US20130072854A1 - Microbubble complexes and methods of use - Google Patents

Microbubble complexes and methods of use Download PDF

Info

Publication number
US20130072854A1
US20130072854A1 US13/528,399 US201213528399A US2013072854A1 US 20130072854 A1 US20130072854 A1 US 20130072854A1 US 201213528399 A US201213528399 A US 201213528399A US 2013072854 A1 US2013072854 A1 US 2013072854A1
Authority
US
United States
Prior art keywords
complex
microbubble
ligand
sirna
albumin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/528,399
Inventor
Praveena Mohan
Anup Sood
James Edward Rothman
John Donald Burczak
Hae Won Lim
Lisa Anne Lowery
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
General Electric Co
Original Assignee
General Electric Co
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 General Electric Co filed Critical General Electric Co
Priority to US13/528,399 priority Critical patent/US20130072854A1/en
Assigned to GENERAL ELECTRIC COMPANY reassignment GENERAL ELECTRIC COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROTHMAN, JAMES EDWARD, LIM, HAE WON, BURCZAK, JOHN DONALD, LOWERY, LISA ANNE, MOHAN, Praveena, SOOD, ANUP
Priority to MX2014003358A priority patent/MX347719B/en
Priority to CA2849037A priority patent/CA2849037A1/en
Priority to BR112014006412A priority patent/BR112014006412A2/en
Priority to ES12759459.6T priority patent/ES2621821T3/en
Priority to KR1020147006830A priority patent/KR20140063697A/en
Priority to JP2014530263A priority patent/JP6059725B2/en
Priority to AU2012311659A priority patent/AU2012311659B2/en
Priority to EP12759459.6A priority patent/EP2758081B1/en
Priority to RU2014108245A priority patent/RU2613321C2/en
Priority to CN201280045451.6A priority patent/CN103957940B/en
Priority to PCT/EP2012/068288 priority patent/WO2013041500A1/en
Priority to CN201710959069.0A priority patent/CN107737335B/en
Publication of US20130072854A1 publication Critical patent/US20130072854A1/en
Priority to HK15101059.9A priority patent/HK1200362A1/en
Priority to US15/219,419 priority patent/US10004760B2/en
Priority to US16/016,963 priority patent/US11007212B2/en
Priority to HK18110916.0A priority patent/HK1251462A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • 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
    • A61K47/554Medicinal 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 a steroid plant sterol, glycyrrhetic acid, enoxolone or bile 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
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • 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
    • A61K47/6925Medicinal 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 microcapsule, nanocapsule, microbubble or nanobubble
    • 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
    • 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
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M37/0092Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin using ultrasonic, sonic or infrasonic vibrations, e.g. phonophoresis
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • 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
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M37/00Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin
    • A61M2037/0007Other apparatus for introducing media into the body; Percutany, i.e. introducing medicines into the body by diffusion through the skin having means for enhancing the permeation of substances through the epidermis, e.g. using suction or depression, electric or magnetic fields, sound waves or chemical agents
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Definitions

  • the invention relates generally to novel binding of therapeutic agents to albumin microbubble pharmaceuticals using an attachment of albumin affinity ligands to the agents.
  • the binding provides a method of microbubble-assisted delivery of therapeutic agents to targeted cells or tissue of interest, either in vitro or in vivo.
  • Ultrasound-mediated destruction of microbubbles carrying drugs has been found to be useful as a noninvasive drug delivery system.
  • Drugs or other therapeutic agents can be incorporated into the microbubbles in a number of different ways, including binding of the drug to the microbubble shell and attachment of ligands.
  • perfluorocarbon-filled microbubbles are sufficiently stable for circulating in the vasculature as blood pool agents; they act as carriers of these agents until the site of interest is reached.
  • Ultrasound applied over the skin surface can then be used to burst the microbubbles at this site, causing localized release of the drug or therapeutic agents on site specific locations.
  • albumin microbubbles have been used and delivered to a specific organ target by site-directed acoustic ultrasound.
  • Albumin is a major protein in blood, and its natural physiological role is to bind and carry a wide variety of lipophilic/poorly soluble ligands throughout the body. These ligands, which may have an affinity to albumin, include fatty acids and other biosynthetic and catabolic products that are hydrophobic in nature.
  • albumin microbubbles have been used to carry a variety of therapeutic agents based on proteins and other biologics including, oligonucleotides (ODN) and polynucleotides such as antisense ODN, with sequences complementary to a specific targeted messenger RNA (mRNA) sequence.
  • ODN oligonucleotides
  • mRNA messenger RNA
  • microbubble-nucleic acid complexes may be generated from unmodified ODN that are mixed with albumin or lipid components during microbubble shell formation or alternatively, the complex formation can be performed by mixing preformed microbubbles with an ODN of interest.
  • the ODN acts as a mechanistic intervention in the processes of gene translation or an earlier processing event.
  • the advantage of this approach is the potential for gene-specific actions which should be reflected in a relatively low dose and minimal non-targeted side effects.
  • Non-targeted delivery of these drugs can cause systemic toxicity and has prevented the use of many of these drugs all together or at higher doses required for good efficacy.
  • Attempts to deliver these as pro-drugs in many cases have reduced this problem, however, selective uptake in the targeted tissue is not always easy to achieve as most of the uptake mechanisms in the diseased tissue are also present in the normal tissue. Enhancing the uptake of these drugs in selective tissues by non-natural mechanisms as disclosed herein, therefore can add considerable value.
  • compositions and methods for increased binding of therapeutic drugs to microbubbles using the affinity of ligand-therapeutic compositions towards the albumin shell are provided herein.
  • Systemic circulation of the microbubbles carrying the therapeutic composition can be easily visualized through ultrasound imaging.
  • Therapeutic agent is released from the microbubbles using a trigger of high energy pulsed ultrasound specific to the site of treatment.
  • the cavitation of microbubbles causes sonoporation of the neighboring cells/tissue.
  • a microbubble complex comprising a microbubble having an outer shell comprising a mixture of native and denatured albumin and a hollow core encapsulating a perfluorocarbon gas, a therapeutic agent selected from the group comprising a small molecule chemotherapeutic agent, a peptide, a carbohydrate, or an oligonucleotide, and a bifunctional linker having one end attached to the therapeutic agent and the other attached to the ligand through reaction of a reactive group on the ligand.
  • the ligand is bound to the outer shell of the microbubble through hydrophobic interactions.
  • a method for delivering the aforementioned microbubble complex to a tissue target is disclosed.
  • the method comprising the steps of providing the microbubble complex, administrating the microbubble complex to a subject wherein the subject is the source of the tissue target, and administering ultrasonic energy to the subject, wherein the ultrasound energy is sufficient to cause cavitation of the microbubble complex in the tissue target.
  • FIG. 1 is a representation of non-covalent binding of siRNA-ligand to albumin microbubbles.
  • FIG. 2 is representative micrograph of a gel shift assay for a mixture of cholesterol conjugated siRNA (2 pmoles) and varying amounts of Optison (0, 9, 22 and 46 pmoles for i, ii, iii and iv respectively).
  • FIG. 3 is a. representative micrograph of a gel shift assay for Cy3-siRNA (4 pmoles) mixed with varying concentrations of either Optison or native HSA which shows no shift in gel assay.
  • FIG. 4 is a. representative micrograph of a gel assay for Cy3-cholesterol-siRNA (2 pmoles) mixed with varying concentrations of either Optison, native HSA or denatured HSA.
  • FIG. 5 is a graphical representation of binding properties of cholesterol-siRNA to Optison and native HSA;
  • A Relative fluorescence of cholesterol-siRNA bands is measured from the gel shift assay
  • B fraction bound of siRNA calculated from relative fluorescence and plotted against the albumin concentration.
  • FIG. 6 is a graphical representation of the uptake of siRNA by U-87 tumor cells in opticell is quantified by measuring cell cy3-fluorescence.
  • FIG. 7 are micrographs of fluorescence images showing a comparison of siRNA transfection between a lipid transfection reagent (RNAifect) and Optison.
  • FIG. 8 is a graphical representation of the mean cell fluorescence values and standard error of siRNA transfection between a lipid transfection reagent (RNAifect) and Optison.
  • FIG. 9 is a graphical representation of the fraction bound of fluorescein-myristate to Optison and native HSA as calculated from anisotropy values.
  • FIGS. 10 A and B are the fluorescein bound to Optison (0, 8, 40 and 200 pmoles for i, ii, iii and iv respectively) and visualized on the gel as dark bands for fluorescein-myristate (63 pmoles) and fluorescein-stearate (180 pmoles) respectively.
  • the invention relates generally to microbubble-assisted delivery of a therapeutic agent to cells or tissue of interest, either in vitro or in vivo.
  • the therapeutic agent may be comprised of a small molecule chemotherapeutic agent, a peptide, a carbohydrate, or an oligonucleotide, and a bifunctional linker having one end attached to the therapeutic agent and the other attached to the ligand through reaction of a reactive group on the ligand.
  • the therapeutic agent may be an oligonucleotide (ODN).
  • Oligonucleotides refers to nucleic acid polymers that are formed by bond cleavage of longer nucleic acids or are synthesized using building blocks, protected phosphoramidites of natural or chemically modified nucleosides or, to a lesser extent, of non-nucleosidic compounds.
  • the length of the oligonucleotide may vary from a short nucleic acid polymer of fifty or fewer base pairs to more than 200 base pairs.
  • ODN also refers to polynucleotides having more than 200 base pairs.
  • antisense ODN refer to single strands of DNA or RNA that are complementary to a chosen sequence. In the case of antisense RNA, antisense RNA prevents protein translation of certain messenger RNA strands by binding to them. Antisense DNA can be used to target a specific, complementary (coding or non-coding) RNA.
  • small interfering RNA is a class of double-stranded RNA molecules, typically 20-25 nucleotides in length, that play a variety of roles in biology including the RNA interference (RNAi) pathway, where it interferes with the expression of a specific gene, as an antiviral mechanism, or in shaping the chromatin structure of a genome.
  • RNAi RNA interference
  • the therapeutic agent may be a cytotoxin.
  • cytotoxin refers to a substance that has a toxic effect on cells.
  • a cytotoxin may cause undergo necrosis, in which they lose membrane integrity and die as a result of cell lysis.
  • a cytotoxin may be associated with antibody-dependent cell mediated cytotoxicity wherein a cell is marked by an antibody and acted upon by certain lymphocytes.
  • cytotoxic agents examples include taxol; nitrogen mustards, such as mechlorethamine, cyclophosphamide, melphalan, uracil mustard and chlorambucil; ethylenimine derivatives, such as thiotepa; alkyl sulfonates, such as busulfan; nitrosoureas, such as carmustine, lomustine, semustine and streptozocin; triazenes, such as dacarbazine; folic acid analogs, such as methotrexate; pyrimidine analogs, such as fluorouracil, cytarabine and azaribine; purine analogs, such as mercaptopurine and thioguanine; vinca alkaloids, such as vinblastine and vincristine; antibiotics, such as dactinomycin
  • Drugs that interfere with intracellular protein synthesis, protein synthesis inhibitors, can also be coupled to the ligand; such drugs are known to these skilled in the art and include puromycin, cycloheximide, and ribonuclease.
  • the protein includes, but is not limited to, enzymes, soluble and serum proteins, proteins expressed on a surface of a cell, non-immunoglobulin proteins, intracellular proteins, and segment of proteins that are or can be made water-soluble, either individually or in combinations thereof as well as any derivatives of the proteins.
  • the protein includes such as, but not limited to, cysteine proteases, glutathione S transferase, epoxide hydrolase (EH), thiolase, NAD/NADP-dependent oxidoreductase, enoyl coA hydratase, aldehyde dehydrogenase, hydroxypyruvate reductase, tissue transglutaminase (tTG), formiminotransferase cyclodeaminase (FTCD), aminolevulinate-dehydratase (ADD), creatin kinase, carboxylesterase (LCE), monoacylglycerol (MAG) lipase, metalloproteases (MP), phosphotases (protein tyrosine phosphotases, PTP), proteosome, FK506 binding protein (FKBP12), mammalian target of Rapamycin (mTOR; alternatively known as FKBP-rapamycin binding domain (
  • the protein is substantially free of a cofactor.
  • “Substantially free of a cofactor” includes proteins that do not require any additional cofactor, chemical, chemical modification, or physical modification to be naturally stable under physiological conditions and room temperature and pressure in solution or as a solid, and can bind its corresponding ligand in vivo.
  • an albumin microbubble may be utilized to carry a therapeutic agent in systemic delivery. Tissue targeted ultrasound acoustic energy may then be used to cavitate the albumin microbubble and deliver the therapeutic agent into the intracellular environment.
  • the microbubble complex may be administered intravenously or into the peritoneum (intraperitoneally) of a subject whose cells or tissues are to be targeted. Once the microbubble complex is carried through the subject to the targeted cell, the ultrasound acoustic energy is delivered.
  • visualization of the targeted cells may occur prior to delivering the ultrasound while in still other embodiments visualization may be performed in real time and the cavitation monitored.
  • the albumin outer shell of the microbubble is comprised of both native and denatured albumin held together by mostly cysteine to cysteine bonds.
  • the primary composition of the albumin shell is mostly in the native form wherein the denatured portion allows for increased cysteine bond attachments.
  • the relative amount of denatured albumin to native albumin ranges from approximately 0.5 to 30 wt %. In other embodiments the relative amount is in the range of approximately 1% to 15 wt %.
  • the mixture of native and denatured albumin provides a balance of shell elasticity needed for cavitation, with increased reactive binding sites on the microbubble surface.
  • the microbubbles are formed by sonication of perfluorocarbon gas in the presence of pre-heated albumin solution.
  • a small part of the albumin molecules rearrange during sonication of pre-heated albumin solution and crosslinking occurs through disulfide linkages between albumin molecules.
  • These albumin molecules are believed to be similar in structure to an F form of albumin which has more hydrophobic residues exposed. This allows increased binding sites for hydrophobic interactions.
  • the microbubble may be filled with an insoluble perfluorocarbon gas, such as but not limited to, perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluoropentane, or a combination thereof.
  • an insoluble perfluorocarbon gas such as but not limited to, perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluoropentane, or a combination thereof.
  • the microbubbles are about 1 to about 5 microns in diameter, the size being optimized to allow circulation through the blood stream.
  • the therapeutic-microbubble complexes comprise a therapeutic agent modified with a linker having a reactive group capable of binding with a ligand having affinity towards albumin.
  • the therapeutic agent may be coupled to albumin though the ligand.
  • the linker includes any linking moiety that attaches the ligand to the therapeutic agent through a first moiety.
  • the linker can be as short as one carbon or a long polymeric species such as polyethylene glycol, tetraethylene glycol (TEG), polylysine or other polymeric species normally used in the pharmaceutical industry for modulating pharmacokinetic and biodistribution characteristics of therapeutic agents.
  • Other linkers of varying length include C1-C250 length with one or more heteroatoms selected from O, S, N, P, and optionally substituted with halogen atoms.
  • the linker comprises at least one of an oligomeric or polymeric species made of natural or synthetic monomers, oligomeric or polymeric moiety selected from a pharmacologically acceptable oligomer or polymer composition, an oligo- or poly-amino acid, peptide, saccharide, a nucleotide, and an organic moiety with 1-250 carbon atoms, either individually or in combination thereof.
  • the organic moiety with 1-250 carbon may contain one or more heteroatoms such as O, S, N or P and optionally substituted with halogen atoms at one or more places.
  • the first moiety may simply be an extension of the linker, formed by the reaction of a reactive species on the linker with a reactive group on the therapeutic agent.
  • reactive species and the reactive group include, but are not limited to, activated esters (such as N-hydroxysuccinimide ester, pentafluorophenyl ester), a phosphoramidite, an isocyanate, an isothiocyanate, an aldehyde, an acid chloride, a sulfonyl chloride, a maleimide an alkyl halide, an amine, a phosphine, a phosphate, an alcohol or a thiol with the proviso that the reactive species and reactive group are matched to undergo a reaction yielding covalently linked conjugates.
  • the reactive moiety may be a primary amine functionality, and as such, the amine modified therapeutic agent may be conjugated to the affinity ligand through reaction of a carboxyl moiety of the ligand.
  • the reactive moiety may be an alcohol attached to the ligand through a phosphate group.
  • the affinity ligand includes fatty acids, steroids, small aromatic compounds or a combination thereof.
  • albumin binding molecules may be found in US patent application publication number 2010/0172844, published Jul. 8, 2010.
  • the affinity ligand is a fatty acid, including but not limited to myristoyl, lithocolic-oleyl, docosanyl, lauroyl, steoroyl, palmitoyl, oleoyl, or linoleoyl.
  • the lipophilic molecule is a steroid or modified steroid including, cholesterol, cholic acid, lithocholic acid, or chenodeoxycholic acid.
  • the high affinity molecule is selected from 4-p-iodophenyl-butyric acid and analogs or derivatives thereof.
  • the therapeutic agent comprises siRNA
  • the linker comprises tetraethylene glycol
  • the ligand comprises cholesterol.
  • the therapeutic-albumin complexes may be prepared either by sonicating ligand-modified therapeutic agent with albumin or lipid in the presence of perfluorocarbon or by mixing preformed bubbles with ligand modified therapeutic agents.
  • these molecules may be attached to therapeutic agents of interest during therapeutic agent synthesis.
  • the phosphoramidites of cholesterol may be used to incorporate cholesterol during DNA or RNA synthesis on a nucleic acid synthesizer, or post synthesis by incorporating a reacting moiety.
  • the therapeutic agent is a modified ODN which may be prepared enzymatically by using modified nucleoside triphosphates; modified either with the ligand itself or with a reactive functionality for post synthesis modification with the ligand.
  • Ligand attachment may be at one or both termini, internal to the nucleic acid sequence or at multiple positions depending upon the ODN use. In certain embodiments, where siRNA is the ODN, attachment may be through the 3′ OH position.
  • the therapeutic agent in addition to the ligand, may be selectively modified to protect from nucleases.
  • stabilizing modification may include phosphorothioate modification, or 2′-OMe modification.
  • the microbubble complexes may be incubated with cells or the tissue of interest or injected into the body, preferably intravenously, and then cavitated with ultrasound energy at desired site and at a predetermined time or during live imaging.
  • the microbubble complex may be viewed during systemic travel in the blood circulation under normal ultrasound diagnostic imaging.
  • a series of pulsed acoustic energy waves are sent to the tumor. This creates inertial cavitation on the microbubble, which collapses the microbubble. Cavitation of the microbubble occurs where the acoustic energy is maximally located. This direction is achieved on the ultrasound probe by parameters related to mechanical index force, optimal ultrasound acoustic distance, and dimensions of the ultrasound acoustic sweep. The force generated can then potentially form micropores within the cellular plasma membrane.
  • the pulsed energy is administered at a frequency of about 0.5 to about 5 MHz.
  • micropores may facilitate the entrance of ODN into the cellular cytoplasmic environment.
  • ODN is siRNA
  • siRNA in the intracellular environment will utilize the host machinery to silence mRNA and later protein synthesis.
  • mRNA message acts as an angiogenesis promoting proteins including vascular endothelial growth factor (VEGF)
  • VEGF vascular endothelial growth factor
  • the reduction of VEGF expression in a tumor may halt or slow tumor growth.
  • VEGF vascular endothelial growth factor
  • a bolus of the microbubble complex may be mixed to an optimal ratio from previous therapeutic investigations. Once the mixture of the complex is established, the bolus drug is injected systemically by venous route.
  • the bolus may be monitored in the first pass blood kinetics.
  • the microbubble resonance and thus enhanced ultrasound contrast may be monitored with an ultrasound probe using low diagnostic levels of acoustic energy.
  • the bolus arrives on organ target.
  • Cardiovascular tissue perfusion may assist in delivering the bolus into deep microvessels with small lumen diameters.
  • sufficient energy may be provided for the microbubble to undergo inertial cavitation.
  • the siRNA contents may be delivered across the plasma membrane and into the diseased cell. While in the cytoplasmic intracellular environment the siRNA can have a therapeutic effect.
  • siRNA entrance into the cell may occur by various mechanisms.
  • the siRNA may enter the cell by a: a microjetting force from the collapsing microbubble which can push siRNA into the cytoplasm.
  • the mechanism may include microjetting energy or sonoluminescence energy which creates temporary micropores within the plasma membrane to allow for passive diffusion of the siRNA into the cell, or during microbubble resonance before actual cavitation the microbubble bumping into the plasma membrane may push the siRNA in the cell.
  • microbubble delivery has potential applications in the treatment of a wide variety of diseases, which can include cancer, inflammatory, infectious, cardiovascular, metabolic, autoimmune, and central nervous system diseases. Many of these diseases cannot currently be effectively treated by virtue of targeting molecular mechanisms not accessible to conventional small molecule drugs and monoclonal antibodies.
  • FIG. 1 is a representation illustration of binding of siRNA to albumin encapsulated microbubbles to form a microbubble-siRNA complex.
  • the target siRNA for VEGF silencing (vascular endothelial growth factor) were synthesized by IDTDNA technologies.
  • IDTDNA provided lipid modifications such as cholesteryl TEG on the siRNA (Chol-siRNA) as well as dye conjugation.
  • Sense strand 5′-Cy3/GCAUUUGUUUGUCCAAGAUmUmU/3′-Lipid Antisense strand; 5′/mAmArA rUrCrU rUrGrG rArCrA rArArC rArArA rUrGrC/3′
  • Cyanine dye, Cy3 on the siRNA has an excitation wavelength of 550 nm and a peak emission of 580 nm.
  • the siRNA has been labeled with a cy3 dye for easy visualization of siRNA during binding assays and other characterization techniques.
  • OptisonTM GE Healthcare, Chalfont St. Giles, United Kingdom, 10 mg/ml albumin
  • Lyophilized human serum albumin (HSA) powder was dissolved in 1 ⁇ phosphate buffered saline (PBS) to make a stock solution of 10 mg/mL.
  • PBS phosphate buffered saline
  • Both Optison and native albumin solution dilutions were prepared with 1 ⁇ PBS.
  • Denatured HSA solution was prepared by heating native HSA solution to 80° C. for 20 minutes.
  • the stock solutions of cy3-siRNA and cy3-siRNA-cholesterol, 20 ⁇ M were prepared in RNAse free water and stored at ⁇ 20° C. 4 pmoles of cy3-siRNA and 2 pmoles of cholesterol-siRNA solutions were mixed with varying amounts of Optison solution, native HSA and denatured HSA solution, ranging from 0 to 50 pmoles.
  • the reaction buffer was 1 ⁇ PBS, pH 7.4. The reaction mixture was incubated under dark at 25° C. for 45 minutes. After incubation, ten ⁇ L of siRNA mixtures was mixed with 2 ⁇ L of Novex® Hi-Density TBE Sample Buffer (5 ⁇ ) (Invitrogen, Carlsbad, Calif., USA).
  • the fluorescence of Cy3 attached to siRNA can be visualized as distinct siRNA bands on the gel.
  • siRNA and albumin solution When a mixture of siRNA and albumin solution was run on the gel, the mobility of albumin bound-siRNA is slower than free-siRNA resulting in two bands on the gel.
  • sypro ruby stain from EMSA kit (Molecular Probes, Eugene, Oreg., USA) was used to observe the albumin bands in the gel.
  • FIG. 2 is a gel shift assay for a mixture of cholesterol conjugated siRNA (2 pmoles) and varying amounts of Optison (0, 9, 22 and 46 pmoles for i, ii, iii and iv respectively). Fluorescence imaging of the gel shows distinct bands for siRNA (lower band sections) and albumin (upper band sections).
  • FIG. 3 is a fluorescence image of a gel for Cy3-siRNA (4 pmoles) mixed with varying concentrations of either Optison or native HSA shows no shift in gel assay.
  • the dark bands on the gel are the cy3-fluorescence on siRNA.
  • FIG. 4 shows the gel images for binding of chol-siRNA with Optison, native HSA and denatured HSA. Chol-siRNA bound to both native HSA and Optison solution, while the binding significantly decreased for the same amount of denatured HSA.
  • the fluorescence intensity of siRNA in each lane was estimated manually by drawing a box around the bands. Background, equivalent to the average intensity value of the gel, was subtracted from the intensity value of each siRNA band. Fluorescence intensities of bound siRNA over a wide range of albumin concentrations were calculated. Relative fluorescence, R was calculated as:
  • Fbound is fluorescence intensity of bound-siRNA band and Ffree is fluorescence intensity of free-siRNA band. Relative fluorescence was plotted against albumin concentration. This is shown in FIG. 5 , which is a graphical comparison of binding properties of cholesterol-siRNA to Optison and native HSA as described below
  • FIG. 5 graph A shows that at this concentration range, the amount of chol-siRNA bound to Optison solution was higher than the binding to native HSA. To estimate binding constants, higher concentration of albumin was used to allow saturation of amount of siRNA bound to albumin. Fraction bound, x is determined as:
  • Fsat is the fluorescence intensity of maximum bound-siRNA under saturation conditions.
  • Equation 3 was solved using a non-linear fit to determine kd and n for binding of chol-siRNA to both Optison and native HSA (Table 1).
  • Microsoft Excel's solver tool was utilized for this non-linear fit, and the sum of squared errors (SSE) was found to be 0.07 and 0.06 for Optison and native HSA respectively.
  • SSE squared errors
  • MATBIII rat mammary carcinoma and U-87 human glioblastoma cells were cultured in McCoy's 5A Medium (modified) (1 ⁇ ) (Invitrogen, Carlsbad, Calif., USA) and Eagle's Minimum Essential Medium (EMEM) (ATCC, Manassas, Va.) respectively. Both the media solutions were supplemented with 10% heat deactivated fetal bovine serum (FBS) (Fisher Scientific, Springfield, N.J.) and 1% penicillin-streptomycin (Sigma Aldrich, St Louis, Mo.). The cells were maintained at 37° C. in a humidified atmosphere with 5% CO2.
  • FBS heat deactivated fetal bovine serum
  • penicillin-streptomycin Sigma Aldrich, St Louis, Mo.
  • MATB-III and U-87 cells were grown in 10 mL capacity Opticell units (Nalge Nunc International, Rochester, N.Y.) to 90% confluence.
  • the media in OptiCell was replaced with 10 mL fresh media containing 40 pmoles of either cy3-siRNA or cholesterol-siRNA.
  • the opticell was left in the incubator for 24 hours at 37° C. Separately, the cells were either treated with siRNA solution mixed with Optison microbubbles (300 ⁇ L) or a lipid transfection reagent (90 ⁇ L) (RNAifect, Qiagen, Valencia, Calif.).
  • Vivid i imagers with a cardiac sector probe (3S) was used to rupture the microbubbles and deliver the siRNA drug from microbubbles.
  • the opticell was immobilized in a water bath, and the ultrasound probe was attached to a motion arm that spanned the entire length of the opticell.
  • the tip of the probe was immersed in water, and the distance between the probe and opticell surface was 3 cm that allowed sonication of the entire width of the opticell.
  • the microbubbles in opticell were treated with a mechanical index, MI>1.3 continuous sonication.
  • the probe moved at a speed of 1 m/s over the entire length of the opticell. After sonication, the cells were incubated for 24 hours at 37° C.
  • the cells treated with RNAifect were also kept in the incubator for 24 hours. After incubation, the cells were imaged using a fluorescence microscope (Zeiss Axio Imager.Z1, Carl Zeiss). The filter used for cy3 was DsRed/Cy3 (546 ex/620 em). In the region of interest (ROI) in the fluorescent images, cell fluorescence was measured and the mean values of cell fluorescence were calculated. ImageJ was used to process the images and calculate fluorescence intensities.
  • ROI region of interest
  • FIG. 6 The effect of the delivery system is illustrated in FIG. 6 for U-87 cells incubated with either cy3-siRNA or chol-siRNA.
  • the delivery of siRNA into the tumor cells is represented by average cell cy3-fluorescence.
  • mean fluorescence values and standard errors are reported in.
  • FIGS. 7 and 8 show representative images of the cells after treatment.
  • FIG. 8 reports the mean cell fluorescence for all groups with standard errors represented as error bars.
  • Fatty acid NHS ester (2 equivalents, 5.37 mg Myristic acid NHS ester or 6.38 mg Stearic acid NHS ester was taken in a 50:50 mixture of DMSO and dichloromethane (100 ul) and mixed with a solution of Fluorescein cadaverine (FL-Cadaverine, 5 mg, 1 equivalent, in 50 ul DMSO). To this diisopropylethyl amine (3.8 equivalents) was added and mixture was vortexed to give a clear solution. Samples were kept in the dark at room temperature. After 4.5 h, reaction was checked by HPLC and was found to be complete.
  • Crude product was diluted with DMSO to ⁇ 2 ml and purified on AKTA purifier using Xterra MS C18, 19 ⁇ 100 mm column and a gradient of 0-100% B in 18.75 column volumes at a flow rate of 10 ml/min.
  • Solvent A and B were as described above for the analytical method.
  • Product was collected in multiple fractions and each fraction was analyzed by analytical HPLC. Only the purest fraction in each case ( ⁇ 90% purity) was used for binding studies (starting material itself was ⁇ 86% pure, remaining likely a regioisomer with same spectral properties). This fraction was concentrated to dryness at room temperature. Residue was suspended in water ( ⁇ 2 ml) and extracted with dichloromethane (3 ⁇ 2 ml). Organic extracts were combined, dried over anhydrous sodium sulfate and concentrated to dryness.
  • the stock solution of fluorescein-myristate was prepared in 1 ⁇ PBS.
  • the concentration of fluorescein was kept low for the fluorescence polarization assay, at 126 nM. Varying concentrations of either Optison or HSA solutions, ranging from 0 to 15 ⁇ M albumin concentrations, were added to the fluorescein myristate solution.
  • the reaction buffer was 1 ⁇ PBS, pH 7.4. The reaction mixture was incubated under dark at 25° C. for 15 minutes. After incubation, the changes in raw anisotropy values of fluorescein were measured using a microplate reader (SpectraMax 5, Molecular Devices, Sunnyvale, Calif.).
  • FIG. 10 which shows the fluorescein bound to Optison (0, 8, 40 and 200 pmoles for i, ii, iii and iv respectively) is visualized on the gel as dark bands for fluorescein-myristate ( FIG. 10A ) (63 pmoles) and fluorescein-stearate ( FIG. 10B ) (180 pmoles).
  • conjugating a fatty acid such as myristate to a therapeutic compound can increase the binding of such therapeutic compounds to the albumin shell microbubbles.
  • the dissociation constant of fluorescein-myristate binding to Optison was lower than that of binding to native HSA. This suggests better hydrophobic binding properties of microbubble shell that has both native and partially denatured albumin.
  • mice were obtained from Charles River Laboratories (Wilmington, Mass.). Animals were housed in accordance with the Guide for the Care and Use of Laboratory Animals as adopted by the National Institutes of Health. Lewis lung carcinoma cells (LLC) were inoculated subcutaneously to the right flank of anaesthetized mice (3.5 ⁇ 106 cells/100 ⁇ l/mouse).
  • LLC Lewis lung carcinoma cells
  • mice On the fourth day after inoculation, the mice were treated with anti-VEGF siRNA (Sigma Life Sciences, St. Louis, Mo.)—microbubble mixtures, a siRNA dose of 1.0 mg/kg for subcutaneous injections and a dose of 2.0 mg/kg for tail-vein injections.
  • the injection mixture contained 100 ⁇ L of microbubble solution and 100 ⁇ L of siRNA in RNAse free water.
  • the tumors were sonicated using Vivid i imagers with a cardiac sector probe (3S). The energies were delivered in a pulsatile form with the peak MI at 1.3. Control group did not receive any treatments.
  • mice After 24 hours from the treatment day, the mice were euthanized and the tumors were extracted. The tumors were frozen immediately and stored at ⁇ 20° C. The tumors were thawed out at room temperature on the day of VEGF measurement. The tumors were then lysed in RIPA buffer (protease inhibitors added) using a lysing matrix tube (Lysing matrix tube A, RP Biomedical). The lysate collected from the samples were then diluted, and measured for total protein using a protein kit (Pierce BCA reagent protein assay kit) and for VEGF using an ELISA kit (Mouse VEGF ELISA kit, RayBiotech, Norcross, Ga.).

Abstract

The present invention relates to a microbubble complex comprising a microbubble having an outer shell comprising a mixture of native and denatured albumin encapsulating a perfluorocarbon gas, a therapeutic agent, a bifunctional linker having one end attached to the therapeutic agent and the other attached to a ligand and wherein the ligand is bound to the other shell of the microbubble through hydrophobic interactions. Also included are methods for delivering the aforementioned microbubble complex to a tissue target.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part to U.S. patent application Ser. No. 13/235,890 filed Sep. 19, 2011 the disclosure of which is incorporated herein by reference in its entirety.
  • BACKGROUND
  • The invention relates generally to novel binding of therapeutic agents to albumin microbubble pharmaceuticals using an attachment of albumin affinity ligands to the agents. The binding provides a method of microbubble-assisted delivery of therapeutic agents to targeted cells or tissue of interest, either in vitro or in vivo.
  • Ultrasound-mediated destruction of microbubbles carrying drugs has been found to be useful as a noninvasive drug delivery system. Drugs or other therapeutic agents can be incorporated into the microbubbles in a number of different ways, including binding of the drug to the microbubble shell and attachment of ligands. For example, perfluorocarbon-filled microbubbles are sufficiently stable for circulating in the vasculature as blood pool agents; they act as carriers of these agents until the site of interest is reached. Ultrasound applied over the skin surface can then be used to burst the microbubbles at this site, causing localized release of the drug or therapeutic agents on site specific locations.
  • More specifically, albumin microbubbles have been used and delivered to a specific organ target by site-directed acoustic ultrasound. Albumin is a major protein in blood, and its natural physiological role is to bind and carry a wide variety of lipophilic/poorly soluble ligands throughout the body. These ligands, which may have an affinity to albumin, include fatty acids and other biosynthetic and catabolic products that are hydrophobic in nature. As such albumin microbubbles have been used to carry a variety of therapeutic agents based on proteins and other biologics including, oligonucleotides (ODN) and polynucleotides such as antisense ODN, with sequences complementary to a specific targeted messenger RNA (mRNA) sequence. These microbubble-nucleic acid complexes may be generated from unmodified ODN that are mixed with albumin or lipid components during microbubble shell formation or alternatively, the complex formation can be performed by mixing preformed microbubbles with an ODN of interest. In both cases, the ODN acts as a mechanistic intervention in the processes of gene translation or an earlier processing event. The advantage of this approach is the potential for gene-specific actions which should be reflected in a relatively low dose and minimal non-targeted side effects.
  • However, a key barrier to translating the potent biology of ODN into drugs is known to be at the level of drug delivery with efficacy and safety. For example, ODN delivery with chemical formulation, viral vectors, and particle delivery have been hampered with clinical safety related problems before therapeutic efficacy can be attained. Furthermore, the use of albumin microbubbles as a carrier of ODNs such as siRNA is limited due to the limited binding of the ODNs to the albumin microbubble as well as the stability of the albumin-ODN complex. Due to negative shell surface potential of albumin, the negatively charged shorter nucleic acids do not bind very well to the microbubble and gene transfection efficiencies using these complexes are generally suboptimal.
  • Thus there is a need to improve the binding of the therapeutic agents to the microbubble as well as improving the stability and efficacy of the microbubble complex.
  • Furthermore there is a need to reduce toxicity in the selective delivery of highly cytotoxic drugs. Non-targeted delivery of these drugs can cause systemic toxicity and has prevented the use of many of these drugs all together or at higher doses required for good efficacy. Attempts to deliver these as pro-drugs in many cases have reduced this problem, however, selective uptake in the targeted tissue is not always easy to achieve as most of the uptake mechanisms in the diseased tissue are also present in the normal tissue. Enhancing the uptake of these drugs in selective tissues by non-natural mechanisms as disclosed herein, therefore can add considerable value.
  • BRIEF DESCRIPTION
  • Provided herein are novel compositions and methods for increased binding of therapeutic drugs to microbubbles using the affinity of ligand-therapeutic compositions towards the albumin shell.
  • Systemic circulation of the microbubbles carrying the therapeutic composition can be easily visualized through ultrasound imaging. Therapeutic agent is released from the microbubbles using a trigger of high energy pulsed ultrasound specific to the site of treatment. The cavitation of microbubbles causes sonoporation of the neighboring cells/tissue.
  • In one embodiment a microbubble complex is disclosed comprising a microbubble having an outer shell comprising a mixture of native and denatured albumin and a hollow core encapsulating a perfluorocarbon gas, a therapeutic agent selected from the group comprising a small molecule chemotherapeutic agent, a peptide, a carbohydrate, or an oligonucleotide, and a bifunctional linker having one end attached to the therapeutic agent and the other attached to the ligand through reaction of a reactive group on the ligand. The ligand is bound to the outer shell of the microbubble through hydrophobic interactions.
  • In another embodiment a method is for delivering the aforementioned microbubble complex to a tissue target is disclosed. The method comprising the steps of providing the microbubble complex, administrating the microbubble complex to a subject wherein the subject is the source of the tissue target, and administering ultrasonic energy to the subject, wherein the ultrasound energy is sufficient to cause cavitation of the microbubble complex in the tissue target.
  • BRIEF DESCRIPTION OF THE FIGURES
  • These and other features, aspects, and advantages of the present invention will become better understood when the following detailed description is read with reference to the accompanying figures wherein:
  • FIG. 1 is a representation of non-covalent binding of siRNA-ligand to albumin microbubbles.
  • FIG. 2 is representative micrograph of a gel shift assay for a mixture of cholesterol conjugated siRNA (2 pmoles) and varying amounts of Optison (0, 9, 22 and 46 pmoles for i, ii, iii and iv respectively).
  • FIG. 3 is a. representative micrograph of a gel shift assay for Cy3-siRNA (4 pmoles) mixed with varying concentrations of either Optison or native HSA which shows no shift in gel assay.
  • FIG. 4 is a. representative micrograph of a gel assay for Cy3-cholesterol-siRNA (2 pmoles) mixed with varying concentrations of either Optison, native HSA or denatured HSA.
  • FIG. 5 is a graphical representation of binding properties of cholesterol-siRNA to Optison and native HSA; (A) Relative fluorescence of cholesterol-siRNA bands is measured from the gel shift assay (B) fraction bound of siRNA calculated from relative fluorescence and plotted against the albumin concentration.
  • FIG. 6 is a graphical representation of the uptake of siRNA by U-87 tumor cells in opticell is quantified by measuring cell cy3-fluorescence.
  • FIG. 7 are micrographs of fluorescence images showing a comparison of siRNA transfection between a lipid transfection reagent (RNAifect) and Optison.
  • FIG. 8 is a graphical representation of the mean cell fluorescence values and standard error of siRNA transfection between a lipid transfection reagent (RNAifect) and Optison.
  • FIG. 9 is a graphical representation of the fraction bound of fluorescein-myristate to Optison and native HSA as calculated from anisotropy values.
  • FIGS. 10 A and B are the fluorescein bound to Optison (0, 8, 40 and 200 pmoles for i, ii, iii and iv respectively) and visualized on the gel as dark bands for fluorescein-myristate (63 pmoles) and fluorescein-stearate (180 pmoles) respectively.
  • DETAILED DESCRIPTION
  • The following detailed description is exemplary and not intended to limit the invention of the application and uses of the invention. Furthermore, there is no intention to be limited by any theory presented in the preceding background of the invention or descriptions of the drawings.
  • The invention relates generally to microbubble-assisted delivery of a therapeutic agent to cells or tissue of interest, either in vitro or in vivo.
  • In certain embodiment the therapeutic agent may be comprised of a small molecule chemotherapeutic agent, a peptide, a carbohydrate, or an oligonucleotide, and a bifunctional linker having one end attached to the therapeutic agent and the other attached to the ligand through reaction of a reactive group on the ligand. In certain embodiment the therapeutic agent may be an oligonucleotide (ODN). Oligonucleotides refers to nucleic acid polymers that are formed by bond cleavage of longer nucleic acids or are synthesized using building blocks, protected phosphoramidites of natural or chemically modified nucleosides or, to a lesser extent, of non-nucleosidic compounds. The length of the oligonucleotide may vary from a short nucleic acid polymer of fifty or fewer base pairs to more than 200 base pairs. As used herein, ODN also refers to polynucleotides having more than 200 base pairs. Also included are antisense ODN which refer to single strands of DNA or RNA that are complementary to a chosen sequence. In the case of antisense RNA, antisense RNA prevents protein translation of certain messenger RNA strands by binding to them. Antisense DNA can be used to target a specific, complementary (coding or non-coding) RNA. Also included are small interfering RNA (siRNA), sometimes known as short interfering RNA or silencing RNA, is a class of double-stranded RNA molecules, typically 20-25 nucleotides in length, that play a variety of roles in biology including the RNA interference (RNAi) pathway, where it interferes with the expression of a specific gene, as an antiviral mechanism, or in shaping the chromatin structure of a genome.
  • In certain embodiments, the therapeutic agent may be a cytotoxin. As used herein cytotoxin refers to a substance that has a toxic effect on cells. For example a cytotoxin may cause undergo necrosis, in which they lose membrane integrity and die as a result of cell lysis. In other examples a cytotoxin may be associated with antibody-dependent cell mediated cytotoxicity wherein a cell is marked by an antibody and acted upon by certain lymphocytes.
  • Examples of cytotoxic agents are listed in Goodman and Gilman's “The Pharmacological Basis of Therapeutics,” Tenth Edition, McGraw-Hill, New York, 2001. These include taxol; nitrogen mustards, such as mechlorethamine, cyclophosphamide, melphalan, uracil mustard and chlorambucil; ethylenimine derivatives, such as thiotepa; alkyl sulfonates, such as busulfan; nitrosoureas, such as carmustine, lomustine, semustine and streptozocin; triazenes, such as dacarbazine; folic acid analogs, such as methotrexate; pyrimidine analogs, such as fluorouracil, cytarabine and azaribine; purine analogs, such as mercaptopurine and thioguanine; vinca alkaloids, such as vinblastine and vincristine; antibiotics, such as dactinomycin, daunorubicin, doxorubicin, bleomycin, mithramycin and mitomycin; enzymes, such as L-asparaginase; platinum coordination complexes, such as cisplatin; substituted urea, such as hydroxyurea; methyl hydrazine derivatives, such as procarbazine; adrenocortical suppressants, such as mitotane; hormones and antagonists, such as adrenocortisteroids (prednisone), progestins (hydroxyprogesterone caproate, medroprogesterone acetate and megestrol acetate), estrogens (diethylstilbestrol and ethinyl estradiol), antiestrogens (tamoxifen), and androgens (testosterone propionate and fluoxymesterone).
  • Drugs that interfere with intracellular protein synthesis, protein synthesis inhibitors, can also be coupled to the ligand; such drugs are known to these skilled in the art and include puromycin, cycloheximide, and ribonuclease.
  • In one embodiment, the protein includes, but is not limited to, enzymes, soluble and serum proteins, proteins expressed on a surface of a cell, non-immunoglobulin proteins, intracellular proteins, and segment of proteins that are or can be made water-soluble, either individually or in combinations thereof as well as any derivatives of the proteins.
  • In a particular embodiment, the protein includes such as, but not limited to, cysteine proteases, glutathione S transferase, epoxide hydrolase (EH), thiolase, NAD/NADP-dependent oxidoreductase, enoyl coA hydratase, aldehyde dehydrogenase, hydroxypyruvate reductase, tissue transglutaminase (tTG), formiminotransferase cyclodeaminase (FTCD), aminolevulinate-dehydratase (ADD), creatin kinase, carboxylesterase (LCE), monoacylglycerol (MAG) lipase, metalloproteases (MP), phosphotases (protein tyrosine phosphotases, PTP), proteosome, FK506 binding protein (FKBP12), mammalian target of Rapamycin (mTOR; alternatively known as FKBP-rapamycin binding domain (FRB)), serine hydrolase (superfamily), ubiquitin-binding protein, -galactosidase, nucleotide binding enzymes, protein kinases, GTP-binding proteins, cutinase, adenylo succinate synthase, adenylo succinate lyase, glutamate dehydrogenase, dihydrofolate reductase, fatty acid synthase, aspartate transcarbamylase, acetylcholinesterase, HMG cholate reductase, and cyclo-oxygenase (COX-1 and COX-2), either individually or in combinations thereof. Also included are any derivatives of any of the proteins.
  • In another example, the protein is substantially free of a cofactor. “Substantially free of a cofactor” includes proteins that do not require any additional cofactor, chemical, chemical modification, or physical modification to be naturally stable under physiological conditions and room temperature and pressure in solution or as a solid, and can bind its corresponding ligand in vivo.
  • In one embodiment, an albumin microbubble may be utilized to carry a therapeutic agent in systemic delivery. Tissue targeted ultrasound acoustic energy may then be used to cavitate the albumin microbubble and deliver the therapeutic agent into the intracellular environment. For example the microbubble complex may be administered intravenously or into the peritoneum (intraperitoneally) of a subject whose cells or tissues are to be targeted. Once the microbubble complex is carried through the subject to the targeted cell, the ultrasound acoustic energy is delivered. In certain embodiments, visualization of the targeted cells may occur prior to delivering the ultrasound while in still other embodiments visualization may be performed in real time and the cavitation monitored.
  • In certain embodiments, the albumin outer shell of the microbubble is comprised of both native and denatured albumin held together by mostly cysteine to cysteine bonds. In certain embodiments, the primary composition of the albumin shell is mostly in the native form wherein the denatured portion allows for increased cysteine bond attachments. In certain embodiments the relative amount of denatured albumin to native albumin ranges from approximately 0.5 to 30 wt %. In other embodiments the relative amount is in the range of approximately 1% to 15 wt %. The mixture of native and denatured albumin provides a balance of shell elasticity needed for cavitation, with increased reactive binding sites on the microbubble surface. The microbubbles are formed by sonication of perfluorocarbon gas in the presence of pre-heated albumin solution. A small part of the albumin molecules rearrange during sonication of pre-heated albumin solution and crosslinking occurs through disulfide linkages between albumin molecules. These albumin molecules are believed to be similar in structure to an F form of albumin which has more hydrophobic residues exposed. This allows increased binding sites for hydrophobic interactions.
  • In certain embodiments, the microbubble may be filled with an insoluble perfluorocarbon gas, such as but not limited to, perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluoropentane, or a combination thereof. In certain embodiments, the microbubbles are about 1 to about 5 microns in diameter, the size being optimized to allow circulation through the blood stream.
  • In certain embodiments, the therapeutic-microbubble complexes comprise a therapeutic agent modified with a linker having a reactive group capable of binding with a ligand having affinity towards albumin. As such, the therapeutic agent may be coupled to albumin though the ligand.
  • The linker includes any linking moiety that attaches the ligand to the therapeutic agent through a first moiety. The linker can be as short as one carbon or a long polymeric species such as polyethylene glycol, tetraethylene glycol (TEG), polylysine or other polymeric species normally used in the pharmaceutical industry for modulating pharmacokinetic and biodistribution characteristics of therapeutic agents. Other linkers of varying length include C1-C250 length with one or more heteroatoms selected from O, S, N, P, and optionally substituted with halogen atoms. In a particular embodiment, the linker comprises at least one of an oligomeric or polymeric species made of natural or synthetic monomers, oligomeric or polymeric moiety selected from a pharmacologically acceptable oligomer or polymer composition, an oligo- or poly-amino acid, peptide, saccharide, a nucleotide, and an organic moiety with 1-250 carbon atoms, either individually or in combination thereof. The organic moiety with 1-250 carbon may contain one or more heteroatoms such as O, S, N or P and optionally substituted with halogen atoms at one or more places.
  • The first moiety may simply be an extension of the linker, formed by the reaction of a reactive species on the linker with a reactive group on the therapeutic agent. Examples of reactive species and the reactive group include, but are not limited to, activated esters (such as N-hydroxysuccinimide ester, pentafluorophenyl ester), a phosphoramidite, an isocyanate, an isothiocyanate, an aldehyde, an acid chloride, a sulfonyl chloride, a maleimide an alkyl halide, an amine, a phosphine, a phosphate, an alcohol or a thiol with the proviso that the reactive species and reactive group are matched to undergo a reaction yielding covalently linked conjugates.
  • In certain embodiments, the reactive moiety may be a primary amine functionality, and as such, the amine modified therapeutic agent may be conjugated to the affinity ligand through reaction of a carboxyl moiety of the ligand. In certain other embodiments, the reactive moiety may be an alcohol attached to the ligand through a phosphate group.
  • The affinity ligand includes fatty acids, steroids, small aromatic compounds or a combination thereof. Examples of albumin binding molecules may be found in US patent application publication number 2010/0172844, published Jul. 8, 2010.
  • For example in certain embodiments the affinity ligand is a fatty acid, including but not limited to myristoyl, lithocolic-oleyl, docosanyl, lauroyl, steoroyl, palmitoyl, oleoyl, or linoleoyl. In other embodiments, the lipophilic molecule is a steroid or modified steroid including, cholesterol, cholic acid, lithocholic acid, or chenodeoxycholic acid. In other embodiment, the high affinity molecule is selected from 4-p-iodophenyl-butyric acid and analogs or derivatives thereof. In still other embodiments, the therapeutic agent comprises siRNA, the linker comprises tetraethylene glycol, and the ligand comprises cholesterol.
  • In certain embodiments, the therapeutic-albumin complexes may be prepared either by sonicating ligand-modified therapeutic agent with albumin or lipid in the presence of perfluorocarbon or by mixing preformed bubbles with ligand modified therapeutic agents. In certain embodiments, these molecules may be attached to therapeutic agents of interest during therapeutic agent synthesis. For example the phosphoramidites of cholesterol may be used to incorporate cholesterol during DNA or RNA synthesis on a nucleic acid synthesizer, or post synthesis by incorporating a reacting moiety.
  • In certain embodiments, the therapeutic agent is a modified ODN which may be prepared enzymatically by using modified nucleoside triphosphates; modified either with the ligand itself or with a reactive functionality for post synthesis modification with the ligand. Ligand attachment may be at one or both termini, internal to the nucleic acid sequence or at multiple positions depending upon the ODN use. In certain embodiments, where siRNA is the ODN, attachment may be through the 3′ OH position.
  • In certain embodiments, in addition to the ligand, the therapeutic agent, including where the therapeutic agent is ODN, may be selectively modified to protect from nucleases. In certain embodiments, stabilizing modification may include phosphorothioate modification, or 2′-OMe modification.
  • In certain embodiments, the microbubble complexes may be incubated with cells or the tissue of interest or injected into the body, preferably intravenously, and then cavitated with ultrasound energy at desired site and at a predetermined time or during live imaging.
  • In certain embodiments, the microbubble complex may be viewed during systemic travel in the blood circulation under normal ultrasound diagnostic imaging. When the bubble arrives on tissue target, in this case the tumor, a series of pulsed acoustic energy waves are sent to the tumor. This creates inertial cavitation on the microbubble, which collapses the microbubble. Cavitation of the microbubble occurs where the acoustic energy is maximally located. This direction is achieved on the ultrasound probe by parameters related to mechanical index force, optimal ultrasound acoustic distance, and dimensions of the ultrasound acoustic sweep. The force generated can then potentially form micropores within the cellular plasma membrane. Typically the pulsed energy is administered at a frequency of about 0.5 to about 5 MHz.
  • These micropores, along with the microjetting force created under inertial cavitation, may facilitate the entrance of ODN into the cellular cytoplasmic environment. For example when the ODN is siRNA, siRNA in the intracellular environment will utilize the host machinery to silence mRNA and later protein synthesis. Similarly, where mRNA message acts as an angiogenesis promoting proteins including vascular endothelial growth factor (VEGF), the reduction of VEGF expression in a tumor may halt or slow tumor growth. After microbubble cavitation the dense gas of the microbubble center is exhaled out of the body and the albumin shell is metabolized and excreted via the liver elimination pathway.
  • In an exemplary embodiment, a bolus of the microbubble complex may be mixed to an optimal ratio from previous therapeutic investigations. Once the mixture of the complex is established, the bolus drug is injected systemically by venous route.
  • For example in the use of a siRNA-microbubble bolus, the bolus may be monitored in the first pass blood kinetics. The microbubble resonance and thus enhanced ultrasound contrast may be monitored with an ultrasound probe using low diagnostic levels of acoustic energy. During circulation the bolus arrives on organ target. Cardiovascular tissue perfusion may assist in delivering the bolus into deep microvessels with small lumen diameters. By supplying pulsed acoustic energy, sufficient energy may be provided for the microbubble to undergo inertial cavitation. Once the microbubble cavitation is complete the siRNA contents may be delivered across the plasma membrane and into the diseased cell. While in the cytoplasmic intracellular environment the siRNA can have a therapeutic effect.
  • During microbubble cavitation siRNA entrance into the cell may occur by various mechanisms. For example the siRNA may enter the cell by a: a microjetting force from the collapsing microbubble which can push siRNA into the cytoplasm. Alternatively the mechanism may include microjetting energy or sonoluminescence energy which creates temporary micropores within the plasma membrane to allow for passive diffusion of the siRNA into the cell, or during microbubble resonance before actual cavitation the microbubble bumping into the plasma membrane may push the siRNA in the cell.
  • As such, the mechanism of microbubble delivery has potential applications in the treatment of a wide variety of diseases, which can include cancer, inflammatory, infectious, cardiovascular, metabolic, autoimmune, and central nervous system diseases. Many of these diseases cannot currently be effectively treated by virtue of targeting molecular mechanisms not accessible to conventional small molecule drugs and monoclonal antibodies.
  • EXPERIMENTAL Example 1
  • FIG. 1 is a representation illustration of binding of siRNA to albumin encapsulated microbubbles to form a microbubble-siRNA complex.
  • The target siRNA for VEGF silencing (vascular endothelial growth factor) were synthesized by IDTDNA technologies. IDTDNA provided lipid modifications such as cholesteryl TEG on the siRNA (Chol-siRNA) as well as dye conjugation.
  • Sense strand: 5′-Cy3/GCAUUUGUUUGUCCAAGAUmUmU/3′-Lipid
    Antisense strand; 5′/mAmArA rUrCrU rUrGrG rArCrA rArArC rArArA
    rUrGrC/3′
  • Cyanine dye, Cy3 on the siRNA has an excitation wavelength of 550 nm and a peak emission of 580 nm. The siRNA has been labeled with a cy3 dye for easy visualization of siRNA during binding assays and other characterization techniques. Optison™ (GE Healthcare, Chalfont St. Giles, United Kingdom, 10 mg/ml albumin) was centrifuged; the top layer was discarded and the excess albumin solution in the bottom was used for the binding studies. Lyophilized human serum albumin (HSA) powder (Sigma Aldrich, St. Louis Mo.) was dissolved in 1× phosphate buffered saline (PBS) to make a stock solution of 10 mg/mL. Both Optison and native albumin solution dilutions were prepared with 1×PBS. Denatured HSA solution was prepared by heating native HSA solution to 80° C. for 20 minutes.
  • Binding Reaction:
  • The stock solutions of cy3-siRNA and cy3-siRNA-cholesterol, 20 μM were prepared in RNAse free water and stored at −20° C. 4 pmoles of cy3-siRNA and 2 pmoles of cholesterol-siRNA solutions were mixed with varying amounts of Optison solution, native HSA and denatured HSA solution, ranging from 0 to 50 pmoles. The reaction buffer was 1×PBS, pH 7.4. The reaction mixture was incubated under dark at 25° C. for 45 minutes. After incubation, ten μL of siRNA mixtures was mixed with 2 μL of Novex® Hi-Density TBE Sample Buffer (5×) (Invitrogen, Carlsbad, Calif., USA).
  • Gel Electrophoresis:
  • All the reactions were loaded onto precast 6% nondenaturing polyacrylamide gels (Invitrogen, Carlsbad, Calif., USA). The gel was run at 100 V for 45 min in 0.5× Novex TBE Running buffer (Invitrogen, Carlsbad, Calif., USA). The gels containing either DNA, protein or both were imaged for cy3 fluorescence using a typhoon scanner (Typhoon™ 9410, GE Healthcare).
  • Results:
  • The fluorescence of Cy3 attached to siRNA can be visualized as distinct siRNA bands on the gel. When a mixture of siRNA and albumin solution was run on the gel, the mobility of albumin bound-siRNA is slower than free-siRNA resulting in two bands on the gel. In preliminary trials, sypro ruby stain from EMSA kit (Molecular Probes, Eugene, Oreg., USA) was used to observe the albumin bands in the gel. An example is shown in FIG. 2 which is a gel shift assay for a mixture of cholesterol conjugated siRNA (2 pmoles) and varying amounts of Optison (0, 9, 22 and 46 pmoles for i, ii, iii and iv respectively). Fluorescence imaging of the gel shows distinct bands for siRNA (lower band sections) and albumin (upper band sections).
  • Cy3-siRNA
  • When the mixture of cy3-siRNA and native HSA/Optison was run on the gel, there was no bound-siRNA visualized for increasing concentrations of albumin. There was no significant binding of cy3-siRNA with either native HSA or Optison solution. This is shown in FIG. 3 which is a fluorescence image of a gel for Cy3-siRNA (4 pmoles) mixed with varying concentrations of either Optison or native HSA shows no shift in gel assay. The dark bands on the gel are the cy3-fluorescence on siRNA. There is no significant binding of cy3-siRNA to both Optison and native HSA.
  • Chol-siRNA
  • FIG. 4 shows the gel images for binding of chol-siRNA with Optison, native HSA and denatured HSA. Chol-siRNA bound to both native HSA and Optison solution, while the binding significantly decreased for the same amount of denatured HSA. The fluorescence intensity of siRNA in each lane was estimated manually by drawing a box around the bands. Background, equivalent to the average intensity value of the gel, was subtracted from the intensity value of each siRNA band. Fluorescence intensities of bound siRNA over a wide range of albumin concentrations were calculated. Relative fluorescence, R was calculated as:

  • R=(Fbound−Ffree)/Ffree  (1)
  • Fbound is fluorescence intensity of bound-siRNA band and Ffree is fluorescence intensity of free-siRNA band. Relative fluorescence was plotted against albumin concentration. This is shown in FIG. 5, which is a graphical comparison of binding properties of cholesterol-siRNA to Optison and native HSA as described below
  • At low albumin concentration ranging from 0 to 15 μM, linear dependence of bound fluorescence on albumin concentration was visualized. FIG. 5, graph A shows that at this concentration range, the amount of chol-siRNA bound to Optison solution was higher than the binding to native HSA. To estimate binding constants, higher concentration of albumin was used to allow saturation of amount of siRNA bound to albumin. Fraction bound, x is determined as:

  • x=(Fbound−Ffree)/(Fsat−Ffree)  (2)
  • Fsat is the fluorescence intensity of maximum bound-siRNA under saturation conditions.
  • Fraction bound was plotted against increasing albumin concentrations, as shown in FIG. 5 graph B, and the data points were fitted to the following equilibrium equation;

  • x=n*[Albumin]/(kd+[Albumin])  (3)
  • kd is the dissociation constant, n is the number of binding sites and [Albumin] is the total albumin concentration for the respective samples. Equation 3 was solved using a non-linear fit to determine kd and n for binding of chol-siRNA to both Optison and native HSA (Table 1). Microsoft Excel's solver tool was utilized for this non-linear fit, and the sum of squared errors (SSE) was found to be 0.07 and 0.06 for Optison and native HSA respectively. The binding constant of chol-siRNA was similar for both Optison and native HSA.
  • Example 2 Delivery of siRNA to Tumor Cells Cell Culture:
  • MATBIII rat mammary carcinoma and U-87 human glioblastoma cells were cultured in McCoy's 5A Medium (modified) (1×) (Invitrogen, Carlsbad, Calif., USA) and Eagle's Minimum Essential Medium (EMEM) (ATCC, Manassas, Va.) respectively. Both the media solutions were supplemented with 10% heat deactivated fetal bovine serum (FBS) (Fisher Scientific, Springfield, N.J.) and 1% penicillin-streptomycin (Sigma Aldrich, St Louis, Mo.). The cells were maintained at 37° C. in a humidified atmosphere with 5% CO2.
  • Sonication of Substrate-Attached Cells:
  • MATB-III and U-87 cells were grown in 10 mL capacity Opticell units (Nalge Nunc International, Rochester, N.Y.) to 90% confluence. The media in OptiCell was replaced with 10 mL fresh media containing 40 pmoles of either cy3-siRNA or cholesterol-siRNA. The opticell was left in the incubator for 24 hours at 37° C. Separately, the cells were either treated with siRNA solution mixed with Optison microbubbles (300 μL) or a lipid transfection reagent (90 μL) (RNAifect, Qiagen, Valencia, Calif.). For siRNA/Optison mixtures, Vivid i imagers with a cardiac sector probe (3S) was used to rupture the microbubbles and deliver the siRNA drug from microbubbles. The opticell was immobilized in a water bath, and the ultrasound probe was attached to a motion arm that spanned the entire length of the opticell. The tip of the probe was immersed in water, and the distance between the probe and opticell surface was 3 cm that allowed sonication of the entire width of the opticell. The microbubbles in opticell were treated with a mechanical index, MI>1.3 continuous sonication. The probe moved at a speed of 1 m/s over the entire length of the opticell. After sonication, the cells were incubated for 24 hours at 37° C. Similarly, the cells treated with RNAifect were also kept in the incubator for 24 hours. After incubation, the cells were imaged using a fluorescence microscope (Zeiss Axio Imager.Z1, Carl Zeiss). The filter used for cy3 was DsRed/Cy3 (546 ex/620 em). In the region of interest (ROI) in the fluorescent images, cell fluorescence was measured and the mean values of cell fluorescence were calculated. ImageJ was used to process the images and calculate fluorescence intensities.
  • The data are reported as mean+1.0 standard error (SE) for N=4. The statistical significance of the differences between the groups was evaluated using two-sample t-test and the statistical analyses were carried out using Minitab® 12 (Minitab Inc, State College, Pa. USA).
  • Results:
  • The effect of the delivery system is illustrated in FIG. 6 for U-87 cells incubated with either cy3-siRNA or chol-siRNA. The delivery of siRNA into the tumor cells is represented by average cell cy3-fluorescence. For each group, mean fluorescence values and standard errors are reported in. Cell sonication substantially enhanced cy3-siRNA penetration into the cell. Due to the effects of sonoporation, average cell fluorescence for Optison/ultrasound treated cells was 39% more than untreated cells. For cholesterol-siRNA, there was a 53% increase in average cell fluorescence after treatment with Optison/ultrasound. Significant differences between the groups were evaluated using two sample t-tests (p=0.032 for cy3-siRNA and p=0.059 for cholesterol-siRNA).
  • Similarly for MATBIII cells, the effect of Optison/ultrasound treatment was compared to a commercially available lipid transfection reagent. The cells were treated with either Cy3-siRNA or chol-siRNA in combination with either RNAifect or Optison/ultrasound delivery agents and the results are shown in FIGS. 7 and 8. FIG. 7 shows representative images of the cells after treatment. FIG. 8 reports the mean cell fluorescence for all groups with standard errors represented as error bars. For cy3-siRNA, the average cell fluorescence was higher for Optison/ultrasound treatment (two sample t-test, p=0.007). This is primarily due to sonoporation of the cells in the presence of microbubbles. There was no significant difference between RNAifect and Optison/ultrasound delivery of chol-siRNA into cells. Although the average cell fluorescence was similar, the lipid transfection reagent was found to be toxic to the tumor cells as evidenced by the irregular shape of the cells in FIG. 7. It should be noted that the same amount of transfection reagent and siRNA was used in both unmodified and cholesterol-siRNA. While the transfection reagent was toxic in both the cases, it was higher for chol-siRNA. Example 3
  • Preliminary binding studies of therapeutic-fatty acid conjugates to microbubbles were evaluated using fluorescein-fatty acid conjugates.
  • Conjugation Method;
  • Fatty acid NHS ester (2 equivalents, 5.37 mg Myristic acid NHS ester or 6.38 mg Stearic acid NHS ester was taken in a 50:50 mixture of DMSO and dichloromethane (100 ul) and mixed with a solution of Fluorescein cadaverine (FL-Cadaverine, 5 mg, 1 equivalent, in 50 ul DMSO). To this diisopropylethyl amine (3.8 equivalents) was added and mixture was vortexed to give a clear solution. Samples were kept in the dark at room temperature. After 4.5 h, reaction was checked by HPLC and was found to be complete. A large shift in retention time was observed for both conjugates (Retention times FL-Cadaverine 4.7 min, FL-Cadaverine stearate 12.1 minute and FL-Cadaverine Myristate 9.9 min, column X-Bridge Shield RP 18, 4.6×50 mm column, particle size 5 um, gradient method 0-100% B in 15 min and 100% B for 5 min, solvent A 0.1M TEAA, pH 7.0 and solvent B 100% acetonitrile, flow rate 1 ml/min) as expected. Crude product was diluted with DMSO to ˜2 ml and purified on AKTA purifier using Xterra MS C18, 19×100 mm column and a gradient of 0-100% B in 18.75 column volumes at a flow rate of 10 ml/min. Solvent A and B were as described above for the analytical method. Product was collected in multiple fractions and each fraction was analyzed by analytical HPLC. Only the purest fraction in each case (˜90% purity) was used for binding studies (starting material itself was ˜86% pure, remaining likely a regioisomer with same spectral properties). This fraction was concentrated to dryness at room temperature. Residue was suspended in water (˜2 ml) and extracted with dichloromethane (3×2 ml). Organic extracts were combined, dried over anhydrous sodium sulfate and concentrated to dryness.
  • Fluorescence Polarization Assay
  • The stock solution of fluorescein-myristate was prepared in 1×PBS. The concentration of fluorescein was kept low for the fluorescence polarization assay, at 126 nM. Varying concentrations of either Optison or HSA solutions, ranging from 0 to 15 μM albumin concentrations, were added to the fluorescein myristate solution. The reaction buffer was 1×PBS, pH 7.4. The reaction mixture was incubated under dark at 25° C. for 15 minutes. After incubation, the changes in raw anisotropy values of fluorescein were measured using a microplate reader (SpectraMax 5, Molecular Devices, Sunnyvale, Calif.).
  • The samples were measured in Corning 96-well plates (black plate with a clear bottom) (Sigma Aldrich, St Louis, Mo.). Fluorescein was excited at 470 nm, and emission was measured at 540 nm. Fraction bound (x) was calculated using the same equation as before (Equation 2), but replacing fluorescence values with anisotropy values. The fraction bound calculated was then plotted against albumin concentration as shown in FIG. 9. The data are reported as mean+1.0 standard error (SE) for N=3. Equation 3 was used to determine kd and n for fluorescein-myristate binding to both Optison and native HSA (Table 2). This is represented also in FIG. 10 which shows the fluorescein bound to Optison (0, 8, 40 and 200 pmoles for i, ii, iii and iv respectively) is visualized on the gel as dark bands for fluorescein-myristate (FIG. 10A) (63 pmoles) and fluorescein-stearate (FIG. 10B) (180 pmoles).
  • When the fluorescein without the myristate conjugation was tested for its binding properties to albumin, no significant changes in anisotropy was observed. It is well known that the fatty acids have stronger binding properties than cholesterol, and is also confirmed here with the lower dissociation constants, kd, observed for fluorescein-myristate conjugate (Table 1 and Table 2).
  • TABLE 1
    Number of binding sites and dissociation constants for
    binding of cholesterol-siRNA to Optison and native HSA
    Optison Native HSA
    Number of binding sites, n 1.16 1.13
    Dissociation constant, kd (μM) 7.16 6.4
  • TABLE 2
    Number of binding sites and dissociation constants for binding
    of fluorescein-myristate to Optison and native HSA
    Optison Native HSA
    Number of binding sites, n 1.03 1.02
    Dissociation constant, kd (μM) 0.238 0.378
  • Therefore, conjugating a fatty acid such as myristate to a therapeutic compound can increase the binding of such therapeutic compounds to the albumin shell microbubbles. The dissociation constant of fluorescein-myristate binding to Optison was lower than that of binding to native HSA. This suggests better hydrophobic binding properties of microbubble shell that has both native and partially denatured albumin.
  • Example 4 Stability of siRNA In Vivo
  • The stability of therapeutic compounds such as siRNA is very low once injected into the body. A comparison between subcutaneous and tail-vein injection of a mixture of albumin microbubbles and native siRNA (without conjugates) was studied.
  • Eleven to fourteen weeks (body weight ˜30 g) old nu/nu mice were obtained from Charles River Laboratories (Wilmington, Mass.). Animals were housed in accordance with the Guide for the Care and Use of Laboratory Animals as adopted by the National Institutes of Health. Lewis lung carcinoma cells (LLC) were inoculated subcutaneously to the right flank of anaesthetized mice (3.5×106 cells/100 μl/mouse).
  • On the fourth day after inoculation, the mice were treated with anti-VEGF siRNA (Sigma Life Sciences, St. Louis, Mo.)—microbubble mixtures, a siRNA dose of 1.0 mg/kg for subcutaneous injections and a dose of 2.0 mg/kg for tail-vein injections. The injection mixture contained 100 μL of microbubble solution and 100 μL of siRNA in RNAse free water. After injection, the tumors were sonicated using Vivid i imagers with a cardiac sector probe (3S). The energies were delivered in a pulsatile form with the peak MI at 1.3. Control group did not receive any treatments.
  • After 24 hours from the treatment day, the mice were euthanized and the tumors were extracted. The tumors were frozen immediately and stored at −20° C. The tumors were thawed out at room temperature on the day of VEGF measurement. The tumors were then lysed in RIPA buffer (protease inhibitors added) using a lysing matrix tube (Lysing matrix tube A, RP Biomedical). The lysate collected from the samples were then diluted, and measured for total protein using a protein kit (Pierce BCA reagent protein assay kit) and for VEGF using an ELISA kit (Mouse VEGF ELISA kit, RayBiotech, Norcross, Ga.).
  • The results are reported in Table 3 as mean pg VEGF/mg protein for control and different treatment groups. The subcutaneous injection of 1.0 mg/kg siRNA-microbubble mixture resulted in an approximately 39% decrease in VEGF when compared to the control group (two-sample t-test; p=0.0096). While there was only a minor difference between the control group and 2.0 mg/kg tail-vein injection of siRNA-microbubble mixtures, this may be due to lack or less efficient binding of unmodified siRNA to the microbubble.
  • TABLE 3
    The effect of siRNA delivery to tumors;
    mean pg VEGF/mg total protein.
    Mean pg VEGF/ Std
    Conditions mg protein error 95% CI n
    control 296.19 24.89 48.79 5
    Optison/siRNA SQ 1 mg/kg 181.97 24.38 47.78 7
    Optison/siRNA TV 2 mg/kg 255.1 51.06 100.07 3
  • The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects as illustrative rather than limiting on the invention described herein. The scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.

Claims (34)

1. A microbubble complex comprising:
a microbubble having an outer shell comprising a mixture of native and denatured albumin and a hollow core encapsulating a perfluorocarbon gas;
a therapeutic agent selected from a group comprising a small molecule chemotherapeutic agent, peptide, carbohydrate, oligonucleotide, cytotoxin, protein synthesis inhibitor, or combination thereof;
a bifunctional linker having one end attached to the therapeutic agent and the other attached to a ligand through reaction of a reactive group on said ligand; and
wherein the ligand is bound to the outer shell of the microbubble through hydrophobic interactions.
2. The complex of claim 1 wherein the therapeutic agent is an oligonucleotide.
3. The complex of claim 2 wherein the oligonucleotide is a naturally occurring or modified DNA or RNA.
4. The complex of claim 3 wherein the RNA is a small interfering RNA.
5. The complex of claim 1 wherein the bifunctional linker comprises a oligo- or poly-amino acid, peptide, saccharide, nucleotide, organic moiety having approximately 1 to 250 carbon atoms, or a combination thereof.
6. The complex of claim 1 wherein the bifunctional linker comprises tetraethylene glycol (TEG) or polyethylene glycol.
7. The complex of claim 1 wherein the reactive group comprises an activated ester, phosphoramidite, isocyanate, isothiocyanate, aldehyde, acid chloride, sulfonyl chloride, maleimide, alkyl halide, amine, phosphine, phosphate, alcohol or thiol.
8. The complex of claim 7 wherein the reactive group is an amine.
9. The complex of claim 1 wherein the ligand is a fatty acid, steroid, or a combination thereof.
10. The complex of claim 9 wherein the fatty acid is myristoyl, lithocolic-oleyl, docosanyl, lauroyl, steoroyl, palmitoyl, oleoyl, linoleoyl, or a combination thereof.
11. The complex of claim 9 wherein the steroid is cholesterol, cholic acid, lithocholic acid, chenodeoxycholic acid or a combination thereof.
12. The complex of claim 1 wherein the ligand is 4-iodophenyl butyric acid or an analog or derivative thereof.
13. The complex of claim 1 wherein the amount of denature to native albumin is in the range of approximately 0.5 to 30 wt %.
14. The complex of claim 13 wherein the range of denature to native albumin is approximately 1 to 15 wt %.
15. The complex of claim 1 wherein the therapeutic agent comprises siRNA, the linker comprises tetraethylene glycol, and the ligand comprises cholesterol.
16. A method for delivering a microbubble complex to a tissue target comprising the steps of:
providing a microbubble complex, said complex comprising;
a microbubble having an outer shell comprising a mixture of native and denatured albumin and a hollow core encapsulating a perfluorocarbon gas;
a therapeutic agent selected from a group comprising a small molecule chemotherapeutic agent, peptide, carbohydrate, oligonucleotide, cytotoxin, protein synthesis inhibitor, or combination thereof;
a bifunctional linker having one end attached to the therapeutic agent and the other attached to a ligand through reaction of a reactive group on said ligand; and
wherein the ligand is bound to the outer shell of the microbubble through hydrophobic interactions;
administrating the microbubble complex to a subject wherein the subject is the source of the tissue target; and
administering ultrasonic energy to the subject, wherein said energy is sufficient to cause cavitation of the microbubble complex in the tissue target.
17. The method of claim 16 wherein the tissue target is in vivo and administrating the microbubble complex comprises intravenous or intraperitoneal injection of the microbubble complex.
18. The method of claim 16 further comprising the step of visualizing the microbubble complex at the tissue target prior to administering the ultrasonic energy for cavitation of the microbubble complex.
19. The method of claim 16 wherein the visualizing and administering the ultrasonic energy are performed in real time.
20. The method of claim 16 wherein the tissue target is in vitro.
21. The method of claim 16 wherein the therapeutic agent is an oligonucleotide.
22. The method of claim 21 wherein the oligonucleotide is a naturally occurring or modified DNA or RNA.
23. The method of claim 22 wherein the RNA is a small interfering RNA.
24. The method of claim 16 wherein the bifunctional linker comprises a oligo- or poly-amino acid, peptide, saccharide, nucleotide, organic moiety having approximately 1 to 250 carbon atoms, or a combination thereof.
25. The method of claim 16 wherein the bifunctional linker comprises tetraethylene glycol (TEG) or polyethylene glycol.
26. The method of claim 16 wherein the reactive group comprises an activated ester, phosphoramidite, isocyanate, isothiocyanate, aldehyde, acid chloride, sulfonyl chloride, maleimide, alkyl halide, amine, phosphine, phosphate, a alcohol or thiol.
27. The method of claim 26 wherein the reactive group is an amine.
28. The method of claim 16 wherein the ligand is a fatty acid, steroid, or a combination thereof.
29. The method of claim 28 wherein the fatty acid is myristoyl, lithocolic-oleyl, docosanyl, lauroyl, steoroyl, palmitoyl, oleoyl, linoleoyl, or a combination thereof.
30. The method of claim 28 wherein the steroid is cholesterol, cholic acid, lithocholic acid, chenodeoxycholic acid or a combination thereof.
31. The method of claim 16 wherein the ligand is 4-iodophenyl butyric acid or an analog or derivative thereof.
32. The method of claim 16 wherein the amount of denature to native albumin is in the range of approximately 0.5 to 30 wt %.
33. The method of claim 32 wherein the range of denature to native albumin is approximately 1 to 15 wt %.
34. The method of claim 16 wherein the therapeutic agent comprises siRNA, the linker comprises tetraethylene glycol, and the ligand comprises cholesterol.
US13/528,399 2011-09-19 2012-06-20 Microbubble complexes and methods of use Abandoned US20130072854A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
US13/528,399 US20130072854A1 (en) 2011-09-19 2012-06-20 Microbubble complexes and methods of use
CN201710959069.0A CN107737335B (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
EP12759459.6A EP2758081B1 (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
CN201280045451.6A CN103957940B (en) 2011-09-19 2012-09-18 Microvesicle compound and application method
BR112014006412A BR112014006412A2 (en) 2011-09-19 2012-09-18 microbubble complex, and method for administering a microbubble complex to a target tissue
ES12759459.6T ES2621821T3 (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
KR1020147006830A KR20140063697A (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
JP2014530263A JP6059725B2 (en) 2011-09-19 2012-09-18 Microbubble composite and method of use
AU2012311659A AU2012311659B2 (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
MX2014003358A MX347719B (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use.
RU2014108245A RU2613321C2 (en) 2011-09-19 2012-09-18 Complexes based on micro-bubbles and methods of application
CA2849037A CA2849037A1 (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
PCT/EP2012/068288 WO2013041500A1 (en) 2011-09-19 2012-09-18 Microbubble complexes and methods of use
HK15101059.9A HK1200362A1 (en) 2011-09-19 2015-02-02 Microbubble complexes and methods of use
US15/219,419 US10004760B2 (en) 2011-09-19 2016-07-26 Microbubble complexes and methods of use
US16/016,963 US11007212B2 (en) 2011-09-19 2018-06-25 Microbubble complexes and methods of use
HK18110916.0A HK1251462A1 (en) 2011-09-19 2018-08-24 Microbubble complexes and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201113235890A 2011-09-19 2011-09-19
US13/528,399 US20130072854A1 (en) 2011-09-19 2012-06-20 Microbubble complexes and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US201113235890A Continuation-In-Part 2011-09-19 2011-09-19

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/219,419 Continuation US10004760B2 (en) 2011-09-19 2016-07-26 Microbubble complexes and methods of use

Publications (1)

Publication Number Publication Date
US20130072854A1 true US20130072854A1 (en) 2013-03-21

Family

ID=46852013

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/528,399 Abandoned US20130072854A1 (en) 2011-09-19 2012-06-20 Microbubble complexes and methods of use
US15/219,419 Active US10004760B2 (en) 2011-09-19 2016-07-26 Microbubble complexes and methods of use
US16/016,963 Active 2032-01-06 US11007212B2 (en) 2011-09-19 2018-06-25 Microbubble complexes and methods of use

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/219,419 Active US10004760B2 (en) 2011-09-19 2016-07-26 Microbubble complexes and methods of use
US16/016,963 Active 2032-01-06 US11007212B2 (en) 2011-09-19 2018-06-25 Microbubble complexes and methods of use

Country Status (13)

Country Link
US (3) US20130072854A1 (en)
EP (1) EP2758081B1 (en)
JP (1) JP6059725B2 (en)
KR (1) KR20140063697A (en)
CN (2) CN103957940B (en)
AU (1) AU2012311659B2 (en)
BR (1) BR112014006412A2 (en)
CA (1) CA2849037A1 (en)
ES (1) ES2621821T3 (en)
HK (2) HK1200362A1 (en)
MX (1) MX347719B (en)
RU (1) RU2613321C2 (en)
WO (1) WO2013041500A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016079338A1 (en) * 2014-11-21 2016-05-26 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US9457082B2 (en) 2013-09-26 2016-10-04 Samsung Electronics Co., Ltd. Liposome including complex of hydrophobic active ingredient and polypeptide and use of the liposome
US9743909B1 (en) * 2013-05-15 2017-08-29 University Of Washington Through Its Center For Commercialization Imaging bubbles in a medium
US10136835B1 (en) 2012-05-02 2018-11-27 University Of Washington Through Its Center For Commercialization Determining a presence of an object
US11382987B2 (en) * 2015-11-23 2022-07-12 University Of Ulster Microbubble-chemotherapeutic agent complex for sonodynamic therapy

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201511459D0 (en) * 2015-06-30 2015-08-12 Cell Therapy Ltd Sirna microbubble
KR102245539B1 (en) * 2018-02-12 2021-04-29 주식회사 지앤피바이오사이언스 Composition for increasing expression level of growth factor genes containing core-shell structured microparticles as effective component
KR102398743B1 (en) * 2020-09-03 2022-05-16 충남대학교병원 Composition for treating the dysuresia comprising microbubbles to be ultrasonic-induced

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030157025A1 (en) * 1995-06-07 2003-08-21 Unger Evan C. Novel methods of imaging and treatment with targeted compositions
US20060193869A1 (en) * 2004-12-17 2006-08-31 Franck Barrat Methods and compositions for induction or promotion of immune tolerance
US20070087022A1 (en) * 1996-10-01 2007-04-19 Abraxis Bioscience, Inc. Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20070148168A1 (en) * 2005-10-28 2007-06-28 Sabbadini Roger A Compositions and methods for the treatment and prevention of fibrotic, inflammatory and neovascularization conditions
US20080319375A1 (en) * 2007-06-06 2008-12-25 Biovaluation & Analysis, Inc. Materials, Methods, and Systems for Cavitation-mediated Ultrasonic Drug Delivery in vivo

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NO940711D0 (en) 1994-03-01 1994-03-01 Nycomed Imaging As Preparation of gas-filled microcapsules and contrast agents for diagnostic imaging
JP4774135B2 (en) * 1996-03-12 2011-09-14 ボード オブ リージェンツ オブ ザ ユニバーシティ オブ ネブラスカ Targeted site-specific drug delivery compositions and uses thereof
US6245747B1 (en) 1996-03-12 2001-06-12 The Board Of Regents Of The University Of Nebraska Targeted site specific antisense oligodeoxynucleotide delivery method
DE69724599T2 (en) * 1996-03-12 2004-08-05 The Board Of Regents Of The University Of Nebraska, Lincoln COMPOSITION FOR TARGET-SPECIFIC ADMINISTRATION OF A MEDICINAL PRODUCT AND METHOD FOR USE
CA2270120A1 (en) * 1996-10-28 1998-05-07 Pal Rongved Improvements in or relating to diagnostic/therapeutic agents
US6261537B1 (en) 1996-10-28 2001-07-17 Nycomed Imaging As Diagnostic/therapeutic agents having microbubbles coupled to one or more vectors
NZ335799A (en) * 1996-10-28 2000-11-24 Nycomed Imaging As Improvements in or relating to diagnostic/therapeutic agents
JPH111811A (en) 1997-06-05 1999-01-06 Aderans Co Ltd Stopper for wig and sheet for attaching stopper for wig
WO2000042988A1 (en) * 1999-01-21 2000-07-27 University Of Virginia Method of identifying and treating inflamed tissue
CN1433478A (en) * 1999-12-30 2003-07-30 诺瓦提斯公司 Novel colloid synthetic vectors for gene therapy
JP2004510830A (en) * 2000-10-11 2004-04-08 ターゲサム・インコーポレーテッド Targeted therapeutic agents
US20050148530A1 (en) 2002-02-20 2005-07-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20030147812A1 (en) * 2001-12-11 2003-08-07 Friedrich Ueberle Device and methods for initiating chemical reactions and for the targeted delivery of drugs or other agents
AU2002241270A1 (en) * 2002-03-22 2003-10-08 Anges Mg, Inc. Compositions for delivering biologically active drug and method of using the same
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
WO2005007810A2 (en) * 2003-06-16 2005-01-27 Grinstaff Mark W Functional synthetic molecules and macromolecules for gene delivery
EP1644475A4 (en) 2003-06-20 2009-06-03 Isis Pharmaceuticals Inc Double stranded compositions comprising a 3'-endo modified strand for use in gene modulation
JP2005314381A (en) * 2004-03-30 2005-11-10 Anges Mg Inc Prophylactic/therapeutic/ameliorating agent for proliferative nephropathy
US7893244B2 (en) 2005-04-12 2011-02-22 Intradigm Corporation Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases
JP4992071B2 (en) * 2005-07-29 2012-08-08 国立大学法人 鹿児島大学 Composition and device for introduction of bioactive agents into ocular tissue
CN100551440C (en) * 2006-08-30 2009-10-21 中国人民解放军第三军医大学第二附属医院 A kind of therapeutic type ultrasonic microbubble that is used for tumour ultrasonic therapy and preparation method thereof
GB0621973D0 (en) * 2006-11-03 2006-12-13 Philogen Spa Binding molecules and uses thereof
US20080114287A1 (en) 2006-11-14 2008-05-15 Kar Neng Lai Ultrasound Microbubble Mediated Genes Delivery System
CA2702028A1 (en) 2007-10-02 2009-04-09 Rxi Pharmaceuticals Corp. Tripartite rnai constructs
KR101198354B1 (en) 2007-10-17 2013-03-14 주식회사 삼양바이오팜 Ldl-like cationic nanoparticles for deliverying nucleic acid gene, method for preparing thereof and method for deliverying nucleic acid gene using the same
BRPI0704944A8 (en) 2007-11-30 2017-08-15 V & M Do Brasil S/A FORGED SEAMLESS TUBE AXLE FOR RAILWAY VEHICLES AND MANUFACTURING PROCESS OF FORGED SEAMLESS TUBE AXLE FOR RAILWAY VEHICLES

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030157025A1 (en) * 1995-06-07 2003-08-21 Unger Evan C. Novel methods of imaging and treatment with targeted compositions
US20070087022A1 (en) * 1996-10-01 2007-04-19 Abraxis Bioscience, Inc. Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20060193869A1 (en) * 2004-12-17 2006-08-31 Franck Barrat Methods and compositions for induction or promotion of immune tolerance
US20070148168A1 (en) * 2005-10-28 2007-06-28 Sabbadini Roger A Compositions and methods for the treatment and prevention of fibrotic, inflammatory and neovascularization conditions
US20080319375A1 (en) * 2007-06-06 2008-12-25 Biovaluation & Analysis, Inc. Materials, Methods, and Systems for Cavitation-mediated Ultrasonic Drug Delivery in vivo

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Sirsi et al (Microbubble compositions, properties and biomedical applications, November 2009) *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10136835B1 (en) 2012-05-02 2018-11-27 University Of Washington Through Its Center For Commercialization Determining a presence of an object
US11096604B2 (en) 2012-05-02 2021-08-24 University Of Washington Through Its Center For Commercialization Determining a presence of an object
US9743909B1 (en) * 2013-05-15 2017-08-29 University Of Washington Through Its Center For Commercialization Imaging bubbles in a medium
US9457082B2 (en) 2013-09-26 2016-10-04 Samsung Electronics Co., Ltd. Liposome including complex of hydrophobic active ingredient and polypeptide and use of the liposome
WO2016079338A1 (en) * 2014-11-21 2016-05-26 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US10052394B2 (en) 2014-11-21 2018-08-21 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US20180344880A1 (en) * 2014-11-21 2018-12-06 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US10751429B2 (en) * 2014-11-21 2020-08-25 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US11007285B2 (en) 2014-11-21 2021-05-18 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US11382987B2 (en) * 2015-11-23 2022-07-12 University Of Ulster Microbubble-chemotherapeutic agent complex for sonodynamic therapy

Also Published As

Publication number Publication date
RU2014108245A (en) 2015-10-27
EP2758081B1 (en) 2017-02-08
JP2014526493A (en) 2014-10-06
AU2012311659A1 (en) 2014-04-10
AU2012311659B2 (en) 2017-06-15
US20170007636A1 (en) 2017-01-12
CN103957940A (en) 2014-07-30
CN103957940B (en) 2017-11-17
CN107737335B (en) 2023-08-29
RU2613321C2 (en) 2017-03-15
CA2849037A1 (en) 2013-03-28
US11007212B2 (en) 2021-05-18
CN107737335A (en) 2018-02-27
KR20140063697A (en) 2014-05-27
MX2014003358A (en) 2014-08-01
JP6059725B2 (en) 2017-01-11
BR112014006412A2 (en) 2017-04-04
WO2013041500A1 (en) 2013-03-28
EP2758081A1 (en) 2014-07-30
US10004760B2 (en) 2018-06-26
MX347719B (en) 2017-05-10
US20190060347A1 (en) 2019-02-28
HK1251462A1 (en) 2019-02-01
HK1200362A1 (en) 2015-08-07
ES2621821T3 (en) 2017-07-05

Similar Documents

Publication Publication Date Title
US11007212B2 (en) Microbubble complexes and methods of use
Dong et al. Strategies, design, and chemistry in siRNA delivery systems
Al-Jawadi et al. Ultrasound-responsive lipid microbubbles for drug delivery: A review of preparation techniques to optimise formulation size, stability and drug loading
Tai et al. A ribonucleoprotein octamer for targeted siRNA delivery
AU2013256341B2 (en) Novel tetraGalNAc and peptide containing conjugates and methods for delivery of oligonucleotides
Du et al. Ultrasound responsive magnetic mesoporous silica nanoparticle-loaded microbubbles for efficient gene delivery
Zhou et al. SPANosomes as delivery vehicles for small interfering RNA (siRNA)
JP2009029810A (en) Composition and method for altering biodistribution of biological agent
US20210162067A1 (en) Composition and Methods of Controllable Co-Coupling Polypeptide Nanoparticle Delivery System for Nucleic Acid Therapeutics
BR112016000160B1 (en) improved nanoparticle-type oligonucleotide structure having high efficiency and method for preparing the same
Zou et al. Fluorinated DNA micelles: synthesis and properties
Allemailem et al. Recent advances in understanding oligonucleotide aptamers and their applications as therapeutic agents
US20200246484A1 (en) Spherical nucleic acids (snas) with sheddable peg layers
Xu et al. Rational design and combinatorial chemistry of ionizable lipids for RNA delivery
US20130108686A1 (en) Method for the delivery of oligonucleotides
Yang et al. Synthetic conjugated Oligoelectrolytes are effective siRNA transfection carriers: relevance to pancreatic cancer gene therapy
Li et al. Dual pH-and glutathione-responsive CO2-generating nanodrug delivery system for contrast-enhanced ultrasonography and therapy of prostate cancer
Raikwar et al. Opportunities in ultrasonic drug delivery to tumor
US20090142391A1 (en) Conjugated RNAi Therapeutics
Bristow et al. Aptamer-Targeted Dendrimersomes Assembled from Azido-Modified Janus Dendrimers “Clicked” to DNA
Nishad et al. Enhancement of bioavailability of therapeutics using drug conjugation approach: an in-depth review
Liao et al. Imaging-Assisted Antisense Oligonucleotide Delivery for Tumor-Targeted Gene Therapy
An Utility of two distinct macromolecular carriers for nucleic acid delivery
Stimulated CHEMICAL AND MOLECULAR CONJUGATES
Larsen Non-viral gene delivery: An inside-out approach

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENERAL ELECTRIC COMPANY, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOHAN, PRAVEENA;SOOD, ANUP;ROTHMAN, JAMES EDWARD;AND OTHERS;SIGNING DATES FROM 20120611 TO 20120614;REEL/FRAME:028416/0462

STCB Information on status: application discontinuation

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