WO1998000172A2 - Compositions and methods for altering the biodistribution of biological agents - Google Patents

Compositions and methods for altering the biodistribution of biological agents Download PDF

Info

Publication number
WO1998000172A2
WO1998000172A2 PCT/US1997/010766 US9710766W WO9800172A2 WO 1998000172 A2 WO1998000172 A2 WO 1998000172A2 US 9710766 W US9710766 W US 9710766W WO 9800172 A2 WO9800172 A2 WO 9800172A2
Authority
WO
WIPO (PCT)
Prior art keywords
albumin
microbubbles
protein
ougonucleotide
composition
Prior art date
Application number
PCT/US1997/010766
Other languages
French (fr)
Other versions
WO1998000172A3 (en
Inventor
Thomas Porter
Patrick L. Iversen
Original Assignee
Board Of Regents Of The University Of Nebraska
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 Board Of Regents Of The University Of Nebraska filed Critical Board Of Regents Of The University Of Nebraska
Priority to JP50420598A priority Critical patent/JP4201348B2/en
Priority to AT97932228T priority patent/ATE260677T1/en
Priority to DE69727958T priority patent/DE69727958T2/en
Priority to CA002258882A priority patent/CA2258882C/en
Priority to EP97932228A priority patent/EP0938341B1/en
Publication of WO1998000172A2 publication Critical patent/WO1998000172A2/en
Publication of WO1998000172A3 publication Critical patent/WO1998000172A3/en

Links

Classifications

    • 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
    • A61K47/50Medicinal 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
    • A61K49/00Preparations for testing in vivo
    • A61K49/22Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations
    • A61K49/222Echographic preparations; Ultrasound imaging preparations ; Optoacoustic imaging preparations characterised by a special physical form, e.g. emulsions, liposomes
    • A61K49/223Microbubbles, hollow microspheres, free gas bubbles, gas microspheres
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics

Definitions

  • TITLE COMPOSITIONS AND METHODS FOR ALTERING THE "
  • This invention relates to a new and improved pharmaceutical composition and method for delivery of bioactive substances.
  • the methods and composition of the invention can be used with several agents and can achieve site specific delivery of a biologically active substances. This can allow for lower doses and for improved efficacy with drugs particularly agents such as oligonucleotides which are plagued with problems in achieving therapeutic concentrations at targeted organs.
  • Drug delivery techniques are employed in the formulation of all drug therapy to augment drug availability, to reduce drug dose, and consequently to reduce drug-induced side effects. These techniques serve to control, regulate, and target the release of drugs in the body. The goals have been to provide less frequent drug administration, to maintain constant and continuous therapeutic levels of a drug in the systemic circulation or at a specific target organ site, to achieve a reduction in undesirable side effects, and to promote a reduction in the amount and dose concentration required to realize the desired therapeutic benefit.
  • drug delivery systems have included drug carriers based upon proteins, polysaccharides, synthetic polymers, erythrocytes, DNA and liposomes.
  • New generation biologicals such as monoclonal antibodies, gene therapy vectors, anti-cancer drugs such as Taxol, viral based drugs, and oligo and poly nucleotides have presented several problems with regard to delivery. In fact drug delivery may be the primary hurdle to achieving mainstream therapeutic use of these biologies whose initial potential seemed unlimited but whose therapeutic parameters have prevented realization of full benefit.
  • Synthetic oligodeoxyribonucleotides which are chemically modified to confer nuclease resistance represent a fundamentally different approach to drug therapy.
  • the most common applications to date are antisense oligos with " sequences complementary to a specific targeted mRNA sequence.
  • An antisense oligonucleotide approach to therapy involves a remarkably simple and specific drug design concept in which the oligo causes a mechanistic intervention in the processes of 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.
  • Phosphorothioate analogs of polynucleotides have chiral internucleoside linkages in which one of the non-bridging ligands is sulfur.
  • the phosphorothioate analog is currently the most commonly employed analogue in biological studies including both in vitro and in vivo.
  • the most apparent disadvantage of phosphorothioate oligonucleotides include the high cost of preparation of sufficient amounts of high quality material and non-specific binding to proteins. Hence, the primary advantage of antisense approach (low dose and minimal side effects) fall short of expectations.
  • oligonucleotides and polynucleotides have focused on two key challenges; transfection of oligonucleotides into cells and alteration of distribution of oligonucleotides in vivo.
  • Transfection involves the enhancement of in vitro cellular uptake.
  • Biological approaches to improve uptake have included viral vectors such as reconstituted viruses and pseudo virions, and chemicals such as liposomes.
  • Methods to improve biodistribution have focused on such things as cationic lipids, which are postulated to increase cellular uptake of drugs due to the positively charged hpid attraction to the negatively charged surfaces of most cells.
  • Lipofection and DC-cholesterol liposomes have been reported to enhance gene transfer into vascular cells in vivo when administered by catheter.
  • Cationic Hpid DNA complexes have also been reported to result in effective gene transfer into mouse lungs after intratracheal administration.
  • Cationic liposomal delivery of oligonucleotides has also been accomplished however, altered distribution to the lung and liver was experienced.
  • Asialoglycoprotein poly(L)-lysine complexes have met with limited success as well as complexation with Sendai virus coat protein containing liposomes. Toxicity and biodistribution, however, have remained significant issues.
  • One object of this invention is to provide a novel composition of matter to deliver a pharmaceutical agent to a targeted site in vivo.
  • -Another object of the invention is to provide a method for delivering a pharmaceutical agent increasing drug bioavailabihty and decreasing toxicity.
  • compositions and methods can be used to deliver agents such as therapeutics or diagnostics which have been plagued with delivery problems such as oligonucleotides as well as traditional agents and can drastically reduce the effective dosages of each, increasing the therapeutic index and improving bioavailability. This in turn can reduce drug cytotoxicity and side effects.
  • the invention employs conjugation of the biologic agent with a filmogenic protein which is formed as a protein shell microbubble encapsulating an insoluble gas.
  • the composition is prepared as an aqueous suspension of a plurality of the microbubbles for parenteral administration. Conjugation of the biologic with albumin or other such protein encapsulated microbubbles can allow for targeted delivery of the biologic to alternate including those which traditionally interact with the protein. DESCRIPTION OF THE FIGURES
  • Figure 1 is a Lineweaver-Burke plot of the binding data for PESDA microbubbles with PS-ODN.
  • Ultrasonic imaging has long been used as a diagnostic tool to aid in therapeutic procedures It is based on the principle that waves of sound energy can be focused upon an area of interest and reflected to produce an image Generally an ultrasonic transducer is placed on a body surface overlying the area to be imaged and ultrasonic energy, produced by generating and receiving sound waves, is transmitted The ultrasonic energy is reflected back to the transducer where it is translated into an ultrasonic image The amount of characteristics of the reflected energy depend upon the acoustic properties of the tissues, and contrast agents which are echogenic are preferably used to create ultrasonic energy in the area of interest and improve the imaging received.
  • Contrast echocardiography has been used to delineate intracardiac structures, assess valvular competence, and demonstrate intracardiac shunts
  • MCE Myocardial contrast echocardiography
  • Ultrasonic vibration has also been used at therapeutic levels in the medical field to increase the absorption of various medicaments.
  • percutaneous absorption of a medicament is enhanced by ultrasonic vibration.
  • U.S. Patent Nos. 4,953,565 and 5,007,438 also disclose a technique of percutaneous absorption of medicaments by the aid of ultrasonic vibration.
  • U.S. Patent No. 5,315,998 discloses a booster for drug therapy comprising microbubbles in combination ultrasonic energy to allow the medicament to diffuse and penetrate. This discloses the use of therapeutic levels of ultrasound for up to 20 minutes in contrast to the invention which uses diagnostic levels of ultrasound with exposure for much shorter time periods to achieve release of conjugated bioactive agents.
  • the pharmaceutical composition of the invention comprises a liquid suspension containing microbubbles of an insoluble gas having a diameter of 0.1 to 10 microns.
  • the microbubbles are formed by entrapping microbubbles of a gas into a liquid.
  • the microbubbles are made of various insoluble gases such as fluorocarbon or sulfur hexafluoride gas.
  • the liquid includes any Uquid which can form microbubbles.
  • any insoluble gas can be used. It must be gaseous at body temperature and be nontoxic. The gas must also form stable microbubbles of average size of between about .1 and 10 microns in diameter when the pharmaceutical composition is sonicated to form microbubbles.
  • perfluorocarbon gases such as perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluoropentane are preferred.
  • perfluoropropane and perfluorobutane are especially preferred because of their demonstrated safety for intraocular injection in humans. They have been used in human studies for intraocular injections to stabilize retinal detachments (Wong and Thompson, Opthamology 95:609-613). Treatment with intraocular perfluoropropane is considered to be the standard of care for treatment of this disorder.
  • the gases must also have a diffusion coefficient and blood solubility lower than nitrogen or oxygen which diffuse once in the internal atmosphere of the blood vessel.
  • the agent of the invention is formulated in a pharmaceutically effective dosage form for peripheral administration to the host.
  • a host is a human host, although other mammalian hosts such as canine or equine can also be subject to this therapy.
  • the pharmaceutical liquid composition of the invention uses a liquid wherein the microbubbles are stabilized by a filmogenic protein coating.
  • Suitable proteins include naturally occurring proteins such as albumin, human gamma globulin, human apotransferin, Beta lactose and urease.
  • the invention preferably employs a naturally occurring protein but synthetic proteins may also be used. Preferred is human serum albumin.
  • an aqueous solution containing a mixture of a pharmaceutically accepted saccharide e.g., dextrose in combination with the earlier described protein.
  • the pharmaceutical liquid composition of the invention is the sonicated mixture of commercially available albumin (human), U.S.P. solution (generally supplied as 5% or 25% by weight sterile aqueous solutions), and commercially available dextrose, U.S.P. for intravenous administration.
  • the mixture is sonicated under ambient conditions i.e. room air temperature and pressure and is perfused with an insoluble gas (99.9% by weight) during sonication.
  • the pharmaceutical Uquid composition includes a two-fold to eight-fold dilution of 5% to 50% by weight of dextrose and a 2% to 10% by weight of human serum albumin.
  • exemplary of other saccharide solutions of the invention are aqueous monosaccharide solution (e.g. having the formula su ch as the hexose sugars, dextrose or fructose or mixtures thereof), aqueous disaccharide solution (e.g. having a formula 0 ⁇ 2 ⁇ 22 ⁇ 11 such as sucrose, lactose or maltose or mixtures thereof), or aqueous polysaccharide solution (e.g. soluble starches having the formula C(5H ⁇ ⁇ 5(n) wherein n is a whole number integer between 20 and about 200 such as amylase or dextran or mixtures thereof.
  • the microbubbles are formed by sonication, typically with a sonicating horn. Sonication by ultrasonic energy causes cavitation within the dextrose albumin solution at sites of particulate matter or gas in the fluid. These cavitation sites eventually resonate and produce smaU microbubbles (about 7 microns in size) which are non-coUapsing and stable. In general, sonication conditions which produce concentrations of greater than about 4X10 8 m of between about 5 and about 6 micron microbubbles are preferred. Generally the mixture will be sonicated for about 80 seconds, while being perfused with an insoluble gas.
  • a second method of preparation includes hand agitating 15 ⁇ 2 ml of sonicated dextrose albumin with 8 ⁇ 2 ml of perfluorocarbon gas prior to sonication. Sonication then proceeds for 80 + 5 seconds.
  • microbubble sizes are particularly ideal since a microbubble must have a mean diameter of less than 10 microns and greater than .1 to be sufficient for transpulmonary passage, and must be stable enough to prevent significant diffusion of gases within the microbubble following intravenous injection and during transit to the target site.
  • the microbubbles are next incubated with the medicament so that the medicament becomes conjugated with the microbubble.
  • filmogenic proteins in the form of microbubbles as previously used in contrast agents retain their abiUty to bind medicaments. This is surprising because traditionally it was thought that in the formation of microbubble contrast agents the protein sphere was made of denatured protein.
  • AppUcant has demonstrated that when an insoluble gas instead of air is used for the microbubble, much of the sonication energy is absorbed by the gas and the protein retains its binding activity. Air filled microbubbles do not retain their binding capabilities and cannot be used m the method of the invention.
  • the therapy involves the use of a pharmaceutical composition conjugated to a protein microbubble of a diameter of about .1 to 10 microns.
  • the invention uses agents traditionaUy used in diagnostic ultrasound imaging.
  • Therapeutic agents useful in the present invention are selected via their abihty to bind with the filmogenic protein.
  • the filmogenic protein is albumin
  • the therapeutic or diagnostic agent can include ohgonucleotides (such as antisense or antigen ohgos), polynucleotides (such as retroviral, adenoviral, plasmid vectors or probes), or ribozymes all of which can bind with albumin and as such can form a conjugation with the microbubble
  • ohgonucleotides such as antisense or antigen ohgos
  • polynucleotides such as retroviral, adenoviral, plasmid vectors or probes
  • ribozymes all of which can bind with albumin and as such can form a conjugation with the microbubble
  • a Ust of drugs which bind to albumin at site 1 (which retains its binding capacity) and thus would be useful in the methods and compositions of the present invention in the albumin embodiment
  • Nonsteroidal anti inflammatory drug represents patient-to-patient variability
  • Other drugs which bind with albumin particularly at site 1 would also be useful in this embodiment and can be ascertained by those of skill in the art through Drug Interaction and Pharmacology tests standard to those of skill m the art such as "Drug Information or "Facts and Comparisons" pubUshed by Berney OUn updated every quarter. Other such references are widely available in the are.
  • protein coated microbubbles of insoluble gas have been found to form stable conjugates with ohgonucleotides
  • the ohgo conjugated bubbles are then introduced to the animal and the protein coating directs the conjugated agent to sites of interaction Ultimately as the bubble dissipates the agent will be released at the tissue site.
  • Therapeutic sites can include such things as the location of a specific tumor, an organ which due to differential gene activation expresses a particular gene product, the site of an injury or thrombosis, a site for further processing and distribution of the therapeutic etc.
  • the target site is selected based upon the bioprocessmg of the filmogenic protein
  • the kidneys and Uver take up albumin and albumin microbubbles can be used to specifically direct the administration of conjugated bioactive agents to these areas
  • the metaboUsm and bioprocessmg of other filmogenic proteins can be easily obtained through standard pharmacologic texts such as "Basic and CUnical Pharmacology" by Bertram G. Katzung the relevant disclosure of which is incorporated by reference
  • the method preferred for practicing the deUvery therapy of the invention involves obtaining a pharmaceutical Uquid agent of the invention, introducing said agent into a host by intravenous injection, intravenously (i.v. infusion), percutaneously or intramuscularly.
  • the microbubble is then processed in the animal and is taken up and interacted with according to the filmogenic protein which coats the microbubble.
  • the bubble dissipates deUvering the bioactive at the site of processing of the protein.
  • microbubble conjugation of bioactive agents can be used in targeted deUvery protocols with deUvery of the biologic upon appUcation of ultrasound to the target site, causing cavitation of the microbubble and ultimate release of the biologic at the site in interaction with the ultrasound field.
  • appUcant has now discovered that appUcation of ultrasound is not necessary for the targeted deUvery of biologies to sites of bioprocessmg of the protein coating. The protein traffics the microbubble and conjugate to sites of processing and as the bubbles dissipate the oUgo or other biologic is released to interact with the site allowing for a fraction of the biologic to achieve the same biological effect.
  • the agent of the invention is a perfluorocarbon enhanced sonicated dextrose albumin solution comprised of a sonicated three-fold dilution of 5% human serum albumin with 5% dextrose.
  • the solution is perfused with perfluorocarbon gas for about 80 seconds which lowers the solubility and diffusivity of the microbubble gas.
  • the resulting microbubbles are concentrated at room temperature for at least about 120 ⁇ 5 minutes wherein the excess solution settles in the sonicating syringe.
  • the microbubbles are then exposed to a therapeutic agent and aUowed to interact such that the agent becomes conjugated to the microbubbles.
  • the conjugated microbubbles are transferred to a sterile syringe and injected parenteraUy into a mammal, preferably near the target site of activity of the agent.
  • Methods of ultrasonic imaging in which microbubbles formed by sonicating an aqueous protein solution are injected into a mammal to alter the acoustic properties of a predetermined area which is then ultrasonically scanned to obtain an image for use in medical procedures is well known. For example see U.S. Patent No. 4,572,203, U.S. Patent No. 4,718,433 and U.S. Patent No. 4,774,958, the contents of each of which are incorporated herein by reference.
  • contrast agents as a pharmaceutical composition as part of a targeted deUvery system that is the novel improvement of this invention.
  • the invention has been shown to drasticaUy improve the efficiency and therapeutic activity by altering biodistribution of several drugs including, most notably, anti-sense oUgonucleotides which have been traditionally plagued with ineffective pharmacologic parameters, including high clearance rate and toxicity.
  • microbubble-therapeutic agent therapy can reduce any toxic effects of persons who perhaps could not tolerate certain therapeutics at doses and concentrations necessary to achieve a beneficial result.
  • the protein substance such as human serum albumin is easily metaboUzed within the body and excreted outside and hence is not harmful to the human body. Further gas trapped within the microbubbles is extremely smaU and is easily dissolved in blood fluid, perfluoropropane and perfluorobutane have long been known to be safe in humans. Both have been used in humans for intra ocular injections to stabiUze retinal detachments. Wong and Thompson, Ophthalmology 95:609-613. Thus the anti thrombosis agents of the invention are extremely safe and nontoxic for patients.
  • the invention is particularly useful for deUvery of nucleotide sequences in the form of gene therapy vectors, or anti-sense of anti-gene type strategies to ultimately alter gene expressions in target ceUs.
  • Antisense oUgonucleotides represent potential tools in research and therapy by virtue of their ahility to specifically inhibit synthesis of target proteins. A major theoretical advantage of these ohgos is their potential specificity for binding to one site in the cell.
  • a synthetic oUgonucleotide of at least 6 nucleotides, preferably complementary to DNA (antigene) or RNA (antisense), which interferes with the process of transcription or translation of endogenous proteins is presented.
  • any of the known methods for oUgonucleotide synthesis can be used to prepare the oUgonucleotides. They are most conveniently prepared using any of the commercially available, automated nucleic acid synthesizers, such as appUed biosystems, Inc., DNA synthesizer (Model 380B). According to manufacturers protocols using phosphoroamidite chemistry. After biosystems (Foster City, CA). Phosphorothioate oUgonucleotides were synthesized and purified according to the methods described in Stek and Zahn J.
  • Modified or related nucleotides of the present invention can include one or more modifications of the nucleic acid bases, sugar moieties, internucleoside phosphate Unkages, or combinations of modifications at these sites.
  • the internucleoside phosphate Unkages can be phosphorothioate, phosphoramidate; methylphosphonate, phosphorodithioate and combinations of such similar Unkages (to produce mix backbone modified oUgonucleotides).
  • Modifications may be internal or at the end(s) of the oUgonucleotide molecule and can include additions to the molecule of the internucleoside phosphate Unkages, such as cholesterol, diamine compounds with varying numbers of carbon residues between the amino groups, and terminal ribose, deoxyriboase and phosphate modifications which cleave, or crossUnk to the opposite chains or to associated enzymes or other proteins which bind to the genome.
  • internucleoside phosphate Unkages such as cholesterol, diamine compounds with varying numbers of carbon residues between the amino groups, and terminal ribose, deoxyriboase and phosphate modifications which cleave, or crossUnk to the opposite chains or to associated enzymes or other proteins which bind to the genome.
  • the modification is a phosphorothioate oUgonucleotide.
  • the following examples are for illustration purposes only and are not intended to Umit this invention in any way. It will be appreciated by those of skill in the art, that numerous other protein-bioactive agent combinations can be used in the invention and are even contemplated herein.
  • the filmogenic protein is transferrin
  • the bioactive agent could be any transferrin binding pharmacologic.
  • Chain extension syntheses were performed on a 1 ⁇ mole column support on an ABI Model 391 DNA synthesizer (Perkin Elmer, Foster City, CA) or provided by Lynx Therapeutics, Inc. (Hayward CA).
  • the 1 micromole synthesis employed cyanoethyl phosphoroamidites and sulfurization with tetraethylthiuram disulfide as per ABI user BuUetin 58.
  • Radiolabeled ohgonucleotides were synthesized as hydrogen phosphonate material by Glen Research (Bethesda, MD).
  • the uniformly ⁇ S- labeled PS-ODN with sequences 5'-TAT GCT GTG CCG GGG TCT TCG GGC 3' (24-mer complementary to c-myb) (SEQ ID NO:2) and 5* TTAGGG 3' (SEQ ID NO:3) were incubated in a final volume of 0.5 ml with the perfluorocarbon- exposed sonicated dextrose albumin microbubble solution for 30 minutes at 37 °C The solutions were aUowed to stand so that the bubbles could rise to the top and 100 microhters were removed from the clear solution at the bottom and 100 microhters were removed from the top containing the microbubbles
  • Radioactive 24-mer PS-ODN was added to a washed solution of PESDA and room air sonicated dextrose albumin (RA-SDA) microbubbles at a concentration of 5nM
  • Non-radioactive PS-ODN 20-mer was added to tubes containing radioactive 24-mer in a series of increasing concentrations (0, 3 3, 10, 32.7, 94 5, 167, and 626 ⁇ )
  • the suspension of bubbles is mixed by inversion and incubated at 37°C for 60 minutes
  • Measurement of Radioactivity Radioactivity in solutions were determined by Uquid scintillation counting in a Uquid scintillation counter (model LSC7500, Beckman Instruments GmbH, Kunststoff, Germany).
  • the sample volume was lOO ⁇ l to which 5 ml of Hydrocount biodegradable scintillation cocktail was added and mixed. Samples were counted immediately after each experiment and then again 24 hours later in order to reduce the influence of chemfluminescence and of quenching. Flow cytometry
  • the uniformity of room air versus perfluorocarbon-containing sonicated dextrose albumin microbubble binding of PS-ODN was determined by flow cytometry. A solution of microbubbles was washed in a 1000 fold excess volume of sterile saline.
  • Group A control in which 100 ⁇ l of microbubbles were added to a 900 ⁇ L of saUne
  • Group B in which 100 ⁇ 1 of microbubbles were added to 900 ⁇ L of saUne and 2 ⁇ L of FITC-labeled 20-mer was added (final 20-mer concentration is 151 nM)
  • group C in which 100 ⁇ L of microbubbles were added to 800 ⁇ L of saUne, 2 ⁇ L of FITC-labeled 20-mer and 100 ⁇ L of unlabeled 20-mer(final concentration is 151nM).
  • the incubations were all conducted for 20 minutes at room temperature.
  • Washed microbubble suspensions were diluted in sterile saUne (Baxter) and then incubated with FITC-labeled PS-ODN.
  • Flow cytometric analysis was performed using a FACStar Plus (Becton Dickinson) equipped with t 100 mW air-cooled argon laser and the Lysis II acquisition and analysis software.
  • List mode data were employed for a minimum of 10 ⁇ coUected microbubbles and independent analysis a for each sample.
  • Study Protocol A variable flow microsphere scanning chamber was developed for the study which is similar to that we have described previously Mor-Avi V., et al "StabiUty of Albunex microspheres under ultrasonic irradiation; and in vitro study.
  • This system consists of a circular scanning chamber connected to a Masterflex flow system (Microgon, Inc., Madison Hills California) The scanning chamber was enclosed on each side by water-filled chambers and bound on each side by acousticaUy transparent material The PS-ODN-labeled PESDA microbubbles (0 1 milliUters) were injected as a bolus over one second proximal to the scanning chamber which then flowed through plastic tubing into a tap water-filled scanning chamber at a controlled flow rate of 100 ml/mm.
  • the scanner (2.0 Megahertz) frequency, 1.2 Megapascals peak negative pressure) was set to either deUver ultrasound at a conventional 30 Hertz frame rate or was shut off FoUowmg passage through the scanning chamber, the solution was then passed through the same size plastic tubing into a graduated cyUnder The first 10 milliliters was discarded Following this, the next 10 milliliters was aUowed to enter into a coUection tube The coUection tube containing the effluent microbubbles was aUowed to stand in order to separate microbubbles on the top from whatever free oUgonucleotide existed in the lower portion of the sample.
  • the observed binding data are presented as a Lineweaver Burke plot in Figure 1.
  • the equiUbrium dissociation constant K m (calculated for the 7 concentrations which were run in duphcate) for the binding to the microbubbles was 1.76 X 10" 5 M.
  • albumin binding characteristics particularly as they relate to oUgonucleotides see Kumar, Shashi et al "Characterization of Binding Sites, Extent of Binding, and Drug Interactions of OUgonucleotides with Albumin” Antisense Research and Development 5: 131-139 (1995) the disclosure of which is hereby incorporated ⁇ by reference.
  • PS-ODN binds to the albumin in PESDA microbubbles, indicating that the binding site 1 on albumin is biologicaUy active foUowing production of these bubbles by electromechanical sonication. This binding site affinity is lost when the electromechanical sonication is performed only with room sir. Further, removal of albumin not associated with PESDA microbubbles by washing shows a significant partitioning of the PS-ODNs with the microbubbles (Table 1).
  • antisense phosphorothioate oUgonucleotides were designed to the cytochrome P450 IIB 1 gene sequence to alter the metaboUsm of Phenobarbital.
  • the oUgonucleotides were conjugated to perfluoropropane exposed sonicated dextrose albumin microbubbles (PESDA) as earUer described and deUvered to rats intravenously.
  • PESDA perfluoropropane exposed sonicated dextrose albumin microbubbles
  • the oUgonucleotide was synthesized according to the rat cytochrome P450 IIB 1 known sequence and had the foUowing sequence: GGAGCAAGATACTGGGCTCCAT (SEQ ID NO:5) AAAGAAGAGAGAGCAGGGAG (SEQ ID NO:6) Male Sprague-Dawley rats (Sasco, Omaha), were used and weighed between 210 to 290 grams for aU studies. They were housed in animal quarters at the University of Kansas Medical Center, AAALAC approved animal resource facility. The animals were exposed to 12 hour Ught/dark cycle " and aUowed access to Purina rat chow and tap water ad Ubitum.
  • Rats in groups with PB were injected intraperitoneaUy with phenobarbital (MaJlinckrodt, St. Louis) at 80 ml kg/day x 2 days.
  • the PB injections were given simultaneously with the ODN-microbubble injections.
  • Phosphorothioate ODN injections were 1 ml/kg/day x 2 days. Sleep times were measured 48 hours after the first injection.
  • the rats were injected intraperitoneaUy with 100 ml/kg hexobarbital (Sigma, St. Louis), paired fresh daily. The volume of this injection is 1 ml/kg body weight.
  • Each rat was injected with 100 mg/kg of hexobarbital intraperitoneaUy.
  • Livers were perfused with 12 ml of 4% saUne via the portal vein and then removed from the animal.
  • the Uvers were minced, homogenized in .25 M sucrose (Sigma) and centrifuged at 8000 x g for 20 minutes at 4°C in a Sorvall RC2-B centrifuge (Dupont, Wilmington, DE). The supernatant was saved and resuspended in a .25 M sucrose and centrifuged at 100,000 x g for 45 minutes at 4°C in a SorvaU OTD55B ultracentrifuge (Dupont).
  • the peUet was resuspended in 1.15% KCL (Sigma) and centrifuged at 100,000 x g for 1 hour at 4°C with the final peUet resuspended in an equal volume buffer (10 mM Tns-acetate, 1 mM EDTA, 20% glycerol; Sigma) and frozen at -80°C.
  • Protein concentrations were determined by Bradford assay (Bradford, 1976) 80 ⁇ l ahquots of homogenate were added to a 96 weU plate (Becton, Dickinson Labware, Lincoln Park, NJ) 20 ⁇ l of Bradford reagent (Bio-Rad Richmond, CA) was then added and the plates read at 595 nm on the microplate reader (Molecular Devices, Newport MN) The data was compared to standard curve generated with known concentrations of bovine serum albumin (Sigma)
  • CYP IIB 1 content was determined by pentoxyresorufin 0-dealkylation (PROD) activity (Burke et al 1985)
  • PROD pentoxyresorufin 0-dealkylation
  • 1 mg protein in 1 ml .1 M potassium phosphate buffer, 1 ml 2 ⁇ M 5- pentoxyresorufin (Pierce, Rockford, IL), and 17 ⁇ l 60 mM NADPH were mixed and incubated for 10 minutes at 37°C The mixture was then added to a 2 ml cuvette and read on a RF5000U spectrofluorophotometer (Shimadzu, Columbia, MD) using an excitation wavelength of 530 nm and emission wavelength of 585 nm Concentrations of unknowns were calculated from a standard curve of resorufin (Pierce, Rockford, IL) standards Results were recorded in nmol resorufin/mg protem/mm
  • CYP IIB1 protein Direct measurement of CYP IIB1 protein was determined by an ELISA assay using an antibody directed the CYP IIB l protein (Schuurs and Van Weeman, 1977) 50 ⁇ g of Uver per weU were plated in 100 ⁇ l 35% sodium bicarbonate buffer overnight on a 96 weU nunc-immuno plate (InterMed, Skokie, IL) The microsomes were washed 3x with 1% bovine serum albumin in PBS (PBS/BSA) and incubated for 1 hr at 37°C with 200 ⁇ l PBS/BSA The PBS/BSA was removed and 50 ⁇ l of CYP IIB l antibody (Oxygene, DaUas) was added and incubated for 1 hour at 37°C The microsomes were washed 5x with sahne/tween 20 (Sigma) and had 50 ⁇ l horseradish peroxidase antibody (Bio- rad) added The microsomes were incuba
  • 100 mg/kg HB was injected i.p. to each animal at the end of 2 days of treatment with PB and/or the ODNs.
  • Control rats had a sleep time of about 23 minutes.
  • PB had a significant reduction in sleep time to about 11.414.5 minutes.
  • PB stimulates CYP IIB l mRNA, as a result, hexobarbital which is hydroxylated by CYP IIBl is more quickly metaboUzed l o and its sedative effect reduced.
  • ADDRESSEE Zarley, McKee, Thomte, Voorhees & Sease
  • B STREET: 801 Grand Suite 3200

Abstract

The invention relates to a new and improved pharmaceutical composition and method for delivery of therapeutic agents. The methods and composition of the invention can be used with several therapeutic agents and can achieve site specific delivery of a therapeutic or diagnostic substance. This can allow for lower doses and for improved efficacy with drugs which traditionally reach targeted sites and can result in improved utility for agents such as oligonucleotides and polynucleotides which are plagued with problems with biodistribution.

Description

TITLE: COMPOSITIONS AND METHODS FOR ALTERING THE "
BIODISTRIBUTION OF BIOLOGICAL AGENTS
FIELD OF THE INVENTION This invention relates to a new and improved pharmaceutical composition and method for delivery of bioactive substances. The methods and composition of the invention can be used with several agents and can achieve site specific delivery of a biologically active substances. This can allow for lower doses and for improved efficacy with drugs particularly agents such as oligonucleotides which are plagued with problems in achieving therapeutic concentrations at targeted organs.
BACKGROUND OF THE INVENTION
Drug delivery techniques are employed in the formulation of all drug therapy to augment drug availability, to reduce drug dose, and consequently to reduce drug-induced side effects. These techniques serve to control, regulate, and target the release of drugs in the body. The goals have been to provide less frequent drug administration, to maintain constant and continuous therapeutic levels of a drug in the systemic circulation or at a specific target organ site, to achieve a reduction in undesirable side effects, and to promote a reduction in the amount and dose concentration required to realize the desired therapeutic benefit.
To date, drug delivery systems have included drug carriers based upon proteins, polysaccharides, synthetic polymers, erythrocytes, DNA and liposomes. New generation biologicals such as monoclonal antibodies, gene therapy vectors, anti-cancer drugs such as Taxol, viral based drugs, and oligo and poly nucleotides have presented several problems with regard to delivery. In fact drug delivery may be the primary hurdle to achieving mainstream therapeutic use of these biologies whose initial potential seemed unlimited but whose therapeutic parameters have prevented realization of full benefit.
Synthetic oligodeoxyribonucleotides which are chemically modified to confer nuclease resistance represent a fundamentally different approach to drug therapy. The most common applications to date are antisense oligos with" sequences complementary to a specific targeted mRNA sequence. An antisense oligonucleotide approach to therapy involves a remarkably simple and specific drug design concept in which the oligo causes a mechanistic intervention in the processes of 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.
Phosphorothioate analogs of polynucleotides have chiral internucleoside linkages in which one of the non-bridging ligands is sulfur. The phosphorothioate analog is currently the most commonly employed analogue in biological studies including both in vitro and in vivo. The most apparent disadvantage of phosphorothioate oligonucleotides include the high cost of preparation of sufficient amounts of high quality material and non-specific binding to proteins. Hence, the primary advantage of antisense approach (low dose and minimal side effects) fall short of expectations.
Drug delivery efforts with regard to oligonucleotides and polynucleotides have focused on two key challenges; transfection of oligonucleotides into cells and alteration of distribution of oligonucleotides in vivo.
Transfection involves the enhancement of in vitro cellular uptake. Biological approaches to improve uptake have included viral vectors such as reconstituted viruses and pseudo virions, and chemicals such as liposomes. Methods to improve biodistribution have focused on such things as cationic lipids, which are postulated to increase cellular uptake of drugs due to the positively charged hpid attraction to the negatively charged surfaces of most cells.
Lipofection and DC-cholesterol liposomes have been reported to enhance gene transfer into vascular cells in vivo when administered by catheter. Cationic Hpid DNA complexes have also been reported to result in effective gene transfer into mouse lungs after intratracheal administration. Cationic liposomal delivery of oligonucleotides has also been accomplished however, altered distribution to the lung and liver was experienced. Asialoglycoprotein poly(L)-lysine complexes have met with limited success as well as complexation with Sendai virus coat protein containing liposomes. Toxicity and biodistribution, however, have remained significant issues.
From the foregoing it can be seen that a targeted drug delivery system for delivery of biologies, particularly poly and oligo nucleotides is needed for these drugs to achieve their fullest potential. One object of this invention is to provide a novel composition of matter to deliver a pharmaceutical agent to a targeted site in vivo.
-Another object of the invention is to provide a method for delivering a pharmaceutical agent increasing drug bioavailabihty and decreasing toxicity.
Other objects of the inventions will become apparent from the description of the invention which follows.
SUMMARY OF THE INVENTION
According to the invention a new biologically active agent delivery method and composition are disclosed. The compositions and methods can be used to deliver agents such as therapeutics or diagnostics which have been plagued with delivery problems such as oligonucleotides as well as traditional agents and can drastically reduce the effective dosages of each, increasing the therapeutic index and improving bioavailability. This in turn can reduce drug cytotoxicity and side effects. The invention employs conjugation of the biologic agent with a filmogenic protein which is formed as a protein shell microbubble encapsulating an insoluble gas. The composition is prepared as an aqueous suspension of a plurality of the microbubbles for parenteral administration. Conjugation of the biologic with albumin or other such protein encapsulated microbubbles can allow for targeted delivery of the biologic to alternate including those which traditionally interact with the protein. DESCRIPTION OF THE FIGURES
Figure 1 is a Lineweaver-Burke plot of the binding data for PESDA microbubbles with PS-ODN. The equilibrium dissociation constant Km (calculated for the 7 concentrations which were run in duplicate) for the binding to the microbubbles was 1 76 X 10"5M (r2 = 0.999, Y-int = 0 0566, 7 concentrations) This is nearly within the range observed for binding a 15mer PS-ODN with sequence δ'dCAACGTTGAGGGGCAT)^' (SEQ ID NO 1) to human serum albumin in solution of 3 7 - 4 8 X 10-5M previously reported Snmvasan SK et al, "Characterization of binding sites, extent of binding, and drug interactions of oligonucleotides with albumin A tisen Res Dev 5 131, 1995
DETAILED DESCRIPTION OF THE INVENTION Ultrasonic imaging has long been used as a diagnostic tool to aid in therapeutic procedures It is based on the principle that waves of sound energy can be focused upon an area of interest and reflected to produce an image Generally an ultrasonic transducer is placed on a body surface overlying the area to be imaged and ultrasonic energy, produced by generating and receiving sound waves, is transmitted The ultrasonic energy is reflected back to the transducer where it is translated into an ultrasonic image The amount of characteristics of the reflected energy depend upon the acoustic properties of the tissues, and contrast agents which are echogenic are preferably used to create ultrasonic energy in the area of interest and improve the imaging received. For a discussion of contrast echographic instrumentation, see, DeJong and, "Acoustic Properties of Ultrasound Contrast Agents", CIP-GEGEVENS KONINKLIJKE BIBLIOTHEEK, DENHAG (1993), pp. 120 et seq.
Contrast echocardiography has been used to delineate intracardiac structures, assess valvular competence, and demonstrate intracardiac shunts
Myocardial contrast echocardiography (MCE) has been used to measure coronary blood flow reserve in humans MCE has been found to be a safe and useful technique for evaluating relative changes in myocardial perfusion and delineating areas at risk.
Ultrasonic vibration has also been used at therapeutic levels in the medical field to increase the absorption of various medicaments. For example in Japanese Patent Kokai number 115591/1977 discloses that percutaneous absorption of a medicament is enhanced by ultrasonic vibration. U.S. Patent Nos. 4,953,565 and 5,007,438 also disclose a technique of percutaneous absorption of medicaments by the aid of ultrasonic vibration. U.S. Patent No. 5,315,998 discloses a booster for drug therapy comprising microbubbles in combination ultrasonic energy to allow the medicament to diffuse and penetrate. This discloses the use of therapeutic levels of ultrasound for up to 20 minutes in contrast to the invention which uses diagnostic levels of ultrasound with exposure for much shorter time periods to achieve release of conjugated bioactive agents. Applicant has demonstrated that traditional diagnostic ultrasound therapy contrast agents can be used as a specific targeted delivery device to release therapeutic agents at the specifically designated sites of interest thereby altering drug distribution. Surprisingly, this objective can be accomplished with the contrast agent alone and without the use of any diagnostic ultrasound.
The pharmaceutical composition of the invention comprises a liquid suspension containing microbubbles of an insoluble gas having a diameter of 0.1 to 10 microns. The microbubbles are formed by entrapping microbubbles of a gas into a liquid. The microbubbles are made of various insoluble gases such as fluorocarbon or sulfur hexafluoride gas. The liquid includes any Uquid which can form microbubbles. Generally any insoluble gas can be used. It must be gaseous at body temperature and be nontoxic. The gas must also form stable microbubbles of average size of between about .1 and 10 microns in diameter when the pharmaceutical composition is sonicated to form microbubbles. Generally perfluorocarbon gases such as perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, perfluoropentane are preferred. Of these gases, perfluoropropane and perfluorobutane are especially preferred because of their demonstrated safety for intraocular injection in humans. They have been used in human studies for intraocular injections to stabilize retinal detachments (Wong and Thompson, Opthamology 95:609-613). Treatment with intraocular perfluoropropane is considered to be the standard of care for treatment of this disorder. The gases must also have a diffusion coefficient and blood solubility lower than nitrogen or oxygen which diffuse once in the internal atmosphere of the blood vessel.
Other inert gases such as sulfur hexafluoride are also useful in the invention provided they have a diffusion coefficient and blood solubility lower than nitrogen or oxygen. The agent of the invention is formulated in a pharmaceutically effective dosage form for peripheral administration to the host. Generally such host is a human host, although other mammalian hosts such as canine or equine can also be subject to this therapy. The pharmaceutical liquid composition of the invention uses a liquid wherein the microbubbles are stabilized by a filmogenic protein coating. Suitable proteins include naturally occurring proteins such as albumin, human gamma globulin, human apotransferin, Beta lactose and urease. The invention preferably employs a naturally occurring protein but synthetic proteins may also be used. Preferred is human serum albumin.
It is also optional to use an aqueous solution containing a mixture of a pharmaceutically accepted saccharide e.g., dextrose, in combination with the earlier described protein. In a preferred embodiment the pharmaceutical liquid composition of the invention is the sonicated mixture of commercially available albumin (human), U.S.P. solution (generally supplied as 5% or 25% by weight sterile aqueous solutions), and commercially available dextrose, U.S.P. for intravenous administration. The mixture is sonicated under ambient conditions i.e. room air temperature and pressure and is perfused with an insoluble gas (99.9% by weight) during sonication. In a most preferred embodiment the pharmaceutical Uquid composition includes a two-fold to eight-fold dilution of 5% to 50% by weight of dextrose and a 2% to 10% by weight of human serum albumin. Exemplary of other saccharide solutions of the invention are aqueous monosaccharide solution (e.g. having the formula
Figure imgf000009_0001
such as the hexose sugars, dextrose or fructose or mixtures thereof), aqueous disaccharide solution (e.g. having a formula 0^2^22^11 such as sucrose, lactose or maltose or mixtures thereof), or aqueous polysaccharide solution (e.g. soluble starches having the formula C(5Hι øθ5(n) wherein n is a whole number integer between 20 and about 200 such as amylase or dextran or mixtures thereof.
The microbubbles are formed by sonication, typically with a sonicating horn. Sonication by ultrasonic energy causes cavitation within the dextrose albumin solution at sites of particulate matter or gas in the fluid. These cavitation sites eventually resonate and produce smaU microbubbles (about 7 microns in size) which are non-coUapsing and stable. In general, sonication conditions which produce concentrations of greater than about 4X108m of between about 5 and about 6 micron microbubbles are preferred. Generally the mixture will be sonicated for about 80 seconds, while being perfused with an insoluble gas.
A second method of preparation includes hand agitating 15 ± 2 ml of sonicated dextrose albumin with 8 ± 2 ml of perfluorocarbon gas prior to sonication. Sonication then proceeds for 80 + 5 seconds.
These microbubble sizes are particularly ideal since a microbubble must have a mean diameter of less than 10 microns and greater than .1 to be sufficient for transpulmonary passage, and must be stable enough to prevent significant diffusion of gases within the microbubble following intravenous injection and during transit to the target site. The microbubbles are next incubated with the medicament so that the medicament becomes conjugated with the microbubble. Quite unexpectedly it was demonstrated that filmogenic proteins in the form of microbubbles as previously used in contrast agents retain their abiUty to bind medicaments. This is surprising because traditionally it was thought that in the formation of microbubble contrast agents the protein sphere was made of denatured protein. AppUcant has demonstrated that when an insoluble gas instead of air is used for the microbubble, much of the sonication energy is absorbed by the gas and the protein retains its binding activity. Air filled microbubbles do not retain their binding capabilities and cannot be used m the method of the invention.
The therapy involves the use of a pharmaceutical composition conjugated to a protein microbubble of a diameter of about .1 to 10 microns. The invention uses agents traditionaUy used in diagnostic ultrasound imaging.
Therapeutic agents useful in the present invention are selected via their abihty to bind with the filmogenic protein. For example if the filmogenic protein is albumin, the therapeutic or diagnostic agent can include ohgonucleotides (such as antisense or antigen ohgos), polynucleotides (such as retroviral, adenoviral, plasmid vectors or probes), or ribozymes all of which can bind with albumin and as such can form a conjugation with the microbubble A Ust of drugs which bind to albumin at site 1 (which retains its binding capacity) and thus would be useful in the methods and compositions of the present invention in the albumin embodiment foUows
Drug % Albumin Binding Drug Class
Naproxen 99.7 NSAID®
Piroxicam 99 3 NSAID®
Warfarin 99 0 Anticoagulant
Furosemide 98.8 Loop diuretic
Phenylbutazone 96 1 NSAID©
Valproic Acid 93.0 Antiepileptic
Sulfϊsoxazole 91 4 Sufonimide Antibiotic
Ceftriaxone 90-95* Third Generation cephalosponn antibiotic
Miconazole 90.7-93.1* Antrfungal
Phenytom 89.0 Antiepileptic
ΘNonsteroidal anti inflammatory drug *Represents patient-to-patient variability Other drugs which bind with albumin particularly at site 1 would also be useful in this embodiment and can be ascertained by those of skill in the art through Drug Interaction and Pharmacology tests standard to those of skill m the art such as "Drug Information or "Facts and Comparisons" pubUshed by Berney OUn updated every quarter. Other such references are widely available in the are. Assays for determination of appropriate protein- therapeutic combinations are disclosed herein and can be sued to test any combination for its ability to work with the method of the invention According to a preferred embodiment of the invention, protein coated microbubbles of insoluble gas have been found to form stable conjugates with ohgonucleotides The ohgo conjugated bubbles are then introduced to the animal and the protein coating directs the conjugated agent to sites of interaction Ultimately as the bubble dissipates the agent will be released at the tissue site.
This is of particular relevance to oUgonucleotide and polynucleotide therapy as the primary hurdle to effective anti-sense, anti-gene, or even gene therapy employing viral or plasmid nucleotide deUvery is the abύity of the therapeutic to reach the target site at high enough concentrations to achieve a therapeutic effect Therapeutic sites can include such things as the location of a specific tumor, an organ which due to differential gene activation expresses a particular gene product, the site of an injury or thrombosis, a site for further processing and distribution of the therapeutic etc. Generally the target site is selected based upon the bioprocessmg of the filmogenic protein For example the kidneys and Uver take up albumin and albumin microbubbles can be used to specifically direct the administration of conjugated bioactive agents to these areas The metaboUsm and bioprocessmg of other filmogenic proteins can be easily obtained through standard pharmacologic texts such as "Basic and CUnical Pharmacology" by Bertram G. Katzung the relevant disclosure of which is incorporated by reference The method preferred for practicing the deUvery therapy of the invention involves obtaining a pharmaceutical Uquid agent of the invention, introducing said agent into a host by intravenous injection, intravenously (i.v. infusion), percutaneously or intramuscularly. The microbubble is then processed in the animal and is taken up and interacted with according to the filmogenic protein which coats the microbubble. Ultimately the bubble dissipates deUvering the bioactive at the site of processing of the protein.
It has been previously shown by appUcants that microbubble conjugation of bioactive agents can be used in targeted deUvery protocols with deUvery of the biologic upon appUcation of ultrasound to the target site, causing cavitation of the microbubble and ultimate release of the biologic at the site in interaction with the ultrasound field. Quite unexpectedly, appUcant has now discovered that appUcation of ultrasound is not necessary for the targeted deUvery of biologies to sites of bioprocessmg of the protein coating. The protein traffics the microbubble and conjugate to sites of processing and as the bubbles dissipate the oUgo or other biologic is released to interact with the site allowing for a fraction of the biologic to achieve the same biological effect.
In a preferred embodiment the agent of the invention is a perfluorocarbon enhanced sonicated dextrose albumin solution comprised of a sonicated three-fold dilution of 5% human serum albumin with 5% dextrose. During sonication, the solution is perfused with perfluorocarbon gas for about 80 seconds which lowers the solubility and diffusivity of the microbubble gas. The resulting microbubbles are concentrated at room temperature for at least about 120 ± 5 minutes wherein the excess solution settles in the sonicating syringe. The microbubbles are then exposed to a therapeutic agent and aUowed to interact such that the agent becomes conjugated to the microbubbles. Next the conjugated microbubbles are transferred to a sterile syringe and injected parenteraUy into a mammal, preferably near the target site of activity of the agent. Methods of ultrasonic imaging in which microbubbles formed by sonicating an aqueous protein solution are injected into a mammal to alter the acoustic properties of a predetermined area which is then ultrasonically scanned to obtain an image for use in medical procedures is well known. For example see U.S. Patent No. 4,572,203, U.S. Patent No. 4,718,433 and U.S. Patent No. 4,774,958, the contents of each of which are incorporated herein by reference.
It is the use of these types of contrast agents as a pharmaceutical composition as part of a targeted deUvery system that is the novel improvement of this invention.
The invention has been shown to drasticaUy improve the efficiency and therapeutic activity by altering biodistribution of several drugs including, most notably, anti-sense oUgonucleotides which have been traditionally plagued with ineffective pharmacologic parameters, including high clearance rate and toxicity.
This is particularly significant as the microbubble-therapeutic agent therapy can reduce any toxic effects of persons who perhaps could not tolerate certain therapeutics at doses and concentrations necessary to achieve a beneficial result. The protein substance such as human serum albumin is easily metaboUzed within the body and excreted outside and hence is not harmful to the human body. Further gas trapped within the microbubbles is extremely smaU and is easily dissolved in blood fluid, perfluoropropane and perfluorobutane have long been known to be safe in humans. Both have been used in humans for intra ocular injections to stabiUze retinal detachments. Wong and Thompson, Ophthalmology 95:609-613. Thus the anti thrombosis agents of the invention are extremely safe and nontoxic for patients.
The invention is particularly useful for deUvery of nucleotide sequences in the form of gene therapy vectors, or anti-sense of anti-gene type strategies to ultimately alter gene expressions in target ceUs. Antisense oUgonucleotides represent potential tools in research and therapy by virtue of their ahility to specifically inhibit synthesis of target proteins. A major theoretical advantage of these ohgos is their potential specificity for binding to one site in the cell. According to one embodiment of the invention a synthetic oUgonucleotide of at least 6 nucleotides, preferably complementary to DNA (antigene) or RNA (antisense), which interferes with the process of transcription or translation of endogenous proteins is presented.
Any of the known methods for oUgonucleotide synthesis can be used to prepare the oUgonucleotides. They are most conveniently prepared using any of the commercially available, automated nucleic acid synthesizers, such as appUed biosystems, Inc., DNA synthesizer (Model 380B). According to manufacturers protocols using phosphoroamidite chemistry. After biosystems (Foster City, CA). Phosphorothioate oUgonucleotides were synthesized and purified according to the methods described in Stek and Zahn J. Chromatography, 326:263-280 and in AppUed Biosystems, DNA Synthesizer, User BuUetin, Models 380A-380B-381A-391-EP, December 1989. The oUgo is introduced to cells by methods which are known to those of skill in the art. See Iverson, et al., "Anti-Cancer Drug Design", 1991, 6531-6538, incorporated herein by reference. Traditional Umitations of oUgonucleotide therapy have been preparation of the oUgonucleotide analogue which is substantially resistant to the endo and exonucleases found in the blood and cells of the body. While unmodified oUgos have been shown to be effective, several modifications to these oUgos has helped alleviate this problem. Modified or related nucleotides of the present invention can include one or more modifications of the nucleic acid bases, sugar moieties, internucleoside phosphate Unkages, or combinations of modifications at these sites. The internucleoside phosphate Unkages can be phosphorothioate, phosphoramidate; methylphosphonate, phosphorodithioate and combinations of such similar Unkages (to produce mix backbone modified oUgonucleotides).
Modifications may be internal or at the end(s) of the oUgonucleotide molecule and can include additions to the molecule of the internucleoside phosphate Unkages, such as cholesterol, diamine compounds with varying numbers of carbon residues between the amino groups, and terminal ribose, deoxyriboase and phosphate modifications which cleave, or crossUnk to the opposite chains or to associated enzymes or other proteins which bind to the genome.
These modifications traditionally help shield the oUgo from enzymatic degradation within the cell. Any of the above modifications can be used with the method of the invention. However, in preferred embodiment the modification is a phosphorothioate oUgonucleotide. The following examples are for illustration purposes only and are not intended to Umit this invention in any way. It will be appreciated by those of skill in the art, that numerous other protein-bioactive agent combinations can be used in the invention and are even contemplated herein. For example, if the filmogenic protein is transferrin, the bioactive agent could be any transferrin binding pharmacologic.
In aU the foUowing examples, all parts and percentages are by weight unless otherwise mentioned, all dilutions are by volume.
EXAMPLE 1 Phosphorothioate oligonucleotide synthesis
Chain extension syntheses were performed on a 1 μmole column support on an ABI Model 391 DNA synthesizer (Perkin Elmer, Foster City, CA) or provided by Lynx Therapeutics, Inc. (Hayward CA). The 1 micromole synthesis employed cyanoethyl phosphoroamidites and sulfurization with tetraethylthiuram disulfide as per ABI user BuUetin 58.
Radiolabeled ohgonucleotides were synthesized as hydrogen phosphonate material by Glen Research (Bethesda, MD). The uniformly ^S- labeled PS-ODN with sequences 5'-TAT GCT GTG CCG GGG TCT TCG GGC 3' (24-mer complementary to c-myb) (SEQ ID NO:2) and 5* TTAGGG 3' (SEQ ID NO:3) were incubated in a final volume of 0.5 ml with the perfluorocarbon- exposed sonicated dextrose albumin microbubble solution for 30 minutes at 37 °C The solutions were aUowed to stand so that the bubbles could rise to the top and 100 microhters were removed from the clear solution at the bottom and 100 microhters were removed from the top containing the microbubbles
Preparation of Microbubble Agent
Five percent human serum albumin and five percent dextrose were obtained from a commercial source Three parts of 5% dextrose and one part 5% human serum albumin (total 16 milbliters) were drawn into a 35-mιLuUter Monojet syringe Each dextrose albumin sample was hand agitated with 8±2 milUhters of either a fluorocarbon gas (decafluorobutane, molecular weight 238 grams/mole) or 8±2 milUhters of room air, and the sample was then exposed to electromechanical sonication at 20 kilohertz for 80±5 seconds The mean size of four consecutive samples of the perfluorocarbon-exposed sonicated dextrose albumin (PESDA) microbubbles produced in this manner, as measured with hemocytometry was 4 6±0 4 microns, and mean concentration, as measured by a Coulter counter was 1 4x10^ bubbles/milliUter The solution of microbubbles was then washed in a 1000 times volume excess of 5% dextrose to remove albumin which was not associated with the microbubbles The microbubbles were allowed four hours to rise The lower solution was then removed leaving the washed foam The washed foam was then mixed with 0 9% sodium chloride Binding Assays
The radioactive 24-mer PS-ODN was added to a washed solution of PESDA and room air sonicated dextrose albumin (RA-SDA) microbubbles at a concentration of 5nM Non-radioactive PS-ODN 20-mer was added to tubes containing radioactive 24-mer in a series of increasing concentrations (0, 3 3, 10, 32.7, 94 5, 167, and 626 μ ) The suspension of bubbles is mixed by inversion and incubated at 37°C for 60 minutes Measurement of Radioactivity Radioactivity in solutions were determined by Uquid scintillation counting in a Uquid scintillation counter (model LSC7500, Beckman Instruments GmbH, Munich, Germany). The sample volume was lOOμl to which 5 ml of Hydrocount biodegradable scintillation cocktail was added and mixed. Samples were counted immediately after each experiment and then again 24 hours later in order to reduce the influence of chemfluminescence and of quenching. Flow cytometry
The uniformity of room air versus perfluorocarbon-containing sonicated dextrose albumin microbubble binding of PS-ODN was determined by flow cytometry. A solution of microbubbles was washed in a 1000 fold excess volume of sterile saline. Three groups of samples were prepared in triplicate as follows; Group A (control) in which 100 μl of microbubbles were added to a 900 μL of saUne, Group B in which 100 μ1 of microbubbles were added to 900μ L of saUne and 2μL of FITC-labeled 20-mer was added (final 20-mer concentration is 151 nM), and group C in which 100 μL of microbubbles were added to 800 μL of saUne, 2μL of FITC-labeled 20-mer and 100 μL of unlabeled 20-mer(final concentration is 151nM). The incubations were all conducted for 20 minutes at room temperature.
Washed microbubble suspensions were diluted in sterile saUne (Baxter) and then incubated with FITC-labeled PS-ODN. Flow cytometric analysis was performed using a FACStar Plus (Becton Dickinson) equipped with t 100 mW air-cooled argon laser and the Lysis II acquisition and analysis software. List mode data were employed for a minimum of 10^ coUected microbubbles and independent analysis a for each sample. Study Protocol A variable flow microsphere scanning chamber was developed for the study which is similar to that we have described previously Mor-Avi V., et al "StabiUty of Albunex microspheres under ultrasonic irradiation; and in vitro study. J Am Soc Echocardiology 7:S29, 1994. This system consists of a circular scanning chamber connected to a Masterflex flow system (Microgon, Inc., Laguna Hills California) The scanning chamber was enclosed on each side by water-filled chambers and bound on each side by acousticaUy transparent material The PS-ODN-labeled PESDA microbubbles (0 1 milliUters) were injected as a bolus over one second proximal to the scanning chamber which then flowed through plastic tubing into a tap water-filled scanning chamber at a controlled flow rate of 100 ml/mm. As the bubbles passed through the scanning chamber, the scanner(2.0 Megahertz) frequency, 1.2 Megapascals peak negative pressure) was set to either deUver ultrasound at a conventional 30 Hertz frame rate or was shut off FoUowmg passage through the scanning chamber, the solution was then passed through the same size plastic tubing into a graduated cyUnder The first 10 milliliters was discarded Following this, the next 10 milliliters was aUowed to enter into a coUection tube The coUection tube containing the effluent microbubbles was aUowed to stand in order to separate microbubbles on the top from whatever free oUgonucleotide existed in the lower portion of the sample. Drops from both the upper and lower operation of the effluent were then placed upon a hemocytometer slide and analyzed using a 10X magnification Photographs of these shdes were then made and the number of microbubbles over a 36 square centimeter field were then hand-counted The upper and lower layers of the remaining effluent were then used for analysis of oUgonucleotide content using flow cytometry in the same manner described below Microbubble samples exposed to the various oUgonucleotide solution were mixed 15(v/v) with a solution of formamide and EDTA and heated to 95°
C for 5 minutes. These samples were then examined on an AppUed
Biosystems Model 373A DNA sequencer with e 20% polyacrylamide gel The data were acquired with GeneScanner software so that fluorescence intensity area under the curve could be determined
EXAMPLE 2
Phosphorothioate OUgonucleotide Binding of PESDA versus RA-SDA Microbubbles
The partitioning of PS-ODN to PESDA microbubbles top layer) and non- bubble washed (albumin-free) and unwashed (non-bubble albumin containing) lower layers as counted by liquid scintillation counting are demonstrated in Table 1 TABLE 1 OLIGONUCLEOTIDES BINDINGTOALBUMIN OF PESDA MICROBUBBLES
BUBBLES INTHE PRESENCE OF FREEALBUMIN
N TOP BOTTOM RATIO cpm/μl cpm/μl T/B
TTAGGG 6 125+6.4 92.316.4 1.35 c-myb 6 94.1117.6 77.311.2 1.35
WASHED BUBBLES (NO FREE ALBUMIN)
N TOP BOTTOM RATIO cpm/μl cpm/μl T/B
TTAGGG 6 210110.8 126+8.7 1.67 c-myb 6 200.3137.4 92.7+15.7 2.16
These data indicate that albumin in the unwashed solution which is not associated with the microbubble wifl bind to the PS-ODN so that the partitioning of PS-ODN is equivalent between microbubbles(top layer) and the surrounding solution (lower layer; p=HS). Removal of non -microbubble associated albumin (Washed Bubbles in Table 1) does not show a significant partitioning of the PS-ODNs with the PESDA microbubbles (1.67 for TTAGGG PS-ODN and 2.16 for c-myb PS-ODN). The recovery of total radioactivity in the experiments reported in Table 1 is 96% of the radioactivity added which is not significantly different from 100%.
The affinity of binding of PS-ODN to washed microbubbles was evaluated by addition of increasing amounts of excess non-radioactive PS=ODN as a competing Ugand for binding sites. In this case a 20mer PS- ODN with sequence 5'-d(CCC TGC TCC CCC CTG GCT CQ-3' (SEQ ID NO:4) was employed to displace the radioactive 24mer. Albumin protein concentrations in the washed microbubble experiments was 0.281 0.04 mg/ml as determined by the Bradford Assay Bradford M et al "A Rapid and Sensitive Method for the quantification of microgram quantities of protein utiUzing the principle of protein-dye binding" anal. Bioche,. 72:248, 1976. The observed binding data are presented as a Lineweaver Burke plot in Figure 1. The equiUbrium dissociation constant Km (calculated for the 7 concentrations which were run in duphcate) for the binding to the microbubbles was 1.76 X 10"5 M.
The distribution of FITC-labeled microbubbles is provided in table 2
TABLE 2 DISTRIBUTION OF OLIGONUCLEOTIDE (PS-ODN) BOUND MICROBUBBLES
Control PS-ODN lδlnM FITC PS-ODN Excess Unlabeled OE
No. PE MI PE MI PE MI
1 99.5 2.38 98.9 2109.8 97.8 1753.1
2 99.3 4.07 99.1 2142.3 98.7 1710.9
3 99.4 3.52 99.1 2258.5 99.3 1832.2
Figure imgf000020_0001
PE=percent events MI=mean intensity SE=standard error lindicates this mean is significantly different form control, P<0.001 ^indicates this mean is significantly different form 151nM, P<0.001
The significant decrease in mean fluorescence intensity in the samples containing excess unlabeled PS-ODN indicates the binding to microbubbles is saturable. Consequently, since the binding is saturable, the nonspecific interactions of PS-ODN with the microbubble surface are Umited. A Gaussian distribution of PS-ODN to washed PESDA microbubbles indicated that the albumin on these microbubbles had retained its binding site for the oUgonucleotide. The absence of a Gaussian distribution for washed RA-SDA indicated loss of albumin binding site 1 for this oUgonucleotide occurred during sonication of these microbubbles. For a discussion of albumin binding characteristics particularly as they relate to oUgonucleotides see Kumar, Shashi et al "Characterization of Binding Sites, Extent of Binding, and Drug Interactions of OUgonucleotides with Albumin" Antisense Research and Development 5: 131-139 (1995) the disclosure of which is hereby incorporated ~ by reference.
From the foregoing it can be seen that, PS-ODN binds to the albumin in PESDA microbubbles, indicating that the binding site 1 on albumin is biologicaUy active foUowing production of these bubbles by electromechanical sonication. This binding site affinity is lost when the electromechanical sonication is performed only with room sir. Further, removal of albumin not associated with PESDA microbubbles by washing shows a significant partitioning of the PS-ODNs with the microbubbles (Table 1). These observations demonstrate that albumin denaturation does not occur with perfluorocarbon-containing dextrose albumin solutions during sonication as has been suggested with sonication in the presence of air. The retained bioactivity of albumin(especiaUy at site 1) in PESDA microbubbles was confirmed by the affinity of binding of PS-ODN to washed PESDA microbubbles in the presence of increasing amounts of excess non-radioactive PS-ODN as a competing Ugand for binding sites (Table 2). The significant decrease in mean fluorescence intensity in the samples containing excess unlabeled PS=-ODN indicates the binding to microbubbles is saturable.
EXAMPLE 3
ALTERED BIODISTRIBUTION VIA MICROBUBBLE DELIVERY OF ANTISENSE OLIGOS
According to the invention antisense phosphorothioate oUgonucleotides were designed to the cytochrome P450 IIB 1 gene sequence to alter the metaboUsm of Phenobarbital. The oUgonucleotides were conjugated to perfluoropropane exposed sonicated dextrose albumin microbubbles (PESDA) as earUer described and deUvered to rats intravenously. The oUgonucleotide was synthesized according to the rat cytochrome P450 IIB 1 known sequence and had the foUowing sequence: GGAGCAAGATACTGGGCTCCAT (SEQ ID NO:5) AAAGAAGAGAGAGAGCAGGGAG (SEQ ID NO:6) Male Sprague-Dawley rats (Sasco, Omaha), were used and weighed between 210 to 290 grams for aU studies. They were housed in animal quarters at the University of Nebraska Medical Center, AAALAC approved animal resource facility. The animals were exposed to 12 hour Ught/dark cycle" and aUowed access to Purina rat chow and tap water ad Ubitum.
Rats in groups with PB were injected intraperitoneaUy with phenobarbital (MaJlinckrodt, St. Louis) at 80 ml kg/day x 2 days. The PB injections were given simultaneously with the ODN-microbubble injections. Phosphorothioate ODN injections were 1 ml/kg/day x 2 days. Sleep times were measured 48 hours after the first injection. The rats were injected intraperitoneaUy with 100 ml/kg hexobarbital (Sigma, St. Louis), paired fresh daily. The volume of this injection is 1 ml/kg body weight. Each rat was injected with 100 mg/kg of hexobarbital intraperitoneaUy.
The animals were placed on their backs to insure that they were still under sedation from the hexobarbital. Sleep time is defined as the time they are placed on their backs to the time when they roU over. The sleep times Usted are the mean of each animal in the group + standard deviation. Results indicate that deUvery of the oUgonucleotide conjugated microbubbles greatly improved efficacy of the drug. Rats given l/20th dose of ohgo experienced a sleep time of more than 50 minutes. This is compared to non microbubble conjugated oUgo with an approximate sleep time of 13 minutes Rats were ultimately sacrificed using ethyl ether and microsomes were prepared as described by FrankUn and Estabrook (1971). Livers were perfused with 12 ml of 4% saUne via the portal vein and then removed from the animal. The Uvers were minced, homogenized in .25 M sucrose (Sigma) and centrifuged at 8000 x g for 20 minutes at 4°C in a Sorvall RC2-B centrifuge (Dupont, Wilmington, DE). The supernatant was saved and resuspended in a .25 M sucrose and centrifuged at 100,000 x g for 45 minutes at 4°C in a SorvaU OTD55B ultracentrifuge (Dupont). The peUet was resuspended in 1.15% KCL (Sigma) and centrifuged at 100,000 x g for 1 hour at 4°C with the final peUet resuspended in an equal volume buffer (10 mM Tns-acetate, 1 mM EDTA, 20% glycerol; Sigma) and frozen at -80°C. Protein concentrations were determined by Bradford assay (Bradford, 1976) 80 μl ahquots of homogenate were added to a 96 weU plate (Becton, Dickinson Labware, Lincoln Park, NJ) 20 μl of Bradford reagent (Bio-Rad Richmond, CA) was then added and the plates read at 595 nm on the microplate reader (Molecular Devices, Newport MN) The data was compared to standard curve generated with known concentrations of bovine serum albumin (Sigma)
CYP IIB 1 content was determined by pentoxyresorufin 0-dealkylation (PROD) activity (Burke et al 1985) For each microsomal sample, 1 mg protein in 1 ml .1 M potassium phosphate buffer, 1 ml 2 μM 5- pentoxyresorufin (Pierce, Rockford, IL), and 17 μl 60 mM NADPH were mixed and incubated for 10 minutes at 37°C The mixture was then added to a 2 ml cuvette and read on a RF5000U spectrofluorophotometer (Shimadzu, Columbia, MD) using an excitation wavelength of 530 nm and emission wavelength of 585 nm Concentrations of unknowns were calculated from a standard curve of resorufin (Pierce, Rockford, IL) standards Results were recorded in nmol resorufin/mg protem/mm
Direct measurement of CYP IIB1 protein was determined by an ELISA assay using an antibody directed the CYP IIB l protein (Schuurs and Van Weeman, 1977) 50 μg of Uver per weU were plated in 100 μl 35% sodium bicarbonate buffer overnight on a 96 weU nunc-immuno plate (InterMed, Skokie, IL) The microsomes were washed 3x with 1% bovine serum albumin in PBS (PBS/BSA) and incubated for 1 hr at 37°C with 200 μl PBS/BSA The PBS/BSA was removed and 50 μl of CYP IIB l antibody (Oxygene, DaUas) was added and incubated for 1 hour at 37°C The microsomes were washed 5x with sahne/tween 20 (Sigma) and had 50 μl horseradish peroxidase antibody (Bio- rad) added The microsomes were incubated for 1 hour at 37°C, washed 5x with sahne/tween 20 and twice with 85% saUne. 100 μl of horseradish peroxidase substrate (Kirkegaard & Perry Labs, Gaithersburg, MD) was added and the plate read continuously in a microplate reader (Molecular Devices) at 405 nm for 1 hour. Results were recorded as horseradish peroxidase activity in mOD/min.
Results demonstrated that the oUgo conjugated microbubbles directed the oUgo to the Uver and kidney. These are site of phenobarbitol metaboUsm. 5 As described earUer, 100 mg/kg HB was injected i.p. to each animal at the end of 2 days of treatment with PB and/or the ODNs. Control rats had a sleep time of about 23 minutes. PB had a significant reduction in sleep time to about 11.414.5 minutes. PB stimulates CYP IIB l mRNA, as a result, hexobarbital which is hydroxylated by CYP IIBl is more quickly metaboUzed l o and its sedative effect reduced.
SEQUENCE LISTING
(1) GENERAL INFORMATION: d) APPLICANT: Board of Regents of the University of Nebraska
(n) TITLE OF INVENTION: Compositions and Methods for Altering the Biodistribution of Biological Agents
(m) NUMBER OF SEQUENCES: 6
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Zarley, McKee, Thomte, Voorhees & Sease (B) STREET: 801 Grand Suite 3200
( C ) CI TY : Des Homes
( D ) STATE : I owa
(E) COUNTRY: United States
(F) ZIP: 50309
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: Patentln Release #1.0, Version #1.30
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE: ( C) CLASSI FI CATION :
( vm ) ATTORNEY/AGENT INFORMATI ON :
(A) NAME : Nebel , Heidi S .
( B ) REGI STRATI ON NUMBER : 37 , 719 (C) REFERENCE/DOCKET NUMBER: P2013 63107A dx) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE- 515-288-3667
(B) TELEFAX: 515-288-1338
(2) INFORMATION FOR SEQ ID NO : 1 :
(l) SEQUENCE CHARACTERISTICS: (A) LENGTH: 15 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS : single
(D) TOPOLOGY: linear (ll) MOLECULE TYPE: CDNA
(m) HYPOTHETICAL: NO
(iv) ANTI -SENSE: YES
(xi ) SEQUENCE DESCRIPTION. SEQ ID NO.l: AACGTTGAGG GGCAT 15
(2) INFORMATION FOR SEQ ID NO : 2 : (ι) SEQUENCE CHARACTERISTICS: (A) LENGTH: 24 base pairs (B) TYPE nucleic acid
(C) STRANDEDNESS single
(D) TOPOLOGY linear
(n) MOLECULE TYPE cDNA (m) HYPOTHETICAL NO (iv) ANTI -SENSE YES
(xi ) SEQUENCE DESCRIPTION SEQ ID NO 2
TATGCTGTGC CGGGGTCTTC GGGC 24
(2) INFORMATION FOR SEQ ID NO 3 (l) SEQUENCE CHARACTERISTICS
(A) LENGTH 6 base pairs
(B) TYPE nucleic acid
(C) STRANDEDNESS single
(D) TOPOLOGY linear
(n) MOLECULE TYPE cDNA
(m) HYPOTHETICAL NO (iv) ANTI -SENSE YES
(xi) SEQUENCE DESCRIPTION SEQ ID NO 3
TTAGGG 6
(2) INFORMATION FOR SEQ ID NO 4
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 20 base pairs
(B) TYPE nucleic acid
(C) STRANDEDNESS single (D) TOPOLOGY linear
(ll) MOLECULE TYPE cDNA
(ill) HYPOTHETICAL NO
(iv) ANTI -SENSE YES
(xi) SEQUENCE DESCRIPTION SEQ ID NO 4
CCCTGCTCCC CCCTGGCTCC 2C (2) INFORMATION FOR SEQ ID NO 5
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 22 base pairs
(B) TYPE nucleic acid (C) STRANDEDNESS single
(D) TOPOLOGY linear ( 11 ) MOLECULE TYPE cDNA
( m ) HYPOTHETI CAL NO
( iv ) ANTI - SENSE YES
(xi) SEQUENCE DESCRIPTION SEQ ID NO 5
GGAGCAAGAT ACTGGGCTCC AT 22 (2) INFORMATION FOR SEQ ID NO 6
(l) SEQUENCE CHARACTERISTICS
(A) LENGTH 22 base pairs
(B) TYPE nucleic acid (C) STRANDEDNESS single
(D) TOPOLOGY linear
(n) MOLECULE TYPE cDNA (in) HYPOTHETICAL NO
(iv) ANTI -SENSE YES
(xi) SEQUENCE DESCRIPTION SEQ ID NO 6 AAAGAAGAGA GAGAGCAGGG AG 22

Claims

What is claimed is:
1. A method for delivering a biological agent to specific tissue sites comprising: forming a solution of a plurality of protein encapsulated, insoluble gas-filled microbubbles, said microbubbles conjugated to said biological agent; administering said solution to an animal; so that said protein directs the microbubble-conjugated agent to sites of bioprocessing of said protein and upon dissipation of the microbubble releases said agent.
2. The method of claim 1 wherein said protein is selected from the group consisting of albumin, human gamma, globulin, human apotransferin, beta lactose and urease.
3. The method of claim 1 wherein said protein is albumin.
4. The method of claim 1 wherein said insoluble gas is selected from the group consisting of perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, and perfluoropentane.
5. The method of claim 4 wherein said gas is perfluoropropane.
6. The method of claim 1 wherein said microbubbles are formed by the steps of: mixing an aqueous solution comprising about 2% to about 10% by weight of human serum albumin diluted about two-fold to about eight-fold with 5% to 50% by weight of dextrose; and exposing said solution to a sonication horn to create cavitation at p articulate sites in said solution thereby generating stable microbubbles from about .1 to 10 microns in diameter.
7. The method of claim 6 wherein said dilution of albumin with dextrose is a three-fold dilution.
8. The method of claim 6 wherein said human serum albumin is a 5% by weight solution.
9. The method of claim 6 wherein said dextrose is a 5% by weight solution.
10. The method of claim 1 wherein said protein is albumin and said biological agent is selected from the group consisting of: an oUgonucleotide, a polynucleotide, a ribozyme, naproxen, piroxicam, warfarin, furosemide, phenylbutazone, valproic acid, sulfisoxazole, ceftriaxone, miconazole.
11. The method of claim 10 wherein said biological agent is an oUgonucleotide.
12. The method of claim 11 wherein said oUgonucleotide is a phosphorothioate oUgonucleotide.
13. The method of claim 12 wherein said phosphorothioate oUgonucleotide is an antisense oUgonucleotide.
14. The method of claim 2 wherein said target site is the Uver and the kidney of said animal.
15. A composition for deUvery of a biological agent to a target site comprising: an aqueous suspension comprising a pluraUty of protein encapsulated insoluble gas-filled microbubbles and; a biological agent conjugated to said protein.
16. The composition of claim 15 wherein said protein is selected from the group consisting of albumin, human gamma globuUn, human apotransferin, beta lactose and urease.
17. The composition of claim 15 wherein said protein is albumin.
18. The composition of claim 15 wherein said gas is a perfluorocarbon gas.
19. The composition of claim 15 wherein said gas is selected from the group consisting of perfluoromethane, perfluoroethane, perfluoropropane, perfluorobutane, and perfluoropentane.
20. The composition of claim 19 wherein said gas is perfluorobutane.
21. The composition of claim 19 wherein said gas is perfluoropropane.
22. The composition of claim 15 wherein said protein is albumin and said biological agent is selected from the group consisting of: an oUgonucleotide, a polynucleotide, a ribozyme, naproxen, piroxicam, warfarin, furosemide, phenylbutazone, valproic acid, sulfisoxazole, ceftriaxone, miconazole.
23. The composition of claim 22 wherein said biological agent is an oUgonucleotide.
24. A composition for deUvery of nucleotide based biological agents to a target site comprising: a pluraUty of albumin encapsulated insoluble gas-filled microbubbles and a nucleotide based biological agent conjugated to said albumin microbubbles.
25. The composition of claim 24 wherein said microbubbles are .1 to 10 microns in diameter.
26. The composition of claim 24 wherein said gas is a perfluorocarbon gas.
27. A method for deUvering nucleotide based biological agents to the kidney and Uver of animals comprising: forming a solution comprising a pluraUty of albumin encapsulated insoluble gas-filled microbubbles, said microbubbles conjugated to said nucleotide based biological agent; and administering said solution to an animal; so that said albumin encapsulated microbubble is taken up by said Uver and said kidney and upon dissipation of the microbubble, releases said biological agent.
28. The method of claim 1 wherein said nucleic acid biological agent is selected from the group consisting of an antisense oUgonucleotide, antigene oUgonucleotide, oUgonucleotide probe, or a nucleotide vector.
PCT/US1997/010766 1996-06-28 1997-06-20 Compositions and methods for altering the biodistribution of biological agents WO1998000172A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP50420598A JP4201348B2 (en) 1996-06-28 1997-06-20 Compositions and methods for modifying biodistribution of biological agents
AT97932228T ATE260677T1 (en) 1996-06-28 1997-06-20 COMPOSITIONS AND METHODS FOR ALTERING THE BIODISTRIBUTION OF BIOLOGICAL AGENTS
DE69727958T DE69727958T2 (en) 1996-06-28 1997-06-20 Compositions and methods for altering biodistribution of biological agents
CA002258882A CA2258882C (en) 1996-06-28 1997-06-20 Compositions and methods for altering the biodistribution of biological agents
EP97932228A EP0938341B1 (en) 1996-06-28 1997-06-20 Compositions and methods for altering the biodistribution of biological agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/670,999 1996-06-28
US08/670,999 US5849727A (en) 1996-06-28 1996-06-28 Compositions and methods for altering the biodistribution of biological agents

Publications (2)

Publication Number Publication Date
WO1998000172A2 true WO1998000172A2 (en) 1998-01-08
WO1998000172A3 WO1998000172A3 (en) 1998-05-14

Family

ID=24692746

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/010766 WO1998000172A2 (en) 1996-06-28 1997-06-20 Compositions and methods for altering the biodistribution of biological agents

Country Status (7)

Country Link
US (5) US5849727A (en)
EP (1) EP0938341B1 (en)
JP (2) JP4201348B2 (en)
AT (1) ATE260677T1 (en)
CA (1) CA2258882C (en)
DE (1) DE69727958T2 (en)
WO (1) WO1998000172A2 (en)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998018500A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018498A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018495A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018501A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO2000002588A1 (en) * 1998-07-13 2000-01-20 The Board Of Regents Of The University Of Nebraska Targeted site specific drug delivery compositions and method of use
US6245747B1 (en) 1996-03-12 2001-06-12 The Board Of Regents Of The University Of Nebraska Targeted site specific antisense oligodeoxynucleotide delivery method
US6261537B1 (en) 1996-10-28 2001-07-17 Nycomed Imaging As Diagnostic/therapeutic agents having microbubbles coupled to one or more vectors
US6264917B1 (en) 1996-10-28 2001-07-24 Nycomed Imaging As Targeted ultrasound contrast agents
US6331289B1 (en) 1996-10-28 2001-12-18 Nycomed Imaging As Targeted diagnostic/therapeutic agents having more than one different vectors
JP2002145784A (en) * 2000-11-10 2002-05-22 Ryuichi Morishita Biologically active medicament-introducing composition and method for using the same
WO2003080123A1 (en) * 2002-03-22 2003-10-02 Anges Mg, Inc. Compositions for delivering biologically active drug and method of using the same
WO2003092741A1 (en) * 2002-05-03 2003-11-13 Avi Biopharma, Inc. Delivery of microparticle-conjugated drugs for inhibition of stenosis
JP2004523465A (en) * 2000-05-17 2004-08-05 エイブイアイ バイオファーマ, インコーポレイテッド Antisense enzyme inhibitors to improve drug pharmacokinetics
US6822086B1 (en) 1999-08-09 2004-11-23 The General Hospital Corporation Drug-carrier complexes and methods of use thereof
DE102004057196A1 (en) * 2004-11-26 2006-06-01 Rösner Research GmbH & Co.KG Method for producing albumin conjugates with non-steroidal anti-inflammatory drugs (NSAIDs)
WO2006129080A1 (en) * 2005-05-31 2006-12-07 The Institute Of Cancer Research: Royal Cancer Hospital Materials and methods for transducing cells with a viral vector
JP2010095541A (en) * 1998-07-13 2010-04-30 Board Of Regents Of The Univ Of Nebraska Targeted site specific drug delivery composition and method of use
US7754238B2 (en) * 2002-05-03 2010-07-13 Avi Biopharma, Inc. Delivery of microparticle-conjugated drugs for inhibition of stenosis
US8012118B2 (en) 2006-03-08 2011-09-06 Fresenius Medical Care Holdings, Inc. Artificial kidney dialysis system
US8106098B2 (en) 1999-08-09 2012-01-31 The General Hospital Corporation Protein conjugates with a water-soluble biocompatible, biodegradable polymer
US8241239B2 (en) 2006-08-24 2012-08-14 Fresenius Medical Care Holdings, Inc. Device for removing fluid from blood in a patient
US8715221B2 (en) 2006-03-08 2014-05-06 Fresenius Medical Care Holdings, Inc. Wearable kidney
US8777892B2 (en) 2008-11-03 2014-07-15 Fresenius Medical Care Holdings, Inc. Portable peritoneal dialysis system
WO2014117232A1 (en) * 2013-02-04 2014-08-07 Farret Neto Abdo Medicinal preparation in the form of a foam for the sustained release of medicinal drugs, and system for producing the medicinal preparation
US9545457B2 (en) 1998-01-14 2017-01-17 Lantheus Medical Imaging, Inc. Preparation of a lipid blend and a phospholipid suspension containing the lipid blend

Families Citing this family (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
US6088613A (en) 1989-12-22 2000-07-11 Imarx Pharmaceutical Corp. Method of magnetic resonance focused surgical and therapeutic ultrasound
US5205290A (en) 1991-04-05 1993-04-27 Unger Evan C Low density microspheres and their use as contrast agents for computed tomography
US6743779B1 (en) 1994-11-29 2004-06-01 Imarx Pharmaceutical Corp. Methods for delivering compounds into a cell
US6176842B1 (en) * 1995-03-08 2001-01-23 Ekos Corporation Ultrasound assembly for use with light activated drugs
US6521211B1 (en) 1995-06-07 2003-02-18 Bristol-Myers Squibb Medical Imaging, Inc. Methods of imaging and treatment with targeted compositions
US5648098A (en) * 1995-10-17 1997-07-15 The Board Of Regents Of The University Of Nebraska Thrombolytic agents and methods of treatment for thrombosis
DE69736981D1 (en) 1996-05-01 2007-01-04 Imarx Pharmaceutical Corp IN VITRO PROCESS FOR INTRODUCING NUCLEIC ACIDS INTO A CELL
US5849727A (en) * 1996-06-28 1998-12-15 Board Of Regents Of The University Of Nebraska Compositions and methods for altering the biodistribution of biological agents
US20070036722A1 (en) * 1996-10-28 2007-02-15 Pal Rongved Separation processes
US6090800A (en) 1997-05-06 2000-07-18 Imarx Pharmaceutical Corp. Lipid soluble steroid prodrugs
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6582392B1 (en) 1998-05-01 2003-06-24 Ekos Corporation Ultrasound assembly for use with a catheter
US6676626B1 (en) 1998-05-01 2004-01-13 Ekos Corporation Ultrasound assembly with increased efficacy
US6416740B1 (en) 1997-05-13 2002-07-09 Bristol-Myers Squibb Medical Imaging, Inc. Acoustically active drug delivery systems
US6548047B1 (en) 1997-09-15 2003-04-15 Bristol-Myers Squibb Medical Imaging, Inc. Thermal preactivation of gaseous precursor filled compositions
US20030078227A1 (en) * 1998-07-02 2003-04-24 Greenleaf James F. Site-directed transfection with ultrasound and cavitation nuclei
US6309355B1 (en) * 1998-12-22 2001-10-30 The Regents Of The University Of Michigan Method and assembly for performing ultrasound surgery using cavitation
DE10119522A1 (en) * 2001-04-20 2002-12-05 Innovacell Biotechnologie Gmbh Preparation and application of a suspension composition with an ultrasound contrast medium
JP4426288B2 (en) * 2001-07-10 2010-03-03 ソノジーン,リミテッド ライアビリティー カンパニー Enhanced transfection of liver DNA
US7897382B2 (en) * 2001-10-22 2011-03-01 Alnylam Pharmaceuticals, Inc. Transfection kinetics and structural promoters
EP1444363B1 (en) * 2001-10-22 2006-09-20 Nucleonics, Inc Transfection kinetics and structural promoters
US7220239B2 (en) 2001-12-03 2007-05-22 Ekos Corporation Catheter with multiple ultrasound radiating members
US8226629B1 (en) 2002-04-01 2012-07-24 Ekos Corporation Ultrasonic catheter power control
US20040180438A1 (en) * 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
US20040126400A1 (en) * 2002-05-03 2004-07-01 Iversen Patrick L. Delivery of therapeutic compounds via microparticles or microbubbles
CA2487274A1 (en) * 2002-05-06 2003-11-13 Nucleonics Inc. Spermine chemically linked to lipids and cell-specific targeting molecules as a transfection agent
WO2004011624A2 (en) * 2002-07-31 2004-02-05 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
US6921371B2 (en) 2002-10-14 2005-07-26 Ekos Corporation Ultrasound radiating members for catheter
ES2351976T3 (en) * 2003-04-29 2011-02-14 Avi Biopharma, Inc. COMPOSITIONS TO IMPROVE THE TRANSPORTATION AND ANTI-EFFECTIVE EFFECTIVENESS OF NUCLEIC ACID ANALOGS IN CELLS.
US20050222068A1 (en) * 2003-10-23 2005-10-06 Mourich Dan V Method and antisense composition for selective inhibition of HIV infection in hematopoietic cells
US7025726B2 (en) * 2004-01-22 2006-04-11 The Regents Of The University Of Nebraska Detection of endothelial dysfunction by ultrasonic imaging
US20050288246A1 (en) * 2004-05-24 2005-12-29 Iversen Patrick L Peptide conjugated, inosine-substituted antisense oligomer compound and method
US20060240032A1 (en) * 2005-03-31 2006-10-26 Hinrichs David J Immunomodulating compositions and methods for use in the treatment of human autoimmune diseases
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US10219815B2 (en) 2005-09-22 2019-03-05 The Regents Of The University Of Michigan Histotripsy for thrombolysis
US8057408B2 (en) 2005-09-22 2011-11-15 The Regents Of The University Of Michigan Pulsed cavitational ultrasound therapy
US20070140965A1 (en) * 2005-12-02 2007-06-21 Barnes-Jewish Hospital Methods to ameliorate and image angioplasty-induced vascular injury
ES2471118T3 (en) 2007-06-22 2014-06-25 Ekos Corporation Method and apparatus for the treatment of intracranial hemorrhages
EP2170363B1 (en) * 2007-06-29 2018-08-08 Sarepta Therapeutics, Inc. Tissue specific peptide conjugates and methods
US20100016215A1 (en) 2007-06-29 2010-01-21 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
WO2009043031A2 (en) 2007-09-27 2009-04-02 Children's Medical Center Corporation Microbubbles and methods for oxygen delivery
US9023321B2 (en) * 2008-03-21 2015-05-05 The Board Of Trustees Of The University Of Arkansas Methods for producing microbubbles
EP2376633A1 (en) * 2008-12-17 2011-10-19 AVI BioPharma, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
US20110269665A1 (en) 2009-06-26 2011-11-03 Avi Biopharma, Inc. Compound and method for treating myotonic dystrophy
CA2770452C (en) 2009-08-17 2017-09-19 Histosonics, Inc. Disposable acoustic coupling medium container
CA2770706C (en) 2009-08-26 2017-06-20 Charles A. Cain Devices and methods for using controlled bubble cloud cavitation in fractionating urinary stones
WO2011028603A2 (en) 2009-08-26 2011-03-10 The Regents Of The University Of Michigan Micromanipulator control arm for therapeutic and imaging ultrasound transducers
WO2011025893A1 (en) 2009-08-28 2011-03-03 The Trustees Of Columbia University In The City Of New York Systems, methods, and devices for production of gas-filled microbubbles
US8539813B2 (en) 2009-09-22 2013-09-24 The Regents Of The University Of Michigan Gel phantoms for testing cavitational ultrasound (histotripsy) transducers
WO2011084694A1 (en) 2009-12-17 2011-07-14 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Stabilized stat3 decoy oligonucleotides and uses therefor
EP2637701A4 (en) * 2010-11-12 2014-09-17 Childrens Medical Center Gas-filled microbubbles and systems for gas delivery
US9161948B2 (en) 2011-05-05 2015-10-20 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
KR20120140290A (en) * 2011-06-21 2012-12-31 주식회사 퍼시픽시스템 Method of manufacturing bubble for accelerating functional material and device or transferring functional material
US9144694B2 (en) 2011-08-10 2015-09-29 The Regents Of The University Of Michigan Lesion generation through bone using histotripsy therapy without aberration correction
US10357450B2 (en) 2012-04-06 2019-07-23 Children's Medical Center Corporation Process for forming microbubbles with high oxygen content and uses thereof
US9049783B2 (en) 2012-04-13 2015-06-02 Histosonics, Inc. Systems and methods for obtaining large creepage isolation on printed circuit boards
WO2013166019A1 (en) 2012-04-30 2013-11-07 The Regents Of The University Of Michigan Ultrasound transducer manufacturing using rapid-prototyping method
WO2014055906A1 (en) 2012-10-05 2014-04-10 The Regents Of The University Of Michigan Bubble-induced color doppler feedback during histotripsy
US9101745B2 (en) 2013-03-14 2015-08-11 Sonogene Llc Sonochemical induction of ABCA1 expression and compositions therefor
US10577554B2 (en) 2013-03-15 2020-03-03 Children's Medical Center Corporation Gas-filled stabilized particles and methods of use
AU2014232195A1 (en) * 2013-03-15 2015-10-15 Doheny Eye Institute Management of tractional membranes
US10124126B2 (en) * 2013-04-18 2018-11-13 The Regents Of The University Of Colorado, A Body Corporate System and methods for ventilation through a body cavity
WO2015003154A1 (en) 2013-07-03 2015-01-08 Histosonics, Inc. Articulating arm limiter for cavitational ultrasound therapy system
CN105530869B (en) 2013-07-03 2019-10-29 希斯托索尼克斯公司 The histotripsy excitation sequence optimized is formed to bubble cloud using impact scattering
WO2015027164A1 (en) 2013-08-22 2015-02-26 The Regents Of The University Of Michigan Histotripsy using very short ultrasound pulses
US11305013B2 (en) * 2014-08-26 2022-04-19 Drexel University Surfactant microbubbles and process for preparing and methods of using the same
US10052394B2 (en) 2014-11-21 2018-08-21 General Electric Company Microbubble tether for diagnostic and therapeutic applications
US11000476B2 (en) 2015-05-28 2021-05-11 Pacific System Co., Ltd. Cavitation seed for drug delivery, and drug delivery method using same
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions
CN107708581B (en) 2015-06-10 2021-11-19 Ekos公司 Ultrasonic wave guide tube
ES2948135T3 (en) 2015-06-24 2023-08-31 Univ Michigan Regents Histotripsy therapy systems for the treatment of brain tissue
JP7118063B2 (en) 2016-12-19 2022-08-15 サレプタ セラピューティクス, インコーポレイテッド Exon-skipping oligomeric conjugates for muscular dystrophy
WO2018160752A1 (en) 2017-02-28 2018-09-07 Children's Medical Center Corporation Stimuli-responsive particles encapsulating a gas and methods of use
WO2020113083A1 (en) 2018-11-28 2020-06-04 Histosonics, Inc. Histotripsy systems and methods
WO2021155026A1 (en) 2020-01-28 2021-08-05 The Regents Of The University Of Michigan Systems and methods for histotripsy immunosensitization
WO2021155177A1 (en) 2020-01-29 2021-08-05 Flagship Pioneering Innovations Vi, Llc Compositions comprising linear polyribonucleotides for protein modulation and uses thereof
EP4096682A1 (en) 2020-01-29 2022-12-07 Flagship Pioneering Innovations VI, LLC Compositions for translation and methods of use thereof
US20230104113A1 (en) 2020-01-29 2023-04-06 Flagship Pioneering Innovations Vi, Llc Delivery of compositions comprising circular polyribonucleotides
AU2021275223A1 (en) 2020-05-20 2023-02-02 Flagship Pioneering Innovations Vi, Llc. Coronavirus antigen compositions and their uses
CN116194139A (en) 2020-05-20 2023-05-30 旗舰创业创新第六有限责任公司 Immunogenic compositions and uses thereof
WO2022051629A1 (en) 2020-09-03 2022-03-10 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and uses thereof
AR127073A1 (en) 2021-09-17 2023-12-13 Flagship Pioneering Innovations Vi Llc COMPOSITIONS AND METHODS FOR PRODUCING CIRCULAR POLYRIBONUCLEOTIDES
TW202322826A (en) 2021-10-18 2023-06-16 美商旗艦先鋒創新有限責任公司 Compositions and methods for purifying polyribonucleotides
WO2023097003A2 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and their uses
WO2023096990A1 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovation Vi, Llc Coronavirus immunogen compositions and their uses
WO2023096963A1 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovations Vi, Llc Varicella-zoster virus immunogen compositions and their uses
TW202340461A (en) 2021-12-22 2023-10-16 美商旗艦先鋒創新有限責任公司 Compositions and methods for purifying polyribonucleotides
WO2023122789A1 (en) 2021-12-23 2023-06-29 Flagship Pioneering Innovations Vi, Llc Circular polyribonucleotides encoding antifusogenic polypeptides

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4232755A1 (en) * 1992-09-26 1994-03-31 Schering Ag Microparticle preparations made from biodegradable copolymers
WO1994018954A1 (en) * 1993-02-22 1994-09-01 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of biologics and compositions useful therefor
WO1994028874A1 (en) * 1993-06-11 1994-12-22 Unger Evan C Novel therapeutic delivery systems
WO1995015118A1 (en) * 1993-11-30 1995-06-08 Imarx Pharmaceutical Corp. Gas microspheres for topical and subcutaneous application
WO1996038181A1 (en) * 1995-05-31 1996-12-05 Board Of Regents Of The University Of Nebraska Sonicated dextrose-albumin ultrasound contrast agent, containing perfluorobutane
WO1996038180A1 (en) * 1995-05-30 1996-12-05 Board Of Regents Of The University Of Nebraska Ultrasound contrast agents, containing perfluorocarbon in dextrose-albumin microbubbles
WO1997033474A1 (en) * 1996-03-12 1997-09-18 Board Of Regents Of The University Of Nebraska Targeted site specific drug delivery compositions and method of use

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4718433A (en) * 1983-01-27 1988-01-12 Feinstein Steven B Contrast agents for ultrasonic imaging
US4572203A (en) * 1983-01-27 1986-02-25 Feinstein Steven B Contact agents for ultrasonic imaging
US4634586A (en) * 1984-05-21 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Reagent and method for radioimaging leukocytes
US5040537A (en) * 1987-11-24 1991-08-20 Hitachi, Ltd. Method and apparatus for the measurement and medical treatment using an ultrasonic wave
US4844882A (en) * 1987-12-29 1989-07-04 Molecular Biosystems, Inc. Concentrated stabilized microbubble-type ultrasonic imaging agent
US5410516A (en) * 1988-09-01 1995-04-25 Schering Aktiengesellschaft Ultrasonic processes and circuits for performing them
US4957656A (en) * 1988-09-14 1990-09-18 Molecular Biosystems, Inc. Continuous sonication method for preparing protein encapsulated microbubbles
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5445813A (en) * 1992-11-02 1995-08-29 Bracco International B.V. Stable microbubble suspensions as enhancement agents for ultrasound echography
IN172208B (en) * 1990-04-02 1993-05-01 Sint Sa
US5315997A (en) * 1990-06-19 1994-05-31 Molecular Biosystems, Inc. Method of magnetic resonance imaging using diamagnetic contrast
AU635449B2 (en) * 1990-10-05 1993-03-18 Bracco International B.V. Method for the preparation of stable suspensions of hollow gas-filled microspheres suitable for ultrasonic echography
US5107842A (en) * 1991-02-22 1992-04-28 Molecular Biosystems, Inc. Method of ultrasound imaging of the gastrointestinal tract
EP0732106A3 (en) * 1991-03-22 2003-04-09 Katsuro Tachibana Microbubbles containing booster for therapy of disease with ultrasound
US5409688A (en) * 1991-09-17 1995-04-25 Sonus Pharmaceuticals, Inc. Gaseous ultrasound contrast media
DE69230885T3 (en) * 1991-09-17 2008-01-24 Ge Healthcare As GASOUS ULTRASONIC CONTRASTING AGENTS
US5304325A (en) * 1991-11-13 1994-04-19 Hemagen/Pfc Emulsions containing alkyl- or alkylglycerophosphoryl choline surfactants and methods of use
US5255683A (en) * 1991-12-30 1993-10-26 Sound Science Limited Partnership Methods of and systems for examining tissue perfusion using ultrasonic contrast agents
IL104084A (en) * 1992-01-24 1996-09-12 Bracco Int Bv Long-lasting aqueous suspensions of pressure-resistant gas-filled microvesicles their preparation and contrast agents consisting of them
US5585479A (en) * 1992-07-24 1996-12-17 The United States Of America As Represented By The Secretary Of The Navy Antisense oligonucleotides directed against human ELAM-I RNA
US5302372A (en) * 1992-07-27 1994-04-12 National Science Council Method to opacify left ventricle in echocardiography
SK281535B6 (en) * 1993-01-25 2001-04-09 Sonus Pharmaceuticals, Inc. Contrast medium for diagnostic ultrasound procedures and process for preparing thereof
US5362478A (en) * 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US5401493A (en) * 1993-03-26 1995-03-28 Molecular Biosystems, Inc. Perfluoro-1H,-1H-neopentyl containing contrast agents and method to use same
US5567415A (en) * 1993-05-12 1996-10-22 The Board Of Regents Of The University Of Nebraska Ultrasound contrast agents and methods for their manufacture and use
NZ268826A (en) * 1993-07-02 1996-11-26 Molecular Biosystems Inc Protein encapsulated gas microspheres and their use in ultrasonic imaging
US5385147A (en) * 1993-09-22 1995-01-31 Molecular Biosystems, Inc. Method of ultrasonic imaging of the gastrointestinal tract and upper abdominal organs using an orally administered negative contrast medium
NO940711D0 (en) * 1994-03-01 1994-03-01 Nycomed Imaging As Preparation of gas-filled microcapsules and contrast agents for diagnostic imaging
US5540909A (en) * 1994-09-28 1996-07-30 Alliance Pharmaceutical Corp. Harmonic ultrasound imaging with microbubbles
US5560364A (en) * 1995-05-12 1996-10-01 The Board Of Regents Of The University Of Nebraska Suspended ultra-sound induced microbubble cavitation imaging
US5849727A (en) * 1996-06-28 1998-12-15 Board Of Regents Of The University Of Nebraska Compositions and methods for altering the biodistribution of biological agents

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4232755A1 (en) * 1992-09-26 1994-03-31 Schering Ag Microparticle preparations made from biodegradable copolymers
WO1994018954A1 (en) * 1993-02-22 1994-09-01 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of biologics and compositions useful therefor
WO1994028874A1 (en) * 1993-06-11 1994-12-22 Unger Evan C Novel therapeutic delivery systems
WO1995015118A1 (en) * 1993-11-30 1995-06-08 Imarx Pharmaceutical Corp. Gas microspheres for topical and subcutaneous application
WO1996038180A1 (en) * 1995-05-30 1996-12-05 Board Of Regents Of The University Of Nebraska Ultrasound contrast agents, containing perfluorocarbon in dextrose-albumin microbubbles
WO1996038181A1 (en) * 1995-05-31 1996-12-05 Board Of Regents Of The University Of Nebraska Sonicated dextrose-albumin ultrasound contrast agent, containing perfluorobutane
WO1997033474A1 (en) * 1996-03-12 1997-09-18 Board Of Regents Of The University Of Nebraska Targeted site specific drug delivery compositions and method of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PORTER T R ET AL: "Interaction of diagnostic ultrasound with synthetic oligonucleotide-labeled perfluorocarbon-exposed sonicated dextrose albumin microbubbles." JOURNAL OF ULTRASOUND IN MEDICINE, vol. 15, no. 8, August 1996, pages 577-584, XP002054859 *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6245747B1 (en) 1996-03-12 2001-06-12 The Board Of Regents Of The University Of Nebraska Targeted site specific antisense oligodeoxynucleotide delivery method
US6261537B1 (en) 1996-10-28 2001-07-17 Nycomed Imaging As Diagnostic/therapeutic agents having microbubbles coupled to one or more vectors
WO1998018501A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
US6331289B1 (en) 1996-10-28 2001-12-18 Nycomed Imaging As Targeted diagnostic/therapeutic agents having more than one different vectors
WO1998018498A3 (en) * 1996-10-28 1998-07-16 Marsden John Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018500A3 (en) * 1996-10-28 1998-07-23 Mardsen John Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018495A3 (en) * 1996-10-28 1998-07-30 Marsden John Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018501A3 (en) * 1996-10-28 1998-07-30 Marsden John Christopher Improvements in or relating to diagnostic/therapeutic agents
US6264917B1 (en) 1996-10-28 2001-07-24 Nycomed Imaging As Targeted ultrasound contrast agents
US6680047B2 (en) 1996-10-28 2004-01-20 Amersham Health As Diagnostic/therapeutic agents
WO1998018500A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018498A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
WO1998018495A2 (en) * 1996-10-28 1998-05-07 Marsden, John, Christopher Improvements in or relating to diagnostic/therapeutic agents
US9545457B2 (en) 1998-01-14 2017-01-17 Lantheus Medical Imaging, Inc. Preparation of a lipid blend and a phospholipid suspension containing the lipid blend
JP2010095541A (en) * 1998-07-13 2010-04-30 Board Of Regents Of The Univ Of Nebraska Targeted site specific drug delivery composition and method of use
JP2002520298A (en) * 1998-07-13 2002-07-09 ザ・ボード・オブ・リージェンツ・オブ・ザ・ユニバーシティー・オブ・ネブラスカ Targeted site-specific drug delivery compositions and methods of use
WO2000002588A1 (en) * 1998-07-13 2000-01-20 The Board Of Regents Of The University Of Nebraska Targeted site specific drug delivery compositions and method of use
US8106098B2 (en) 1999-08-09 2012-01-31 The General Hospital Corporation Protein conjugates with a water-soluble biocompatible, biodegradable polymer
US6822086B1 (en) 1999-08-09 2004-11-23 The General Hospital Corporation Drug-carrier complexes and methods of use thereof
US8247383B2 (en) 1999-08-09 2012-08-21 The General Hospital Corporation Drug-carrier complexes and methods of use thereof
JP2004523465A (en) * 2000-05-17 2004-08-05 エイブイアイ バイオファーマ, インコーポレイテッド Antisense enzyme inhibitors to improve drug pharmacokinetics
JP2002145784A (en) * 2000-11-10 2002-05-22 Ryuichi Morishita Biologically active medicament-introducing composition and method for using the same
WO2003080123A1 (en) * 2002-03-22 2003-10-02 Anges Mg, Inc. Compositions for delivering biologically active drug and method of using the same
WO2003092741A1 (en) * 2002-05-03 2003-11-13 Avi Biopharma, Inc. Delivery of microparticle-conjugated drugs for inhibition of stenosis
US7754238B2 (en) * 2002-05-03 2010-07-13 Avi Biopharma, Inc. Delivery of microparticle-conjugated drugs for inhibition of stenosis
DE102004057196A1 (en) * 2004-11-26 2006-06-01 Rösner Research GmbH & Co.KG Method for producing albumin conjugates with non-steroidal anti-inflammatory drugs (NSAIDs)
WO2006129080A1 (en) * 2005-05-31 2006-12-07 The Institute Of Cancer Research: Royal Cancer Hospital Materials and methods for transducing cells with a viral vector
US8012118B2 (en) 2006-03-08 2011-09-06 Fresenius Medical Care Holdings, Inc. Artificial kidney dialysis system
US8715221B2 (en) 2006-03-08 2014-05-06 Fresenius Medical Care Holdings, Inc. Wearable kidney
US8241239B2 (en) 2006-08-24 2012-08-14 Fresenius Medical Care Holdings, Inc. Device for removing fluid from blood in a patient
US9138521B2 (en) 2006-08-24 2015-09-22 Fresenius Medical Care Holdings, Inc. Device for removing fluid from blood in a patient
US8777892B2 (en) 2008-11-03 2014-07-15 Fresenius Medical Care Holdings, Inc. Portable peritoneal dialysis system
WO2014117232A1 (en) * 2013-02-04 2014-08-07 Farret Neto Abdo Medicinal preparation in the form of a foam for the sustained release of medicinal drugs, and system for producing the medicinal preparation

Also Published As

Publication number Publication date
US20040057946A1 (en) 2004-03-25
ATE260677T1 (en) 2004-03-15
US7198949B2 (en) 2007-04-03
US5849727A (en) 1998-12-15
DE69727958D1 (en) 2004-04-08
CA2258882C (en) 2009-03-17
US7115583B2 (en) 2006-10-03
US6537814B1 (en) 2003-03-25
US6117858A (en) 2000-09-12
US20040013662A1 (en) 2004-01-22
JP2000515499A (en) 2000-11-21
JP2009029810A (en) 2009-02-12
DE69727958T2 (en) 2005-04-07
EP0938341B1 (en) 2004-03-03
WO1998000172A3 (en) 1998-05-14
CA2258882A1 (en) 1998-01-08
JP4201348B2 (en) 2008-12-24
EP0938341A2 (en) 1999-09-01

Similar Documents

Publication Publication Date Title
CA2258882C (en) Compositions and methods for altering the biodistribution of biological agents
US6245747B1 (en) Targeted site specific antisense oligodeoxynucleotide delivery method
JP2007297414A (en) Targeted site specific drug delivery composition and use thereof
JP5049958B2 (en) Pharmaceutical composition containing gas filled microcapsules for ultrasound mediated delivery
Porter et al. Interaction of diagnostic ultrasound with synthetic oligonucleotide‐labeled perfluorocarbon‐exposed sonicated dextrose albumin microbubbles.
AU2005273865B2 (en) Gas-filled microvesicles composition for contrast imaging
AU736301B2 (en) Methods for delivering compounds into a cell
US6743779B1 (en) Methods for delivering compounds into a cell
CA2337361C (en) Targeted site specific drug delivery compositions and method of use
JP2001503407A (en) Improved diagnostic / therapeutic agents
JP4774135B2 (en) Targeted site-specific drug delivery compositions and uses thereof
KR100649035B1 (en) Targeted site specific drug delivery compositions and method of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

ENP Entry into the national phase

Ref document number: 2258882

Country of ref document: CA

Ref country code: CA

Ref document number: 2258882

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997932228

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997932228

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1997932228

Country of ref document: EP