WO2007021236A1 - A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle - Google Patents

A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle Download PDF

Info

Publication number
WO2007021236A1
WO2007021236A1 PCT/SE2006/000960 SE2006000960W WO2007021236A1 WO 2007021236 A1 WO2007021236 A1 WO 2007021236A1 SE 2006000960 W SE2006000960 W SE 2006000960W WO 2007021236 A1 WO2007021236 A1 WO 2007021236A1
Authority
WO
WIPO (PCT)
Prior art keywords
particle
cell
substance
membrane
particle according
Prior art date
Application number
PCT/SE2006/000960
Other languages
French (fr)
Inventor
Sarah Fredriksson
Sylvia Koivunen
Fredrik Olsson
Hanna-Karin Toftevall
Original Assignee
Genovis Ab
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 Genovis Ab filed Critical Genovis Ab
Priority to JP2008526907A priority Critical patent/JP4965570B2/en
Priority to US11/990,522 priority patent/US8557289B2/en
Priority to EP06784101A priority patent/EP1940513A4/en
Priority to CA002619118A priority patent/CA2619118A1/en
Priority to KR1020087006690A priority patent/KR101331891B1/en
Priority to CN200680030233XA priority patent/CN101242870B/en
Priority to AU2006280519A priority patent/AU2006280519B2/en
Publication of WO2007021236A1 publication Critical patent/WO2007021236A1/en
Priority to IL189559A priority patent/IL189559A/en
Priority to HK08113799.8A priority patent/HK1120452A1/en

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82BNANOSTRUCTURES FORMED BY MANIPULATION OF INDIVIDUAL ATOMS, MOLECULES, OR LIMITED COLLECTIONS OF ATOMS OR MOLECULES AS DISCRETE UNITS; MANUFACTURE OR TREATMENT THEREOF
    • B82B3/00Manufacture or treatment of nanostructures by manipulation of individual atoms or molecules, or limited collections of atoms or molecules as discrete units
    • 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
    • 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
    • A61K41/0028Disruption, e.g. by heat or ultrasounds, sonophysical or sonochemical activation, e.g. thermosensitive or heat-sensitive liposomes, disruption of calculi with a medicinal preparation and ultrasounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0052Thermotherapy; Hyperthermia; Magnetic induction; Induction heating therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal 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 colloid or an emulsion
    • A61K47/6907Medicinal 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 colloid or an emulsion the form being a microemulsion, nanoemulsion or micelle
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/02Magnetotherapy using magnetic fields produced by coils, including single turn loops or electromagnets

Definitions

  • the present invention relates to a particle suitable for the delivery of a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance, to the use of said particle in different applications as well as a method of delivering a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance .
  • Biomolecule delivery methods are indispensable tools for many Life scientistss. Transfection, siRNA evaluation and screening of various cell labeling methods are examples where efficient delivery to the target cell culture is crucial. Successful biomolecule non-viral delivery methods for in vitro purposes have to overcome a series of hindrance and avoid toxic effects on the target cell and still efficiently deliver functional biomolecules of choice within the cell.
  • the first step in the delivery process is for the biomolecule to reach the surfaces of the target cells .
  • the biomolecule should be delivered to the cell independent of media components in the cell culture and reach both adherent cell lines and cells in suspension. Once delivered at the cell surface the biomolecule has to either transverse the cell membrane/cell wall or enter the cell through an endocytosis pathway.
  • both the cytosol and the endosome contain various degrading enzymes .
  • Efficient delivery methods should preferably minimize cellular degrading of the biomolecule in question, whether it constitutes a DNA, RNA, protein or other molecule.
  • the biomolecule Once in the cytosol the biomolecule should preferably be targeted to a special intracellular compartment such as for instance the cell nucleus .
  • the present invetion relates in one aspect to a particle suitable for the delivery of a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance comprising: a superparamagnetic core capable of generating heat in a magnetic alternating field encapsulated in a heat sensitive coating whereto said substance and a membrane disruptive agent are associated.
  • the invention in a further aspect relates to the use of a particle for modifying the genetic code and/or metabolism of the host cell.
  • the invention relates to the use of a particle for releasing said substance from endosomal or lysosomal organelles.
  • the invention relates to a method of delivering a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance in combination with heat induced release of a membrane disruptive agent comprising:
  • Fig. 1 TEM image of an example of iron oxide core of the described nanoparticle .
  • Fig. 2 Heat effects
  • A The ' relation between the specific absorption r_ate (SAR-value) of the nanoparticles and the applied magnetic field strength in an alternating field with the frequency of 10 MHz.
  • B This graph illustrates the increase in temperature within a 100 ⁇ l sample of 7 mg Fe /ml in 0.15 M NaCl subjected to an alternating magnetic field of 10 MHz, 2.9 mT . The values have been corrected for the temperature increase in a reference sample without particles. Temperature measurements were performed with a fiber optic probe (IPITEK)
  • Fig. 3 Schematic illustration of experimental setup.
  • Fig. 4 Investigation of DNA binding capacity of themagnetic nanoparticles, surface-modified with amino groups, Linear PEI and Branched PEI. Different amounts of particles where incubated with plasmid DNA (phRLSV40-luc, 3.7 kb) forlO minutes in TE buffer. After addition of PicoGreen fluorescence was measured using a microplate fluorescence reader equipped with a FITC filter. Reduced Fluorescence indicates condensed DNA.
  • Fig. 5 Result from the test on high gradient magnetic separation of cells treated with particle as described in example 7.
  • Fig. 6 Results from transfection of K562 cells as described in example 10.
  • the sample marked as ⁇ cells' is a control of background generated in cells not transfected.
  • the present invention provides a particle suitable for biomolecule delivery, which combine endosomal release within a diversity of possible target cells with delivery of a variaty of biomolecules with a high efficiency.
  • the invention described here is a particle designed for biomolecule delivery in vitro or ex vivo which make use of the thermal energy released from the superparamagnetic core upon exposure to an alternating magnetic field to release membrane disruptive molecules such as lipids, cholesterol or detergents from the heat sensitive coating of said particle in order to induce chemical membrane disrupture.
  • membrane disruptive molecules such as lipids, cholesterol or detergents from the heat sensitive coating of said particle in order to induce chemical membrane disrupture.
  • Both the substance (e.g. a biomolecule) that is to be delivered hereinafter called cargo molecule
  • cargo molecule molecules that can promote disruption of cell or organelle membranes like endosome membrane are released during the heating of the superparamagnetic core of said particle.
  • the terms “substance” and "cargo molecule” are used interchangeably.
  • the substances or cargo molecules are delivered to the target cells through the particles of the invention.
  • An example of a substance or cargo molecule is a biomolecule.
  • Applications of magnetic particles in biomedicine and biotechnology are continuously growing.
  • Today magnetic nanoparticles are used for purification, quantification, transfection, hyperthermia, drug delivery, imaging by MRI among others .
  • Superparamagnetic particles entrapped together with prodrugs inside liposomes for drug delivery purposes have been described in US2003211045, where heating of the magnetic particles inside the liposomes will convert the prodrug into drug.
  • Yet another particle where superparamagnetic cores are incapsulted in organic compounds and vesicle forming lipids are described in US5441746, where the particles are intented for inductive heating of the superparamagnetic core to promote killing of cancer cells.
  • the present invention describes a superparamagnetic particle that in addition to generating heat in an alternating magnetic field is designated for non-viral biomolecule delivery in vitro or ex vivo.
  • the particle In addition to carrying the cargo molecule to the cell surface where the particle/cargo molecule complex is taken up by the cell through endocytosis, the particle is enhancing endosomal release of the cargo molecule when exposed to an alternating magnetic field.
  • the surface of the particle is hence not homogenous, but instead carries a combination of heat sensitive structures that will be released or change configuration upon pulses of heating.
  • Each particle of plurality described by the invention carries multiple coupling and or attachment sites for the cargo molecule, one or more membrane disrupting agents, recognition molecules which can recognize and bind to the target cells and signal-molecules (such as peptides) and fluorescent markers for imaging of the delivery process.
  • the size of the superparamagnetic core is between 1 and 100 nm, preferably between 8 and 15 nanometers .
  • said superparamagnetic core consists of magnetite, maghemite, ironoxidehydrate, ferrites of the general formula MeOxFe2O3 where Me is a bivalent metal selected from the group consisting of Co, Ni, Mn, Be, Mg, Ca, Ba, Sr, Cu, Zn, Pt, Al, Cr, Bi and combinations thereof.
  • the heat sensitive coating comprises a micelle like structure in direct contact with said core, wherein said micelle like structure is built up by at least one hydrophobic molecule and at least one molecule containing both an hydrophobic and a hydrophilic moiety.
  • the heat sensitive coating is attached to said substance and membrane disruptive agent with single or multiple disulfide bonds, peroxide bonds or azo bonds or combinations thereof.
  • said hydrophilic moiety structure contains polymers, lipids, synthetic molecules, proteins or combinations thereof.
  • At least one recognition molecule is also associated to the heat sensitive coating, wherein said recognition molecule is chosen from the group consisting of antibodies, fragments of antibodies and lectins.
  • the membrane disruptive agent is chosen from the group consisting of detergents, cholesterol, hydrophobic proteins and surfactants and mixtures thereof.
  • said substances or cargo molecules are selected from the group consisting of DNA molecules, RNA molecules, proteins, peptides, lipids, chemical preparations, organic compounds, detergents, synthetic polymers and combinations thereof.
  • said magnetic field has an alternating field direction of a frequency in the range of 0.1 to 5 MHz.
  • the magnetic field may have a field strength in the range of 1 to 100 mT .
  • said magnetic field is exposed repeatedly as a series of pulses over time each pulse duration preferably between 1 and 600 s, and even more preferably between 1 and 45 seconds.
  • the magnetic field may have an alternating field which is non-homogeneous and has an alternating gradient field direction, the direction of said alternating magnetic field being generated by two coils, and said sample is inserted between the coils.
  • the coils may be supplied with either the positive or negative part of the supplied alternating current.
  • the present invention constitutes a superparamagnetic core encapsulated in coating layer or layers, where the inner layer is in direct contact with the superparamagnetic core, see example 1 and 2.
  • the superparamagnetic core of the particle fulfils three desirable features; it is traceable using MRI, cells harbouring such particles can be sorted using magnetic separation technologies, see example 2 and 7 and most important; it can be made to generate heat.
  • An alternating magnetic field induces heat within the magnetic core of each particle due to both a rotation of the particle and to a rotation of the magnetic momentum within the particle, see example 3. Both of these phenomenon result in heat losses. Which one of them that dominates in a given moment depends on the particle size, field frequency and particle coating.
  • the preferred size of the core of the particle is between 8 and 15 nanometer, since cores in this size interval makes it possible to use frequencies in the lower mega hertz range of the alternating magnetic field and hence avoid eddy heating of target cells and surrounding growth media.
  • the coating of the particles should be kept as thin as possible to keep the size of the particle as a whole preferably below 50 nanometers. Small particles promote high surfaces and advantageous kinetics for binding to various cell surface, see examples 9. If the particle is small enough not to preciptate in a test tube due to gravity it is capable of interacting with both cells in suspension and adherent cell lines .
  • the coating of the particle is of great importance to keep the particles well separated from each other in a stable colloidal solution.
  • the stability in various buffers and cell culture media is crucial for the usefulness and effectiveness of the nanoparticle both in terms of surface modification, using various biochemical coupling technologies, as well as in terms of the effect on living cells.
  • amino-PEG can be introduced on the surface of the particles enabling coupling by NHS (N- hudroxysulfosuccinimide ester), SPDP (N-succinimidyl 3- (2 pyridyldithio) propionate) , glutaric aldehyde and other bioconjugate techniques.
  • the cargo molecule to be delivered to cells in vitro or ex vivo is attached to the coating covalently, electrostatically or through van der Waals interactions.
  • the cargo molecule being a DNA or RNA molecule electrostatic interactions between the negative backbone of DNA or RNA molecules and positively charged molecules like DOTAB integrated within the outer layer of the coating or positively charged molecules like polylysin, PEI (polyetylenimin) , protamine, arginin- or lysine rich peptides covalently coupled to a amino-PEG molecule in the outer surface layer of the heat sensitive coating is readily utilized. Additional molecules like florescent markers or target cell recognition molecules are attached by the same procedures as the cargo molecule.
  • the particle When the particle is complex bound with a cargo molecule, like DNA or RNA molecules for gene transfer purposes, the particle should serve as a stable and nontoxic carrier. Additionally, the particles should preferably protect their cargo-molecules from degradation as well as promote release of the very same molecule at the target site within the cell. By complexing the cargo molecule close to the particle surface a steric hindrance for degrading enzymes is introduced which in turn is favourable to prevent enzymatic degradation of the cargo molecule .
  • the nanoparticle can carry signal-molecules (such as peptides) and fluorescent markers.
  • the present invention can, in addition to carrying different cargo molecules, induce a heat release effect from the surface of the nanoparticle. The idea behind heat induced delivery is to increase the rate of molecular diffusion in close vicinity to the particle and to promote the release of molecules attached to the surface of the particles .
  • the inner layer consists of a hydrophobic molecule, for instance a fatty acid like oleic acid, or a mixture of hydrophobic molecules.
  • the second layer consists of a mixture of molecules each of them is characterized by one hydrophobic part and a hydrophilic part.
  • the hydrophobic part is building up a double layer, a micelle like structure, with the first layer while the hydrophilic part is directed outward into the surrounding media giving the particle stability in water based buffers and growth media.
  • This hydrophilic part is further utilized to attach the cargo molecule and any recognition molecule or signal molecule additionally attached to the surface of the particle coating.
  • phase transition temperature is defined as the temperature required to induce a change in the lipid physical state from the ordered gel phase, where the hydrocarbon chains are fully extended and closely packed, to the disordered liquid crystalline phase, where the hydrocarbon chains are randomly oriented and fluid.
  • the superparamagnetic core of the particle can be made to generate heat within an alternating field, the heating procedure can be made very precise in volume in very close vicinity to the particle.
  • the target cell as a whole will hence not be heated.
  • helper molecules like cholesterol, DOPE (dioleoyl-phosphatidylethanolamine) lipids is transported as parts/members of the surface outer layer.
  • Detergents like Triton X and SDS (sodium dodecyl sulfate) and similar molecules can also be parts/members of the outer coating layer. When these detergents are integrated in the particle they are less toxic to the target cells, but when released from the particle they are promoting disrupture of endosomal membrane (s) , see further in example 10.
  • endosomal membrane see further in example 10.
  • temperature sensitive covalent bridges between molecules in the outer coating layer and the cargo molecules it is possible to even more strongly promote the release of the cargo molecule from the nanoparticle for subsequent escape from the endosomal compartment, see example 4 and 5.
  • the particle according to the described invention is used for substance delivery into cell cultures in vitro or ex vivo.
  • the desired substance or substances is bound to the surface of the particle through electrostatic interactions, covalently or through van der Waal interactions as a cargo molecule.
  • the particle/cargo molecule complexes are mixed with the cell growth medium.
  • the surface of the particle may carry affinity molecules that can bind to certain targets on the surface of the target cells or have a net positive charge which promote the particle/cargo complex to interact with the target cells negative surface areas.
  • the particle is rapidly encapsulated by endocytos by the target cells and the early endosomes can then be transported into the cells and subsequently processed by the cells into late endosomes and transported to Golgi and endoplasmatic reticulum organelle compartments within the cells.
  • the magnetic field is then applied in pulses of a few seconds to minutes with pauses of equal time durations. Two to hundreds of pulses can be applied and the repeated heating cause leaking from the particle coating molecules and hence a release of both cargo molecule associated them and membrane disruptive mole- cules .
  • Membrane disruptive agents insert themselves in the endosomal membrane and hence causing rupture of the endosomes.
  • a heat sensitive covalent bond like for instance a disulfide bridge, can promote the release effect additionally when heated upon.
  • the incubation time of cells and particle before exposure to the magnetic field have an impact on the delivery procedure and is individually optimized for each specific cell line.
  • the magnetic field can be applied more than one occasion to improve the delivery process over time with an incubation time of a less than an hour to several hours in between each treatment .
  • Treated cells are automatically labeled and traceable in MRI and in addition fluorescent markers can be added to the particle coating to provide the possibility to follow the procedure in a florescent microscope.
  • the cells can be washed and sorted using high gradient magnetic field separation methods at any time and cells can be moved under a microscope when exposed to movements of a permanent magnet .
  • Particles are purified from excess reagents by washing repeatedly with ethanol, methanol, chloroform or a combination thereof and finally resuspended in chloroform or hexane .
  • EXAMPLE 2 Poly (ethyleneglycol) -lipid surface coating
  • PEG-lipid e.g 1 , 2-Dipalmitoyl-sn-Glycero-3-Phosphoethanolamine-N- [Methoxy (Polyethylene glycol) -2000] , 1, 2-Distearoyl-sn- Glycero-3-Phospho
  • Chloroform is evaporated and particles are further dried for 1-4 hours. Particles are then hydrated using water, NaCl or common physiological buffer and mildly sonicated to receive a clear solution that can be sterile filtered through a 100 nm filter. Particles are purified from lipids and PEG- lipids by high gradient magnetic separation and stored at 4 0 C. However, particles may be stored at room temperature for weeks without noticeable change in properties.
  • Particles remain colloidal stable for weeks in common buffers with physiological salt concentrations. Particles remain stable even in complex growth media containing 10% FCS (serum) for more than 3 days although upper time limits have not been tested, they may stay in solution for even longer time periods .
  • FCS serum
  • the particle suspension was easily purified from coupling chemicals and/or salts by filtration through a magnetic column (magnetic high gradient separation) or by size exclusion chromatography.
  • the particle suspension is applied to a column containing a magnetic matrix, which was exposed to a high permanent magnetic field of 1.2T .
  • the particles are retained in the magnetic matrix as long as the matrix is exposed to a high magnetic field.
  • the magnetic field was removed the particles was eluted with water or desired buffer solution. It was also possible to concentrate the ferrofluid on the magnetic column by eluting a smaller volume.
  • Size exclusion chromatography The particle suspension is applied to a column packed with SephacrylTM S-500 High Resolution gel.
  • the ferrofluid is separated from coupling chemicals and/or salts by size exclusion.
  • the ferrofluid is eluted with water or desired buffer solution. Size and stability
  • the average diameter of the iron oxide cores of the particles was estimated to 11 ⁇ 1 niti using TEM (trans electron microscope) , see figure 1.
  • the diameter of the whole particle varies depending on composition of the surface layer of lipids and characteristics of the carrier liquid.
  • the magnetic nanosparticles are easily- sterile filtered and colloidal stable in common physiological buffers and hence it was possible to utilize size exclusion chromatography, dialysis and high magnetic field separation during the process of production and further modification of the particles .
  • the amino groups on the surface of the particles were analysed using Fluram® with a standard protocol and the amount of amino groups could be varied between a few hundred up to several thousands amino groups per particle without affecting the stability of the particles. Furthermore, the amino groups are accessible to covalent coupling using common and well-established coupling chemistry for addition of other functional groups or molecules .
  • EXAMPLE 3 Heat losses in alternating magnetic fields
  • the cores of the particles are superparamagnetic and when such a particle is subjected to an alternating magnetic field heat is lost to the surrounding medium.
  • the frequency dependence of the specific absorption rate (SAR) of the particles was tested and evaluated. SAR values of more than 500 watt/g was achieved at a frequency of 10 MHz and field strength of 2.8 mT, as illustrated in figure 2.
  • SAR values of more than 500 watt/g was achieved at a frequency of 10 MHz and field strength of 2.8 mT, as illustrated in figure 2.
  • EXAMPLE 4 The effect of heat on release of molecules from the surface of the particles
  • TAT-peptide covalently coupled to the amino groups on the surface of the particle were treated as described above. This example was set up to find out if the release effect can be increased by covalently creating a bond that is heat sensitive between the amino group and another molecule.
  • the peptide was coupled to the particle with SPDP as mediator generating a disulfide bridge between the particle and the TAT-peptide. Disulfide bridges are known to be relatively heat sensitive. The results can be summarized as follows:
  • the adherent cell line COS-7 was grown in DMEM, 10 % FCS and 5 % Streptomycin/penicillin.
  • the suspension cell line K562 was grown in RPMI, 10 % FCS and 5 % CO2. Cells were passaged two times per week. Proliferation COS-7 and K562 cells were harvested and seeded in a 96-well plate with 10 5 cells per well in 100 ⁇ l complete DMEM or RPMI, respectively. Cells for a proliferation standard were seeded with 10 4 to 10 5 cells/well in triplicates. Particles with amino groups, linear PEI or branched PEI were diluted in an appropriate buffer to different Fe concentrations.
  • the proliferation assay was preformed after 24 hours incubation. It could be concluded that it seems like the proliferation decreases if the cells that carries relatively high concentrations of particles are exposed to an alternating magnetic field. Furthermore, modification of particles with PEI (polyethylenimin) renders them more toxic to cells, and K562 are more sensitive to the particles than COS-7. EXAMPLE 7 Separation and washing of cells
  • K562 cells were seeded in a 24 well micrititer plate with 1 ml of medium per well. Particle carrying DOTAB/DOPE/ phospholipids in mixed coating were added to the cells in the following concentrations: Samplel: 1 picogram Fe/cell, incubated two hours Sample2 : 2 picogram Fe/cell, incubated two hours Sample3 : 2 picogram Fe/cell, incubated two hours, no magnet where used in separation coloumn.
  • Sample4 2 picogram Fe/cell, incubated 24 hours
  • Sample5 2 picogram Fe/cell, incubated 48 hours
  • Sample ⁇ 2 picograra Fe/cell, incubated 72 hours
  • Sample7 0 picogram Fe/cell, incubated two hours
  • COS-7 cells were incubated for 12 hours with magnetic nanoparticles (100 pg iron/cell) labelled with Texas-Red and covalently modified with a FITC labelled HIV-I TAT-peptide.
  • This peptide can in a native state target and enter the cell nucleus.
  • Both the Texas-Red label (on the phospholipids) and the FITC labelled HIV-I TAT-peptide co-localize in the cell cytoplasm.
  • Nanoparticles with or without covalently coupled linear PEI (polyethyleneimin) or branched PEI to the amino PEG on the particle surface was diluted in buffer to the same particle concentration. Particles were mixed with the same amount of DNA (100 ng) in different particle to DNA ratios (0.1-500). A DNA standard was prepared (10-500 ng/ml). The solutions were incubated 10 min in room temperature then PicoGreen® was added and fluorescence was measured (Ex 485/Em 520) . The results are presented in Figure 5 and it can be concluded that all of the particles were capable of binding DNA and that particles with surface that exposed branched PEI bound DNA most effectively. EXAMPLE 10 Transfection with particles carrying a mixture of SDS , DOPE and DOTAB in comparison with particles carrying PEG- NH2 only.
  • Cells (K562) were incubated for 3 hours together with particles with DOTAB/DOPE/SDS coating carrying texas red and plasmid coding for luciferase (phRL-SV40) and as a refernce cells were incubated in the same way with particels with Amnio-PEG (NH2) coating carrying plasmid coding for luciferase (phRL-SV40) . After incubation, cells were treated in AMF (alternating magnetic field) repeatedly with 30 second pulses and 30 second rest in between pulses.
  • AMF alternating magnetic field

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Nanotechnology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Medical Informatics (AREA)
  • Immunology (AREA)
  • Ceramic Engineering (AREA)
  • Inorganic Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Manufacturing & Machinery (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

A nano sized particle for in vitro or ex vivo biomolecule delivery to cell cultures through heat induced endosomal release comprising a superparamagnetic core coated in a heat sensitive coating, comprising membrane disruptive components and binding sites for attachment of biomolecules and markers to be delivered. A method for introducing the release effect of a plurality of said biomolecule and endosomal disrupture molecules by applying an alternating field to a cell culture harbouring the particle described by the invention.

Description

A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
Technical field of the invention
The present invention relates to a particle suitable for the delivery of a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance, to the use of said particle in different applications as well as a method of delivering a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance . Background of the invention
Reliable biomolecule delivery methods are indispensable tools for many Life Scientists. Transfection, siRNA evaluation and screening of various cell labeling methods are examples where efficient delivery to the target cell culture is crucial. Successful biomolecule non-viral delivery methods for in vitro purposes have to overcome a series of hindrance and avoid toxic effects on the target cell and still efficiently deliver functional biomolecules of choice within the cell. The first step in the delivery process is for the biomolecule to reach the surfaces of the target cells . The biomolecule should be delivered to the cell independent of media components in the cell culture and reach both adherent cell lines and cells in suspension. Once delivered at the cell surface the biomolecule has to either transverse the cell membrane/cell wall or enter the cell through an endocytosis pathway. Once within the endosome pH is low and both the cytosol and the endosome contain various degrading enzymes . Efficient delivery methods should preferably minimize cellular degrading of the biomolecule in question, whether it constitutes a DNA, RNA, protein or other molecule. To achieve a successful delivery process where the biomolecule enter the cell through endocytosis it is desirable to promote endosomal release into the cytosol of as many biomolecules as possible from each endosomal compartment. Once in the cytosol the biomolecule should preferably be targeted to a special intracellular compartment such as for instance the cell nucleus .
The basic technologies for all non-viral biomolecule delivery methods available today can be divided into two approaches. Physical methods like electroporation, microinjection and gene bombardment which deliver the biomolecule through the cell membrane into the cytosol. The chemical/synthetical methods use the target cell endocytosis. Both technologies have specific features as well as limitations and are well established methods. The increased number of high throughput preclinical cell based in vitro studies on stem cells, primary cell lines as well as cultured standard laboratory cell lines requires efficient, robust and cost effective biomolecule delivery methods. To meet these needs it is desirable to improve delivery efficiency of chemical and synthetic delivery method. One approach to increase the efficiency is to actively promote endosomal release. Summary of the invention
The present invetion relates in one aspect to a particle suitable for the delivery of a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance comprising: a superparamagnetic core capable of generating heat in a magnetic alternating field encapsulated in a heat sensitive coating whereto said substance and a membrane disruptive agent are associated. The invention in a further aspect relates to the use of a particle for modifying the genetic code and/or metabolism of the host cell.
In a yet further aspect the invention relates to the use of a particle for releasing said substance from endosomal or lysosomal organelles. In a yet further aspect the invention relates to a method of delivering a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance in combination with heat induced release of a membrane disruptive agent comprising:
- providing a sample comprising membrane enclosed cells or cell organelles and a plurality of particles according to the invention; - applying a magnetic alternating field to said sample, whereby thermal energy of the particle core causes the heat sensitive coating of said particle to decompose, which cause the disruptive agent to disrupt the membrane of the enclosed cell or cell organelle and release said substance.
Brief description of the drawings
Fig. 1: TEM image of an example of iron oxide core of the described nanoparticle . Fig. 2 Heat effects (A) The' relation between the specific absorption r_ate (SAR-value) of the nanoparticles and the applied magnetic field strength in an alternating field with the frequency of 10 MHz. (B) This graph illustrates the increase in temperature within a 100 μl sample of 7 mg Fe /ml in 0.15 M NaCl subjected to an alternating magnetic field of 10 MHz, 2.9 mT . The values have been corrected for the temperature increase in a reference sample without particles. Temperature measurements were performed with a fiber optic probe (IPITEK) Fig. 3: Schematic illustration of experimental setup.
Fig. 4 Investigation of DNA binding capacity of themagnetic nanoparticles, surface-modified with amino groups, Linear PEI and Branched PEI. Different amounts of particles where incubated with plasmid DNA (phRLSV40-luc, 3.7 kb) forlO minutes in TE buffer. After addition of PicoGreen fluorescence was measured using a microplate fluorescence reader equipped with a FITC filter. Reduced Fluorescence indicates condensed DNA.
Fig. 5: Result from the test on high gradient magnetic separation of cells treated with particle as described in example 7.
Fig. 6: Results from transfection of K562 cells as described in example 10. The sample marked as Λcells' is a control of background generated in cells not transfected. Detailed description of the invention
With the foregoing in mind, the present invention provides a particle suitable for biomolecule delivery, which combine endosomal release within a diversity of possible target cells with delivery of a variaty of biomolecules with a high efficiency.
The present invention will now be described more fully hereinafter with references to the accompanying drawings, in which preferred embodiments of the invention are shown. This invention may, however, be embodied in many different forms and should not be construed as limited to the illustrated embodiments set forth herein. Rather, these embodiments are provided so that this disclosure will be thorough and will fully convey the scope of the invention to those skilled in the art. We have earlier described in WO 01/18168 a method for introduction or extraction of bioparticles into or from biological membrane-enveloped structures by applying an alternating magnetic field to a sample of super- paramagneic particles attached to the membrane enclosed cell or organelle, whereby an increase in the thermal energy causes heating of the superparamagnetic core of the particle, which in turn causes temporary openings in the cell membrane or organelle membrane.
The invention described here is a particle designed for biomolecule delivery in vitro or ex vivo which make use of the thermal energy released from the superparamagnetic core upon exposure to an alternating magnetic field to release membrane disruptive molecules such as lipids, cholesterol or detergents from the heat sensitive coating of said particle in order to induce chemical membrane disrupture. Both the substance (e.g. a biomolecule) that is to be delivered (herinafter called cargo molecule) and molecules that can promote disruption of cell or organelle membranes like endosome membrane are released during the heating of the superparamagnetic core of said particle. In the context of the present application the terms "substance" and "cargo molecule" are used interchangeably. The substances or cargo molecules are delivered to the target cells through the particles of the invention. An example of a substance or cargo molecule is a biomolecule. Applications of magnetic particles in biomedicine and biotechnology are continuously growing. Today magnetic nanoparticles are used for purification, quantification, transfection, hyperthermia, drug delivery, imaging by MRI among others . Superparamagnetic particles entrapped together with prodrugs inside liposomes for drug delivery purposes have been described in US2003211045, where heating of the magnetic particles inside the liposomes will convert the prodrug into drug. Yet another particle where superparamagnetic cores are incapsulted in organic compounds and vesicle forming lipids are described in US5441746, where the particles are intented for inductive heating of the superparamagnetic core to promote killing of cancer cells. In contrast to these earlier described lipid encapsulated superparamagnetic cores the present invention describes a superparamagnetic particle that in addition to generating heat in an alternating magnetic field is designated for non-viral biomolecule delivery in vitro or ex vivo. In addition to carrying the cargo molecule to the cell surface where the particle/cargo molecule complex is taken up by the cell through endocytosis, the particle is enhancing endosomal release of the cargo molecule when exposed to an alternating magnetic field. The surface of the particle is hence not homogenous, but instead carries a combination of heat sensitive structures that will be released or change configuration upon pulses of heating. Each particle of plurality described by the invention carries multiple coupling and or attachment sites for the cargo molecule, one or more membrane disrupting agents, recognition molecules which can recognize and bind to the target cells and signal-molecules (such as peptides) and fluorescent markers for imaging of the delivery process.
In an embodiment of the invention the size of the superparamagnetic core is between 1 and 100 nm, preferably between 8 and 15 nanometers . In a further embodiment of the invention said superparamagnetic core consists of magnetite, maghemite, ironoxidehydrate, ferrites of the general formula MeOxFe2O3 where Me is a bivalent metal selected from the group consisting of Co, Ni, Mn, Be, Mg, Ca, Ba, Sr, Cu, Zn, Pt, Al, Cr, Bi and combinations thereof.
In a further embodiment of the invention the heat sensitive coating comprises a micelle like structure in direct contact with said core, wherein said micelle like structure is built up by at least one hydrophobic molecule and at least one molecule containing both an hydrophobic and a hydrophilic moiety. The heat sensitive coating is attached to said substance and membrane disruptive agent with single or multiple disulfide bonds, peroxide bonds or azo bonds or combinations thereof. In an embodiment of the invention said hydrophilic moiety structure contains polymers, lipids, synthetic molecules, proteins or combinations thereof.
In a yet further embodiment of the invention at least one recognition molecule is also associated to the heat sensitive coating, wherein said recognition molecule is chosen from the group consisting of antibodies, fragments of antibodies and lectins. In a further embodiment of the invention the membrane disruptive agent is chosen from the group consisting of detergents, cholesterol, hydrophobic proteins and surfactants and mixtures thereof. In a further embodiment of the invention said substances or cargo molecules are selected from the group consisting of DNA molecules, RNA molecules, proteins, peptides, lipids, chemical preparations, organic compounds, detergents, synthetic polymers and combinations thereof.
In an embodiment of the method of the invention said magnetic field has an alternating field direction of a frequency in the range of 0.1 to 5 MHz. The magnetic field may have a field strength in the range of 1 to 100 mT . In a yet further embodiment of the invention said magnetic field is exposed repeatedly as a series of pulses over time each pulse duration preferably between 1 and 600 s, and even more preferably between 1 and 45 seconds. The magnetic field may have an alternating field which is non-homogeneous and has an alternating gradient field direction, the direction of said alternating magnetic field being generated by two coils, and said sample is inserted between the coils. The coils may be supplied with either the positive or negative part of the supplied alternating current.
The present invention constitutes a superparamagnetic core encapsulated in coating layer or layers, where the inner layer is in direct contact with the superparamagnetic core, see example 1 and 2. The superparamagnetic core of the particle fulfils three desirable features; it is traceable using MRI, cells harbouring such particles can be sorted using magnetic separation technologies, see example 2 and 7 and most important; it can be made to generate heat. An alternating magnetic field induces heat within the magnetic core of each particle due to both a rotation of the particle and to a rotation of the magnetic momentum within the particle, see example 3. Both of these phenomenon result in heat losses. Which one of them that dominates in a given moment depends on the particle size, field frequency and particle coating. The preferred size of the core of the particle is between 8 and 15 nanometer, since cores in this size interval makes it possible to use frequencies in the lower mega hertz range of the alternating magnetic field and hence avoid eddy heating of target cells and surrounding growth media. The coating of the particles should be kept as thin as possible to keep the size of the particle as a whole preferably below 50 nanometers. Small particles promote high surfaces and advantageous kinetics for binding to various cell surface, see examples 9. If the particle is small enough not to preciptate in a test tube due to gravity it is capable of interacting with both cells in suspension and adherent cell lines .
The coating of the particle is of great importance to keep the particles well separated from each other in a stable colloidal solution. The stability in various buffers and cell culture media is crucial for the usefulness and effectiveness of the nanoparticle both in terms of surface modification, using various biochemical coupling technologies, as well as in terms of the effect on living cells. For coupling chemistry of the cargo molecule and/or cell recognition molecules to the surface of the particle amino-PEG can be introduced on the surface of the particles enabling coupling by NHS (N- hudroxysulfosuccinimide ester), SPDP (N-succinimidyl 3- (2 pyridyldithio) propionate) , glutaric aldehyde and other bioconjugate techniques. The cargo molecule to be delivered to cells in vitro or ex vivo is attached to the coating covalently, electrostatically or through van der Waals interactions. In the case of the cargo molecule being a DNA or RNA molecule electrostatic interactions between the negative backbone of DNA or RNA molecules and positively charged molecules like DOTAB integrated within the outer layer of the coating or positively charged molecules like polylysin, PEI (polyetylenimin) , protamine, arginin- or lysine rich peptides covalently coupled to a amino-PEG molecule in the outer surface layer of the heat sensitive coating is readily utilized. Additional molecules like florescent markers or target cell recognition molecules are attached by the same procedures as the cargo molecule.
When the particle is complex bound with a cargo molecule, like DNA or RNA molecules for gene transfer purposes, the particle should serve as a stable and nontoxic carrier. Additionally, the particles should preferably protect their cargo-molecules from degradation as well as promote release of the very same molecule at the target site within the cell. By complexing the cargo molecule close to the particle surface a steric hindrance for degrading enzymes is introduced which in turn is favourable to prevent enzymatic degradation of the cargo molecule . In addition to cargo molecules the nanoparticle can carry signal-molecules (such as peptides) and fluorescent markers. The present invention can, in addition to carrying different cargo molecules, induce a heat release effect from the surface of the nanoparticle. The idea behind heat induced delivery is to increase the rate of molecular diffusion in close vicinity to the particle and to promote the release of molecules attached to the surface of the particles .
Two layers of molecules build up the coating. The inner layer consists of a hydrophobic molecule, for instance a fatty acid like oleic acid, or a mixture of hydrophobic molecules. The second layer consists of a mixture of molecules each of them is characterized by one hydrophobic part and a hydrophilic part. The hydrophobic part is building up a double layer, a micelle like structure, with the first layer while the hydrophilic part is directed outward into the surrounding media giving the particle stability in water based buffers and growth media. This hydrophilic part is further utilized to attach the cargo molecule and any recognition molecule or signal molecule additionally attached to the surface of the particle coating.
When exposing the coating to temperatures corresponding to the coating lipids transition temperatures the coating described above is responding with interior molecular rearrangements or disruptions due to leading to a leakage of the coating from the particle, which in turn releases the substances attached to said coating. The phase transition temperature is defined as the temperature required to induce a change in the lipid physical state from the ordered gel phase, where the hydrocarbon chains are fully extended and closely packed, to the disordered liquid crystalline phase, where the hydrocarbon chains are randomly oriented and fluid. There are several factors, which directly affect the phase transition temperature including hydrocarbon length, unsaturation, charge, and head group species.
Since the superparamagnetic core of the particle can be made to generate heat within an alternating field, the heating procedure can be made very precise in volume in very close vicinity to the particle. The target cell as a whole will hence not be heated.
By including various molecules in the outer lipid layer a multipurpose heat release nanoparticle is built up. Known helper molecules like cholesterol, DOPE (dioleoyl-phosphatidylethanolamine) lipids is transported as parts/members of the surface outer layer. Detergents like Triton X and SDS (sodium dodecyl sulfate) and similar molecules can also be parts/members of the outer coating layer. When these detergents are integrated in the particle they are less toxic to the target cells, but when released from the particle they are promoting disrupture of endosomal membrane (s) , see further in example 10. In addition, by choosing temperature sensitive covalent bridges between molecules in the outer coating layer and the cargo molecules it is possible to even more strongly promote the release of the cargo molecule from the nanoparticle for subsequent escape from the endosomal compartment, see example 4 and 5.
In one embodiment of the invention described is a method where the particle according to the described invention is used for substance delivery into cell cultures in vitro or ex vivo. The desired substance or substances is bound to the surface of the particle through electrostatic interactions, covalently or through van der Waal interactions as a cargo molecule. The particle/cargo molecule complexes are mixed with the cell growth medium. The surface of the particle may carry affinity molecules that can bind to certain targets on the surface of the target cells or have a net positive charge which promote the particle/cargo complex to interact with the target cells negative surface areas. The particle is rapidly encapsulated by endocytos by the target cells and the early endosomes can then be transported into the cells and subsequently processed by the cells into late endosomes and transported to Golgi and endoplasmatic reticulum organelle compartments within the cells. The magnetic field is then applied in pulses of a few seconds to minutes with pauses of equal time durations. Two to hundreds of pulses can be applied and the repeated heating cause leaking from the particle coating molecules and hence a release of both cargo molecule associated them and membrane disruptive mole- cules . Membrane disruptive agents insert themselves in the endosomal membrane and hence causing rupture of the endosomes. If the cargo molecule is covalently coupled to the particle coating a heat sensitive covalent bond like for instance a disulfide bridge, can promote the release effect additionally when heated upon. The incubation time of cells and particle before exposure to the magnetic field have an impact on the delivery procedure and is individually optimized for each specific cell line. In addition the magnetic field can be applied more than one occasion to improve the delivery process over time with an incubation time of a less than an hour to several hours in between each treatment .
Treated cells are automatically labeled and traceable in MRI and in addition fluorescent markers can be added to the particle coating to provide the possibility to follow the procedure in a florescent microscope. In addition a the cells can be washed and sorted using high gradient magnetic field separation methods at any time and cells can be moved under a microscope when exposed to movements of a permanent magnet . EXAMPLE 1
Thermal decomposition of iron carboxylate salt
For production of monodisperse particles 11+1 nm the , following procedure was used. 360 mg of Iron (III) Oxide was grounded to smaller particles and added to 5 g of oleic acid and 11 g of 1-Octadecene . The solution is mixed and transferred to a three neck bottle flask, placed in a heating mantel with magnetic stirrer and heated until temperature reaches at least 3200C and 1- Octadecen starts boiling. During a one hour reaction time temperature is monitored and should preferably not drop down below the above mentioned temperature. After one hour incubation the solution was allowed to cool to room temperature. Particles are purified from excess reagents by washing repeatedly with ethanol, methanol, chloroform or a combination thereof and finally resuspended in chloroform or hexane . EXAMPLE 2 Poly (ethyleneglycol) -lipid surface coating
Briefly, particles were suspended in chloroform, and subsequentially dried under vacuum. A chloroform solution containing between 30-60% mol phospholipids (e.g 1,2- Dipalmitoyl-sΩ-Glycero-3-Phosphocholine or others similar in chemical structure) and 40% mol or more PEG-lipid (e.g 1 , 2-Dipalmitoyl-sn-Glycero-3-Phosphoethanolamine-N- [Methoxy (Polyethylene glycol) -2000] , 1, 2-Distearoyl-sn- Glycero-3-Phosphoethanolamine-N- [Amino (Polyethylene Glycol) 2000] or similar PEG-lipids, or others similar in chemical structure) are added to the dried particles which then returns to solution. Chloroform is evaporated and particles are further dried for 1-4 hours. Particles are then hydrated using water, NaCl or common physiological buffer and mildly sonicated to receive a clear solution that can be sterile filtered through a 100 nm filter. Particles are purified from lipids and PEG- lipids by high gradient magnetic separation and stored at 40C. However, particles may be stored at room temperature for weeks without noticeable change in properties.
Particles remain colloidal stable for weeks in common buffers with physiological salt concentrations. Particles remain stable even in complex growth media containing 10% FCS (serum) for more than 3 days although upper time limits have not been tested, they may stay in solution for even longer time periods .
The particle suspension was easily purified from coupling chemicals and/or salts by filtration through a magnetic column (magnetic high gradient separation) or by size exclusion chromatography.
Magnetic high gradient separation column
The particle suspension is applied to a column containing a magnetic matrix, which was exposed to a high permanent magnetic field of 1.2T . The particles are retained in the magnetic matrix as long as the matrix is exposed to a high magnetic field. When the magnetic field was removed the particles was eluted with water or desired buffer solution. It was also possible to concentrate the ferrofluid on the magnetic column by eluting a smaller volume.
Size exclusion chromatography The particle suspension is applied to a column packed with Sephacryl™ S-500 High Resolution gel. The ferrofluid is separated from coupling chemicals and/or salts by size exclusion. The ferrofluid is eluted with water or desired buffer solution. Size and stability
The average diameter of the iron oxide cores of the particles was estimated to 11 ± 1 niti using TEM (trans electron microscope) , see figure 1. The diameter of the whole particle varies depending on composition of the surface layer of lipids and characteristics of the carrier liquid. The magnetic nanosparticles are easily- sterile filtered and colloidal stable in common physiological buffers and hence it was possible to utilize size exclusion chromatography, dialysis and high magnetic field separation during the process of production and further modification of the particles . The amino groups on the surface of the particles were analysed using Fluram® with a standard protocol and the amount of amino groups could be varied between a few hundred up to several thousands amino groups per particle without affecting the stability of the particles. Furthermore, the amino groups are accessible to covalent coupling using common and well-established coupling chemistry for addition of other functional groups or molecules . EXAMPLE 3 Heat losses in alternating magnetic fields
The cores of the particles are superparamagnetic and when such a particle is subjected to an alternating magnetic field heat is lost to the surrounding medium. The frequency dependence of the specific absorption rate (SAR) of the particles was tested and evaluated. SAR values of more than 500 watt/g was achieved at a frequency of 10 MHz and field strength of 2.8 mT, as illustrated in figure 2. EXAMPLE 4 The effect of heat on release of molecules from the surface of the particles
Samples containing particles carrying phospholipids and amino-PEG- lipids with the amino groups fluorescently labeled with Fluram® were treated for 5 min in 10 MHz, 2,9 itiT followed by magnetic separation to get a first indication on the heat on the surface of the particles and possible heat release in the magnetic field. In comparison tests were also done with 5 min treatment of samples in temperatures ranging from 40 to 95 "C in a conventional water bath. The release of Fluram®-labeled amino groups was measured in the flow through from magnetic separation of the particles. The results can be summarized as follows: 1 . Between 2 and 6 % of the amino groups are released from the particle upon heating.
2. The amount of released group peaked between 50-60°C and then the amount of release decreased at higher temperatures . 3. The sample treated 5 min in the magnetic field released between 2 and 3 % of the amino groups . The amino-PEG-nn leaves the particle at relatively low temperature increases. This feature makes the particle suitable for slow release. If a molecule is coupled covalently to the amino group a slow and repeated release can be performed with a repeated treatment in an alternating magnetic field. EXAMPLE 7
To find out if both the amino-PEG-nn and the phospholipids are released at the same rate and temperature samples with particles carrying FITC labeled phospholipids were treated 5 min in temperatures ranging from 40 to 95 °C in a conventional water bath. The results can be summarized as follows: 1. Up to 25 % of the FITC labeled phospholipids are released from the particle upon heating above 80 "C for 5 minutes. 2. Samples treated at temperatures above 80 °C flocculated after treatment indicating colloidal instability.
This example illustrates that the phospholipids in comparison to the PEG-lipid is more reluctant to leave the iron oxide core and that heating above 80 °C has a severe impact on the colloidal stability of the particle suspension . EXAMPLE 5 Samples with particles carrying FITC labeled HIV
TAT-peptide covalently coupled to the amino groups on the surface of the particle were treated as described above. This example was set up to find out if the release effect can be increased by covalently creating a bond that is heat sensitive between the amino group and another molecule. The peptide was coupled to the particle with SPDP as mediator generating a disulfide bridge between the particle and the TAT-peptide. Disulfide bridges are known to be relatively heat sensitive. The results can be summarized as follows:
1. Between 3 and 13 % of the TAT-peptide was released from the particle upon heating.
2. The release effect increased with temperature. In comparison with the results presented in example 2 this is an increased release effect probably due to heat induced breakage of the disulfide bridges.
3. The samples treated with the magnetic field released between 3 and 5 % of the TAT- peptides . EXAMPLE 6
Cell growth
The adherent cell line COS-7 was grown in DMEM, 10 % FCS and 5 % Streptomycin/penicillin. The suspension cell line K562 was grown in RPMI, 10 % FCS and 5 % CO2. Cells were passaged two times per week. Proliferation COS-7 and K562 cells were harvested and seeded in a 96-well plate with 105 cells per well in 100 μl complete DMEM or RPMI, respectively. Cells for a proliferation standard were seeded with 104 to 105 cells/well in triplicates. Particles with amino groups, linear PEI or branched PEI were diluted in an appropriate buffer to different Fe concentrations. Each dilution of the different particles was added in quadruples to the cells to give a final Fe concentration between 50 and 250 pg Fe per cell. Additionally, tests on COS-7 cell proliferation upon exposure of particle/cell complex to an alternating magnetic field 10 MHz, 2.9 mT were performed using a experimental setup illustrated in figure 3. A water bath was used to cool the coil and keep the sample at desired temperature. The temperature within the sample is continuously monitored using an optical fibre thermometer immune to RF magnetic field.
The proliferation assay was preformed after 24 hours incubation. It could be concluded that it seems like the proliferation decreases if the cells that carries relatively high concentrations of particles are exposed to an alternating magnetic field. Furthermore, modification of particles with PEI (polyethylenimin) renders them more toxic to cells, and K562 are more sensitive to the particles than COS-7. EXAMPLE 7 Separation and washing of cells
K562 cells were seeded in a 24 well micrititer plate with 1 ml of medium per well. Particle carrying DOTAB/DOPE/ phospholipids in mixed coating were added to the cells in the following concentrations: Samplel: 1 picogram Fe/cell, incubated two hours Sample2 : 2 picogram Fe/cell, incubated two hours Sample3 : 2 picogram Fe/cell, incubated two hours, no magnet where used in separation coloumn.
Sample4 : 2 picogram Fe/cell, incubated 24 hours Sample5 : 2 picogram Fe/cell, incubated 48 hours Sampleδ : 2 picograra Fe/cell, incubated 72 hours Sample7 : 0 picogram Fe/cell, incubated two hours
Cells were removed from the culture dishes and washed using short centrifugation in buffer (PBS+BSA) . Live cells were marked with Calcein AM folowwed by subsequent washing and then applied to a high gradient magnetic separation coloumn (Milteyi) . Each sample was treated in a coloumn, sample nr 3 was separated without applying the external static magnetic field. The cells were washed with three coloumn volumes of buffer and eluated by releasing the column from the permanent magnet according to suppliers manual . Calcein flourescens were detected in the various fractions . Fractions : Flow through (FT)
Washings 1, 2, 3 (Wl, W2, W3) Elution (E)
From the result presented in figure 4 it is clearly seen that addition of 2 picograms FE/cell is enough to give treated cell enough magnetic material to be able to use high gradient magnetic field for separation and washing and the effect is lasting for up to 72 hours. EXAMPLE 8 Cellular uptake and distribution of particles Fluorescence microscopy of COS-7 cells incubated for 1 hour and 24 hours with superparamagnetic nanoparticles were utilized to follow the particles on there transport through the cell. It could be seen that the particles are readily and relatively quickly taken up by the cell and distributed evenly through the cell.
In another test COS-7 cells were incubated for 12 hours with magnetic nanoparticles (100 pg iron/cell) labelled with Texas-Red and covalently modified with a FITC labelled HIV-I TAT-peptide. This peptide can in a native state target and enter the cell nucleus. Both the Texas-Red label (on the phospholipids) and the FITC labelled HIV-I TAT-peptide co-localize in the cell cytoplasm.
EXAMPLE 9
Plasmid DNA binding capacity Nanoparticles with or without covalently coupled linear PEI (polyethyleneimin) or branched PEI to the amino PEG on the particle surface was diluted in buffer to the same particle concentration. Particles were mixed with the same amount of DNA (100 ng) in different particle to DNA ratios (0.1-500). A DNA standard was prepared (10-500 ng/ml). The solutions were incubated 10 min in room temperature then PicoGreen® was added and fluorescence was measured (Ex 485/Em 520) . The results are presented in Figure 5 and it can be concluded that all of the particles were capable of binding DNA and that particles with surface that exposed branched PEI bound DNA most effectively. EXAMPLE 10 Transfection with particles carrying a mixture of SDS , DOPE and DOTAB in comparison with particles carrying PEG- NH2 only.
Cells (K562) were incubated for 3 hours together with particles with DOTAB/DOPE/SDS coating carrying texas red and plasmid coding for luciferase (phRL-SV40) and as a refernce cells were incubated in the same way with particels with Amnio-PEG (NH2) coating carrying plasmid coding for luciferase (phRL-SV40) . After incubation, cells were treated in AMF (alternating magnetic field) repeatedly with 30 second pulses and 30 second rest in between pulses.
After AMF exposure cells were seeded into new culture dishes and proliferation an luminescens were read after 48 hours post treatment. As seen in figure 6 the particles carrying detergent and helper lipids (DOTAB/DOPE/SDS ) shows an substantially higher expression rate.

Claims

1. A particle suitable for the delivery of a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance comprising: a superparamagnetic core capable of generating heat in a magnetic alternating field encapsulated in a heat sensitive coating whereto said substance and a membrane disruptive agent are associated.
2. A particle according to claim 1, wherein the size of the superparamagnetic core is between 1 and 100 ran, preferably between 8 and 15 nanometers .
3. A particle according to claim 1 or 2, wherein said superparamagnetic core consists of magnetite, maghemite, ironoxidehydrate, ferrites of the general formula MeOxFe2O3 where Me is a bivalent metal selected from the group consisting of Co, Ni, Mn, Be, Mg, Ca, Ba, Sr, Cu, Zn, Pt, Al, Cr, Bi and combinations thereof.
4. A particle according to any one of claims 1-3, wherein said heat sensitive coating comprises a micelle like structure in direct contact with said core.
5. A particle according to claim 4, wherein said micelle like structure is built up by at least one hydrophobic molecule and at least one molecule containing both an hydrophobic and a hydrophilic moiety.
6. A particle according to claim 4 or 5, wherein said heat sensitive coating is attached to said substance and membrane disruptive agent with single or multiple disulfide bonds, peroxide bonds or azo bonds or combinations thereof.
7. A particle according to claim 5, wherein said hydrophilic moiety structure contains polymers, lipids, synthetic molecules, proteins or combinations thereof.
8. A particle according to any one of claims 1-7, wherein a recognition molecule is also associated to the heat sensitive coating.
9. A particle according to claims 8, wherein said recognition molecule is chosen from the group consisting of antibodies, fragments of antibodies and lectins.
10. A particle according to any one of claims 1-9, wherein said membrane disruptive agent is chosen from the group consisting of detergents, cholesterol, hydrophobic proteins and surfactants and mixtures thereof.
11. A particle according to any one of claims 1-10, wherein said substances are selected from the group consisting of DNA molecules, RNA molecules, proteins, peptides, lipids, chemical preparations, organic compounds, detergents, synthetic polymers and combinations thereof.
12. Use of a particle according to any one of claims 1-11, for modifying the genetic code and/or metabolism of the host cell.
13. Use of a particle according to any one of claims 1-11, for releasing said substance from endosomal or lysosomal organelles .
14. A method of delivering a substance into or out of a membrane enclosed cell or cell organelle through heat induced release of said substance in combination with heat induced release of a membrane disruptive agent comprising: - providing a sample comprising membrane enclosed cells or cell organelles and a plurality of particles according to any one of claims 1-11;
- applying a magnetic alternating field to said sample, whereby thermal energy of the particle core causes the heat sensitive coating of said particle to decompose, which cause the disruptive agent to disrupt the membrane of the enclosed cell or cell organelle and release said substance .
15. A method according to claim 14, wherein said magnetic field has an alternating field direction of a frequency in the range of 0.1 to 5 MHz.
16. A method according to claim 14 or 15, wherein said magnetic field has a field strength in the range of 1 to 100 iriT.
17. A method according to claim 16, wherein said magnetic field is exposed repeatedly as a series of pulses over time each pulse duration preferably between 1 and 600 s, and even more preferably between 1 and 45 seconds .
18. A method according to any one of claims 14-17, wherein said magnetic field has an alternating field which is non-homogeneous and has an alternating gradient field direction, the direction of said alternating magnetic field being generated by two coils, and said sample is inserted between the coils .
19. A method according to claim 18, wherein said coils are supplied with either the positive or negative part of the supplied alternating current.
20. A method according to any one of claims 14-19, wherein said substances are selected from the group consisting of DNA molecules, RNA molecules, proteins, peptides, lipids, chemical preparations, organic compounds, detergents, synthetic polymers and combinations thereof.
21. A method according to any of claims 14-20, wherein said membrane disruptive agent is chosen from the group consisting of detergents, cholesterol, hydrophobic proteins and surfactants and mixtures thereof.
PCT/SE2006/000960 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle WO2007021236A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2008526907A JP4965570B2 (en) 2005-08-19 2006-08-21 Nanoparticles suitable for delivering biomolecules into and out of cells or organelles surrounded by membranes
US11/990,522 US8557289B2 (en) 2005-08-19 2006-08-21 Nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
EP06784101A EP1940513A4 (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
CA002619118A CA2619118A1 (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
KR1020087006690A KR101331891B1 (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
CN200680030233XA CN101242870B (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
AU2006280519A AU2006280519B2 (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
IL189559A IL189559A (en) 2005-08-19 2008-02-17 Nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
HK08113799.8A HK1120452A1 (en) 2005-08-19 2008-12-19 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US70945405P 2005-08-19 2005-08-19
US60/709,454 2005-08-19

Publications (1)

Publication Number Publication Date
WO2007021236A1 true WO2007021236A1 (en) 2007-02-22

Family

ID=37757824

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/SE2006/000960 WO2007021236A1 (en) 2005-08-19 2006-08-21 A nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle

Country Status (10)

Country Link
US (1) US8557289B2 (en)
EP (1) EP1940513A4 (en)
JP (1) JP4965570B2 (en)
KR (1) KR101331891B1 (en)
CN (1) CN101242870B (en)
AU (1) AU2006280519B2 (en)
CA (1) CA2619118A1 (en)
HK (1) HK1120452A1 (en)
IL (1) IL189559A (en)
WO (1) WO2007021236A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073851A2 (en) * 2006-12-08 2008-06-19 Massachusetts Institute Of Technology Remotely triggered release from heatable surfaces
WO2009135937A2 (en) * 2008-05-09 2009-11-12 General Electric Company Novel pei-peg graft copolymer coating of iron oxide nanoparticles for inflammation imaging
US20110008304A1 (en) * 2007-07-09 2011-01-13 Kansas State University Research Foundation Use of cells to facilitate targeted delivery of nanoparticle therapies
WO2011147926A3 (en) * 2010-05-26 2012-05-31 Erik Reimhult Magnetically responsive membrane structures
WO2014124322A1 (en) * 2013-02-08 2014-08-14 University Of Louisville Research Foundation, Inc. Nanoparticles for drug delivery
EP3162361A1 (en) 2015-11-02 2017-05-03 Universität für Bodenkultur Wien Improved magnetically reactive vesicular bodies

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101974152B (en) * 2010-09-28 2012-07-11 山东大学 Lipoid-cation polymer and preparation method thereof
US20210030467A1 (en) * 2018-02-12 2021-02-04 Flagship Pioneering Innovations V, Inc. Devices and methods for delivering material into a biological tissue or cell
WO2020160499A1 (en) * 2019-02-01 2020-08-06 University Of Washington Noncovalent tags and related methods for cytosolic delivery of macrobiomolecules

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001018168A1 (en) 1999-09-08 2001-03-15 Genovis Ab Device for introducing pores into biological materials
WO2004024910A1 (en) * 2002-09-12 2004-03-25 Genovis Ab Particle for magnetically induced membrane transport
WO2005013897A2 (en) * 2003-07-10 2005-02-17 Triton Biosystems, Inc. Magnetic nanoparticle compositions, and methods related thereto
WO2005065282A2 (en) * 2003-12-31 2005-07-21 The Regents Of The University Of California Remote magnetically induced treatment of cancer

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0772776B1 (en) * 1994-07-27 2000-03-22 Herbert Dr. Pilgrimm Superparamagnetic particles, process for their production and their use
US20030211045A1 (en) * 2001-02-05 2003-11-13 Danuta Leszcyznska Magnetoliposome composition for targeted treatment of biological tissue and associated methods
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
SE522099C2 (en) 2002-06-07 2004-01-13 Genovis Ab Device for multiple simultaneous gene transfer
CN1537646A (en) * 2003-10-22 2004-10-20 高春平 Tumour local comprehensive therepy method and device
CA2553647A1 (en) * 2004-01-20 2005-08-04 Alnis Biosciences, Inc. Articles comprising magnetic material and bioactive agents

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001018168A1 (en) 1999-09-08 2001-03-15 Genovis Ab Device for introducing pores into biological materials
WO2004024910A1 (en) * 2002-09-12 2004-03-25 Genovis Ab Particle for magnetically induced membrane transport
WO2005013897A2 (en) * 2003-07-10 2005-02-17 Triton Biosystems, Inc. Magnetic nanoparticle compositions, and methods related thereto
WO2005065282A2 (en) * 2003-12-31 2005-07-21 The Regents Of The University Of California Remote magnetically induced treatment of cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GUPTA A.K. ET AL.: "Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications", BIOMATERIALS, vol. 26, no. 18, 2005, pages 3995 - 4021, XP004697279 *
See also references of EP1940513A4

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073851A2 (en) * 2006-12-08 2008-06-19 Massachusetts Institute Of Technology Remotely triggered release from heatable surfaces
WO2008073851A3 (en) * 2006-12-08 2009-05-22 Massachusetts Inst Technology Remotely triggered release from heatable surfaces
US20110008304A1 (en) * 2007-07-09 2011-01-13 Kansas State University Research Foundation Use of cells to facilitate targeted delivery of nanoparticle therapies
WO2009135937A2 (en) * 2008-05-09 2009-11-12 General Electric Company Novel pei-peg graft copolymer coating of iron oxide nanoparticles for inflammation imaging
WO2009135937A3 (en) * 2008-05-09 2010-12-23 General Electric Company Novel pei-peg graft copolymer coating of iron oxide nanoparticles for inflammation imaging
WO2011147926A3 (en) * 2010-05-26 2012-05-31 Erik Reimhult Magnetically responsive membrane structures
WO2014124322A1 (en) * 2013-02-08 2014-08-14 University Of Louisville Research Foundation, Inc. Nanoparticles for drug delivery
EP3162361A1 (en) 2015-11-02 2017-05-03 Universität für Bodenkultur Wien Improved magnetically reactive vesicular bodies

Also Published As

Publication number Publication date
IL189559A0 (en) 2008-08-07
AU2006280519A1 (en) 2007-02-22
EP1940513A4 (en) 2013-03-13
JP4965570B2 (en) 2012-07-04
CA2619118A1 (en) 2007-02-22
IL189559A (en) 2011-07-31
HK1120452A1 (en) 2009-04-03
KR20080053305A (en) 2008-06-12
JP2009504181A (en) 2009-02-05
US8557289B2 (en) 2013-10-15
KR101331891B1 (en) 2013-11-21
US20090304796A1 (en) 2009-12-10
CN101242870B (en) 2013-01-09
EP1940513A1 (en) 2008-07-09
AU2006280519B2 (en) 2011-06-16
CN101242870A (en) 2008-08-13

Similar Documents

Publication Publication Date Title
US8557289B2 (en) Nanoparticle suitable for delivery of a biomolecule into or out of a membrane enclosed cell or cell organelle
Rayamajhi et al. Surface functionalization strategies of extracellular vesicles
Kurosaki et al. Ternary complexes of pDNA, polyethylenimine, and γ-polyglutamic acid for gene delivery systems
Arsianti et al. Assembly of polyethylenimine-based magnetic iron oxide vectors: insights into gene delivery
Chou et al. Strategies for the intracellular delivery of nanoparticles
Wang et al. A magnetic nanoparticle-based multiple-gene delivery system for transfection of porcine kidney cells
US7935517B2 (en) Nanospearing for molecular transportation into cells
Yuan et al. Ternary nanoparticles of anionic lipid nanoparticles/protamine/DNA for gene delivery
Park et al. Multimodal magnetic nanoclusters for gene delivery, directed migration, and tracking of stem cells
KR101777837B1 (en) Effecive Method for Transfering Genes into Cells Using Photothermal Effects of Gold Nanoparticles
JP2010538607A (en) Magnetic delivery device
AU2018305148B2 (en) Biodegradable multilayer nanocapsules for the delivery of biologically active agents in target cells
Vankayala et al. A general strategy to achieve ultra-high gene transfection efficiency using lipid-nanoparticle composites
Xie et al. Low aggregation magnetic polyethyleneimine complexes with different saturation magnetization for efficient gene transfection in vitro and in vivo
Olszewska‐Widdrat et al. Bacteriophage‐templated assembly of magnetic nanoparticles and their actuation potential
WO2006072593A2 (en) Device for magnet assisted transfer of chemical compounds into cells and method for magnet assisted transfer of proteins into cells
EP1546321A1 (en) Particle for magnetically induced membrane transport
WO2022246058A1 (en) Partial endosomal magnetolysis for cytosolic delivery of biologics
Karki et al. Ultrasound-Mediated Targeted Drug Delivery in T-Cells
WO2024073796A1 (en) Cell permeant particles
Ang Magnetic nanoparticle assisted gene delivery
Vernon Enhancing magnetic nanoparticle-based DNA transfection: Intracellular-active cassette features
Prow et al. Gene Delivery, Expression, and Recovery of DNA Tethered Nanoparticles
Çakmak Immunofunctionalization of magnetic poly (glycidyl methacrylate) microspheres and investigation of their effectiveness in animal cell purification
Santhakumaran et al. and TJ Thomas

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680030233.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2619118

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006280519

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 189559

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2008526907

Country of ref document: JP

Ref document number: 816/CHENP/2008

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006280519

Country of ref document: AU

Date of ref document: 20060821

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2006784101

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020087006690

Country of ref document: KR

Ref document number: 2006784101

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11990522

Country of ref document: US