographique.
PCT/IN2013/000143 2013-03-12 2013-03-12 Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament WO2014141288A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IN2013/000143 WO2014141288A1 (fr) 2013-03-12 2013-03-12 Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IN2013/000143 WO2014141288A1 (fr) 2013-03-12 2013-03-12 Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament

Publications (1)

Publication Number Publication Date
WO2014141288A1 true WO2014141288A1 (fr) 2014-09-18

Family

ID=48703628

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2013/000143 WO2014141288A1 (fr) 2013-03-12 2013-03-12 Art, procédé, manière, processus et systèmes d'un nano-biominéral pour imagerie de contraste multimodale et administration de médicament

Country Status (1)

Country Link
WO (1) WO2014141288A1 (fr)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104288784A (zh) * 2014-10-24 2015-01-21 浙江理工大学 纳米羟基磷灰石-基因-药物复合物及制备方法和应用
WO2015181138A1 (fr) * 2014-05-28 2015-12-03 Evonik Röhm Gmbh Nanoparticule
CN105327366A (zh) * 2015-11-17 2016-02-17 上海纳米技术及应用国家工程研究中心有限公司 一种用于肿瘤成像与治疗的金纳米花的制备
CN105327364A (zh) * 2015-12-15 2016-02-17 浙江理工大学 一种纳米羟基磷灰石-siRNA复合物及其制备方法
WO2017096342A1 (fr) * 2015-12-03 2017-06-08 Amrita Vishwa Vidyapeetham Nanoparticules dopées sensibles aux ondes radio pour thérapie guidée par image
WO2017152127A1 (fr) * 2016-03-03 2017-09-08 Amrita Vishwa Vidyapeetham Implant composite à contraste irm et tdm pour régénération et thérapie de tissu guidées par image
WO2017201375A1 (fr) * 2016-05-19 2017-11-23 Amrita Vishwa Vidyapeetham Microbilles radio-opaques radio-marquées non iodées à contraste irm pour radio-embolisation
CN108866046A (zh) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 一种分离、纯化样品中外泌体和/或游离核酸的方法
CN109125744A (zh) * 2018-08-24 2019-01-04 浙江大学 一种具有mri与ct双模态成像功能的钆掺杂氧化铪纳米颗粒的制备方法
CN109395078A (zh) * 2018-09-28 2019-03-01 浙江大学 一种MoO2-ICG多功能纳米颗粒及其制备方法与应用
CN110646401A (zh) * 2019-10-12 2020-01-03 福建师范大学 基于羟基磷灰石纳米粒子吸附蛋白的sers检测方法
CN110898234A (zh) * 2019-12-25 2020-03-24 河南大学 一种二维铋纳米复合材料及其制备方法和应用
CN112451665A (zh) * 2020-11-26 2021-03-09 山西医科大学 一种新型nir-ⅱ诊疗一体化硅-碳纳米探针及其制备方法

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5342609A (en) 1991-10-22 1994-08-30 Mallinckrodt Medical, Inc. Microfluidization of calcium/oxyanion-containing particles
US5344640A (en) 1991-10-22 1994-09-06 Mallinckrodt Medical, Inc. Preparation of apatite particles for medical diagnostic imaging
US20100092364A1 (en) * 2008-10-10 2010-04-15 Rajendra Kumar Kasinath Surface Functionalized Colloidally Stable Spheroidal Nano-apatites Exhibiting Intrinsic Multi-functionality
US20100311902A1 (en) * 2005-09-16 2010-12-09 Pentax Corporation Nanoparticles comprising calcium phosphate ethylene imine compositions and methods of production thereof
US20110129413A1 (en) * 2009-11-06 2011-06-02 Morgan Thomas T Bioconjugation of Calcium Phosphosilicate Nanoparticles For Selective Targeting of Cells In Vivo
WO2011151631A1 (fr) * 2010-06-04 2011-12-08 King's College London Nanoparticules et leurs utilisations en imagerie moléculaire

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5342609A (en) 1991-10-22 1994-08-30 Mallinckrodt Medical, Inc. Microfluidization of calcium/oxyanion-containing particles
US5344640A (en) 1991-10-22 1994-09-06 Mallinckrodt Medical, Inc. Preparation of apatite particles for medical diagnostic imaging
US5468465A (en) 1991-10-22 1995-11-21 Mallinckrodt Medical, Inc. Treated apatite particles for x-ray diagnostic imaging
US5690908A (en) 1991-10-22 1997-11-25 Mallinckrodt Medical, Inc. Treated apatite particles for medical diagnostic imaging
US20100311902A1 (en) * 2005-09-16 2010-12-09 Pentax Corporation Nanoparticles comprising calcium phosphate ethylene imine compositions and methods of production thereof
US20100092364A1 (en) * 2008-10-10 2010-04-15 Rajendra Kumar Kasinath Surface Functionalized Colloidally Stable Spheroidal Nano-apatites Exhibiting Intrinsic Multi-functionality
US20110129413A1 (en) * 2009-11-06 2011-06-02 Morgan Thomas T Bioconjugation of Calcium Phosphosilicate Nanoparticles For Selective Targeting of Cells In Vivo
WO2011151631A1 (fr) * 2010-06-04 2011-12-08 King's College London Nanoparticules et leurs utilisations en imagerie moléculaire

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ARAVIND A ET AL: "Aptamer conjugated paclitaxel and magnetic fluid loaded fluorescently tagged PLGA nanoparticles for targeted cancer therapy", JOURNAL OF MAGNETISM AND MAGNETIC MATERIALS 2013 ELSEVIER NLD, vol. 344, 2013, pages 116 - 123, XP002715928, DOI: 10.1016/J.JMMM.2013.05.036 *
ASHOKAN A ET AL: "A molecular receptor targeted, hydroxyapatite nanocrystal based multi-modal contrast agent", BIOMATERIALS, ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, vol. 31, no. 9, 1 March 2010 (2010-03-01), pages 2606 - 2616, XP026884285, ISSN: 0142-9612, [retrieved on 20100201], DOI: 10.1016/J.BIOMATERIALS.2009.11.113 *
ASHOKAN A ET AL: "Multifunctional calcium phosphate nano-contrast agent for combined nuclear, magnetic and near-infrared invivo imaging", BIOMATERIALS SEPTEMBER 2013 ELSEVIER LTD GBR, vol. 34, no. 29, September 2013 (2013-09-01), pages 7143 - 7157, XP002715927, DOI: 10.1016/J.BIOMATERIALS.2013.05.077 *
ASHOKAN ANUSHA ET AL: "Development and haematotoxicological evaluation of doped hydroxyapatite based multimodal nanocontrast agent for near-infrared, magnetic resonance and X-ray contrast imaging", NANOTOXICOLOGY, vol. 6, no. 6, September 2012 (2012-09-01), pages 652 - 666, XP008165707 *
CHEN FENG ET AL: "Multifunctional Eu3+/Gd3+ dual-doped calcium phosphate vesicle-like nanospheres for sustained drug release and imaging", BIOMATERIALS, vol. 33, no. 27, September 2012 (2012-09-01), pages 6447 - 6455, XP002715923 *
FENG CHEN ET AL: "The photoluminescence, drug delivery and imaging properties of multifunctional Eu<3+>/Gd<3+> dual-doped hydroxyapatite nanorods", BIOMATERIALS ELSEVIER SCIENCE LTD. UK, vol. 32, no. 34, December 2011 (2011-12-01), pages 9031 - 9039, XP002715924, ISSN: 0142-9612 *
LI Z ET AL: "Aptamer-capped multifunctional mesoporous strontium hydroxyapatite nanovehicle for cancer-cell-responsive drug delivery and imaging", BIOMACROMOLECULES 20121210 AMERICAN CHEMICAL SOCIETY USA, vol. 13, no. 12, 10 December 2012 (2012-12-10), pages 4257 - 4263, XP002715926, DOI: 10.1021/BM301563Q *
RAFAEL TORRES MARTIN DE ROSALES ET AL: "99m Tc-Bisphosphonate-Iron Oxide Nanoparticle Conjugates for Dual-Modality Biomedical Imaging", BIOCONJUGATE CHEMISTRY, vol. 22, no. 3, 16 March 2011 (2011-03-16), pages 455 - 465, XP055003271, ISSN: 1043-1802, DOI: 10.1021/bc100483k *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10172807B2 (en) 2014-05-28 2019-01-08 Evonik Röhm Gmbh Nanoparticle
WO2015181138A1 (fr) * 2014-05-28 2015-12-03 Evonik Röhm Gmbh Nanoparticule
CN104288784B (zh) * 2014-10-24 2017-11-24 浙江理工大学 纳米羟基磷灰石‑基因‑药物复合物及制备方法和应用
CN104288784A (zh) * 2014-10-24 2015-01-21 浙江理工大学 纳米羟基磷灰石-基因-药物复合物及制备方法和应用
CN105327366A (zh) * 2015-11-17 2016-02-17 上海纳米技术及应用国家工程研究中心有限公司 一种用于肿瘤成像与治疗的金纳米花的制备
CN105327366B (zh) * 2015-11-17 2018-08-17 上海纳米技术及应用国家工程研究中心有限公司 一种用于肿瘤成像与治疗的金纳米花的制备
WO2017096342A1 (fr) * 2015-12-03 2017-06-08 Amrita Vishwa Vidyapeetham Nanoparticules dopées sensibles aux ondes radio pour thérapie guidée par image
US11369681B2 (en) 2015-12-03 2022-06-28 Amrita Vishwa Vidyapeetham Radio-wave responsive doped nanoparticles for image-guided therapeutics
AU2016365412B2 (en) * 2015-12-03 2022-05-19 Amrita Vishwa Vidyapeetham Radio-wave responsive doped nanoparticles for image-guided therapeutics
US20190000973A1 (en) * 2015-12-03 2019-01-03 Amrita Vishwa Vidyapeetham Radio-wave responsive doped nanoparticles for image-guided therapeutics
CN105327364A (zh) * 2015-12-15 2016-02-17 浙江理工大学 一种纳米羟基磷灰石-siRNA复合物及其制备方法
CN105327364B (zh) * 2015-12-15 2019-03-12 浙江理工大学 一种纳米羟基磷灰石-siRNA复合物及其制备方法
US10806805B2 (en) 2016-03-03 2020-10-20 Amrita Vishwa Vidyapeetham MRI and CT contrast-enabled composite implants for image-guided tissue regeneration and therapy
WO2017152127A1 (fr) * 2016-03-03 2017-09-08 Amrita Vishwa Vidyapeetham Implant composite à contraste irm et tdm pour régénération et thérapie de tissu guidées par image
EP3458112A4 (fr) * 2016-05-19 2019-12-25 Amrita Vishwa Vidyapeetham Microbilles radio-opaques radio-marquées non iodées à contraste irm pour radio-embolisation
WO2017201375A1 (fr) * 2016-05-19 2017-11-23 Amrita Vishwa Vidyapeetham Microbilles radio-opaques radio-marquées non iodées à contraste irm pour radio-embolisation
CN108866046A (zh) * 2018-07-25 2018-11-23 辽宁润基生物科技有限公司 一种分离、纯化样品中外泌体和/或游离核酸的方法
CN109125744A (zh) * 2018-08-24 2019-01-04 浙江大学 一种具有mri与ct双模态成像功能的钆掺杂氧化铪纳米颗粒的制备方法
CN109125744B (zh) * 2018-08-24 2021-04-23 浙江大学 一种具有mri与ct双模态成像功能的钆掺杂氧化铪纳米颗粒的制备方法
CN109395078A (zh) * 2018-09-28 2019-03-01 浙江大学 一种MoO2-ICG多功能纳米颗粒及其制备方法与应用
CN109395078B (zh) * 2018-09-28 2021-06-25 浙江大学 一种MoO2-ICG多功能纳米颗粒及其制备方法与应用
CN110646401A (zh) * 2019-10-12 2020-01-03 福建师范大学 基于羟基磷灰石纳米粒子吸附蛋白的sers检测方法
CN110898234A (zh) * 2019-12-25 2020-03-24 河南大学 一种二维铋纳米复合材料及其制备方法和应用
CN112451665A (zh) * 2020-11-26 2021-03-09 山西医科大学 一种新型nir-ⅱ诊疗一体化硅-碳纳米探针及其制备方法
CN112451665B (zh) * 2020-11-26 2022-06-21 山西医科大学 一种nir-ⅱ诊疗一体化硅-碳纳米探针及其制备方法

Similar Documents

Publication Publication Date Title
WO2014141288A1 (fr) Art, procédé, manière, processus et systèmes d&#39;un nano-biominéral pour imagerie de contraste multimodale et administration de médicament
Cui et al. Renal clearable Ag nanodots for in vivo computer tomography imaging and photothermal therapy
Chen et al. Monitoring pH-triggered drug release from radioluminescent nanocapsules with X-ray excited optical luminescence
Kim et al. Recent development of inorganic nanoparticles for biomedical imaging
Parchur et al. Vascular interventional radiology-guided photothermal therapy of colorectal cancer liver metastasis with theranostic gold nanorods
Li et al. Multimodality imaging in nanomedicine and nanotheranostics
Huang et al. Calcium phosphate nanocarriers for drug delivery to tumors: imaging, therapy and theranostics
Sun et al. A pH-responsive yolk-like nanoplatform for tumor targeted dual-mode magnetic resonance imaging and chemotherapy
Fang et al. Multifunctional magnetic nanoparticles for medical imaging applications
Zhen et al. Development of manganese-based nanoparticles as contrast probes for magnetic resonance imaging
JP6619231B2 (ja) 癌を治療するための電離放射線との併用での無機ナノ粒子組成物
AU2016365412B2 (en) Radio-wave responsive doped nanoparticles for image-guided therapeutics
Chakravarty et al. Image-guided drug delivery with single-photon emission computed tomography: a review of literature
K Bakht et al. Scope of nanotechnology-based radiation therapy and thermotherapy methods in cancer treatment
KR100825939B1 (ko) 근적외선 형광체가 결합된 양친성 고분자의 나노 입자를포함하는 암 진단용 조영제
Feng et al. Renal-clearable peptide-functionalized Ba2GdF7 nanoparticles for positive tumor-targeting dual-mode bioimaging
WO2014141287A1 (fr) Technique, procédé, manière, processus et système de nanobiomatériau multifonctionnel destiné à l&#39;imagerie moléculaire et l&#39;administration de médicaments
US20150064107A1 (en) Imaging agent
Gupta et al. Metallic nanoparticles for CT-guided imaging of tumors and their therapeutic applications
WO2021228867A9 (fr) Procédés de radiothérapie guidée par image
Abueva Photo-triggered theranostic nanoparticles in cancer therapy
JP6318096B2 (ja) 希土類酸化物粒子及び特に画像化におけるその使用
Degli Esposti et al. Inorganic nanoparticles for theranostic use
TWI833803B (zh) 含有高z元素之奈米顆粒在製備用於治療腫瘤之藥物之用途
WO2021148973A1 (fr) Nanoparticules multifonctionnelles à base de nanoalliages métalliques à usage diagnostique et thérapeutique

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: 13732636

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13732636

Country of ref document: EP

Kind code of ref document: A1