WO1996024334A1 - Lipid constructs for cytoplasmic delivery of agents - Google Patents

Lipid constructs for cytoplasmic delivery of agents Download PDF

Info

Publication number
WO1996024334A1
WO1996024334A1 PCT/US1996/001960 US9601960W WO9624334A1 WO 1996024334 A1 WO1996024334 A1 WO 1996024334A1 US 9601960 W US9601960 W US 9601960W WO 9624334 A1 WO9624334 A1 WO 9624334A1
Authority
WO
WIPO (PCT)
Prior art keywords
oligonucleotide
lipid
cell
seq
construct
Prior art date
Application number
PCT/US1996/001960
Other languages
French (fr)
Other versions
WO1996024334A9 (en
Inventor
Roxanne Male-Brunne
Richard Proffitt
Original Assignee
Nexstar Pharmaceuticals, Inc.
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 Nexstar Pharmaceuticals, Inc. filed Critical Nexstar Pharmaceuticals, Inc.
Priority to AU49236/96A priority Critical patent/AU4923696A/en
Publication of WO1996024334A1 publication Critical patent/WO1996024334A1/en
Publication of WO1996024334A9 publication Critical patent/WO1996024334A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • A61K9/1272Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers with substantial amounts of non-phosphatidyl, i.e. non-acylglycerophosphate, surfactants as bilayer-forming substances, e.g. cationic lipids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1135Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against oncogenes or tumor suppressor genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y203/00Acyltransferases (2.3)
    • C12Y203/01Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • C12Y203/01028Chloramphenicol O-acetyltransferase (2.3.1.28)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates

Definitions

  • This invention relates generally to pharmaceutical sciences and more particularly to the production of lipid constructs for the delivery of drugs to the cytoplasm of a cell and in particular to vascular smooth muscle tissue.
  • a preferred use of this invention relates to the administration of these constructs for the prevention of restenosis following angioplasty.
  • oligonucleotides are degraded by intracellular and extracellular enzymes such as exonucleases and endonucleases.
  • workers have modified the typical phosphodiester form of oligonucleotides. For example, modifications have included the creation of phosphorothioate and methylphosphonate oligonucleotides. Methylphosphonate oligonucleotides contain uncharged sectors which increase intracellular uptake and resist enzymatic degradation. Phosphorothioate oligonucleotides are negatively charged but are resistant to endonucleases.
  • Lipofectin liposomes formed from a mixture of N-[2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) and dioleoylphosphatidylethanolamine (DOPE), has been used to transduce recombinant genes into coronary arteries in vivo.
  • DOTMA N-[2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOPE dioleoylphosphatidylethanolamine
  • a cationic liposome used to introduce genes into DNA is a liposome preparation containing DC-chol, 3b[N-9N'N'-dimethylaminoethane)- carbamoyl] cholesterol and DOPE.
  • DOPE dimethylaminoethane
  • Other efforts have been directed toward the use of modified retroviruses and cationic liposomes for gene transfer.
  • Liposomes in general have been known. Liposomes are microscopic vesicles made from phospholipids, which form closed, fluid filled spheres when dispersed with aqueous solutions. Phospholipid molecules are polar, having a hydrophilic head and two hydrophobic tails consisting of long fatty acid chains.
  • the tails spontaneously associate to exclude water while the hydrophilic phosphate heads interact with water.
  • the result is a spherical, bilayer membrane in which the fatty acid tails converge in the interior of the newly formed membrane, and the polar heads point in opposite directions toward an aqueous medium.
  • These bilayer membranes thus form closed spheres known as liposomes.
  • the polar heads at the inner surface of the membrane point toward the aqueous interior of the liposome and, at the opposite surface of the spherical membrane, the polar heads interact with the surrounding aqueous medium.
  • Liposomes As the liposomes are formed, water soluble molecules can be incorporated into the aqueous interior, and lipophilic molecules may be incorporated into the lipid bilayer. Liposomes may be either multilamellar, like an onion with liquid separating many lipid bilayers, or unilamellar, with a single bilayer surrounding an aqueous center.
  • liposome preparation techniques which may be employed to produce various types of liposomes. These can be selected depending on the use, the chemical intended to be entrapped, and the type of lipids used to form the bilayer membrane.
  • the requirements which must be considered in producing a liposome preparation are similar to those of other controlled release mechanisms. They are: (1) a high percent of chemical entrapment; (2) increased chemical stability; (3) low drug toxicity; (4) rapid method of production; and (5) a reproducible size distribution.
  • Liposomes can also be formed as unilamellar vesicles (UVs), which generally have a size less than 0.5 ⁇ m ( ⁇ m, also referred to as "microns").
  • UVs unilamellar vesicles
  • Smaller unilamellar vesicles can be formed using a variety of techniques, such as applying a force sufficient to reduce the size of the liposomes and or produce smaller unilamellar vesicles.
  • Such force can be produced by a variety of methods, including homogenization, sonication or extrusion (through filters) of MLVs. These methods results in dispersions of UVs having diameters of up to 0.2 ⁇ m, which appear as clear or translucent suspensions.
  • Other standard methods for the formation of liposomes are know in the art, for example, methods for the commercial production of liposomes include the homogenization procedure described in U.S. Patent No. 4,753,788 to Gamble, a preferred technique, and the method described in U.S.
  • Patent 4,935,171 to Bracken which are incorporated herein by reference.
  • Another method of making unilamellar vesicles is to dissolve phospholipids in ethanol and inject them into a buffer, whereby the lipids will spontaneously rearrange into unilamellar vesicles.
  • This provides a simple method to produce UVs which have internal volumes similar to that of those produced by sonication (0.2-0.5 L/mol of lipid).
  • Another common method for producing small UVs is the detergent removal technique. Phospholipids are solubilized in either ionic or non-ionic detergents such as cholates, Triton X- 100, or n-alkylglucosides. The drug is then mixed with the solubilized lipid-detergent micelles.
  • Liposomes are then removed by one of several techniques: dialysis, gel filtration, affinity chromatography, centrifugation or ultrafiltration. The size distribution and entrapment efficiencies of the UVs produced this way will vary depending on the details of the technique used.
  • the therapeutic uses of liposomes include the delivery of drugs which are normally toxic in the free form. In the liposomal form the toxic drug may be directed away from the sensitive tissue and targeted to selected areas. Liposomes can also be used therapeutically to release drugs, over a prolonged period of time, reducing the frequency of administration. In addition, liposomes can provide a method for forming an aqueous dispersion of hydrophobic drugs for intravenous delivery.
  • liposomes When liposomes are used to target encapsulated drugs to selected host tissues, and away from sensitive tissues, several techniques can be employed. These procedures involve manipulating the size of the liposomes, their net surface charge as well as the route of administration. More specific manipulations have included labeling the liposomes with receptors or antibodies for particular sites in the body. The route of delivery of liposomes can also affect their distribution in the body. Passive delivery of liposomes involves the use of various routes of administration, e.g., intravenous, subcutaneous and topical. Each route produces differences in localization of the liposomes. Two methods used to actively direct the liposomes to selected target areas are binding either antibodies or specific receptor ligands to the surface of the liposomes. Antibodies are known to have a high specificity for their corresponding antigen and have been shown to be capable of being bound to the surface of liposomes, thus increasing the target specificity of the liposome encapsulated drug.
  • liposomes can be very useful in adapting these drugs for intravenous delivery. Furthermore, since liposomes are essentially hollow spheres made up of amphipathic molecules, they can entrap hydrophilic drugs in their aqueous interior space and hydrophobic molecules in their lipid bilayer. Unwanted molecules that remain in the dispersion external to the liposomes, such as unentrapped agents, are removed by column chromatography or ultrafiltration. Although methods for making liposomes are well known in the art, it is not always possible to determine a working formulation without experimentation.
  • United States Patent No. 4,310,505 discloses lipid vesicle formulations containing amino sugar derivatives of cholesterol as cell-surface receptor analogs.
  • the vesicles were found to release their contents in a controlled manner, and in some cases, to be rapidly concentrated in the lymphatic system and/or liver, lungs or spleen of the host. These liposomes were noted to be used in the treatment of lysosomal storage disease.
  • Liposomes containing amino-sugar derivatives and have been shown to localize in aggregates of polymorphonuclear leukocytes. Mauk, M.R. et al., "Vesicle Targeting: Timed Release and Specificity for Leukocytes in Mice by Subcutaneous Injection” Science 207:309-311 (1980).
  • arteriosclerosis a disease associated with the hardening or narrowing of arterial walls and the leading cause of death in Western Society.
  • atherosclerosis a type of arteriosclerosis characterized by lipid deposits in the intima of large and medium size arteries. These deposits or lesions are among three classes: fatty streaks, fibrous plaques and complicated lesions.
  • Fatty streaks probably the earliest lesions of atherosclerosis, are generally characterized by a build-up of lipid-filled smooth muscle cells, macrophage and fibrous tissue in the intima.
  • Advanced atherosclerosis gives rise to fibrous plaques, which are raised lesions and elevated areas of intimal thickening.
  • lesions consist of a central core of extracellular lipid and necrotic cell debris (gruel) covered by a fibromuscular layer containing large numbers of smooth muscle cells, macrophage, and collagen.
  • the third class known as the complicated lesion, is characterized by calcified fibrous plaques. Stenosis, or narrowing, of vascular passages can result from gradual occlusion as the plaques thicken and thrombi form.
  • Atherosclerosis and stenotic vascular lesions are typically treated through the use of percutaneous transluminal coronary angioplasty (PTC A) procedures. These methods, which include balloon angioplasty, the atherectomy catheter, the excimer laser, and the rotablator, are used to dilate the stenosed blood vessels.
  • PTC A percutaneous transluminal coronary angioplasty
  • the efficacy of PTC A is limited by the development of restenosis, or renarrowing, of the treated area following PTC A due to neointima formation.
  • Approximately 25% of successful first- time angioplasty procedures must be repeated within 6 months and an additional 10% of the patients must later undergo coronary bypass surgery because of restenosis.
  • a discussion of the causative elements of restenosis appears in Bone, R.C., ed., "Restenosis after Coronary Angioplasty," Disease- A-Month, 39:616-670 (1993).
  • Neointima formation is a multi- step process involving a complex interaction among several growth factors which promote vascular smooth muscle cell (VSMC) proliferation and migration. In this process, erosion of the intima, the single continuous layer of endothelial cells lining the arterial walls, during angioplasty promotes platelet aggregation.
  • VSMC vascular smooth muscle cell
  • the platelets adhere to the arterial wall and release growth factors (e.g., platelet-derived growth factor, thrombin, basic fibroblast growth factor and transforming growth factor) which initiate the migration of VSMC to the intima and promote the proliferation of VSMC comprising the tunica media.
  • growth factors e.g., platelet-derived growth factor, thrombin, basic fibroblast growth factor and transforming growth factor
  • Growth factor-induced VSMC proliferation involves, in turn, a sequential activation of intracellular proteins which promote cell-cycle progression.
  • the next step of the process is associated with an inflammatory response, evidenced by an invasion of inflammatory cells such as monocytes, macrophage, and other white blood cells.
  • inflammatory cells further induce and stimulate growth of VSMC in the media, which subsequently migrate into the intimal space and produce large amounts of extracellular proteolytic matrix material, forming a neointima layer which represents the lesion. It appears that endothelium then grows over the injured area and restenosis results.
  • drugs considered for inhibiting restenosis include, but are not limited to, oligonucleotides (e.g., antisense gene therapy), protein kinase C, endothelial growth factor and anti-platelet activating agents.
  • oligonucleotides e.g., antisense gene therapy
  • protein kinase C protein kinase C
  • endothelial growth factor e.g., endothelial growth factor
  • anti-platelet activating agents e.g., anti-platelet activating agents.
  • Efforts to prevent restenosis utilizing systemic administration of such drugs have been hampered by generalized toxicity at effective dosages.
  • successful therapy for preventing restenosis has been thwarted by the complexity of the physiological processes responsible for neointima formation as well as the inability to deliver effective quantities of drugs to the site of injury, due to inefficient cellular uptake and lack of specificity (i.e.. targeting) to VSMC.
  • PCNA a nuclear protein required for DNA synthesis by DNA polymerase ⁇
  • cdc2 a serine/threonine protein kinase
  • HVJ Sendai hemagglutinating virus of Japan
  • the invention provides for the delivery of an imaging or therapeutic agent into the cytoplasm of a cell by delivering the lipid construct to a mammal wherein the lipid construct comprises an aminomannose derivatized cholesterol.
  • the delivery of the agents could be done in vitro as well as in vivo. This includes delivery of agents to cells, tissues or organs in vitro and reintroduction of the cells, tissues or organs in vivo. It also includes the transfection of cultured cells in vitro for the purpose of conferring desirable properties in these cells, including enhanced, decreased or novel production of proteins.
  • Lipid constructs for purposes of this invention, are complexes containing lipids, phospholipids, or derivatives thereof comprising a variety of different structures which lipids are known to adopt in aqueous suspension. These structures include, but are not limited to, micelles, liposomes, lipid ribbons or sheets, and may be complexed with a variety of drugs and adjuvants which are known to be pharmaceutically acceptable. Common adjuvants include cholesterol and ⁇ -tocopherol, among others. The lipids may be used alone or in any combination which one skilled in the art would appreciate to provide the characteristics desired for a particular application.
  • the present invention further comprises a method for targeting vascular smooth muscle tissue and cells comprising the delivery of an agent to the cell using a lipid construct comprising an aminomannose derivatized cholesterol.
  • This approach is advantageous over the prior art since it provides targeting but does not involve the use of viral products.
  • the aminomannose-containing lipid constructs exhibit dramatically enhanced in vitro and in vivo targeting to VSMC, compared to identical constructs not containing aminomannose. While not wishing to be bound by any particular theory, it appears that the increased targeting results from specific mannose- receptor-mediated endocytosis or fusion of the lipid constructs.
  • the lipid constructs contain drugs which inhibit neointima formation and are administered to prevent restenosis following PTCA procedures. Since restenosis is caused generally from the accumulation of Vascular Smooth Muscle Cells, the targeting of such cells is critical during PTCA procedures. Also provided herein is a method of delivering a lipid composition into a vascular smooth muscle cell by delivering a lipid construct to a mammal comprising aminomannose derivatized cholesterol.
  • the preferred composition for carrying out the invention includes a lipid construct comprising an aminomannose derivatized cholesterol.
  • Further embodiments of the invention include the construct having a cationic lipid wherein the total cationic lipid is greater than or equal to 5% of total lipid and the lipid construct further comprises a therapeutic or imaging agent.
  • the aminomannose derivatized cholesterol is included in the definition of a cationic lipid.
  • the invention is useful for inhibiting restenosis in a mammal by delivering a lipid construct to the mammal wherein the lipid construct comprises an aminomannose derivatized cholesterol and a cationic lipid wherein the total cationic lipid is greater than or equal to 5% of total lipid wherein the lipid construct further comprises an agent in an amount effective for said inhibition.
  • the 5% cationic lipid could include only aminomannose derivatized cholesterol.
  • Another aspect of the invention comprises the cytoplasmic delivery of an agent into the cytoplasm of a cancer cell.
  • Figure 1 shows a freeze fracture micrograph of a lipid construct- containing oligonucleotides.
  • Figure 2 shows a confocal micrograph of fluorescently labeled chimeric oligonucleotide taken at 2.5 hours.
  • Figures 3A & 3B show a confocal micrograph of DOPE:Amch complexed with labeled chimeric oligonucleotide taken at 2.5 hours and 16 hours respectively.
  • Figures 4A & 4B show a confocal micrograph of DSPC:Chol complexed with labeled chimeric oligonucleotide taken at 2.5 hours and 16 hours respectively.
  • the present invention relates to the formation of a lipid construct comprising an aminomannose derivatized cholesterol.
  • a preferred representative of this compound is 6-(cholest-5-en-3 ⁇ -yloxy)hexyl-6-amino-6- deoxy- 1 -thio- ⁇ -D-mannopyranoside ( Amch).
  • lipids may be used in combination with aminomannose derivatized cholesterol.
  • lipids which may be used successfully include, but are not limited to: dimyristoyl triethylamine propane (DMTAP), dimyristoyl dimethylammonium propane (DMDAP), l,2-dioleoyl-3- trimethylammonium propane (DOTAP), l,2-dioleoyl-3-dimethylammonium propane (DODAP) and dioleoyl-sn-glycero-3-ethyl phosphatidylcholine (DGE), hydrogenated soy and egg phosphatidyl choline (HSPC, HEPC) distearoyl phosphatidylcholine (DSPC), Dimyristoyl phosphatidylcholine (DMPC), dilauryl phoshatidylcholine (DLPC), dioleoyl phosphatidycholine (DOPC), phosphatidyl
  • DMTAP
  • lipid constructs may also be used to treat or prevent cancer or viral infections by facilitating delivery of oligonucleotide therapeutic agents to the cellular cytoplasm.
  • These lipids may be used in any suitable combination with an aminomannose derivatized cholesterol.
  • Preferred lipid formulations are listed in Tables 1 & 2. The following formulations give rise to optimal results in preventing restenosis: DOPE:Amch (1 :1 molar ratio) having a lipid to oligonucleotide ratio (w/w) less than 5:1 and preferably between 1.25: 1 & 0.5:1.
  • the preferred size of these construct is range from 1 Onm to 250nm preferably at 50nm.
  • Preferred lipids used in the constructs comprise cationic lipids, preferably greater than or equal to 5% total.
  • the preferred lipid construct is a unilamellar liposome as shown in Figure 1 wherein the liposome is shown prepared having a formula DOPE:Amch (1 : 1 molar ratio).
  • the lipid constructs preferred for use in delivering oligonucleotides which inhibit tumor growth comprise HSPC:Chol:Amch in a mole ratio of 16:8:1.
  • the preferred lipid constructs are prepared by first hydrating a mixture of lipids in suitable buffer such as 9% sucrose. Prior to hydration, it is preferable that a lipid powder or film of the lipid mixture is formed, for example, by spray-drying or rotary evaporative techniques.
  • One preferred use of the invention is in the treatment of restenosis. and further for the prevention of neointima formation.
  • Agents that may be used include the following: antithrombotic agents, antiplatelet agents, antiproliferative agents, anti-inflammatory agents, calcium antagonists, lipid lowering drugs, and antibodies to growth factors, inositol diphosphate, and antisense compounds.
  • Some of these agents include coumadin, heparin, low molecular weight heparins, hirudin, aspirin, dipyridamole, thromboxane A2, thromboxane A2 synthetase inhibitor, thromboxane A2 receptor blockers, ridogrel, sulfinpyrazone, dextran, prostacyclin or prostacyclin analogues, prostaglandin El, ticlopidine, serotonin antagonists, glycoprotein Ilb/IIIa receptor blockers, angiotensin converting enzyme inhibitors, colchicine, platelet-derived growth factor antagonists, angiopeptin, somatomedin, octreotide, cytostatic, corticosteroids, ibuprofen, nifedipine, nicardipine, diltiazem, verapamil, fish oils, (such as omega 3 fatty acids, eicosapentaenoic acids, and
  • Microparticles have been noted as a potential microcarrier drug delivery system in order to prevent the rapid elimination by the increase network of vasa vasorum and atherosclerotic lesions that cause early outward diffusion. Red blood cells have also been investigated as a microcarrier system.
  • the preferred agents used are oligonucleotides that inhibit neointima formation and prevent restenosis.
  • the preferred size of the oligonucleotides is from 15 mer to 30 mer, although sizes up to 80 mer may be associated with the lipid construct taught in this invention.
  • the present invention is also useful for blocking cell-cycle regulatory genes such as proliferating-cell nuclear antigen (PCNA), p34cdc2 (cdc2), and cdk2 (cyclin-dependent kinases).
  • PCNA proliferating-cell nuclear antigen
  • cdc2 p34cdc2
  • cdk2 cyclin-dependent kinases
  • This invention has particular advantages for complexing aminomannose derivatized cholesterol with oligonucleotide ligands generated by the SELEX ® technology as described in U.S. Patent Number 5,270,163 and incorporated herein by reference. This invention will be more fully understood by references to the following examples, which are intended to be illustrative of the invention, but not limiting thereof.
  • Example 1 Liposomal Targeting and Uptake of Smooth Muscle Cells A. Preparation of Liposomes.
  • a lipid film containing distearoyl phosphatidylcholine (DSPC) and cholesterol (Choi) in a 2: 1 molar ratio was prepared.
  • a second lipid film containing DSPC, Choi and aminomannose derivatized cholesterol (Amch) in a molar ratio of about 2:0.96:0.04 was also prepared.
  • the lipid films were formed by dissolving the lipids in chloroform, mixing and then evaporating off the chloroform.
  • the lipid films were radiolabeled with 3 H-cholesteryl hexadecyl ether and/or fluorescently labeled with the lipophilic fluorescent probe, octadecyl rhodamine.
  • the 3 H- cholesterylhexadecyl ether (5 ⁇ Ci) was added during the formation of the lipid film.
  • the lipids were hydrated in 100 mM NaCl and 25 mM Phosphate buffer pH 7.4 and probe sonicated for approximately 10 minutes to form unilamellar vesicles with an average diameter of 70 nm ⁇ 20 nm.
  • the octadecyl rhodamine was incubated with the liposomes for 20 minutes to fluorescently label the liposomes.
  • the rabbits (6 arteries/group) were sacrificed at 0.5, 1, 4, 24 and 168 hours. Rabbits injected with normal saline or octadecyl rhodamine served as controls (4 arteries/group). The iliac arteries were snap frozen in liquid nitrogen and the rhodamine fluorescence was measured using an Olympus fluorescence microscope.
  • the liposomes were accumulated under the adventitia. At four hours, the liposomes began to distribute in the medial layer. At twenty-four hours, the liposomes were distributed in a heavy homogeneous pattern throughout the width of the arterial wall. At one week, the intima remained stained and the liposomes were redistributed into the adventia with the media being clear. The blood vessels of rabbits are somewhat peculiar in that the intima is extremely thin. Therefore, any intima seen after catheterization is assumed to be neointima, formed as a result of migration of VSMC from the media and consequent release of extracellular matrix. By 24 hours, the liposomes redistribute throughout the vessel. At one week the fluorescent dye is clearly seen within the neointima and not the media. Liposomes associated with VSMC within the media migrated across the internal elastica into the neointima.
  • lipid constructs comprising an aminomannose derivatized cholesterol target vascular smooth muscle cells (VSMC).
  • VSMC vascular smooth muscle cells
  • Continuing fluorescence microscopy confirmed delivery of the lipid construct to VSMC, evidenced by binding of aminomannose derivatized liposomes to the VSMC tissue.
  • Autoradiography confirmed delivery into VSMC, evidenced by intracellular localization of the lipid within the VSMC.
  • Example 2 Liposomes containing greater than 5% cationic lipid were prepared. The lipids were hydrated in sterile-filtered 9% sucrose, heated for 0.5 minutes at 65°C and sonicated. Sizing was done by microtrac analysis.
  • Oligonucleotides (2328) GTCCTCCATAGTTACTCA (SEQ ID NO.: 1), (2342) GATCAGGCGTGCCTCAAA (SEQ ID NO.: 2), (2616) ACTCATTGATACCTCCTG (SEQ ID NO.: 3) and
  • AAACTCCGTGCGGACTAG (SEQ ID NO.: 4) were added to the liposomes and heated at 65°C for 10 minutes.
  • Table 2 lists all cationic formulations, lipid concentrations and liposomes sizes.
  • DMTAP DMPE: Amch (1:1:1) 3.1 1.761410.5416(1%) 0.012310.0090(99%) Fo ⁇ nulation#* Lipid Size( ⁇ __ ⁇ ) Conc.(mgmL)
  • DODAP DOPE:amino (1:1:1) 3.72 0.88810.5061 (11%)
  • DOTAP:DOPE:Amch (1.3:1:1.2) 3.0 1.953410.7815(14%) 0.054110.0867(86%)
  • liposomes were sonicated until greater than 85% of the liposomes were less than 300nm diameter. This allowed sterile filtration using a 450nm cellulose acetate filter without significant liposome loss on the filter.
  • Al liposome formulations except DOT AP:DOPE: Amch (1 : 1 :1), DODAP:DOPE:Amch (1:1 :1) and DOSPA:DOPE:Amch were 10-lOOnm average diameter.
  • Example 3 Lipid constructs were prepared as in Example 2. Electron microscopy were obtained of liposomes prepared with DOPE: Amch (1 : 1, mole ratio) and mixed with oligonucleotides in a 5:1 lipid:oligo ratio and immediately frozen ( Figure 1). Figure 1 clearly indicates that unilamellar liposomes are formed
  • oligonucleotides targeted to the CAT gene were complexed (and combined), with the lipid construct.
  • the sequences of the oligonucleotides were (2520) TAGCTTCCTTAGCTCCTGCAT (SEQ ID NO.: 5) and (2519) TAGCTT CCGCAACTCTTGCAT (SEQ ID NO.: 6), both antisense to a mRNA which codes for chloramphenicol acetyl transferase (CAT).
  • Chloramphenicol is an antibiotic which blocks protein synthesis by inhibiting the peptidyl transferase activity of the 50S ribosomal subunit in prokaryotes; peptidyl transferase catalyzes the formation of peptide bonds.
  • CAT is a bacterial drug-resistance gene which inactivates chloramphenicol by acetylating the drug at one or both of its two hydroxyl groups.
  • CAT assay two expression plasmids containing modified bacterial CAT genes were constructed. These plasmids were pG 1000 and pG1007 as taught in Reynolds, M.A. "Triple-Strand-Forming methylphosphonate oligonucleotides targeted to mRNA efficiently block protein synthesis," Proc Acad. Sci. 91 :12433-12437 at 12434 (1994), with the following modifications; the amplified DNA fragment containing the sequence target site for oligonucleotides 2519 or 2520 was substituted for the polypyrimidine insert. The two plasmids differed from one another by four bases in the translation initiation region targeted by the oligos.
  • This gene is usually not found in eukaryotes; thus there is little or no background CAT activity in eukaryotic cells.
  • Cell extracts were incubated with 3 H or 14 C chloramphenicol and n-butyryl coenzyme A.
  • the modified chloramphenicol partitioned into the xylene fraction while the unmodified chloramphenicol remained in the aqueous phase.
  • Cos 7 cells or A 10 smooth muscle cells were plated the day before the experiment so that confluency was approximately 70% on the day of the experiment.
  • Formulations of lipofectin complexed with plasmid and lipid constructs complexed with phosphorothioate oligonucleotides were diluted in OptiMEM, a serum-free medium. The cells are washed once with OptiMEM and then incubated for 3 hr with 0.5 mL of the diluted liposome-oligo complexes. The liposomes were removed and washed with OptiMEM and transfected for 3 hours with the target plasmid complexed with Lipofectin. The plasmid complexes were removed and the cells were cultured overnight in complete medium. The cells were harvested approximately 16 hours after the addition of the target plasmid. Cell lysates were analyzed for CAT activity to assess inhibition and protein content. The results are displayed in Table 3. TABLE 3
  • a stably inducible model was also evaluated using COS 7 cells and A10 smooth muscle cells. These cells were stably transfected with a plasmid containing an inducible CAT gene using LipofectAmine (DOSPA:DOPE 3 : 1 mole ratio).
  • COS 7 and A10 smooth muscle cells were plated the day before the experiment, treated with the liposomes and washed as described above. The liposome-oligo complexes were removed, washed in OptiMEM and cultured overnight in medium containing CdCl 2 , which induces protein production. The cells were harvested approximately 16 hours later and analyzed for CAT activity. Cells were treated with ⁇ M oligo. The results are displayed in Tables 4 and 5.
  • pG 1041 The plasmid containing an inducible CAT gene is pG 1041 which has a modified chloramphenicol acetyl transferase (CAT) gene inserted into pMEP4 (Clonetech).
  • CAT chloramphenicol acetyl transferase
  • a modified chloramphenicol acetyl transferase (CAT) gene producing wild- type CAT protein was amplified in a PCR reaction using the following primer: 5' primer:
  • the first adenosine of the open reading frame is designated + 1.
  • the coding region nucleotide base changes between wild-type and pG1041 are conservative and do not change the amino acid sequence.
  • lipid constructs comprised of Aminomannose derivatized cholesterol, DOPE:Amch (1 : 1 molar ratio) and DSPC hol (2: 1 molar ratio) were prepared as in Examples 1 & 2.
  • the DOPE: Amch lipid constructs were incubated for 10 minutes at 65 °C with a fluorescently labeled chimeric oligonucleotide having the sequence 5'-GGTATATCCAGTGATC ⁇ C ⁇ CTC-3' (SEQ ID NO.: 13).
  • the lipid to oligonucleotide ratio was 5:1 w/w.
  • DSPCxhol liposomes were prepared by hydrating the lipids with buffer containing the chimeric oligonucleotides and sonication.
  • Six flasks containing 10 6 PI 798 (lymphocarcinoma cells) cells in 10 ml complete Iscoves (10% Fetal Calf Serum, 1% penicillin/streptomycin) were incubated at 37°C, 5% C0 2 . The cells were then incubated with 50 ⁇ g of oligonucleotide, either as free oligo or lipid associated oligo. Flasks 1, 2 and 3 were incubated with free oligo or oligo complexed with one of the two lipid formulations overnight (16 hour incubation).
  • Flasks 4, 5 and 6 were incubated the next morning with free oligo or oligo complexed with one of the two lipid formulations (2.5 hour incubation). Cells were removed at 2.5 and 16 hours. The cells were washed to remove unassociated liposomes/oligonucleotide and examined by confocal microscopy. For each sample, a confocal microscopy series and corresponding bright field image were recorded. That is, a midsection fluorescence (top), bright-field (middle) and 3-d reconstruction (bottom) are displayed in figures 2-4.
  • Figures 2A and 2B show rapid and intense fluorescence diffusing from the edge of the cells into the cytoplams.
  • Amch:DOPE lipid constructs exhibit rapid and concentrated fluorescence into the cytoplasm as shown by intense fluorescence in the cytoplasm whereas the cells incubated with free oligonucleotide and oligonucleotide associated with DSPCxhol constructs shows very little fluorescence at all time periods.
  • EXAMPLE 5 A transfection study was carried out to further verify the ability of aminomannose derivatized cholesterol lipid constructs to deliver oligonucleotides into the nucleus of a cell.
  • LFA is currently used to transfect a plasmid, that is, introduce the plasmid into the nucleus of a cell so that it expresses a protein for which it codes.
  • the plasmid was pG1035 which has a modified chloramphenicol acetyl transferase (CAT) gene inserted into pRc/CMV (Invitrogen).
  • pG1035 was produced as follows. A modified chloramphenicol acetyl transferase (CAT) gene producing wild- type CAT protein was amplified in a PCR reaction using the following primers: 5' primer:
  • LFA is optimized at a lipid:plasma w/w ratio of 6: 1.
  • the following lipid:plasmid constructs were formed: DOPE:Amch (1 : 1 molar ratio) with a lipid plasmid ratio w/w of 10: 1 ; DMPE:Amch (1 : 1 and 1 : 1.5 molar ratios) with lipid plasmid ratio w/w of 10: 1 ; DODAP:DOPE:Amch ( 1 : 1 : 1 ) at lipid plasmid ratios w/w of 15: 1 and 10:1.
  • the DOPE:Amch lipid constructs were 1.14 times more efficacious then LFA.
  • the Aminomannose constructs were effective in transfecting AlO smooth muscle cells.
  • Lipid constructs were formulated as in Example 2 using the following Oligonucleotides: cdc gene (rat) GTCCTCCATAGTTACTCA (SEQ ID NO.: 1); PCNA gene (rat) GATCAGGCGTGCCTCAAA (SEQ ID NO.: 2); rev-AS (2328) ACTCATTGATACCTCCTG (SEQ ID NO.: 3); rev-AS (2342) AAACTCCGTGCGGACTAG (SEQ ID NO.: 4).
  • Ten unheparinized rats were treated with 10 ⁇ M total oligo (antisense or reverse antisense) complexed with DOPE: aminomannose (1 :1) at a 5:1, 1.25:1 or 0.5:1 lipidxligo w/w ratios and sacrificed 2 weeks post-angioplasty.
  • Eight heparinized rats were treated with 10 ⁇ M total oligo (antisense or reverse antisense) complexed with DOPE:aminomannose liposomes (1 : 1) at 5: 1 lipid:oligo w/w ratio and sacrificed 2 weeks post-angioplasty.
  • a No. 2 French Fogarty catheter was used to induce vascular injury in male Spraque-Dawley rats.
  • the rats were anesthetized, and a cannula introduced into the left common carotid via the external carotid artery. After vascular injury of the common carotid, the distal injured segment was transiently isolated by temporary ligatures.
  • the lipid complex formulation (0.25 ml) containing the antisense oligos (5 ⁇ M of each) was infused into the segment and incubated for 15 - 30 minutes at room temperature.
  • Control rats received an identical formulation containing reverse antisense oligos (rev- AS). Following incubation, the infusion cannula was removed and blood flow to the common carotid restored by release of the ligatures.
  • Lipid constructs were prepared as in Example 2 and complexed with oligonucleotides as in Example 6. Rats were heparinized and treated with constructs complexed with cdc2 and PCNA (0.5: 1 lipidxligo w/w and 50 ⁇ M oligo, 0.5:1 lipidxligo w/w and 5 ⁇ M oligo, 0.25:1 lipidxligo w/w and 5 ⁇ M oligo) or reverse cdc2 and PCNA (0.5: 1 lipidxligo w/w and 5 ⁇ M oligo) phosphorothioate oligonucleotides. The results of the study are listed below in Table 6.
  • EXAMPLE 8 Stock solutions of the oligonucleotides were prepared as followed: antisense, c-myc:5'-AACGTTGAGGGGCAT-3' (SEQ ID NO.: 15) and nonsense c-myc:5'-CTGAAGTGGCATGAG-3' (SEQ ID NO.: 16) were dissolved in phosphate buffer to make a stock solution with a concentration of approximately 3.4 mg/ml.
  • a lipid film was prepared containing hydrogenated soy phosphatidylcholine:chol:Amch in a mole ratio of 16:8:1 having a total lipid concentration of 560 milligrams.
  • oligonucleotide stock solution was added to the lipid film and bath sonicated at a temperature of 40° C for 10 seconds.
  • the resulting solution was put through a 4 cycle freeze- thaw procedure using liquid nitrogen.
  • the resulting homogeneous solution was extruded first through a 0.8 ⁇ M filter membrane (3 times) then extruded through a 0.45 ⁇ M filter (3 times) and finally extruded through a 0.2 ⁇ M filter (3 times).
  • the free oligonucleotides were removed by passing through a Sephadex G-50 column with a bed volume of about 18 ml. The vesicle volume was recovered in 5.5 ml of buffer.
  • Control liposomes were prepared as above without oligonucleotides.
  • the mean diameter of the control liposomes was 80.0 ⁇ 30 8.4nm.
  • the mean diameter of the oligonucleotides containing liposomes were 210 ⁇ 80nm.
  • the antisense c-myc and oligonucleotides concentration was 50 ⁇ g/ml.
  • the liposomes containing oligonucleotides were tested against two strains of small cell lung carcinoma.
  • the first strain was a cisplatin (cDDP).
  • sensitive line (GLC4)
  • the second strain was a 5.6x cDDP resistant sub-line (GLC4-cDDP).
  • GLC4 and GLC4-cDDP there is a 40-60x c-myc oncogene amplification with a corresponding c-myc mRNA and protein overexpression.
  • the lipid concentration in the control group was 21.8 mg/ml and the lipid concentration in the liposome preparation containing the oligonucleotides was 10.3 mg/ml.
  • Table 1 disclose that encapsulated oligonucleotides in liposomes allow for a 10 to 20 times reduction of antisense c-myc oligonucleotides concentration needed to exert the same inhibitory effect in the GLC4 and GLC4-cDDP cell lines compared to the unencapsulated antisense c-myc oligonucleotides. Empty liposomes and nonsense oligonucleotides did not have an inhibitory effect.
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • ANTI-SENSE YES
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICA NO
  • ANTI-SENSE NO
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • ANTI-SENSE YES
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • ANTI-SENSE YES
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • ANTI-SENSE YES
  • Met Glu Lys Lys lie Ser Gly Tyr Thr Thr 1 5 10
  • MOLECULE TYPE mRNA
  • HYPOTHETICAL NO
  • MOLECULE TYPE protein
  • HYPOTHETICAL NO
  • Met Glu Lys Lys lie Ser Gly Tyr Thr Thr 1 5 10
  • MOLECULE TYPE mRNA
  • HYPOTHETICAL NO
  • ANTI-SENSE NO
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL NO
  • ANTI-SENSE NO

Abstract

A lipid construct comprising an aminomannose derivatized cholesterol suitable for targeting smooth muscle cells and tissue. Preferred formulations contain 6-(cholest-5-en-3β-yloxyl)hexyl-6-amino-6-deoxy-1-thio-α-D-mannopyranoside in liposome formulations wherein the formulations are delivered generally to arteries using percutaneous transluminal coronary angioplasty procedures. These formulations have applications in the reduction of restenosis.

Description

LIPID CONSTRUCTS FOR CYTOPLASMIC DELIVERY OF AGENTS
FIELD OF THE INVENTION
This invention relates generally to pharmaceutical sciences and more particularly to the production of lipid constructs for the delivery of drugs to the cytoplasm of a cell and in particular to vascular smooth muscle tissue. A preferred use of this invention relates to the administration of these constructs for the prevention of restenosis following angioplasty.
BACKGROUND OF THE INVENTION In order for many drugs and imaging agents to have therapeutic potential it is necessary for them to be delivered to the proper location in the body. In many cases, agents are ineffective because it is impossible, with the available technology, to deliver them to the proper locations within a biological organism. Thus, a main focus in pharmaceutical research is in the delivery of agents to tissues and cells.
An area of development aggressively pursued by researchers is in the delivery of agents not only into a cell but into the cell's cytoplasm and further yet, into the nucleus. This area of research is being pursued in particular for delivery of biological agents such as DNA, RNA, ribozymes and proteins. Not only do these materials hold great promise as therapeutic agents, but some workers believe that in some diseases they may act as the "magic bullet" i.e., curing an illness without deleterious side effects. Examples of therapeutic pursuits include the application of antisense technology. Briefly, the strategy in antisense technology is to deliver an agent such as DNA oligonucleotide that binds to sites on messenger RNA (mRNA) which directs the production of proteins related to disease. Other strategies include the use of triplex agents which bind to the double helix to interfere with transcription, preventing the production of mRNA. In spite of these developments, numerous "delivery" problems have surfaced.
One major problem encountered is that effective delivery of oligonucleotides across cell membranes is difficult, if not impossible. Another problem facing workers is that oligonucleotides are degraded by intracellular and extracellular enzymes such as exonucleases and endonucleases. In order to overcome these problems, workers have modified the typical phosphodiester form of oligonucleotides. For example, modifications have included the creation of phosphorothioate and methylphosphonate oligonucleotides. Methylphosphonate oligonucleotides contain uncharged sectors which increase intracellular uptake and resist enzymatic degradation. Phosphorothioate oligonucleotides are negatively charged but are resistant to endonucleases. Although these modified oligonucleotides show promise as therapeutics, there is no evidence of in vivo success. Further, workers have not provided a delivery vehicle capable of introducing a sufficient quantity of oligonucleotides into the cell such that it has therapeutic effect. Since it is desirable to load a cell with numerous oligonucleotides, workers have begun to focus their efforts on developing a delivery vehicle which not only acts to deliver an agent into the cytoplasm and nucleus of a cell, but also delivers oligonucleotides in large numbers. Cationic liposomes have been used to introduce DNA into vascular cells. Plautz, G.E., et al, "Liposome Mediated Gene Transfer into Vascular Cells", J. Liposome Research 3(2): 179- 199 (1993); Feigner, P.L., et al. "Lipofection: A highly efficient, lipid-mediated DNA-transfection procedure", Proc. Natl Acad. Sci. 84:7413-7417 (1987). For example, Lipofectin, liposomes formed from a mixture of N-[2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA) and dioleoylphosphatidylethanolamine (DOPE), has been used to transduce recombinant genes into coronary arteries in vivo. Another example of a cationic liposome used to introduce genes into DNA is a liposome preparation containing DC-chol, 3b[N-9N'N'-dimethylaminoethane)- carbamoyl] cholesterol and DOPE. Other efforts have been directed toward the use of modified retroviruses and cationic liposomes for gene transfer. Liposomes in general have been known. Liposomes are microscopic vesicles made from phospholipids, which form closed, fluid filled spheres when dispersed with aqueous solutions. Phospholipid molecules are polar, having a hydrophilic head and two hydrophobic tails consisting of long fatty acid chains. Thus, when a sufficient concentration of phospholipid molecules are present in aqueous solutions, the tails spontaneously associate to exclude water while the hydrophilic phosphate heads interact with water. The result is a spherical, bilayer membrane in which the fatty acid tails converge in the interior of the newly formed membrane, and the polar heads point in opposite directions toward an aqueous medium. These bilayer membranes thus form closed spheres known as liposomes. The polar heads at the inner surface of the membrane point toward the aqueous interior of the liposome and, at the opposite surface of the spherical membrane, the polar heads interact with the surrounding aqueous medium. As the liposomes are formed, water soluble molecules can be incorporated into the aqueous interior, and lipophilic molecules may be incorporated into the lipid bilayer. Liposomes may be either multilamellar, like an onion with liquid separating many lipid bilayers, or unilamellar, with a single bilayer surrounding an aqueous center.
Methods for producing liposomes are well known in the art, and there are many types of liposome preparation techniques which may be employed to produce various types of liposomes. These can be selected depending on the use, the chemical intended to be entrapped, and the type of lipids used to form the bilayer membrane. The requirements which must be considered in producing a liposome preparation are similar to those of other controlled release mechanisms. They are: (1) a high percent of chemical entrapment; (2) increased chemical stability; (3) low drug toxicity; (4) rapid method of production; and (5) a reproducible size distribution.
The first method described to encapsulate drugs or other chemicals in liposomes involved the production of multilamellar vesicles (MLVs). Liposomes can also be formed as unilamellar vesicles (UVs), which generally have a size less than 0.5 μm (μm, also referred to as "microns"). There are several techniques known in the art which are used to produce unilamellar liposomes.
Smaller unilamellar vesicles can be formed using a variety of techniques, such as applying a force sufficient to reduce the size of the liposomes and or produce smaller unilamellar vesicles. Such force can be produced by a variety of methods, including homogenization, sonication or extrusion (through filters) of MLVs. These methods results in dispersions of UVs having diameters of up to 0.2 μm, which appear as clear or translucent suspensions. Other standard methods for the formation of liposomes are know in the art, for example, methods for the commercial production of liposomes include the homogenization procedure described in U.S. Patent No. 4,753,788 to Gamble, a preferred technique, and the method described in U.S. Patent 4,935,171 to Bracken, which are incorporated herein by reference. Another method of making unilamellar vesicles is to dissolve phospholipids in ethanol and inject them into a buffer, whereby the lipids will spontaneously rearrange into unilamellar vesicles. This provides a simple method to produce UVs which have internal volumes similar to that of those produced by sonication (0.2-0.5 L/mol of lipid). Another common method for producing small UVs is the detergent removal technique. Phospholipids are solubilized in either ionic or non-ionic detergents such as cholates, Triton X- 100, or n-alkylglucosides. The drug is then mixed with the solubilized lipid-detergent micelles. Detergent is then removed by one of several techniques: dialysis, gel filtration, affinity chromatography, centrifugation or ultrafiltration. The size distribution and entrapment efficiencies of the UVs produced this way will vary depending on the details of the technique used. The therapeutic uses of liposomes include the delivery of drugs which are normally toxic in the free form. In the liposomal form the toxic drug may be directed away from the sensitive tissue and targeted to selected areas. Liposomes can also be used therapeutically to release drugs, over a prolonged period of time, reducing the frequency of administration. In addition, liposomes can provide a method for forming an aqueous dispersion of hydrophobic drugs for intravenous delivery.
When liposomes are used to target encapsulated drugs to selected host tissues, and away from sensitive tissues, several techniques can be employed. These procedures involve manipulating the size of the liposomes, their net surface charge as well as the route of administration. More specific manipulations have included labeling the liposomes with receptors or antibodies for particular sites in the body. The route of delivery of liposomes can also affect their distribution in the body. Passive delivery of liposomes involves the use of various routes of administration, e.g., intravenous, subcutaneous and topical. Each route produces differences in localization of the liposomes. Two methods used to actively direct the liposomes to selected target areas are binding either antibodies or specific receptor ligands to the surface of the liposomes. Antibodies are known to have a high specificity for their corresponding antigen and have been shown to be capable of being bound to the surface of liposomes, thus increasing the target specificity of the liposome encapsulated drug.
Since the chemical composition of many drugs precludes their intravenous administration, liposomes can be very useful in adapting these drugs for intravenous delivery. Furthermore, since liposomes are essentially hollow spheres made up of amphipathic molecules, they can entrap hydrophilic drugs in their aqueous interior space and hydrophobic molecules in their lipid bilayer. Unwanted molecules that remain in the dispersion external to the liposomes, such as unentrapped agents, are removed by column chromatography or ultrafiltration. Although methods for making liposomes are well known in the art, it is not always possible to determine a working formulation without experimentation.
United States Patent No. 4,310,505, incorporated by reference, discloses lipid vesicle formulations containing amino sugar derivatives of cholesterol as cell-surface receptor analogs. The vesicles were found to release their contents in a controlled manner, and in some cases, to be rapidly concentrated in the lymphatic system and/or liver, lungs or spleen of the host. These liposomes were noted to be used in the treatment of lysosomal storage disease. Liposomes containing amino-sugar derivatives and have been shown to localize in aggregates of polymorphonuclear leukocytes. Mauk, M.R. et al., "Vesicle Targeting: Timed Release and Specificity for Leukocytes in Mice by Subcutaneous Injection" Science 207:309-311 (1980).
The delivery problems noted above become manifest in treating a disease such as arteriosclerosis, a disease associated with the hardening or narrowing of arterial walls and the leading cause of death in Western Society. The majority of these deaths is caused by atherosclerosis, a type of arteriosclerosis characterized by lipid deposits in the intima of large and medium size arteries. These deposits or lesions are among three classes: fatty streaks, fibrous plaques and complicated lesions. Fatty streaks, probably the earliest lesions of atherosclerosis, are generally characterized by a build-up of lipid-filled smooth muscle cells, macrophage and fibrous tissue in the intima. Advanced atherosclerosis gives rise to fibrous plaques, which are raised lesions and elevated areas of intimal thickening. These lesions consist of a central core of extracellular lipid and necrotic cell debris (gruel) covered by a fibromuscular layer containing large numbers of smooth muscle cells, macrophage, and collagen. The third class, known as the complicated lesion, is characterized by calcified fibrous plaques. Stenosis, or narrowing, of vascular passages can result from gradual occlusion as the plaques thicken and thrombi form.
Atherosclerosis and stenotic vascular lesions are typically treated through the use of percutaneous transluminal coronary angioplasty (PTC A) procedures. These methods, which include balloon angioplasty, the atherectomy catheter, the excimer laser, and the rotablator, are used to dilate the stenosed blood vessels. However, the efficacy of PTC A is limited by the development of restenosis, or renarrowing, of the treated area following PTC A due to neointima formation. Approximately 25% of successful first- time angioplasty procedures must be repeated within 6 months and an additional 10% of the patients must later undergo coronary bypass surgery because of restenosis. A discussion of the causative elements of restenosis appears in Bone, R.C., ed., "Restenosis after Coronary Angioplasty," Disease- A-Month, 39:616-670 (1993).
PTCA procedures often cause collateral injury to the arterial wall, which triggers a "healing" process involving a series of physiologic events that ultimately results in neointima formation. Neointima formation is a multi- step process involving a complex interaction among several growth factors which promote vascular smooth muscle cell (VSMC) proliferation and migration. In this process, erosion of the intima, the single continuous layer of endothelial cells lining the arterial walls, during angioplasty promotes platelet aggregation. The platelets adhere to the arterial wall and release growth factors (e.g., platelet-derived growth factor, thrombin, basic fibroblast growth factor and transforming growth factor) which initiate the migration of VSMC to the intima and promote the proliferation of VSMC comprising the tunica media. Growth factor-induced VSMC proliferation involves, in turn, a sequential activation of intracellular proteins which promote cell-cycle progression. The next step of the process is associated with an inflammatory response, evidenced by an invasion of inflammatory cells such as monocytes, macrophage, and other white blood cells. These inflammatory cells further induce and stimulate growth of VSMC in the media, which subsequently migrate into the intimal space and produce large amounts of extracellular proteolytic matrix material, forming a neointima layer which represents the lesion. It appears that endothelium then grows over the injured area and restenosis results.
Considerable research has been undertaken to identify agents which prevent or reverse restenosis. For example, many agents have been examined in the prevention of restenosis. A comprehensive review of therapeutics potentially useful in the treatment of restenosis appears in Herrman, Jean-Paul
R. et al. "Pharmacological Approaches to the Prevention of Restenosis Following Angioplasty, The Search for The Holy Grail?" (parts I and II) Drugs 46 (1): 18-52, 46 (2):249-262, 1993.
Other drugs considered for inhibiting restenosis include, but are not limited to, oligonucleotides (e.g., antisense gene therapy), protein kinase C, endothelial growth factor and anti-platelet activating agents. Efforts to prevent restenosis utilizing systemic administration of such drugs, however, have been hampered by generalized toxicity at effective dosages. Furthermore, successful therapy for preventing restenosis has been thwarted by the complexity of the physiological processes responsible for neointima formation as well as the inability to deliver effective quantities of drugs to the site of injury, due to inefficient cellular uptake and lack of specificity (i.e.. targeting) to VSMC. One possible mechanism for the failure of drugs to provide effective prevention of restenosis is that the agents never reach the tissue targeted. For example, the candidate agents may simply be washed away from the treatment areas by blood flow.
Gene therapy, using antisense oligonucleotides which effectively "shut off the genes involved in neointima formation, is a promising approach to the problem of restenosis. However, due to the multiplicity of growth factors involved in neointima formation, selective inhibition of any single growth factor is unlikely to completely prevent lesion formation. Accordingly, works have shown inhibition of neointima hyperplasia in a rat carotid model following angioplasty injury by administration of two antisense oligonucleotides (AS-oligos) which block the cell-cycle regulatory genes for proliferating-cell nuclear antigen (PCNA) and p34cdc2 (cdc2). Morishita, R. et al. "Single intraluminal delivery of antisense cdc2 kinase and proliferating-cell nuclear antigen oligonucleotides results in chronic inhibition of neointimal hyperplasia" Proc. Natl Acad. Sci. 90:84774 (1993). PCNA, a nuclear protein required for DNA synthesis by DNA polymerase Δ, and cdc2, a serine/threonine protein kinase, are primary components of the cell-cycle progression which regulate VSMC proliferation. The AS-oligos were constructed to be directed to the translation initiation sites of PCNA and cdc2. Cellular uptake was enhanced and transfection stability of the AS- oligos increased by complexing the phosphorothioate AS-oligos with liposomes and the protein coat of inactivated Sendai hemagglutinating virus of Japan (HVJ). Morishita et al. observed more rapid uptake and a 10-fold increase in transfection efficiency of AS-oligos or plasmid DNA than standard lipofection or passive uptake methods. However, specific targeting of the HVJ AS-oligos liposome complex to VSMC was not reported. It is generally desired then to develop targeting specificity of drug carriers to VSMC since that would result in greater uptake by the smooth muscle cells at the site of injury and would permit the administration of higher, more effective dosages. Furthermore, it is advantageous to avoid the use of viral proteins in drug carriers as they present a potential biological hazard to humans. Thus, it is an object of the present invention to provide a lipid construct that is superior and more effective in delivering agents into the cytoplasm and nucleus of a cell. It is yet another object of the present invention to provide for the targeting of an agent to vascular smooth muscle tissue. It is still yet a further object of this invention to provide for a less toxic and more efficacious treatment of such diseases as restenosis, viral diseases and cancer.
SUMMARY OF THE INVENTION The invention provides for the delivery of an imaging or therapeutic agent into the cytoplasm of a cell by delivering the lipid construct to a mammal wherein the lipid construct comprises an aminomannose derivatized cholesterol. The delivery of the agents could be done in vitro as well as in vivo. This includes delivery of agents to cells, tissues or organs in vitro and reintroduction of the cells, tissues or organs in vivo. It also includes the transfection of cultured cells in vitro for the purpose of conferring desirable properties in these cells, including enhanced, decreased or novel production of proteins.
Lipid constructs, for purposes of this invention, are complexes containing lipids, phospholipids, or derivatives thereof comprising a variety of different structures which lipids are known to adopt in aqueous suspension. These structures include, but are not limited to, micelles, liposomes, lipid ribbons or sheets, and may be complexed with a variety of drugs and adjuvants which are known to be pharmaceutically acceptable. Common adjuvants include cholesterol and α-tocopherol, among others. The lipids may be used alone or in any combination which one skilled in the art would appreciate to provide the characteristics desired for a particular application. In addition, the technical aspects of lipid complex and liposome formation are well known in the art and any of the methods commonly practiced in the field may be used for the present invention. The present invention further comprises a method for targeting vascular smooth muscle tissue and cells comprising the delivery of an agent to the cell using a lipid construct comprising an aminomannose derivatized cholesterol. This approach is advantageous over the prior art since it provides targeting but does not involve the use of viral products. The aminomannose-containing lipid constructs exhibit dramatically enhanced in vitro and in vivo targeting to VSMC, compared to identical constructs not containing aminomannose. While not wishing to be bound by any particular theory, it appears that the increased targeting results from specific mannose- receptor-mediated endocytosis or fusion of the lipid constructs. In another aspect of the invention, the lipid constructs contain drugs which inhibit neointima formation and are administered to prevent restenosis following PTCA procedures. Since restenosis is caused generally from the accumulation of Vascular Smooth Muscle Cells, the targeting of such cells is critical during PTCA procedures. Also provided herein is a method of delivering a lipid composition into a vascular smooth muscle cell by delivering a lipid construct to a mammal comprising aminomannose derivatized cholesterol.
The preferred composition for carrying out the invention includes a lipid construct comprising an aminomannose derivatized cholesterol. Further embodiments of the invention include the construct having a cationic lipid wherein the total cationic lipid is greater than or equal to 5% of total lipid and the lipid construct further comprises a therapeutic or imaging agent. The aminomannose derivatized cholesterol is included in the definition of a cationic lipid.
The invention is useful for inhibiting restenosis in a mammal by delivering a lipid construct to the mammal wherein the lipid construct comprises an aminomannose derivatized cholesterol and a cationic lipid wherein the total cationic lipid is greater than or equal to 5% of total lipid wherein the lipid construct further comprises an agent in an amount effective for said inhibition. The 5% cationic lipid could include only aminomannose derivatized cholesterol.
Another aspect of the invention comprises the cytoplasmic delivery of an agent into the cytoplasm of a cancer cell.
Brief Description of the Drawings Figure 1 shows a freeze fracture micrograph of a lipid construct- containing oligonucleotides.
Figure 2 shows a confocal micrograph of fluorescently labeled chimeric oligonucleotide taken at 2.5 hours.
Figures 3A & 3B show a confocal micrograph of DOPE:Amch complexed with labeled chimeric oligonucleotide taken at 2.5 hours and 16 hours respectively.
Figures 4A & 4B show a confocal micrograph of DSPC:Chol complexed with labeled chimeric oligonucleotide taken at 2.5 hours and 16 hours respectively.
Detailed Description of the Invention The present invention relates to the formation of a lipid construct comprising an aminomannose derivatized cholesterol. A preferred representative of this compound is 6-(cholest-5-en-3β-yloxy)hexyl-6-amino-6- deoxy- 1 -thio-α-D-mannopyranoside ( Amch).
Of course, any number of lipids may be used in combination with aminomannose derivatized cholesterol. Examples of lipids which may be used successfully include, but are not limited to: dimyristoyl triethylamine propane (DMTAP), dimyristoyl dimethylammonium propane (DMDAP), l,2-dioleoyl-3- trimethylammonium propane (DOTAP), l,2-dioleoyl-3-dimethylammonium propane (DODAP) and dioleoyl-sn-glycero-3-ethyl phosphatidylcholine (DGE), hydrogenated soy and egg phosphatidyl choline (HSPC, HEPC) distearoyl phosphatidylcholine (DSPC), Dimyristoyl phosphatidylcholine (DMPC), dilauryl phoshatidylcholine (DLPC), dioleoyl phosphatidycholine (DOPC), phosphatidyl ethanolamine (PE), dimyristoyl phosphatidylethanolamine (DMPE), dioleoylphosphatidylethanolamine (DOPE), cholesterol, N-[2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), 3b[N-(N'N'-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol). These lipid constructs may also be used to treat or prevent cancer or viral infections by facilitating delivery of oligonucleotide therapeutic agents to the cellular cytoplasm. These lipids may be used in any suitable combination with an aminomannose derivatized cholesterol. Preferred lipid formulations are listed in Tables 1 & 2. The following formulations give rise to optimal results in preventing restenosis: DOPE:Amch (1 :1 molar ratio) having a lipid to oligonucleotide ratio (w/w) less than 5:1 and preferably between 1.25: 1 & 0.5:1. The preferred size of these construct is range from 1 Onm to 250nm preferably at 50nm. Preferred lipids used in the constructs comprise cationic lipids, preferably greater than or equal to 5% total. The preferred lipid construct is a unilamellar liposome as shown in Figure 1 wherein the liposome is shown prepared having a formula DOPE:Amch (1 : 1 molar ratio). The lipid constructs preferred for use in delivering oligonucleotides which inhibit tumor growth comprise HSPC:Chol:Amch in a mole ratio of 16:8:1. The preferred lipid constructs are prepared by first hydrating a mixture of lipids in suitable buffer such as 9% sucrose. Prior to hydration, it is preferable that a lipid powder or film of the lipid mixture is formed, for example, by spray-drying or rotary evaporative techniques.
One preferred use of the invention is in the treatment of restenosis. and further for the prevention of neointima formation. Agents that may be used include the following: antithrombotic agents, antiplatelet agents, antiproliferative agents, anti-inflammatory agents, calcium antagonists, lipid lowering drugs, and antibodies to growth factors, inositol diphosphate, and antisense compounds. Some of these agents include coumadin, heparin, low molecular weight heparins, hirudin, aspirin, dipyridamole, thromboxane A2, thromboxane A2 synthetase inhibitor, thromboxane A2 receptor blockers, ridogrel, sulfinpyrazone, dextran, prostacyclin or prostacyclin analogues, prostaglandin El, ticlopidine, serotonin antagonists, glycoprotein Ilb/IIIa receptor blockers, angiotensin converting enzyme inhibitors, colchicine, platelet-derived growth factor antagonists, angiopeptin, somatomedin, octreotide, cytostatic, corticosteroids, ibuprofen, nifedipine, nicardipine, diltiazem, verapamil, fish oils, (such as omega 3 fatty acids, eicosapentaenoic acids, and docosahexaenoic acids), lovastatin, postangioplasty, colestipol, and antibodies. Microparticles have been noted as a potential microcarrier drug delivery system in order to prevent the rapid elimination by the increase network of vasa vasorum and atherosclerotic lesions that cause early outward diffusion. Red blood cells have also been investigated as a microcarrier system.
The preferred agents used are oligonucleotides that inhibit neointima formation and prevent restenosis. The preferred size of the oligonucleotides is from 15 mer to 30 mer, although sizes up to 80 mer may be associated with the lipid construct taught in this invention. The present invention is also useful for blocking cell-cycle regulatory genes such as proliferating-cell nuclear antigen (PCNA), p34cdc2 (cdc2), and cdk2 (cyclin-dependent kinases). Antisense oligonucleotides directed toward translation sites of these genes provide optimum results. In the treatment of cancer, such antisense oligonucleotides corresponding to the bcl-2 or c-myc mRNA provide significant benefits.
This invention has particular advantages for complexing aminomannose derivatized cholesterol with oligonucleotide ligands generated by the SELEX® technology as described in U.S. Patent Number 5,270,163 and incorporated herein by reference. This invention will be more fully understood by references to the following examples, which are intended to be illustrative of the invention, but not limiting thereof.
Example 1 Liposomal Targeting and Uptake of Smooth Muscle Cells A. Preparation of Liposomes.
Two liposome formulations were prepared. A lipid film containing distearoyl phosphatidylcholine (DSPC) and cholesterol (Choi) in a 2: 1 molar ratio was prepared. A second lipid film containing DSPC, Choi and aminomannose derivatized cholesterol (Amch) in a molar ratio of about 2:0.96:0.04 was also prepared. The lipid films were formed by dissolving the lipids in chloroform, mixing and then evaporating off the chloroform. The lipid films were radiolabeled with 3H-cholesteryl hexadecyl ether and/or fluorescently labeled with the lipophilic fluorescent probe, octadecyl rhodamine. The 3H- cholesterylhexadecyl ether (5 μCi) was added during the formation of the lipid film. The lipids were hydrated in 100 mM NaCl and 25 mM Phosphate buffer pH 7.4 and probe sonicated for approximately 10 minutes to form unilamellar vesicles with an average diameter of 70 nm ± 20 nm. The octadecyl rhodamine was incubated with the liposomes for 20 minutes to fluorescently label the liposomes.
B. In Vitro Measurement of Uptake by Smooth Muscle Cells. The 3H-cholesterylhexadecyl ether labeled liposomes were incubated with confluent rat aortic vascular smooth muscle cells for 5, 30, and 60 minutes. The uptake of the liposomes was measured by scintillation counting and reported in Table 1 as counts per minute (cpm) per microgram of cell protein. All time points were done in triplicate. Fluorescence microscopy and autoradiography were performed to qualitatively detect the association of the liposomes with the vascular smooth muscle cells. TABLE 1
Uptake of Labeled Liposomes by Vascular Smooth Muscle Cells
In CPM/μG Protein
0' 5' 30' 60'
DSPC:Chol 1.35 3.63 4.22 6.51
DSPC:Chol:Amch 1.22 25.17 40.35 52.53
C. In Vivo measurement of Uptake by Smooth Muscle Cells. White New Zealand rabbits were anesthetized and both femoral arteries were exposed. A balloon catheter was advanced into the iliac artery through a cut-down of the femoral artery. The catheter consisted of a metallic plate and tubular extrusions (delivery nipples) occupying one third the circumference of a balloon. The delivery nipples of the balloon deepened about 0.1 mm into the arterial wall. The balloon of the catheter was inflated to 1.5 atm. One ml of aminomannose liposomes was delivered into the catheter against the vessel wall with 100 psi within three seconds. Both iliac arteries were used in all animals. The rabbits (6 arteries/group) were sacrificed at 0.5, 1, 4, 24 and 168 hours. Rabbits injected with normal saline or octadecyl rhodamine served as controls (4 arteries/group). The iliac arteries were snap frozen in liquid nitrogen and the rhodamine fluorescence was measured using an Olympus fluorescence microscope.
At thirty minutes the liposomes were accumulated under the adventitia. At four hours, the liposomes began to distribute in the medial layer. At twenty-four hours, the liposomes were distributed in a heavy homogeneous pattern throughout the width of the arterial wall. At one week, the intima remained stained and the liposomes were redistributed into the adventia with the media being clear. The blood vessels of rabbits are somewhat peculiar in that the intima is extremely thin. Therefore, any intima seen after catheterization is assumed to be neointima, formed as a result of migration of VSMC from the media and consequent release of extracellular matrix. By 24 hours, the liposomes redistribute throughout the vessel. At one week the fluorescent dye is clearly seen within the neointima and not the media. Liposomes associated with VSMC within the media migrated across the internal elastica into the neointima.
The above examples establish that lipid constructs comprising an aminomannose derivatized cholesterol target vascular smooth muscle cells (VSMC). Continuing fluorescence microscopy confirmed delivery of the lipid construct to VSMC, evidenced by binding of aminomannose derivatized liposomes to the VSMC tissue. Autoradiography confirmed delivery into VSMC, evidenced by intracellular localization of the lipid within the VSMC.
Example 2 Liposomes containing greater than 5% cationic lipid were prepared. The lipids were hydrated in sterile-filtered 9% sucrose, heated for 0.5 minutes at 65°C and sonicated. Sizing was done by microtrac analysis.
Oligonucleotides (2328) GTCCTCCATAGTTACTCA (SEQ ID NO.: 1), (2342) GATCAGGCGTGCCTCAAA (SEQ ID NO.: 2), (2616) ACTCATTGATACCTCCTG (SEQ ID NO.: 3) and
(2617) AAACTCCGTGCGGACTAG (SEQ ID NO.: 4) were added to the liposomes and heated at 65°C for 10 minutes. Table 2 lists all cationic formulations, lipid concentrations and liposomes sizes.
TABLE 2 CATIONIC FORMULATIONS
Formulation** Lipid Size(μπ_) Conc.(mgmL)
DOTAP:DOPE:Amch (1:1:0.5) 0.1296 ±0.1446
DODAP:DOPE:Amch (1:1 :0.5) 1.296111.0803(34%) 0.045010.0434 (66%)
DOPE:Amch(l:l) 0.861910.6654(17%) 0.094210.1699(83%)
DMPE:Amch(l:l) 0.273410.1955(44%) 0.071910.0604(58%)
DGE:DOPE:Amch (1:1:0.5) 2.1010.482 (35%) 0.043510.044 (65%)
DGE:DOPE:Amch (1:1:0.5) 1.478510.5654(60%) 0.156610.1430(20%) 0.040310.0230 (20%)
DOPE:Amch(l:l) 0.832510.7217(39%) 0.059410.1035(61%)
DMPE:Amch(l:l) 1.573510.6965(18%) 0.054710.0917(82%)
DMPE:Amch (1:1.5) 1.070910.3329(20%) 0.044510.0445 (74%) 0.009210.0019(6%)
DOPE: Amch (1:1) 2.85 0.037510.0510
DMTAP:DMPE:Amch (1:1:0.5) 3.1 0.015810.0261
DMDAP:DMPE:Amch (1:1:0.5) 3.1 1.6531+1.1338(5%), 0.315210.2193(18%) 0.055110.0542(77%)
DMTAP:DMPE: Amch (1:1:1) 3.1 1.761410.5416(1%) 0.012310.0090(99%) Foπnulation#* Lipid Size(μ__ι) Conc.(mgmL)
DOPE:Amch(l:l) 2.85 0.043110.0437(60%) 0.021810.0053(19%) 0.011610.0020(21%)
DOPE:Amch(l:l) 3.1 1.179510.5194(2%) 0.041610.0761 (98%)
DOTAP:DOPE:Amch (1:1:1) 3.72 0.233310.1677
DODAP:DOPE:amino (1:1:1) 3.72 0.88810.5061 (11%)
0.117210.0988(59%)
0.043710.0179(30%)
DOPE: Amch (1:1) 3.1 0.043810.0719
DcChol:DOPC:Amch (0.25:1:0.25) 4.8 0.313510.1055(2%) 0.044610.0603 (96%)
DcChol:DOPC (0.5:1) 2.5 1.101611.6545(30%) 0.043710.0469 (70%)
DcChol:DOPE:Amch (0.5:1:0.5) 3.1 0.032710.0534
DcChol:DOPE(l:l) 2.7 1.586510.4214(24%) 0.041410.0301 (27%) 0.009510.0018(49%)
DOTAP:DOPE:Amino (1:1:0.5) 3.1 0.049610.0508
DOPE:Amino(l:l) 4.65 0.042810.0718
DOPE:Amino (1:1.5) 3.0 1.499610.4880(12%) 0.052210.0840 (88%)
DOTAP:DOPE:Amch (1:1:1.5) 3.0 1.630610.8100(13%) 0.034310.0512(87%)
DOTAP:DOPE:Amch (1.3:1:1.2) 3.0 1.953410.7815(14%) 0.054110.0867(86%)
DOPE: Amch (1:1.2) 3.1 0.320310.2097 (7%) 0.039610.0407 (93%)
DOTAP:DOPE:Amch (1:1:1.5) 3.1 0.041710.0363
DOPE:Amch(l:l) 3.1 0.062210.0783
DOSPA:DOPE:Amch (3:1:0.5) 0.75 1.016910.2413
DOTMA:DOPE:Amch (1:1:0.5) 0.8 0.095310.1004
Typically, liposomes were sonicated until greater than 85% of the liposomes were less than 300nm diameter. This allowed sterile filtration using a 450nm cellulose acetate filter without significant liposome loss on the filter. Al liposome formulations except DOT AP:DOPE: Amch (1 : 1 :1), DODAP:DOPE:Amch (1:1 :1) and DOSPA:DOPE:Amch were 10-lOOnm average diameter. DOTAP:DOPE:Amch ( 1 : 1 : 1 ) and DODAP:DOPE:Amch( 1 :1 : 1) averaged 100-200nm diameter; DOSPA:DOPE:Amch were greater than 1 μm and could not be sterile filtered.
Example 3 Lipid constructs were prepared as in Example 2. Electron microscopy were obtained of liposomes prepared with DOPE: Amch (1 : 1, mole ratio) and mixed with oligonucleotides in a 5:1 lipid:oligo ratio and immediately frozen (Figure 1). Figure 1 clearly indicates that unilamellar liposomes are formed
(magnified 60,000x ( 1 cm= 167nm) with an average size of 62±51 nm. Two 21 -mer phosphorothioate oligonucleotides targeted to the CAT gene were complexed (and combined), with the lipid construct. The sequences of the oligonucleotides were (2520) TAGCTTCCTTAGCTCCTGCAT (SEQ ID NO.: 5) and (2519) TAGCTT CCGCAACTCTTGCAT (SEQ ID NO.: 6), both antisense to a mRNA which codes for chloramphenicol acetyl transferase (CAT). Incorporation of the oligos into the cytoplasm should inhibit CAT activity for the proper plasmid only if it is a specific antisense effect. The extent of inhibition of restenosis was determined using either of two methods: a chloramphenicol acetyl transferase (CAT) assay or a PCNA/cdc2 assay.
Chloramphenicol is an antibiotic which blocks protein synthesis by inhibiting the peptidyl transferase activity of the 50S ribosomal subunit in prokaryotes; peptidyl transferase catalyzes the formation of peptide bonds. CAT is a bacterial drug-resistance gene which inactivates chloramphenicol by acetylating the drug at one or both of its two hydroxyl groups.
In the CAT assay, two expression plasmids containing modified bacterial CAT genes were constructed. These plasmids were pG 1000 and pG1007 as taught in Reynolds, M.A. "Triple-Strand-Forming methylphosphonate oligonucleotides targeted to mRNA efficiently block protein synthesis," Proc Acad. Sci. 91 :12433-12437 at 12434 (1994), with the following modifications; the amplified DNA fragment containing the sequence target site for oligonucleotides 2519 or 2520 was substituted for the polypyrimidine insert. The two plasmids differed from one another by four bases in the translation initiation region targeted by the oligos. This gene is usually not found in eukaryotes; thus there is little or no background CAT activity in eukaryotic cells. Cell extracts were incubated with 3H or 14C chloramphenicol and n-butyryl coenzyme A. 3H or 14C chloramphenicol + n- butyryl coenzyme A ~CAT~> 3H or 14C chloramphenicol-n-butyryl. The modified chloramphenicol partitioned into the xylene fraction while the unmodified chloramphenicol remained in the aqueous phase.
Cos 7 cells or A 10 smooth muscle cells were plated the day before the experiment so that confluency was approximately 70% on the day of the experiment. Formulations of lipofectin complexed with plasmid and lipid constructs complexed with phosphorothioate oligonucleotides were diluted in OptiMEM, a serum-free medium. The cells are washed once with OptiMEM and then incubated for 3 hr with 0.5 mL of the diluted liposome-oligo complexes. The liposomes were removed and washed with OptiMEM and transfected for 3 hours with the target plasmid complexed with Lipofectin. The plasmid complexes were removed and the cells were cultured overnight in complete medium. The cells were harvested approximately 16 hours after the addition of the target plasmid. Cell lysates were analyzed for CAT activity to assess inhibition and protein content. The results are displayed in Table 3. TABLE 3
Transiently Transfected COS 7 Cells
Formulation Lipid:oligo %Inhib.1 0.5 μM 0.1 μM μM oligo
DOTAP:DOPE:Amch (1:1:0.5) 5:1 94,91 94 64
DODAP:DOPE:Amch (1:1:0.5) 5:1 92,90 93 64
DOPE: Amch (1:1) 10:1 90 85 39
DMPE:Amch(l:l) 10:1 89 85 32
DMPE:Amch(l:l) 5:1 88, 95, 95 79 25
DGE:DOPE:Amch (1:1:0.5) 5:1 84,90 50 11
DMPE:Amch (1:1.5) 10:1 88 72 29
DMTAP:DMPE:Amch (1:1:0.5) 5:1 92 83 24
DMDAP:DMPE:Amch (1:1:0.5) 5:1 84 50 15
DMTAP:DMPE:Amch (1:1:1) 5:1 87 70 41
Lipofectin 2.5:1 93, 86 90 62
A stably inducible model was also evaluated using COS 7 cells and A10 smooth muscle cells. These cells were stably transfected with a plasmid containing an inducible CAT gene using LipofectAmine (DOSPA:DOPE 3 : 1 mole ratio). COS 7 and A10 smooth muscle cells were plated the day before the experiment, treated with the liposomes and washed as described above. The liposome-oligo complexes were removed, washed in OptiMEM and cultured overnight in medium containing CdCl2, which induces protein production. The cells were harvested approximately 16 hours later and analyzed for CAT activity. Cells were treated with μM oligo. The results are displayed in Tables 4 and 5.
The plasmid containing an inducible CAT gene is pG 1041 which has a modified chloramphenicol acetyl transferase (CAT) gene inserted into pMEP4 (Clonetech). pG 1041 was produced as follows:
A modified chloramphenicol acetyl transferase (CAT) gene producing wild- type CAT protein was amplified in a PCR reaction using the following primer: 5' primer:
Hind III 5' cac aag ctt gtc gac teg agt gca gga get aag gaa get ace atg gag aag aag ate act gga tat ace ace 3' (SEQ ID NO.: 7) 3' primer: Not I
5' tta tgc ggc cgc tta cgc ccc gec ctg cca 3' (SEQ ID NO.: 8) The amplified fragment was cut using Hind III and Not I restriction enzymes and ligated into Hind Ill/Not I cut pMEP4 (Clontech vector. The amino terminal protein and 5' nucleotide sequences of the CAT gene in pG1041 compared to wild-type CAT are: Wild-type CAT:
5' +1 3'
Met Glu Lys Lys He Ser Gly Tyr Thr Thr
(SEQ ID NO.: 9) uuuuca gga gcu aag gaa gcu aaa aug gagaaaaaa auc acu ggauau ace ace pG1041,UCAT: (SEQIDNO.: 10)
5' +1 3'
Met Glu Lys Lys He Ser Gly Tyr Thr Thr
(SEQ ID NO.: 11) agu gca gga gcu aag gaa gcu ace aug gag aag aag auc acu gga uau ace ace
(SEQ ID NO.: 12) The first adenosine of the open reading frame is designated + 1. The coding region nucleotide base changes between wild-type and pG1041 are conservative and do not change the amino acid sequence. Amino acid code: Asp = aspartic acid, Arg = arginine, Glu = glutamic acid, Gly = glycine, He = isoleucine, Lys = lysine. Met = methionine, Ser = serine, Thr = threonine, Tyr = tvrosine. Table 4
Stably Transfected COS7 Cells with PS Oligonucleotides
Formulation Lipid:01igo % Inhibition Concentration
DOTAP:DOPE: Amch (1:1: 1) 3.3:1 25%
DODAP:DOPE:Amch (1:1: 1) 3.3:1 20%
DOPE: Amch (1:1) 5:1 0%
LipofectAmine Not Reported 59%
DOPE:Amch(l:l) 5:1 22%
DOPE: Amch (1:1) 1.25:1 16%
DOPE:Amch(l:l) 0.5:1 1%
Lipofectin Not Reported 36%
LipofectAmine Not Reported 60%
DOTAP:DOPE:Amch (1:1:0.5) 5:1 76%
DOPE: Amch (1:1) 5:1 16%
LipofectAmine 8:1 66%
DOPE: Amch (1:1.5) 5:1 35%
DOTAP:DOPE:Amch (1.3 :1:1.2) 5:1 74%
DOTAP:DOPE:Amch (1:1 :1.5) 5:1 73%
Table 5 Stably Transfected AlO Cells with PS Oligonucleotides
Formulation Lipid:01igo % Inhibition Concentration
DOPE: Amch (1:1.5) 5:1 0%
LipofectAmine 6:1 81%
DOTAP:DOPE:Amch (1.3: 1 : 1.2) 5:1 16%
DOTAP:DOPE:Amch (1:1:1.5) 5:1 60%
LipfectAmine 6:1 48%
DOPE: Amch (1:1.5) 5:1 4% ificant inhibition was observed in most formulas. EXAMPLE 4
A confocal microscopy study was undertaken to further verify cytoplasmic delivery of oligonucleotides using lipid constructs comprised of Aminomannose derivatized cholesterol, DOPE:Amch (1 : 1 molar ratio) and DSPC hol (2: 1 molar ratio) were prepared as in Examples 1 & 2. The DOPE: Amch lipid constructs were incubated for 10 minutes at 65 °C with a fluorescently labeled chimeric oligonucleotide having the sequence 5'-GGTATATCCAGTGATCπCπCTC-3' (SEQ ID NO.: 13). The lipid to oligonucleotide ratio was 5:1 w/w. DSPCxhol liposomes were prepared by hydrating the lipids with buffer containing the chimeric oligonucleotides and sonication. Six flasks containing 106 PI 798 (lymphocarcinoma cells) cells in 10 ml complete Iscoves (10% Fetal Calf Serum, 1% penicillin/streptomycin) were incubated at 37°C, 5% C02. The cells were then incubated with 50 μg of oligonucleotide, either as free oligo or lipid associated oligo. Flasks 1, 2 and 3 were incubated with free oligo or oligo complexed with one of the two lipid formulations overnight (16 hour incubation). Flasks 4, 5 and 6 were incubated the next morning with free oligo or oligo complexed with one of the two lipid formulations (2.5 hour incubation). Cells were removed at 2.5 and 16 hours. The cells were washed to remove unassociated liposomes/oligonucleotide and examined by confocal microscopy. For each sample, a confocal microscopy series and corresponding bright field image were recorded. That is, a midsection fluorescence (top), bright-field (middle) and 3-d reconstruction (bottom) are displayed in figures 2-4. Figures 2A and 2B show rapid and intense fluorescence diffusing from the edge of the cells into the cytoplams. As the figures indicate, Amch:DOPE lipid constructs exhibit rapid and concentrated fluorescence into the cytoplasm as shown by intense fluorescence in the cytoplasm whereas the cells incubated with free oligonucleotide and oligonucleotide associated with DSPCxhol constructs shows very little fluorescence at all time periods.
EXAMPLE 5 A transfection study was carried out to further verify the ability of aminomannose derivatized cholesterol lipid constructs to deliver oligonucleotides into the nucleus of a cell. As noted above, LFA is currently used to transfect a plasmid, that is, introduce the plasmid into the nucleus of a cell so that it expresses a protein for which it codes. The plasmid was pG1035 which has a modified chloramphenicol acetyl transferase (CAT) gene inserted into pRc/CMV (Invitrogen). pG1035 was produced as follows. A modified chloramphenicol acetyl transferase (CAT) gene producing wild- type CAT protein was amplified in a PCR reaction using the following primers: 5' primer:
Hind III 5' ate caa get teg acg aga ttt tea gga 3' (SEQ ID NO.: 14) 3' primer: Not I
5' tta tgc ggc cgc tta cgc ccc gec ctg cca 3' (SEQ ID NO.: 8) The amplified fragment was cut using Hind III and Not I restriction enzymes and ligated into Hind Ill/Not I cut pRc/CMB (invitrogen) vector. The amino terminal protein and 5' nucleotide sequence of the CAT gene in pG1035 are: Wild-type CAT:
5' +1 3'
Met Glu Lys Lys He Ser Gly Tyr Thr Thr
(SEQ ID NO.: 9) uuu uca gga gcu aag gaa gcu aaa aug gag aaa aaa auc acu gga uau ace ace (SEQ ID NO.: 10)
It is known that LFA is optimized at a lipid:plasma w/w ratio of 6: 1. The following lipid:plasmid constructs were formed: DOPE:Amch (1 : 1 molar ratio) with a lipid plasmid ratio w/w of 10: 1 ; DMPE:Amch (1 : 1 and 1 : 1.5 molar ratios) with lipid plasmid ratio w/w of 10: 1 ; DODAP:DOPE:Amch ( 1 : 1 : 1 ) at lipid plasmid ratios w/w of 15: 1 and 10:1. The DOPE:Amch lipid constructs were 1.14 times more efficacious then LFA. The DMPE:Amch constructs, 1 : 1 and the 1 : 1.5, were 0.44 and 0.71 as effective as LFA respectively. The DODAP:DOPE: Amch 15 : 1 and 10: 1 formulas, were 0.02 and 0.01 as effective as the LFA respectively. In all cases, the Aminomannose constructs were effective in transfecting AlO smooth muscle cells.
EXAMPLE 6 Lipid constructs were formulated as in Example 2 using the following Oligonucleotides: cdc gene (rat) GTCCTCCATAGTTACTCA (SEQ ID NO.: 1); PCNA gene (rat) GATCAGGCGTGCCTCAAA (SEQ ID NO.: 2); rev-AS (2328) ACTCATTGATACCTCCTG (SEQ ID NO.: 3); rev-AS (2342) AAACTCCGTGCGGACTAG (SEQ ID NO.: 4). Ten unheparinized rats were treated with 10 μM total oligo (antisense or reverse antisense) complexed with DOPE: aminomannose (1 :1) at a 5:1, 1.25:1 or 0.5:1 lipidxligo w/w ratios and sacrificed 2 weeks post-angioplasty. Eight heparinized rats were treated with 10 μM total oligo (antisense or reverse antisense) complexed with DOPE:aminomannose liposomes (1 : 1) at 5: 1 lipid:oligo w/w ratio and sacrificed 2 weeks post-angioplasty. A No. 2 French Fogarty catheter was used to induce vascular injury in male Spraque-Dawley rats. The rats were anesthetized, and a cannula introduced into the left common carotid via the external carotid artery. After vascular injury of the common carotid, the distal injured segment was transiently isolated by temporary ligatures. The lipid complex formulation (0.25 ml) containing the antisense oligos (5 μM of each) was infused into the segment and incubated for 15 - 30 minutes at room temperature. Control rats received an identical formulation containing reverse antisense oligos (rev- AS). Following incubation, the infusion cannula was removed and blood flow to the common carotid restored by release of the ligatures. At 2 weeks after catheterization, rats were sacrificed and vessels perfused-fixed with 4% paraformaldehyde. Sections were analyzed for neointima formation, and data expressed as neointimal/medial area ratios and percent inhibition. Unheparinized rats treated with DOPE: aminomannose lipid constructs formulated at 1.25: 1 or 0.5: 1 lipidxligo w/w showed excellent inhibition of neointima formation as compared to rats treated with liposomes formulated with reverse antisense. Neointima formation was inhibited approximately 45% and 38% by the 0.5: 1 and 1.25:1 constructs respectively. However, liposomes formulated with 5: 1 lipid:oligo w/w were not effective. 60-70% of the unheparinized rats showed moderate to severe thrombosis. Heparinized rats treated with the 5: 1 constructs showed no thrombosis and no reduction in neointima formation when compared to heparinized rats treated with reverse antisense. EXAMPLE 7
Lipid constructs were prepared as in Example 2 and complexed with oligonucleotides as in Example 6. Rats were heparinized and treated with constructs complexed with cdc2 and PCNA (0.5: 1 lipidxligo w/w and 50 μM oligo, 0.5:1 lipidxligo w/w and 5 μM oligo, 0.25:1 lipidxligo w/w and 5 μM oligo) or reverse cdc2 and PCNA (0.5: 1 lipidxligo w/w and 5 μM oligo) phosphorothioate oligonucleotides. The results of the study are listed below in Table 6. Unlike the previous study done with unheparinized rats (Example 6), only 1 rat in the entire study showed signs of thrombosis (rat treated with 0.25: 1 lipid:oligo w/w and 5 μM oligo). All rats treated with antisense oligonucleotides showed a therapeutic response (treated arteries as compared to untreated control arteries). TABLE 6
Neointima Formation in Treated vs. Untreated Control Arteries
Treatment Group Treated Untreated % Reduction t-test
AS 0.5:1, 50 μM 0.17910.035 0.23410.042 22.9% 0.013
AS 0.5: 1 , 5 μM 0.15310.044 0.25010.053 38.7% <W4
AS 0.25:1 , 5 μM 0.15810.028 0.25710.034 38.5% <10"4 rev AS 0.5: 1, 5 μM 0.18110.022 0.18310.018 None 0.834
EXAMPLE 8 Stock solutions of the oligonucleotides were prepared as followed: antisense, c-myc:5'-AACGTTGAGGGGCAT-3' (SEQ ID NO.: 15) and nonsense c-myc:5'-CTGAAGTGGCATGAG-3' (SEQ ID NO.: 16) were dissolved in phosphate buffer to make a stock solution with a concentration of approximately 3.4 mg/ml. A lipid film was prepared containing hydrogenated soy phosphatidylcholine:chol:Amch in a mole ratio of 16:8:1 having a total lipid concentration of 560 milligrams. One ml of the oligonucleotide stock solution was added to the lipid film and bath sonicated at a temperature of 40° C for 10 seconds. The resulting solution was put through a 4 cycle freeze- thaw procedure using liquid nitrogen. The resulting homogeneous solution was extruded first through a 0.8 μM filter membrane (3 times) then extruded through a 0.45 μM filter (3 times) and finally extruded through a 0.2 μM filter (3 times). The free oligonucleotides were removed by passing through a Sephadex G-50 column with a bed volume of about 18 ml. The vesicle volume was recovered in 5.5 ml of buffer. Control liposomes were prepared as above without oligonucleotides. The mean diameter of the control liposomes was 80.0±30 8.4nm. The mean diameter of the oligonucleotides containing liposomes were 210±80nm. The antisense c-myc and oligonucleotides concentration was 50 μg/ml.
The liposomes containing oligonucleotides were tested against two strains of small cell lung carcinoma. The first strain was a cisplatin (cDDP). sensitive line (GLC4) and the second strain was a 5.6x cDDP resistant sub-line (GLC4-cDDP). In GLC4 and GLC4-cDDP there is a 40-60x c-myc oncogene amplification with a corresponding c-myc mRNA and protein overexpression. The lipid concentration in the control group was 21.8 mg/ml and the lipid concentration in the liposome preparation containing the oligonucleotides was 10.3 mg/ml. Both cell lines showed growth inhibition after incubation with 10 and 20 μM antisense c-myc oligonucleotides in SCLC-medium, a defined serum- free culture medium (heat- inactivated for 1 hour at 70° C). Thus, the remaining nucleases were heat-inactivated. In these cell cultures nonsense oligonucleotides did not have any inhibitory effect. The results are listed in Table 7. The results in Table 1 disclose that encapsulated oligonucleotides in liposomes allow for a 10 to 20 times reduction of antisense c-myc oligonucleotides concentration needed to exert the same inhibitory effect in the GLC4 and GLC4-cDDP cell lines compared to the unencapsulated antisense c-myc oligonucleotides. Empty liposomes and nonsense oligonucleotides did not have an inhibitory effect.
TABLE 7
% Growth to control ±sd (n=3) GLC4 GLC4-cDDP control 100 100
10 μM as c-myc n-ODN 83.6±5.9 82.1±7.9
20 μM as c-myc n-ODN 69.5±10.2 55.5±5.9
10/20 μM ns c-myc n-ODN 99.716.1 100.3+6.1
1 μM as c-myc in liposomes 70.3±9.0 62.1±8.6
2.5 μM as c-myc in liposomes 56.9±10.3 31.3±15.5 empty liposomes 104.6±8.3 80.4±14.6
While the invention has been described with reference to specific embodiments and examples, it will be appreciated that various changes and modifications can be made without departing from the invention.
Although this specification has been disclosed and illustrated with reference to particular applications, the principles involved are susceptible to numerous other applications which will be apparent to those skilled in the art. The invention is, therefore, to be limited only as indicated by the scope of the appended claims.
SEQUENCE LI STING
( 1 ) GENERAL INFORMATION :
( i ) APPLICANT :
(A) NAME: NEXSTAR PHARMACEUTICALS, INC.
(B) STREET: 2860 WILDERNESS PLACE
(C) CITY: BOULDER
(D) STATE: CO
(E) COUNTRY: US
(F) POSTAL CODE (ZIP) : 80301
(G) TELEPHONE: (303) 444-5893 (H) TELEFAX: (303) 444-0672
(ii) TITLE OF INVENTION: LIPID CONSTRUCTS FOR CYTOPLASMIC DELIVERY OF AGENTS
(iii) NUMBER OF SEQUENCES: 16
(iv) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.30 (EPO)
(vi) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/386,577
(B) FILING DATE: 10-FEB-1995
(2) INFORMATION FOR SEQ ID NO: 1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1: GTCCTCCATA GTTACTCA 18
(2) INFORMATION FOR SEQ ID NO: 2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: YES
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:
GATCAGGCGT GCCTCAAA 18
(2) INFORMATION FOR SEQ ID NO: 3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICA : NO (iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: ACTCATTGAT ACCTCCTG 18
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 18 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
AAACTCCGTG CGGACTAG 18
(2) INFORMATION FOR SEQ ID NO: 5: (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5: TAGCTTCCTT AGCTCCTGCA T 21
(2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6: TAGCTTCCGC AACTCTTGCA T 21
(2) INFORMATION FOR SEQ ID NO: 7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 72 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7: CACAAGCTTG TCGACTCGAG TGCAGGAGCT AAGGAAGCTA CCATGGAGAA GAAGATCACT 60 GGATATACCA CC 72
(2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8: TTATGCGGCC GCTTACGCCC CGCCCTGCCA 30
(2) INFORMATION FOR SEQ ID NO: 9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: protein
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(v) FRAGMENT TYPE: N-terminal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
Met Glu Lys Lys lie Ser Gly Tyr Thr Thr 1 5 10
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 54 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: mRNA (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10: UUUUCAGGAG CUAAGGAAGC UAAAAUGGAG AAAAAAAUCA CUGGAUAUAC CACC 54
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 10 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: unknown
(D) TOPOLOGY: unknown
(ii) MOLECULE TYPE: protein (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(v) FRAGMENT TYPE: N- erminal
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
Met Glu Lys Lys lie Ser Gly Tyr Thr Thr 1 5 10
(2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 54 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: mRNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12: AGUGCAGGAG CUAAGGAAGC UACCAUGGAG AAGAAGAUCA CUGGAUAUAC CACC 54 (2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13: GGTATATCCA GTGATCTTCT TCTC 24
(2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14: ATCCAAGCTT CGACGAGATT TTCAGGA 27
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15: AACGTTGAGG GGCAT 15
(2) INFORMATION FOR SEQ ID NO: 16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 15 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16: CTGAAGTGGC ATGAG 15

Claims

We claim:
1. A method for delivering a therapeutic or imaging agent into the cytoplasm of a cell comprising the administration to a mammal a lipid construct having an aminomannose-derivatized cholesterol moiety.
2. The method as recited in claim 1 wherein the aminomannose -derivatized moiety is 6-(cholest-5-en-3β-yloxyl)hexyl-6-amino-6-deoxy-l -thio- -D- mannopyranoside.
3. The method as recited in claim 1 wherein the agent is an oligonucleotide.
4. The method as recited in claim 2 wherein the agent is an oligonucleotide.
5. The method of claim 3 wherein the oligonucleotide inhibits the growth of vascular smooth muscle cells.
6 . The method of claim 4 herein the oligonucleotide inhibits the growth of smooth muscle cells.
7. The method of claim 3 where the lipid construct comprises a cationic lipid wherein the total cationic lipid concentration is greater than 5% of total lipid.
8. The method of claim 4 where the lipid construct comprises a cationic lipid wherein the total cationic lipid concentration is greater than 5% of total lipid.
9. The method of claim 5 where the lipid construct comprises a cationic lipid wherein the total cationic lipid concentration is greater than 5% of total lipid.
10. The method of claim 6 where the lipid construct comprises a cationic lipid wherein the total cationic lipid concentration is greater than 5% of total lipid.
1 1. The method of claim 3 where the oligonucleotide inhibits the growth of cancer cells.
12. The method of claim 4 where the oligonucleotide inhibits the growth of cancer cells.
13. The method of claim 11 where the oligonucleotide has the sequence AACGTTGAGGGGCAT (SEQ ID NO.: 15).
14. The method of claim 12 where the oligonucleotide has the sequence AACGTTGAGGGGCAT (SEQ ID NO.: 15).
15. The method of claim 11 where the oligonucleotide has the sequence CTGAAGTGGCATGAG (SEQ ID NO.: 16).
16. The method of claim 12 where the oligonucleotide has the sequence CTGAAGTGGCATGAG (SEQ ID NO.: 16).
17. A lipid construct comprising an oligonucleotide which inhibits the growth of vascular smooth muscle cells wherein the construct comprises 6-(cholest-5- en-3 β-y loxyl)hexy l-6-amino-6-deoxy- 1 -thio-α-D-mannopyranoside and DOPE: Amch in mole ratio of 1 : 1 wherein the lipidxligonucleotide ratio is at or between about 0.5: 1 and 1.25: 1.
18. The construct as recited in claim 17 wherein said oligonucleotide is an antisense oligonucleotide having a size up to 80 mer.
19. The construct as recited in claim 18 wherein said oligonucleotide has a size from 15 mer to 30 mer.
20. The construct as recited in claim 19 wherein said oligonucleotide comprises GTCCTCCATAGTTACTCA (SEA ID NO.: 1).
21. The construct as recited in claim 19 wherein said oligonucleotide comprises GATCAGGCGTGCCTCAAA (SEQ ID NO.: 2).
22. A method for inhibiting the growth of a cancer cell comprising the delivery of a lipid construct having an aminomannose derivatized cholesterol moiety wherein the construct contains an oligonucleotide which inhibits the growth of cancer cells.
23. The method as recited in claim 22 wherein the aminomannose-derivatized moiety is 6-(cholest-5-en3β-ylozyl)hexyl-6-amino-6-deoxy-l-thio- -D mannopyranoside.
24. The method of claim 22 wherein the oligonucleotide is antisense to bcl-1 mRNA.
25. The method of claim 23 wherein the oligonucleotide is antisense to the bcl-1 mRNA.
26. The method of claim 23 wherein the oligonucleotide is antisense to the c- myc mRNA.
27. The method of claim 22 where the oligonucleotide has the sequence AACGTTGAGGGGCAT (SEQ ID NO.: 15).
28. The method of claim 23 where the oligonucleotide has the sequence AACGTTGAGGGGCAT (SEQ ID NO.: 15).
29. The method of claim 3 wherein the oligonucleotide blocks cell-cycle regulatory genes.
30. The method of claim 4 wherein the oligonucleotide blocks cell-cycle regulatory genes.
31. The method of claim 29 wherein the cell-regulatory gene is a proliferating- cell nuclear antigen (PCNA).
32. The method of claim 30 wherein the cell-regulatory gene is a proliferating- cell nuclear antigen (PCNA).
33. The method of claim 29 wherein the cell-regulatory gene is p34cdc2 (cdc2).
34. The method of claim 30 wherein the cell-regulatory gene is p34cdc2(cdc2).
35. The method of claim 29 wherein the cell-regulatory gene is cdk2.
36. The method of claim 30 wherein the cell-regulatory gene is cdk2.
PCT/US1996/001960 1995-02-10 1996-02-08 Lipid constructs for cytoplasmic delivery of agents WO1996024334A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU49236/96A AU4923696A (en) 1995-02-10 1996-02-08 Lipid constructs for cytoplasmic delivery of agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38657795A 1995-02-10 1995-02-10
US08/386,577 1995-02-10

Publications (2)

Publication Number Publication Date
WO1996024334A1 true WO1996024334A1 (en) 1996-08-15
WO1996024334A9 WO1996024334A9 (en) 1996-10-31

Family

ID=23526179

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/001960 WO1996024334A1 (en) 1995-02-10 1996-02-08 Lipid constructs for cytoplasmic delivery of agents

Country Status (2)

Country Link
AU (1) AU4923696A (en)
WO (1) WO1996024334A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997014440A1 (en) * 1995-10-19 1997-04-24 Johnson & Johnson Interventional Systems Conjugation of c-myc antisense oligonucleotides with cholesterol to significantly enhance their inhibitory effect on neointimal hyperplasia
WO1998055490A1 (en) * 1997-06-06 1998-12-10 Rhone-Poulenc Rorer S.A. Novel class of nucleic acid cationic transfecting agents
WO1999012945A2 (en) * 1997-09-08 1999-03-18 Valentis,Inc. Hydrophobic glycosylamine derivatives, compositions, and methods for use
US6350853B1 (en) 1993-04-26 2002-02-26 Peter E. Nielsen Conjugated peptide nucleic acids having enhanced cellular uptake
WO2003018603A1 (en) * 2001-08-29 2003-03-06 Aventis Pharma S.A. Aminoglycoside lipid derivatives for transfection
FR2829136A1 (en) * 2001-08-29 2003-03-07 Aventis Pharma Sa New transfection compounds having an aminoglycoside group linked via a spacer to a lipid such as cholesterol

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4310505A (en) * 1979-11-08 1982-01-12 California Institute Of Technology Lipid vesicles bearing carbohydrate surfaces as lymphatic directed vehicles for therapeutic and diagnostic substances
WO1994015646A1 (en) * 1993-01-07 1994-07-21 Thomas Jefferson University Antisense inhibition of c-myc to modulate the proliferation of smooth muscle cells

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4310505A (en) * 1979-11-08 1982-01-12 California Institute Of Technology Lipid vesicles bearing carbohydrate surfaces as lymphatic directed vehicles for therapeutic and diagnostic substances
WO1994015646A1 (en) * 1993-01-07 1994-07-21 Thomas Jefferson University Antisense inhibition of c-myc to modulate the proliferation of smooth muscle cells

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, Volume 198, No. 1, issued 14 January 1994, ABE et al., "Suppression of Neointimal Smooth Muscle Cell Accumulation in Vivo by Antisense cdc2 and cdk2 Oligonucleotides in Rat Caratoid Artery", pages 16-24. *
GASTROENTEROLOGY, Volume 106, issued April 1994, NAGANO et al., "Growth Inhibition of a Gastric Cancer Cell Line by Antisense Oligonucleotides to c-myc and bcl-2", page A419. *
HUMAN GENE THERAPY, Volume 3, issued 1992, STEWART et al., "Gene Transfer in Vivo With DNA-Liposome Complexes: Safety and Acute Toxicity in Mice", pages 267-275. *
JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY, issued February 1994, MALE et al., "Receptor-Assisted Liposome Uptake Into Vascular Smooth Muscle Cells", page 188A, Abstract No. 891-37. *
JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY, Volume 21, No. 2, issued February 1993, SHI et al., "Maximizing Gene Transfer Into Vascular Smooth Muscle Cells", page 209A, Abstract No. 900-37. *
PROC. NATL. ACAD. SCI. U.S.A., Volume 90, issued September 1993, MORISHITA et al., "Single Intraluminal Delivery of Antisense cdc2 Kinase and Proliferating-Cell Nuclear Antigen Oligonucleotides Results in Chronic Inhibition of Neointimal Hyperplasia", pages 8474-8478. *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6350853B1 (en) 1993-04-26 2002-02-26 Peter E. Nielsen Conjugated peptide nucleic acids having enhanced cellular uptake
US6686442B2 (en) 1993-04-26 2004-02-03 Peter E. Neilsen Conjugated peptide nucleic acids having enhanced cellular uptake
WO1997014440A1 (en) * 1995-10-19 1997-04-24 Johnson & Johnson Interventional Systems Conjugation of c-myc antisense oligonucleotides with cholesterol to significantly enhance their inhibitory effect on neointimal hyperplasia
WO1998055490A1 (en) * 1997-06-06 1998-12-10 Rhone-Poulenc Rorer S.A. Novel class of nucleic acid cationic transfecting agents
FR2764304A1 (en) * 1997-06-06 1998-12-11 Rhone Poulenc Rorer Sa NEW CLASS OF CATIONIC NUCLEIC ACID TRANSFECTANTS
WO1999012945A2 (en) * 1997-09-08 1999-03-18 Valentis,Inc. Hydrophobic glycosylamine derivatives, compositions, and methods for use
WO1999012945A3 (en) * 1997-09-08 1999-08-19 Genemedicine Inc Hydrophobic glycosylamine derivatives, compositions, and methods for use
WO2003018603A1 (en) * 2001-08-29 2003-03-06 Aventis Pharma S.A. Aminoglycoside lipid derivatives for transfection
FR2829136A1 (en) * 2001-08-29 2003-03-07 Aventis Pharma Sa New transfection compounds having an aminoglycoside group linked via a spacer to a lipid such as cholesterol
JP2005504061A (en) * 2001-08-29 2005-02-10 アベンティス・ファーマ・ソシエテ・アノニム Lipid derivatives of aminoglycosides

Also Published As

Publication number Publication date
AU4923696A (en) 1996-08-27

Similar Documents

Publication Publication Date Title
US5660855A (en) Lipid constructs for targeting to vascular smooth muscle tissue
Weissig et al. Cationic bolasomes with delocalized charge centers as mitochondria-specific DNA delivery systems
EP1764091B1 (en) Improvements in or relating to amphoteric liposomes
EP0847272B1 (en) LIPOSOMAL p-ETHOXY OLIGONUCLEOTIDES
US8415461B2 (en) Amphiphilic substances and functionalized lipid vesicles including the same
EP1059092B1 (en) Use of complexes among cationic liposomes and polydeoxyribonucleotides as medicaments
AU710170B2 (en) Cationic virosomes as transfer system for genetic material
US20060165770A1 (en) Lipid particles having asymmetric lipid coating and method of preparing same
US20030219474A1 (en) Inhibition of Bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
US20060216343A1 (en) Pharmaceutical compositions comprising an oligonucleotide as an active agent
JP2002501511A (en) High rate encapsulation of charged therapeutic agents in lipid vesicles
EP1811960A2 (en) Improvements in or relating to pharmaceutical compositions for local administration
WO1998007408A9 (en) Novel liposome complexes for increased systemic delivery
Hu et al. Programmable fusogenic vesicles for intracellular delivery of antisense oligodeoxynucleotides: enhanced cellular uptake and biological effects
JP3910646B2 (en) &#34;Composition for gene transfer into cells&#34;
Fenske et al. Cationic poly (ethyleneglycol) lipids incorporated into pre-formed vesicles enhance binding and uptake to BHK cells
WO1996024334A1 (en) Lipid constructs for cytoplasmic delivery of agents
WO1996024334A9 (en) Lipid constructs for cytoplasmic delivery of agents
Berton et al. Improved oligonucleotide uptake and stability by a new drug carrier, the SupraMolecular BioVector (SMBV)
Zelphati et al. Cationic liposomes as an oligonucleotide carrier: mechanism of action
CA2398945A1 (en) Small oligonucleotides with anti-tumor activity
STERNBERG liposome-DNA complexes for gene therapy
JP4033502B2 (en) Ribozymes, liposome preparations and uses thereof
WO2001042424A2 (en) Sphingolipid-containing cationic liposomes for topical delivery of bioactive material
WO2010110318A1 (en) Therapeutic agent for arteriosclerotic diseases comprising nucleic acid

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UG UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

COP Corrected version of pamphlet

Free format text: PAGES 1/4-4/4,DRAWINGS,REPLACED BY NEW PAGES BEARING THE SAME NUMBER;DUE TO LATE TRANSMITTAL BY THERECEIVING OFFICE

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

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase