WO2003066054A1 - Cationic polymers for use in therapeutic agent delivery - Google Patents

Cationic polymers for use in therapeutic agent delivery Download PDF

Info

Publication number
WO2003066054A1
WO2003066054A1 PCT/US2003/002706 US0302706W WO03066054A1 WO 2003066054 A1 WO2003066054 A1 WO 2003066054A1 US 0302706 W US0302706 W US 0302706W WO 03066054 A1 WO03066054 A1 WO 03066054A1
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
subject
therapeutic
acid molecule
polymer
Prior art date
Application number
PCT/US2003/002706
Other languages
French (fr)
Inventor
Puthupparampil V. Scaria
Aslam M. Ansari
Martin C. Woodle
Original Assignee
Intradigm Corporation
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 Intradigm Corporation filed Critical Intradigm Corporation
Priority to AU2003205380A priority Critical patent/AU2003205380A1/en
Publication of WO2003066054A1 publication Critical patent/WO2003066054A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/765Polymers containing oxygen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/785Polymers containing nitrogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention provides novel polymers for delivering one or more compositions in vivo, as well as methods of using the same.
  • the invention provides methods for delivering one or more therapeutic nucleic acids in vivo.
  • drug and gene delivery vectors e.g., viral vectors, polymers, nanoparticles, and liposomes
  • drug and gene delivery vectors have shown great promise in overcoming the barriers to drug and gene delivery, such as rapid clearance from blood and targeted intracellular delivery of exogenous nucleic acids or drugs
  • delivery systems continue to face other obstacles that limit their therapeutic application.
  • immunogenicity and toxicity of viral gene expression vectors are among the barriers that limit the therapeutic application of gene-based medicines.
  • a patient's immune defense often mounts a response to any administered viral vector particle, as the particles are produced using a natural packaging cell production.
  • This "natural packaging" produces particles virtually identical to those of the virus from which the vector is derived.
  • the produced capsid or envelop thus, is sensitive and susceptible to host immune defenses, which block the delivery of the recombinant genome.
  • Non-viral delivery systems have been developed to overcome the safety problems associated with live vectors.
  • non-viral systems often are limited by low efficiency and a very short persistence.
  • non-viral vectors suffer from unique toxicity problems.
  • "First generation" non-viral vector systems such as poly-(L- lysine)(PLL) and poly-(ethyleneimine) (PEI) are toxic to certain organs in mammals, thereby limiting their use.
  • these polymers adversely affect liver and lung tissues. Accordingly, these toxicity problems need to be managed by chemical modifications .
  • the present invention provides novel polymers for delivery of therapeutic agents to a tissue or cell in vivo with minimized toxicity.
  • the invention also provides methods of administering therapeutic agents to a subject in need thereof on a repeated basis.
  • certain polymers surprisingly can provide for enhanced peptide, protein, drug and nucleic acid (i.e., DNA and RNA and their analogues) delivery, and that such polymers can be administered to a subject to provide an effective amount of a therapeutic agent over an extended period of time.
  • This finding is significant, given the limitations of conventional delivery compositions.
  • the drug, peptide and gene delivery vehicles of the invention are useful in a number of therapeutic applications, including, for example: therapeutic vaccines, preventative vaccines, treatment of inflammatory disorders and many types of malignancies, as well as any other regimen involving repeated administration or expression of a therapeutic agent including nucleic acid molecules and polypeptides.
  • the present invention in one aspect, provides novel bio-degradable or biocompatible cationic polymers that can bind a nucleic acid, polypeptide or small molecule agent.
  • a polymer of the invention preferably binds a nucleic acid, and can deliver the nucleic acid to a targeted cell or tissue.
  • a polymer of the invention can be used in any number of ways to deliver a nucleic acid, polypeptide or other small molecule to a targeted cell or tissue in vivo.
  • a polymer of the invention preferably contains a mixture of ester and amide bonds between various monomer units, as well as substitutions on the polymer backbone, which affect solvent accessibility. These features aid in modulating polymer degradation rate in vivo.
  • a cationic polymer of the invention preferably is able to bind a nucleic acid, which can be effectuated by conventional procedures.
  • an -NH 2 group of a cationic polymer can be protonated to thereby have an affinity for a nucleic acid molecule.
  • a nucleic acid molecule upon binding to a polymer of the invention, condenses into particles or colloids. These condensed particles unravel, however, as the backbone of the cationic polymer is degraded. The kinetics of nucleic acid unraveling is, therefore, directly related to the rate of polymer degradation.
  • the degradation rate of the polymers of the invention (and, hence, the rate of nucleic acid unraveling) can be modulated, for example, by constructing a polymer backbone with internal linkages that are degradable at different rates.
  • Monomer units may be selected such that they provide various functions required for binding and delivery of a therapeutic agent, e.g., a nucleic acid molecule.
  • cationic side chains of the monomers provide nucleic acid binding.
  • a pH-sensitive side chain such as one containing histidine, provides endosome disruption activity, which is desirable for release of the nucleic acid molecule and its complex from endosomal compartments before reaching the lysosomes where they are degraded.
  • Certain amino acids and derivatives thereof e.g., lysine, ornithine and arginine, provide a cationic side chain if linked to a neighboring residue via an amide bond.
  • Other amino acids such as serine and threonine, provide cationic side chains when linked to a neighboring residue via an ester bond.
  • Polymer side chains may also serve as conjugation sites to attach ligands and other molecules.
  • ligands and molecules include steric polymers, fusogenic molecules, nuclear localizing molecules, ligands and ligand-conjugated steric polymers.
  • Functional moieties suitable for attachment to a polymer side chain include: vascular endothelial growth factors, somatostatin and somatostatin analogs, transferring, melanotropin, ApoE and ApoE peptides, von Willebrand's factor and von Willebrand's factor peptides, adeno viral fiber protein and adenoviral fiber protein peptides, PDl and PDl peptides, EGF and EGF peptides, RGD peptides, CCK peptides, antibody and antibody fragments, folate, pyridoxyl and sialyl-LewisX and chemical analogs.
  • Ligands and steric polymers can be attached to the polymers through chemical conjugation at the amine side chains. Accordingly, a polymer of the invention can be represented by one of the following structures:
  • R may be H or CH 3 and n may be 1-500;
  • R may be H or CH 3 ;
  • R 3 may be NH 2 , OH, SH, CN, N 3 ;
  • R 4 may be H, a hydrocarbon, or a hydrophilic polymer, such as poly(ethyleneglycol) ("PEG”), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly-vinylpyrrolidone, polyvinyl alcohol, polyme hyloxazoline, polyethyloxazoline, polyhydroxyethyl- oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers, a ligand conjugated hydrophilic polymer, or ligand, which can be a cell targeting ligand, tissue targeting ligand, fusogenic moiety, or a nuclear localization moiety.
  • R 5 may be H, a hydrocarbon, or a hydrophilic polymer, such as PEG, polymethyloxazoline, polyethyloxazoline, polyacetal, a ligand conjugated hydrophilic polymer, or ligand,
  • X may be a hydrocarbon chain of length 1-10 carbons
  • n may be 1-50;
  • n may be 1-100;
  • n may be 1-20;
  • n may be 1-100
  • X may be hydrocarbon of length 1-10 carbons
  • n may be 1-100
  • n may be 1-100
  • X may hydrocarbon of length 1-10 carbons.
  • the instant hydrocarbon chains may be an alkyl group, an alkenyl group, or an alkynyl group.
  • alkyl refers to a straight-chain or branch-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 1 to about 15 and more preferably from 1 to about 10 carbon atoms.
  • alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, n- hexyl and the like.
  • alkenyl refers to a straight-chain or branched-chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 2-10 carbon atoms and more preferably, from 2-6 carbon atoms.
  • alkenyl radicals include, but are not limited to, ethenyl, E- and Z-propenyl, isopropenyl, E- and Z-butenyl, E- and Z- isobutenyl, E- and Z-pentenyl, E- and Z-hexenyl, E,E-, E,Z-, Z,E- and Z,Z- hexadienyl and the like.
  • alkynyl refers to a straight-chain or branched-chain hydrocarbon radical having one or more triple bonds containing the specified number of carbon atoms, or where no number is specified, preferably from 2 to about 10 carbon atoms.
  • alkynyl radicals include, but are not limited to, ethynyl, propynyl, propargyl, butynyl, pentynyl and the like.
  • Polymers of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active acid or base.
  • appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts.
  • a different process for separation of optical isomers involves the use of a chiral cliromatography column optimally chosen to maximize the separation of the enantiomers.
  • Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting the instant polymers with an optically pure acid in an activated form or an optically pure isocyanate.
  • the synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound.
  • the optically active compounds can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
  • the compounds of the present invention can be used in the form of salts derived from inorganic or organic acids.
  • These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate, 3-
  • the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and other. Water or oil-soluble or dispersible products are thereby obtained.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides such
  • acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid.
  • organic acids such as oxalic acid, maleic acid, succinic acid and citric acid.
  • Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
  • the instant invention contemplates protonated species of the instant polymers, and salts thereof.
  • Two examples of particularly preferred embodiments of the invention are the protonated species:
  • the present invention implements peptide, drug and nucleic acid delivery compositions that do not stimulate an immune response to the same degree typically associated with conventionally available compositions.
  • a "delivery composition” contains, at a minimum, (i) a novel polymer of the invention, and (ii) a therapeutic agent (e.g., nucleic acid, peptide, or small molecule drug) binded thereto.
  • the delivery composition may contain other domains, such as a ligand or tissue-targeting domain, and may also contain a protective polymer, such as poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl-oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
  • PEG poly(ethyleneglycol)
  • polyoxazoline polyacetal
  • hydrolyzed dextran polyacetal polymers polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl-oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
  • the ligand of the instant invention may comprise, for example, a targeting ligand or moiety for targeting specific cells and tissues.
  • a targeting ligand or moiety for targeting specific cells and tissues may contain a fusogenic moiety to facilitate entry into cells.
  • Yet another embodiment may comprises a nuclear targeting moiety.
  • a targeting ligand enhances binding of the polymer to target tissue or cells and permits highly specific interaction of the polymers with the target tissue or cell.
  • the polymer will include a ligand effective for ligand-specific binding to a receptor molecule on a target tissue and cell surface (Woodle et al., Small molecule ligands for targeting long circulating liposomes, in Long Circulating Liposomes: Old drags, new Therapeutics, Woodle and Storm eds., Springer, 1998, p 287-295).
  • the polymer may include two or more targeting moieties, depending on the cell type that is to be targeted.
  • targeting moieties can provide additional selectivity in cell targeting, and also can contribute to higher affinity and/or avidity of binding of the polymer to the target cell.
  • the relative molar ratio of the targeting moieties may be varied to provide optimal targeting efficiency. Methods for optimizing cell binding and selectivity in this fashion are known in the art. The skilled artisan also will recognize that assays for measuring cell selectivity and affinity and efficiency of binding are known in the art and can be used to optimize the nature and quantity of the targeting ligand(s).
  • Suitable ligands include, but are not limited to: vascular endothelial cell growth factor for targeting endothelial cells: FGF2 for targeting vascular lesions and tumors; somatostatin peptides for targeting tumors; transferrin for targeting umors; melanotropin (alpha MSH) peptides for tumor targeting; ApoE and peptides for LDL receptor targeting; von Willebrand's Factor and peptides for targeting exposed collagen; Adenoviral fiber protein and peptides for targeting Coxsackie-adenoviral receptor (CAR) expressing cells; PDl and peptides for targeting Neuropilin 1; EGF and peptides for targeting EGF receptor expressing cells; and RGD peptides for targeting integrin expressing cells, DNA and RNA aptamers.
  • FGF2 for targeting vascular lesions and tumors
  • somatostatin peptides for targeting tumors
  • transferrin for targeting umors
  • kits for treating tumor cells having cell-surface folate receptors include (i) folate, where the polymer is intended for treating tumor cells having cell-surface folate receptors, (ii) pyridoxyl, where the polymer is intended for treating virus-infected CD4+ lymphocytes, or (iii) sialyl- Lewis, where the polymer is intended for treating a region of inflammation.
  • Other peptide ligands may be identified using methods such as phage display (F.
  • the targeting ligand may be somatostatin or a somatostatin analog.
  • Somatostatin has the sequence AGCLNFFWKTFTSC, and contains a disulfide bridge between the cysteine residues.
  • Many somatostatin analogs that bind to the somatostatin receptor are known in the art and are suitable for use in the present invention, such as those described, for example, in U.S. Patent No. 5,776,894, which is incorporated herein by reference in its entirety.
  • these compounds are cyclic due to a disulfide bond between the cysteine residues.
  • these analogs may be derivatized at the free amino group of the phenylalanine residue, for example with a polycationic moiety such as a chain of lysine residues.
  • a polycationic moiety such as a chain of lysine residues.
  • the targeting moiety provides tissue- and cell- specific binding.
  • the ligands may be covalently attached to the polymer so that exposure is adequate for tissue and cell binding.
  • a peptide ligand can be covalently coupled to a polymer such as polyoxazoline.
  • the number of targeting molecules present on the outer layer will vary, depending on factors such as the avidity of the ligand-receptor interaction, the relative abundance of the receptor on the target tissue and cell surface, and the relative abundance of the target tissue and cell. Nevertheless, a targeting molecule coupled with of each polymer usually provides suitable enhancement of cell targeting.
  • An appropriate assay for such binding may be ELISA plate assays, cell culture expression assays, or any other binding assays.
  • the fusogenic moiety promotes fusion of the polymer to the cell membrane of the target cell, facilitating entry of the polymer and therapeutic agents into the cell.
  • the fusogenic moiety comprises a fusion-promoting element.
  • Such elements interact with cell membranes or endosome membranes in a manner that allows transmembrane movement of large molecules or particles, or disrupts the membranes such that the aqueous phases that are separated by the membranes may freely mix.
  • suitable fusogenic moieties include, but are not limited to membrane surfactant peptides, e.g. viral fusion proteins such as hemagglutinin (HA) of influenza virus, or peptides derived from toxins such as PE and ricin.
  • sequences that permit cellular trafficking such as HIV TAT protein and antennapedia or those derived from numerous other species, or synthetic polymers that exhibit pH sensitive properties such as poly(ethylacrylic acid)(Lackey et al., Proc. Int. Symp. Control. Rel. Bioact. Mater. 1999, 26, #6245), N-isopropylacrylamide methacrylic acid copolymers (Meyer et al., FEBS Lett. 421:61 (1999)), or poly(amidoamine)s, (Richardson et al., Proc. Int. Symp. Control. Rel. Bioact. Mater.
  • Suitable membrane surfactant peptides include an influenza hemagglutinin or a viral fusogenic peptide such as the Moloney murine leukemia virus ("MoMuLV” or MLV) envelope (env) protein or vesicular stroma virus (VSV) G-protein.
  • MoMuLV Moloney murine leukemia virus
  • env Moloney murine leukemia virus
  • VSV vesicular stroma virus
  • the membrane-proximal cytoplasmic domain of the MoMuLV env protein may be used. This domain is conserved among a variety viruses and contains a membrane-induced ⁇ -helix.
  • Suitable viral fusogenic peptides for the instant invention may include a fusion peptide from a viral envelope protein ectodomain, a membrane-destabilizing peptide of a viral envelope protein membrane-proximal domain, hydrophobic domain peptide segments of so called viral "fusion" proteins, and an amphiphilic-region containing peptide.
  • Suitable amphiphilic-region containing peptides include, but are not limited to: melittin, the magainins, fusion segments from H.
  • influenza hemagglutinin (HA) protein HIV segment I from the cytoplasmic tail of HIVl gp41, and amphiphilic segments from viral env membrane proteins including those from avian leukosis virus (ALV), bovine leukemia virus (BLV), equine infectious anemia (EIA), feline immunodeficiency virus (FIV), hepatitis virus, herpes simplex virus (HSV) glycoprotein H, human respiratory syncytia virus (hRSV), Mason-Pfizer monkey virus (MPMV), Rous sarcoma vims (RSV), parainfluenza virus (PINF), spleen necrosis viras (SNV), and vesicular stomatitis virus (VSV).
  • ABV avian leukosis virus
  • BLV bovine leukemia virus
  • EIA equine infectious anemia
  • FV feline immunodeficiency virus
  • HSV herpes simplex virus glycoprotein H
  • Suitable peptides include microbial and reptilian cytotoxic peptides.
  • the specific peptides or other molecules having greatest utility can be identified using four kinds of assays: 1) ability to disrupt and induce leakage of aqueous markers from liposomes composed of cell membrane lipids or fragments of cell membranes, 2) ability to induce fusion of liposomes composed of cell membrane lipids or fragments of cell membranes, 3) ability to induce cytoplasmic release of particles added to cells in tissue culture, and 4) ability to enhance plasmid expression by particles in vivo tissues when administered locally or systemically.
  • the fusogenic moiety also may be comprised of a polymer, including peptides and synthetic polymers.
  • the peptide polymer comprises synthetic peptides containing amphipathic aminoacid sequences such as the "GALA'and "KALA'peptides (Wyman TB, Nicol F, Zelphati O, Scoria PV, Plank C, Szoka FC Jr, Biochemistry 1997, 36:3008-3017; Subbarao NK, Parente RA, Szoka FC Jr, Nadasdi L, Pongracz K, Biochemistry 1987 26:2964- 2972 or Wyman supra, Subbarao supra ).
  • peptides include non-natural aminoacids, including D aminoacids and chemical analogues such as peptoids, imidazole-containing polymers.
  • Suitable polymers include molecules containing amino or imidazole moieties with intermittent carboxylic acid functionalities such as ones that form "salt-bridges," either internally or externally, including forms where the bridging is pH sensitive.
  • Other polymers can be used including ones having disulfide bridges either internally or between polymers such that the disulfide bridges block fusogenicity and then bridges are cleaved within the tissue or intracellular compartment so that the fusogenic properties are expressed at those desired sites.
  • a polymer that forms weak electrostatic interactions with a positively charged fusogenic polymer that neutralizes the positive charge could be held in place with disulfide bridges between the two molecules and these disulfides cleaved within an endosome so that the two molecules dissociate, releasing the positive charge and fusogenic activity.
  • Another form of this type of fusogenic agent has the two properties localized onto different segments of the same molecule and thus the bridge is intramolecular so that its dissociation results in a structural change in the molecule.
  • Yet another form of this type of fusogenic agent has a pH sensitive bridge.
  • the fusogenic moiety also may comprise a membrane surfactant polymer- lipid conjugate.
  • the polymer will be either biodegradable or of sufficiently small molecular weight that it can be excreted without metabolism. The skilled artisan will recognize that other fusogenic moieties also may be used without departing from the spirit of the invention.
  • nucleic targeting moiety of the invention is "nuclear targeted,” that is, it contains one or more molecules that facilitate entry of the nucleic acid through the nuclear membrane into the nucleus of the host cell.
  • nuclear targeting may be achieved by incorporating a nuclear membrane transport peptide, or nuclear localization signal (“NLS") peptide, or small molecule that provides the same NLS function, into the core complex. Suitable peptides are described in, for example, U.S.
  • Patent Nos 5,795,587 and 5,670,347 and in patent application WO 9858955 which are hereby incorporated by reference in their entirety, and in Aronsohn et al., J. Drug Targeting 1:163 (1997); Zanta et al., Proc. Nat'l Acad. Sci. USA 96:91-96 (1999); Ciolina et al., Targeting of Plasmid DNA to Importin alpha by Chemical coupling with Nuclear Localization Signal Peptides, in Vector Targeting Strategies for Therapeutic Gene Delivery (Abstracts from Cold Spring Harbor Laboratory 1999 meeting), 1999, p 20; Saphire et al., J. Biol Chem; 273:29764 (1999).
  • a nuclear targeting peptide may be a nuclear localization signal peptide or nuclear membrane transport peptide and it may be comprised of natural aminoacids or non-natural aminoacids including D aminoacids and chemical analogues such as peptoids.
  • the NLS may be comprised of aminoacids or their analogues in a natural sequence or in reverse sequence.
  • Another embodiment provides a steroid receptor-binding NLS moiety that activates nuclear transport of the receptor from the cytoplasm, wherein this transport carries the nucleic acid with the receptor into the nucleus.
  • the NLS is coupled to the polymer in such a manner that the polymer is directed to the cell nucleus where it permits entry of a nucleic acid into the nucleus.
  • incorporation of the NLS moiety into the polymer occurs through association with the nucleic acid, and this association is retained within the cytoplasm. This minimizes loss of the NLS function due to dissociation with the nucleic acid and ensures that a high level of the nucleic acid is delivered to the nucleus. Furthermore, the association with the nucleic acid does not inhibit the intended biological activity within the nucleus once the nucleic acid is delivered.
  • the intended target of the biological activity of the nucleic acid is the cytoplasm or an organelle in the cytoplasm such as ribosomes, the golgi apparatus, or the endoplasmic reticulum.
  • a localization signal is included in the polymer or anchored to it so that it provides direction of the nucleic acid to the intended site where the nucleic acid exerts its activity. Signal peptides that can achieve such targeting are Icnown in the art.
  • the ligand may be conjugated to a hydrophilic polymer.
  • hydrophilic polymer include, but are not limited to poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly-vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl- oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
  • PEG poly(ethyleneglycol)
  • polyoxazoline polyacetal
  • hydrolyzed dextran polyacetal polymers polylactic acid, polyglycolic acid, poly-vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl- oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
  • a stable complex preferably is used for transporting the agent into the cell (including the nucleus).
  • a "stable complex” is a complex that, once delivered in vivo, will not dissociate due to interactions with serum proteins or other types of molecules that may bind or associate with the complex. Once inside the desired subcellular compartment, however, the nucleic acid generally dissociates from the delivery system to become active. Low efficiency of synthetic gene vector systems is due, in part, to difficulties in achieving nucleic acid dissociation from the complex.
  • a highly stable complex is not optimal for nucleic acid delivery. Instead, the complex needs to umavel and free the nucleic acid within the subcellular compartment.
  • the invention has overcome this problem by using biodegradable poly-cations, which effectively can bind nucleic molecules, yet can disintegrate and release the nucleic acid upon exposure to conditions in the target site, such as, e.g., acidic conditions in the endosome, or reducing conditions within the cytoplasm of cells.
  • a polymer of the invention binds to a nucleic acid sequence that encodes a viral genomic sequence, otherwise known as a "viral genome.”
  • the viral genome also contains within its open reading frame a therapeutic gene (i.e., nucleic acid) of interest.
  • nucleic acid i.e. both the viral genomic sequence and therapeutic nucleic acid sequence
  • the "entire" nucleic acid sequence i.e. both the viral genomic sequence and therapeutic nucleic acid sequence
  • the entire nucleic acid sequence can be cloned into a plasmid to generate multiple copies, then the plasmid can be complexed to a novel polymer of the invention.
  • the nucleic acid encoding a viral genome can be self-replicative once administered to a subject, thereby providing an ongoing supply of a therapeutic nucleic acid molecule.
  • Suitable viral genomes are disclosed in U.S. Provisional Application Serial No. 60/330,909, entitled, "therapeutic methods for gene delivery systems," which hereby is incoiporated by reference in its entirety.
  • a novel polymer of the invention is a component of a "synthetic vector," which can be used to affectively deliver an agent to a targeted cell or tissue in vivo.
  • Synthetic vectors for use in the present invention have been disclosed by Woodle et al. (WO 01/49324, filed December 28, 2000), which application is hereby incorporated by reference in its entirety, including the drawings.
  • a "synthetic vector” means a multi-functional synthetic vector which, at a minimum, contains a nucleic acid binding domain and a ligand binding (e.g. tissue targeting) domain, and is complexed with a nucleic acid sequence.
  • a synthetic vector also may contain other domains such as, for example, a hydrophilic polymer domain, endosome disruption or dissociation domain, nuclear targeting domain, and nucleic acid condensing domain.
  • a synthetic vector also can include a protective polymer. The protective polymer reduces non-specific interactions and, in some embodiments, contain an "exposed" component (e.g., a targeting ligand) that provides added specificity.
  • a polymer of the invention is comprised of monomers or oligomers linked with biodegradable or biocompatible linkages.
  • the nucleic acid binding domain, or "complex forming reagent,” is capable of associating with a core nucleic acid complex in a manner that allows assembly of the nucleic acid core complex.
  • the complex forming reagent can be, e.g., a lipid, a synthetic polymer, a natural polymer, a semi-synthetic polymer, a mixture of lipids, a mixture of polymers, a lipid and polymer combination, or a spermine analogue complex.
  • a suitable polymer may contain histidine or an imidazole functional group.
  • WO 01/49324 at, e.g., pages 20-34 disclose suitable DNA binding domains for use in the present invention.
  • a synthetic vector for use in the present invention preferably provides reduced non-specific interactions, yet effectively can engage in ligand-mediated (i.e., specific) cellular binding.
  • a synthetic vector for use in the present invention is able to be complexed to one or more therapeutic nucleic acids, which then can be administered to a subject.
  • the therapeutic nucleic acid is comprised within a nucleic acid sequence encoding a viral genomic sequence; the entire nucleic acid sequence (i.e. both the viral genomic sequence and therapeutic nucleic acid sequence) is complexed to the DNA binding domain of the synthetic vector.
  • the entire nucleic acid sequence can be cloned into a plasmid to generate multiple copies, then the plasmid can be complexed to the DNA binding domain of the synthetic vector.
  • the nucleic acid encoding a viral genome can be self-replicative once administered to a subject, thereby providing an ongoing supply of a therapeutic nucleic acid molecule.
  • a synthetic vector for use in the invention is able to target specific tissues.
  • the cationic surface charge of a synthetic vector can act to target a cell.
  • the ability to bind a target cell can be lost when a steric polymer coat is added to the synthetic vector as a hydrophilic polymer domain, such as a hydrophilic polymer domain disclosed herein.
  • Targeting activity of the synthetic vector can be restored by employing a ligand domain, which can effecmate ligand-mediated binding and cellular uptake of the synthetic vector.
  • a ligand may be conjugated to the distal end of the steric polymer in order to mediate binding with one or more cell surface receptors, acidic conditions in the endosome, or reducing conditions within the cytoplasm of cells.
  • Suitable ligands and tissue-targeting molecules are disclosed herein or in references incorporated herein.
  • the synthetic vectors for use in the invention also account for drawbacks associated with other gene delivery vehicles, such as non-specific interactions, which often result from an electrostatic charge differential between a vector and its environment.
  • a synthetic vector e.g., a condensed cationic reagent-DNA complex, can be made net positive, neutral, or negative depending on the ratio of ' the components in the complex. While electrostatic interactions between a negatively charged cell membrane and a positively charged particle can increase cellular uptake, all cells possess a negative membrane charge. Accordingly, nonspecific interactions can persist in a complex containing a net positive charge.
  • a hydrophilic polymer domain of a synthetic vector preferably is able to minimize undesirable non-specific interactions by controlling the surface properties of the synthetic vector.
  • the hydrophilic polymer may be selected from the group consisting of poly(ethyleneglycol) ("PEG”), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyloxazoline, poly-hydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
  • PEG poly(ethyleneglycol)
  • polyoxazoline polyacetal
  • hydrolyzed dextran polyacetal polymers polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyloxazoline, poly-hydroxypropyloxazoline, and other conventionally
  • a hydrophilic, steric coat can be introduced onto the surface of a synthetic vector by covalently conjugating the polymer to the condensing agent before complexing with therapeutic nucleic acid. This method is preferred over conjugating a hydrophilic, steric polymer to a pre-formed DNA-synthetic vector complex, since chemical reactions carried out after DNA complexing can damage the DNA. Moreover, as the steric barrier is formed, subsequent conjugation reactions are inhibited, which can limit the amount of polymer that can be conjugated to the complex surface.
  • any of the foregoing hydrophilic polymers is conjugated to a spermine or spermadine analog, such as those described in Woodle et al, (WO 01/49324, filed December 28, 2000), which, in torn, interacts with a therapeutic nucleic acid molecule.
  • the chosen hydrophilic polymer also may have complexed thereto a ligand or other tissue targeting moiety.
  • a hydrophilic polymer is conjugated at random to one or more sites on the nucleic acid binding domain, using either a stable covalent linkage or a linkage that can be cleaved.
  • the grafting density can be varied between 2% and 25% of monomer units.
  • Samples having a single molecular weight of the steric polymer can be used.
  • An alternative steric polymer is polyacetal, derived by oxidation and subsequent reduction of dextran.
  • the polymer is linear, possessing one or two alcohol moieties in place of each hexose ring.
  • Polyacetal has been shown to function as a steric polymer for drug delivery and when conjugated to lipids and polycations.
  • a steric polymer layer that can block non-specific binding will increase the serum half-life of a synthetic vector, since (i) minimal non-specific interactions render the particles relatively inert, and (ii) the relatively large size allows the synthetic vector to remain in the blood for prolonged periods.
  • Successful construction and use of a steric polymer layer can be observed from blood pharmacokinetics of the complexes following an intravenous administration (e.g., PEG, PEI and DOTAP: Cholesterol complexes).
  • the present invention provides methods of administering one or more therapeutic drugs or nucleic acid molecules to a subject, using a vehicle comprised of a polymer of the invention, to bring about a therapeutic benefit to the subject.
  • a "therapeutic nucleic acid molecule" or “therapeutic nucleic acid” is any nucleic acid (e.g., DNA or RNA) that, as a nucleic acid or as an expressed nucleic acid or polypeptide, confers a therapeutic benefit to a subject.
  • a therapeutic nucleic acid molecule is administered to a subject as part of, or via, a nucleic acid delivery vehicle.
  • the subject preferably is mammalian such as a mouse, and more preferably is a human being.
  • Gene delivery vehicles for use in the present invention can be used to stimulate an immune response, which may be protective or therapeutic. Accordingly, the gene delivery vehicles can be used to vaccinate a subject against an antigen.
  • the invention provides methods vaccinating or enhancing a physiological response against a pathogen in a subject.
  • This methodology can entail administering to the subject a first, or priming, dosage of a therapeutic nucleic acid molecule that encodes a therapeutic polypeptide, followed by administering to the subject one or more booster dosages of the nucleic acid molecule.
  • the administration regimen can vary, depending on, for example, (i) the subject to whom the therapeutic nucleic acid molecule is administered, and (ii) the pathogen that is involved.
  • a booster dosage of a therapeutic nucleic acid molecule may administered about two weeks after priming, followed by successive booster dosages, which can occur between intervals of constant or increasing duration. It is desirable to administer therapeutic nucleic acid molecules at a periodicity that is appropriate according to the subject's immune response.
  • the administered nucleic acid molecule is comprised within a nucleic acid delivery vehicle of the invention.
  • expression of the therapeutic nucleic acid molecule in the foregoing steps elicits a humoral and/or cellular response in the subject, causing the subject to exhibit a degree of immunity against the pathogen that is greater than before the therapeutic method is carried out.
  • the antigen against which the subject exhibits an increased immunity can be the antigen encoded by the therapeutic nucleic acid molecule.
  • the antigen against which the subject exhibits an increased immunity is distinct from, or in addition to, the antigen expressed by the administered nucleic acid molecule.
  • the antigen encoded by the therapeutic nucleic acid can act to enhance an immune response against another antigen, e.g., a component of a tumor.
  • the route of administration may vary, depending on the therapeutic application (i.e. preventative or therapeutic vaccine) and the type of disorder to be treated.
  • the nucleic acid delivery vehicle may be injected into the skin, muscle, intravenously, directly to the portal, hepatic vein or bile duct, locally to a tumor or to a joint, or orally.
  • the invention also contemplates enhanced delivery of a drug or nucleic acid therapeutic agent by employing oral administration.
  • Enhanced delivery can result from protection of the agent by the polymer and binding to target tissues and cells in the gastrointestinal tract.
  • Nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form.
  • the term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non- naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides.
  • nucleic acid examples include, but are not limited to recombinant plasmids and cosmids, vectors that integrate into the host chromosome, episomal vectors, replication-deficient plasmids, mini-plasmids, a recombinant viral genome, a linear nucleic acid fragment, an antisense agent, a linear polynucleotide, a circular polynucleotide, a ribozyme, a cellular promoter, and viral genomes.
  • the nucleic acid can encode one or more proteins, polypeptides or peptides, or multiple copies of peptides, polypeptides and proteins.
  • the nucleic acid can also be an antisense nucleic acid, and may also be an interfering double stranded RNA (RNAi) or an siRNA, or a polynucleotide encoding a double stranded RNA.
  • RNAi interfering double stranded RNA
  • siRNA siRNA
  • a polynucleotide encoding a double stranded RNA examples of suitable double stranded RNA molecules and vectors encoding such molecules are described in, for example, U.S. Patent No. 6,506,559, which is hereby incorporated by reference in its entirety.
  • nucleic acid analogs include, without limitation, phosphorothioates, phosphorarnidates, methyl phosphonates, chiral methyl phosphonates, 2-0- methyl ribonucleotides, and peptide-nucleic acids (PNAs).
  • a therapeutic nucleic acid molecule(s), which is administered in a gene delivery vehicle expresses an antigenic polypeptide.
  • Expression of an administered therapeutic nucleic acid may be obtained by repeated administration and with strong expression in antigen presenting cells ("APCs"), e.g., through booster dosages.
  • Nucleic acid expression also may occur by virtue of the self-replicative feature of the gene delivery vehicle of which the therapeutic nucleic acid molecule is comprised.
  • the invention provides for selective gene expression through use of tissue selective replication of viral nucleic acid.
  • the invention provides for viral vector replication whereby viral vector particles are produced by the target cells and tissues.
  • the viral vector particles produced provide for tissue selective spread and amplification.
  • Suitable antigens for use in the invention include antigens for infectious agents and for tumor cells that result in the recruitment of antibodies.
  • An administered therapeutic nucleic acid molecule also may induce an immune response.
  • a response can be achieved to intracellular infectious agents including, for example, tuberculosis, Lyme disease, and others.
  • a response can be achieved by expression of antigen, expression of cytokines, and their combination.
  • the invention also provides for expression of HIV antigens and induction of both a protective and a therapeutic immune response for preventing and treating HIV, respectively.
  • the invention additionally provides for the expression of antigens, which elicit a humoral and/or a cellular immune response. This heightened immune response can provide protection from a challenge with infectious agents characterized as having the antigen.
  • the invention utilizes an adenoviral genomic nucleic acid that (i) expresses an antigen under control of a promoter and (ii) targets an APC.
  • the therapeutic nucleic acid encodes a cytokine, which may be expressed with or without an antigen.
  • a cytokine acts to recruit an immune response, which can enhance an immune response to an expressed antigen. Accordingly, cytokine expression can be obtained whereby APCs and other immune response cells are recruited to the vicinity of tumor cells, in which case there is no requirement for co-expression of an antigen by the gene delivery vehicle. Yet, in another embodiment, one or more antigens and cytokines can be co-expressed.
  • the invention contemplates the use of immunostimulatory cytokines, as well as protein analogues exhibiting biological activity similar to an immunostimulatory cytokine, to vaccinate a subject.
  • Suitable cytokines for use in enhancing an immune response include GM-CSF, IL-1, IL-12, IL-15, interferons, B-40, B-7, tumor necrosis factor (TNF)and others.
  • TNF tumor necrosis factor
  • the invention also contemplates utilizing genes that down-regulate immunosupressant cytokines.
  • the invention also provides for expression of "recruitment cytokines" at tumors.
  • Expression of cytokines at tumors giving recruitment of immune response cells can initiate a cellular immune response at the tumor site giving recognition and killing of tumor cells at the site of expression and at distal tumor sites.
  • a preferred embodiment of the invention is comprised of an adenoviral genomic nucleic acid, the nucleic acid exhibiting expression of GM-CSF under a tumor-preferential promoter, further comprised of nucleic acid exhibiting rumor- conditional replication to form adenoviral vector particles exhibiting tumor- conditional replication, and yet further comprised by synthetic vector compositions targeting delivery to tumor lesions.
  • Another preferred embodiment of the invention utilizes an adenoviral genomic nucleic acid encoding a cytokine (e.g., GM-CSF) under regulation of a tumor-conditional promoter. This feature would result in enhanced cytokine expression at the site of a tumor.
  • the adenoviral genomic nucleic acid preferably is administered in conjunction with electroporation to tumor lesions.
  • a gene delivery vehicle also may be used to treat a disorder characterized by inflammation.
  • one or more therapeutic nucleic acid molecules comprised within a nucleic acid delivery vehicle is administered to a subject suffering from a disorder characterized by inflammation, in order to suppress or retard an immune response.
  • Treatable disorders include rheumatoid arthritis, psoriasis, gout and inflammatory bowel disorders.
  • Suitable therapeutic nucleic acids for use in treating inflammation include nucleic acids that encode an inflammation inhibitory cytokine for use in the present invention include, but are not limited to, IL-IRA, soluble TNF receptor, soluble Fas ligand, and others or nucleic acid such as antisense RNA or DNA and interfering RNA that can inhibit expression of inflammatory cytokines.
  • the route and site of administration will vary, depending on the disorder and the location of inflammation.
  • the gene delivery vehicle can be administered into a joint; administration thereto can be in conjunction with electroporation.
  • the nucleic acid delivery vehicles of the invention also can be used to treat a wide variety of diseases and syndromes in a subject, including, but not limited to cancer, cardiovascular diseases, and viral and bacterial infections.
  • Suitable nucleic acids for use in these methods are known in the art, and the present invention provides improved compositions and methods for delivering such nucleic acids to a subject.
  • Nucleic acid delivery vehicle containing viral genome, plasmid, antisense RNA, antisense DNA, or interfering RNA may be administered by various routes of administration into a tumor.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, whether a drag delivery system is utilized and whether the compound is administered as part of a drug combination.
  • the dosage regimen actually employed may vary widely and therefore may deviate from the preferred dosage regimen set forth above.
  • the compounds of the present invention may be administered orally, parenterally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrastemal injection, or infusion techniques.
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drag with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drag.
  • a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drag.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch.
  • Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally by prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more therapeutic agents, such as immunomodulators, antiviral agents or anti- infective agents.

Abstract

The invention provides novel nucleic acid delivery vehicles that can, for example, be administered to a subject repeatedly to effectively deliver one or more therapeutic drugs or nucleic acid molecules or polypeptides to a cell or tissue. The drug or gene delivery vehicles can be used, for instance, as part of the therapeutic and/or preventative vaccine, and/or as well as any other therapeutic regimen that involves an ongoing use of a therapeutic drug or nucleic acid molecule or polypeptide.

Description

CATIONIC POLYMERS FOR USE IN
THERAPEUTIC AGENT DELIVERY
BACKGROUND OF THE INVENTION
1. Field of the Invention
The invention provides novel polymers for delivering one or more compositions in vivo, as well as methods of using the same. In particular, the invention provides methods for delivering one or more therapeutic nucleic acids in vivo.
2. Background of the Invention
Although many drug and gene delivery vectors (e.g., viral vectors, polymers, nanoparticles, and liposomes) have shown great promise in overcoming the barriers to drug and gene delivery, such as rapid clearance from blood and targeted intracellular delivery of exogenous nucleic acids or drugs, such delivery systems continue to face other obstacles that limit their therapeutic application. For instance, immunogenicity and toxicity of viral gene expression vectors are among the barriers that limit the therapeutic application of gene-based medicines. To this end, a patient's immune defense often mounts a response to any administered viral vector particle, as the particles are produced using a natural packaging cell production. This "natural packaging" produces particles virtually identical to those of the virus from which the vector is derived. The produced capsid or envelop, thus, is sensitive and susceptible to host immune defenses, which block the delivery of the recombinant genome.
These drawbacks particularly limit the use of vectors. For instance, vectors that readily elicit an immune response typically are not suitable for therapeutic applications that require repeated administration of the vector. In this regard, the premature clearance of a vector from the body substantially eliminates the ability of using the vector. Another drawback to administering live, attenuated viruses is the considerable safety risk they pose. While efforts have been applied to control viral replication mechanism, certain levels of replication are needed to meet desirable efficacy levels. This dilemma is apparent in HIV vaccines, for failure of controlling replication can result in the transmission of AIDS.
Non-viral delivery systems have been developed to overcome the safety problems associated with live vectors. However, non-viral systems often are limited by low efficiency and a very short persistence.
In addition, non-viral vectors suffer from unique toxicity problems. For example, "First generation" non-viral vector systems, such as poly-(L- lysine)(PLL) and poly-(ethyleneimine) (PEI) are toxic to certain organs in mammals, thereby limiting their use. In particular, these polymers adversely affect liver and lung tissues. Accordingly, these toxicity problems need to be managed by chemical modifications .
There is, accordingly, a need for finding drug and gene delivery systems that: (i) are less toxic than conventionally used vectors, (ii) provide for prolonged persistence in vivo, and/or (iii) provide for selective expression in target tissues, yielding therapeutically effective levels of the therapeutic agent.
SUMMARY OF THE INVENTION
Accordingly, the present invention provides novel polymers for delivery of therapeutic agents to a tissue or cell in vivo with minimized toxicity.
The invention also provides methods of administering therapeutic agents to a subject in need thereof on a repeated basis.
These and other advantages of the invention will be apparent to a skilled worker by reference to the specification and conventional teachings in the art. DETAILED DESCRIPTION OF THE INVENTION
It has been discovered that certain polymers surprisingly can provide for enhanced peptide, protein, drug and nucleic acid (i.e., DNA and RNA and their analogues) delivery, and that such polymers can be administered to a subject to provide an effective amount of a therapeutic agent over an extended period of time. This finding is significant, given the limitations of conventional delivery compositions. As a result, the drug, peptide and gene delivery vehicles of the invention are useful in a number of therapeutic applications, including, for example: therapeutic vaccines, preventative vaccines, treatment of inflammatory disorders and many types of malignancies, as well as any other regimen involving repeated administration or expression of a therapeutic agent including nucleic acid molecules and polypeptides.
The present invention, in one aspect, provides novel bio-degradable or biocompatible cationic polymers that can bind a nucleic acid, polypeptide or small molecule agent. A polymer of the invention preferably binds a nucleic acid, and can deliver the nucleic acid to a targeted cell or tissue. By virtue of its properties, a polymer of the invention can be used in any number of ways to deliver a nucleic acid, polypeptide or other small molecule to a targeted cell or tissue in vivo.
A polymer of the invention preferably contains a mixture of ester and amide bonds between various monomer units, as well as substitutions on the polymer backbone, which affect solvent accessibility. These features aid in modulating polymer degradation rate in vivo.
A cationic polymer of the invention preferably is able to bind a nucleic acid, which can be effectuated by conventional procedures. For instance, an -NH2 group of a cationic polymer can be protonated to thereby have an affinity for a nucleic acid molecule. A nucleic acid molecule, upon binding to a polymer of the invention, condenses into particles or colloids. These condensed particles unravel, however, as the backbone of the cationic polymer is degraded. The kinetics of nucleic acid unraveling is, therefore, directly related to the rate of polymer degradation. The degradation rate of the polymers of the invention (and, hence, the rate of nucleic acid unraveling) can be modulated, for example, by constructing a polymer backbone with internal linkages that are degradable at different rates.
Monomer units may be selected such that they provide various functions required for binding and delivery of a therapeutic agent, e.g., a nucleic acid molecule. For example, cationic side chains of the monomers provide nucleic acid binding. A pH-sensitive side chain, such as one containing histidine, provides endosome disruption activity, which is desirable for release of the nucleic acid molecule and its complex from endosomal compartments before reaching the lysosomes where they are degraded. Certain amino acids and derivatives thereof, e.g., lysine, ornithine and arginine, provide a cationic side chain if linked to a neighboring residue via an amide bond. Other amino acids, such as serine and threonine, provide cationic side chains when linked to a neighboring residue via an ester bond.
Polymer side chains may also serve as conjugation sites to attach ligands and other molecules. Such ligands and molecules include steric polymers, fusogenic molecules, nuclear localizing molecules, ligands and ligand-conjugated steric polymers. Functional moieties suitable for attachment to a polymer side chain (either alone or in comibination) include: vascular endothelial growth factors, somatostatin and somatostatin analogs, transferring, melanotropin, ApoE and ApoE peptides, von Willebrand's factor and von Willebrand's factor peptides, adeno viral fiber protein and adenoviral fiber protein peptides, PDl and PDl peptides, EGF and EGF peptides, RGD peptides, CCK peptides, antibody and antibody fragments, folate, pyridoxyl and sialyl-LewisX and chemical analogs. Ligands and steric polymers can be attached to the polymers through chemical conjugation at the amine side chains. Accordingly, a polymer of the invention can be represented by one of the following structures:
Figure imgf000006_0001
wherein R may be H or CH3 and n may be 1-500; or
Figure imgf000006_0002
wherein
Figure imgf000006_0003
R may be H or CH3;
R3 may be NH2, OH, SH, CN, N3;
R4 may be H, a hydrocarbon, or a hydrophilic polymer, such as poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly-vinylpyrrolidone, polyvinyl alcohol, polyme hyloxazoline, polyethyloxazoline, polyhydroxyethyl- oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers, a ligand conjugated hydrophilic polymer, or ligand, which can be a cell targeting ligand, tissue targeting ligand, fusogenic moiety, or a nuclear localization moiety. R5 may be H, a hydrocarbon, or a hydrophilic polymer, such as PEG, polymethyloxazoline, polyethyloxazoline, polyacetal, a ligand conjugated hydrophilic polymer, or ligand,
X may be a hydrocarbon chain of length 1-10 carbons
n may be 1-50; and
m may be 1-100; or
Figure imgf000007_0001
wherein,
n may be 1-20;
m may be 1-100; and
X may be hydrocarbon of length 1-10 carbons; or
Figure imgf000008_0001
wherein,
n may be 1-100;
m may be 1-100; and
X may hydrocarbon of length 1-10 carbons.
The instant hydrocarbon chains may be an alkyl group, an alkenyl group, or an alkynyl group.
The term "alkyl", alone or in combination with any other term, refers to a straight-chain or branch-chain saturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 1 to about 15 and more preferably from 1 to about 10 carbon atoms. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isoamyl, n- hexyl and the like.
The term "alkenyl", alone or in combination with any other term, refers to a straight-chain or branched-chain mono- or poly-unsaturated aliphatic hydrocarbon radical containing the specified number of carbon atoms, or where no number is specified, preferably from 2-10 carbon atoms and more preferably, from 2-6 carbon atoms. Examples of alkenyl radicals include, but are not limited to, ethenyl, E- and Z-propenyl, isopropenyl, E- and Z-butenyl, E- and Z- isobutenyl, E- and Z-pentenyl, E- and Z-hexenyl, E,E-, E,Z-, Z,E- and Z,Z- hexadienyl and the like.
The term "alkynyl," alone or in combination with any other term, refers to a straight-chain or branched-chain hydrocarbon radical having one or more triple bonds containing the specified number of carbon atoms, or where no number is specified, preferably from 2 to about 10 carbon atoms. Examples of alkynyl radicals include, but are not limited to, ethynyl, propynyl, propargyl, butynyl, pentynyl and the like.
Polymers of the present invention can possess one or more asymmetric carbon atoms and are thus capable of existing in the form of optical isomers as well as in the form of racemic or nonracemic mixtures thereof. The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereoisomeric salts by treatment with an optically active acid or base. Examples of appropriate acids are tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid and then separation of the mixture of diastereoisomers by crystallization followed by liberation of the optically active bases from these salts. A different process for separation of optical isomers involves the use of a chiral cliromatography column optimally chosen to maximize the separation of the enantiomers. Still another available method involves synthesis of covalent diastereoisomeric molecules by reacting the instant polymers with an optically pure acid in an activated form or an optically pure isocyanate. The synthesized diastereoisomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to deliver the enantiomerically pure compound. The optically active compounds can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt. The compounds of the present invention can be used in the form of salts derived from inorganic or organic acids. These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, palmoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, mesylate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and other. Water or oil-soluble or dispersible products are thereby obtained.
Examples of acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid. Other examples include salts with alkali metals or alkaline earth metals, such as sodium, potassium, calcium or magnesium or with organic bases.
The instant invention contemplates protonated species of the instant polymers, and salts thereof. Two examples of particularly preferred embodiments of the invention are the protonated species:
Figure imgf000011_0001
wherein the variables are defined herein. The instant invention also includes salts of these species.
The present invention implements peptide, drug and nucleic acid delivery compositions that do not stimulate an immune response to the same degree typically associated with conventionally available compositions. A "delivery composition" contains, at a minimum, (i) a novel polymer of the invention, and (ii) a therapeutic agent (e.g., nucleic acid, peptide, or small molecule drug) binded thereto. The delivery composition may contain other domains, such as a ligand or tissue-targeting domain, and may also contain a protective polymer, such as poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl-oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers. One or more protective groups can be added by methods described and/or referenced herein. Delivery of the composition can be aided by techniques such as, e.g., electroporation or hydrostatic pressure.
In a particular embodiment, the ligand of the instant invention may comprise, for example, a targeting ligand or moiety for targeting specific cells and tissues. Another embodiment may contain a fusogenic moiety to facilitate entry into cells. Yet another embodiment may comprises a nuclear targeting moiety.
A targeting ligand enhances binding of the polymer to target tissue or cells and permits highly specific interaction of the polymers with the target tissue or cell. In one embodiment, the polymer will include a ligand effective for ligand-specific binding to a receptor molecule on a target tissue and cell surface (Woodle et al., Small molecule ligands for targeting long circulating liposomes, in Long Circulating Liposomes: Old drags, new Therapeutics, Woodle and Storm eds., Springer, 1998, p 287-295). The polymer may include two or more targeting moieties, depending on the cell type that is to be targeted. Use of multiple targeting moieties can provide additional selectivity in cell targeting, and also can contribute to higher affinity and/or avidity of binding of the polymer to the target cell. When more than one targeting moiety is present on the polymer, the relative molar ratio of the targeting moieties may be varied to provide optimal targeting efficiency. Methods for optimizing cell binding and selectivity in this fashion are known in the art. The skilled artisan also will recognize that assays for measuring cell selectivity and affinity and efficiency of binding are known in the art and can be used to optimize the nature and quantity of the targeting ligand(s). Suitable ligands include, but are not limited to: vascular endothelial cell growth factor for targeting endothelial cells: FGF2 for targeting vascular lesions and tumors; somatostatin peptides for targeting tumors; transferrin for targeting umors; melanotropin (alpha MSH) peptides for tumor targeting; ApoE and peptides for LDL receptor targeting; von Willebrand's Factor and peptides for targeting exposed collagen; Adenoviral fiber protein and peptides for targeting Coxsackie-adenoviral receptor (CAR) expressing cells; PDl and peptides for targeting Neuropilin 1; EGF and peptides for targeting EGF receptor expressing cells; and RGD peptides for targeting integrin expressing cells, DNA and RNA aptamers.
Other examples include (i) folate, where the polymer is intended for treating tumor cells having cell-surface folate receptors, (ii) pyridoxyl, where the polymer is intended for treating virus-infected CD4+ lymphocytes, or (iii) sialyl- Lewis, where the polymer is intended for treating a region of inflammation. Other peptide ligands may be identified using methods such as phage display (F. Bartoli et al., Isolation of peptide ligands for tissue-specific cell surface receptors, in Vector Targeting Strategies for Therapeutic Gene Delivery (Abstracts form Cold Spring Harbor Laboratory 1999 meeting), 1999, p4) and microbial display (Georgiou et al., Ultra High Affinity Antibodies from Libraries Displayed on the Surface of Microorganisms and Screened by FACS, in Vector Targeting
Strategies for Therapeutic Gene Delivery (Abstracts form Cold Spring Harbor Laboratory 1999 meeting), 1999, p 3.).
In one embodiment, the targeting ligand may be somatostatin or a somatostatin analog. Somatostatin has the sequence AGCLNFFWKTFTSC, and contains a disulfide bridge between the cysteine residues. Many somatostatin analogs that bind to the somatostatin receptor are known in the art and are suitable for use in the present invention, such as those described, for example, in U.S. Patent No. 5,776,894, which is incorporated herein by reference in its entirety. Particular somatostatin analogs that are useful in the present invention are analogs having the general structure F*CY-(DW)KTCT, where DW is D- tryptophan and F* indicates, that the phenylalanine residue may have either the D- or L-absolute configuration. As in somatostatin itself, these compounds are cyclic due to a disulfide bond between the cysteine residues. Advantageously, these analogs may be derivatized at the free amino group of the phenylalanine residue, for example with a polycationic moiety such as a chain of lysine residues. The skilled artisan will recognize that other somatostatin analogs that are known in the art may advantageously be used in the invention.
Furthermore, methods have been developed to create novel peptide sequences that elicit strong and selective binding for target tissues and cells such as "DNA Shuffling" (W.P.C. Stremmer, Directed Evolution of Enzymes and Pathways by DNA Shuffling, in Vector Targeting Strategies for Therapeutic Gene Delivery (Abstracts form Cold Spring Harbor Laboratory 1999 meeting), 1999, p.5.) and these novel sequence peptides are suitable ligands for the invention. Other chemical forms for ligands are suitable for the invention such as natural carbohydrates which exist in numerous forms and are a commonly- used ligand by cells (Kraling et al., Am. J. Path. 150:1307 (1997) as well as novel chemical species, some of which may be analogues of natural ligands such as D- a ino acids and peptidomimetics and others which are identifed through medicinal chemistry techniques such as combinatorial chemistry (P.D. Kassner et al., Ligand Identification via Expression (LIVE): Direct selection of Targeting Ligands from Combinatorial Libraries, in Vector Targeting Strategies for
Therapeutic Gene Delivery (Abstracts form Cold Spring Harbor Laboratory 1999 meeting), 1999, ρ8.).
The targeting moiety provides tissue- and cell- specific binding. The ligands may be covalently attached to the polymer so that exposure is adequate for tissue and cell binding. For example, a peptide ligand can be covalently coupled to a polymer such as polyoxazoline.
The number of targeting molecules present on the outer layer will vary, depending on factors such as the avidity of the ligand-receptor interaction, the relative abundance of the receptor on the target tissue and cell surface, and the relative abundance of the target tissue and cell. Nevertheless, a targeting molecule coupled with of each polymer usually provides suitable enhancement of cell targeting.
The presence of the targeting moiety leads to the desired enhancement of binding to target tissue and cells. An appropriate assay for such binding may be ELISA plate assays, cell culture expression assays, or any other binding assays.
The fusogenic moiety promotes fusion of the polymer to the cell membrane of the target cell, facilitating entry of the polymer and therapeutic agents into the cell. In one embodiment, the fusogenic moiety comprises a fusion-promoting element. Such elements interact with cell membranes or endosome membranes in a manner that allows transmembrane movement of large molecules or particles, or disrupts the membranes such that the aqueous phases that are separated by the membranes may freely mix. Examples of suitable fusogenic moieties include, but are not limited to membrane surfactant peptides, e.g. viral fusion proteins such as hemagglutinin (HA) of influenza virus, or peptides derived from toxins such as PE and ricin. Other examples include sequences that permit cellular trafficking such as HIV TAT protein and antennapedia or those derived from numerous other species, or synthetic polymers that exhibit pH sensitive properties such as poly(ethylacrylic acid)(Lackey et al., Proc. Int. Symp. Control. Rel. Bioact. Mater. 1999, 26, #6245), N-isopropylacrylamide methacrylic acid copolymers (Meyer et al., FEBS Lett. 421:61 (1999)), or poly(amidoamine)s, (Richardson et al., Proc. Int. Symp. Control. Rel. Bioact. Mater. 1999, 26, #251), and lipidic agents that are released into the aqueous phase upon binding to the target cell or endosome. Suitable membrane surfactant peptides include an influenza hemagglutinin or a viral fusogenic peptide such as the Moloney murine leukemia virus ("MoMuLV" or MLV) envelope (env) protein or vesicular stroma virus (VSV) G-protein. The membrane-proximal cytoplasmic domain of the MoMuLV env protein may be used. This domain is conserved among a variety viruses and contains a membrane-induced α-helix. Suitable viral fusogenic peptides for the instant invention may include a fusion peptide from a viral envelope protein ectodomain, a membrane-destabilizing peptide of a viral envelope protein membrane-proximal domain, hydrophobic domain peptide segments of so called viral "fusion" proteins, and an amphiphilic-region containing peptide. Suitable amphiphilic-region containing peptides include, but are not limited to: melittin, the magainins, fusion segments from H. influenza hemagglutinin (HA) protein, HIV segment I from the cytoplasmic tail of HIVl gp41, and amphiphilic segments from viral env membrane proteins including those from avian leukosis virus (ALV), bovine leukemia virus (BLV), equine infectious anemia (EIA), feline immunodeficiency virus (FIV), hepatitis virus, herpes simplex virus (HSV) glycoprotein H, human respiratory syncytia virus (hRSV), Mason-Pfizer monkey virus (MPMV), Rous sarcoma vims (RSV), parainfluenza virus (PINF), spleen necrosis viras (SNV), and vesicular stomatitis virus (VSV). Other suitable peptides include microbial and reptilian cytotoxic peptides. The specific peptides or other molecules having greatest utility can be identified using four kinds of assays: 1) ability to disrupt and induce leakage of aqueous markers from liposomes composed of cell membrane lipids or fragments of cell membranes, 2) ability to induce fusion of liposomes composed of cell membrane lipids or fragments of cell membranes, 3) ability to induce cytoplasmic release of particles added to cells in tissue culture, and 4) ability to enhance plasmid expression by particles in vivo tissues when administered locally or systemically.
The fusogenic moiety also may be comprised of a polymer, including peptides and synthetic polymers. In one embodiment, the peptide polymer comprises synthetic peptides containing amphipathic aminoacid sequences such as the "GALA'and "KALA'peptides (Wyman TB, Nicol F, Zelphati O, Scoria PV, Plank C, Szoka FC Jr, Biochemistry 1997, 36:3008-3017; Subbarao NK, Parente RA, Szoka FC Jr, Nadasdi L, Pongracz K, Biochemistry 1987 26:2964- 2972 or Wyman supra, Subbarao supra ). Other peptides include non-natural aminoacids, including D aminoacids and chemical analogues such as peptoids, imidazole-containing polymers. Suitable polymers include molecules containing amino or imidazole moieties with intermittent carboxylic acid functionalities such as ones that form "salt-bridges," either internally or externally, including forms where the bridging is pH sensitive. Other polymers can be used including ones having disulfide bridges either internally or between polymers such that the disulfide bridges block fusogenicity and then bridges are cleaved within the tissue or intracellular compartment so that the fusogenic properties are expressed at those desired sites. For example, a polymer that forms weak electrostatic interactions with a positively charged fusogenic polymer that neutralizes the positive charge could be held in place with disulfide bridges between the two molecules and these disulfides cleaved within an endosome so that the two molecules dissociate, releasing the positive charge and fusogenic activity. Another form of this type of fusogenic agent has the two properties localized onto different segments of the same molecule and thus the bridge is intramolecular so that its dissociation results in a structural change in the molecule. Yet another form of this type of fusogenic agent has a pH sensitive bridge.
The fusogenic moiety also may comprise a membrane surfactant polymer- lipid conjugate. Suitable conjugates include Thesit™, Brij 58™, Brij 78TM, Tween 80™, Tween 20™, Cι2E8, Cι4E8, Cι6E8 (CnEn, = hydrocarbon poly(ethylene glycoι)ether where C represents hydrocarbon of carbon length N and E represents poly(ethylene glycol) of degree of polymerization N), Chol-PEG 900, analogues containing polyoxazoline or other hydrophilic polymers substituted for the PEG, and analogues having fluorocarbons substituted for the hydrocarbon. Advantageously, the polymer will be either biodegradable or of sufficiently small molecular weight that it can be excreted without metabolism. The skilled artisan will recognize that other fusogenic moieties also may be used without departing from the spirit of the invention.
A major barrier to efficient transcription and consequent expression of an exogenous nucleic acid moiety is the requirement that the nucleic acid enter the nucleus of the target cell. Advantageously, when the intended biological target of a nucleic acid is the nucleus, the nucleic targeting moiety of the invention is "nuclear targeted," that is, it contains one or more molecules that facilitate entry of the nucleic acid through the nuclear membrane into the nucleus of the host cell. Such nuclear targeting may be achieved by incorporating a nuclear membrane transport peptide, or nuclear localization signal ("NLS") peptide, or small molecule that provides the same NLS function, into the core complex. Suitable peptides are described in, for example, U.S. Patent Nos 5,795,587 and 5,670,347 and in patent application WO 9858955, which are hereby incorporated by reference in their entirety, and in Aronsohn et al., J. Drug Targeting 1:163 (1997); Zanta et al., Proc. Nat'l Acad. Sci. USA 96:91-96 (1999); Ciolina et al., Targeting of Plasmid DNA to Importin alpha by Chemical coupling with Nuclear Localization Signal Peptides, in Vector Targeting Strategies for Therapeutic Gene Delivery (Abstracts from Cold Spring Harbor Laboratory 1999 meeting), 1999, p 20; Saphire et al., J. Biol Chem; 273:29764 (1999). A nuclear targeting peptide may be a nuclear localization signal peptide or nuclear membrane transport peptide and it may be comprised of natural aminoacids or non-natural aminoacids including D aminoacids and chemical analogues such as peptoids. The NLS may be comprised of aminoacids or their analogues in a natural sequence or in reverse sequence. Another embodiment provides a steroid receptor-binding NLS moiety that activates nuclear transport of the receptor from the cytoplasm, wherein this transport carries the nucleic acid with the receptor into the nucleus.
In another embodiment, the NLS is coupled to the polymer in such a manner that the polymer is directed to the cell nucleus where it permits entry of a nucleic acid into the nucleus.
In another embodiment, incorporation of the NLS moiety into the polymer occurs through association with the nucleic acid, and this association is retained within the cytoplasm. This minimizes loss of the NLS function due to dissociation with the nucleic acid and ensures that a high level of the nucleic acid is delivered to the nucleus. Furthermore, the association with the nucleic acid does not inhibit the intended biological activity within the nucleus once the nucleic acid is delivered.
In yet another embodiment, the intended target of the biological activity of the nucleic acid is the cytoplasm or an organelle in the cytoplasm such as ribosomes, the golgi apparatus, or the endoplasmic reticulum. In this embodiment, a localization signal is included in the polymer or anchored to it so that it provides direction of the nucleic acid to the intended site where the nucleic acid exerts its activity. Signal peptides that can achieve such targeting are Icnown in the art.
In one embodiment, the ligand may be conjugated to a hydrophilic polymer. Examples of the hydrophilic polymer include, but are not limited to poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly-vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyl- oxazoline, polyhydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
If a nucleic acid molecule is the desired agent, a stable complex preferably is used for transporting the agent into the cell (including the nucleus). A "stable complex" is a complex that, once delivered in vivo, will not dissociate due to interactions with serum proteins or other types of molecules that may bind or associate with the complex. Once inside the desired subcellular compartment, however, the nucleic acid generally dissociates from the delivery system to become active. Low efficiency of synthetic gene vector systems is due, in part, to difficulties in achieving nucleic acid dissociation from the complex.
Accordingly, a highly stable complex is not optimal for nucleic acid delivery. Instead, the complex needs to umavel and free the nucleic acid within the subcellular compartment. The invention has overcome this problem by using biodegradable poly-cations, which effectively can bind nucleic molecules, yet can disintegrate and release the nucleic acid upon exposure to conditions in the target site, such as, e.g., acidic conditions in the endosome, or reducing conditions within the cytoplasm of cells.
In one embodiment, a polymer of the invention binds to a nucleic acid sequence that encodes a viral genomic sequence, otherwise known as a "viral genome." According to this embodiment the viral genome also contains within its open reading frame a therapeutic gene (i.e., nucleic acid) of interest. In this regard, the "entire" nucleic acid sequence (i.e. both the viral genomic sequence and therapeutic nucleic acid sequence) is complexed to a novel polymer of the invention. Alternatively, the entire nucleic acid sequence can be cloned into a plasmid to generate multiple copies, then the plasmid can be complexed to a novel polymer of the invention. In either embodiment, the nucleic acid encoding a viral genome can be self-replicative once administered to a subject, thereby providing an ongoing supply of a therapeutic nucleic acid molecule. Suitable viral genomes (and analogues thereof) are disclosed in U.S. Provisional Application Serial No. 60/330,909, entitled, "therapeutic methods for gene delivery systems," which hereby is incoiporated by reference in its entirety.
In another embodiment, a novel polymer of the invention is a component of a "synthetic vector," which can be used to affectively deliver an agent to a targeted cell or tissue in vivo. Synthetic vectors for use in the present invention have been disclosed by Woodle et al. (WO 01/49324, filed December 28, 2000), which application is hereby incorporated by reference in its entirety, including the drawings.
As used herein, a "synthetic vector" means a multi-functional synthetic vector which, at a minimum, contains a nucleic acid binding domain and a ligand binding (e.g. tissue targeting) domain, and is complexed with a nucleic acid sequence. A synthetic vector also may contain other domains such as, for example, a hydrophilic polymer domain, endosome disruption or dissociation domain, nuclear targeting domain, and nucleic acid condensing domain. A synthetic vector also can include a protective polymer. The protective polymer reduces non-specific interactions and, in some embodiments, contain an "exposed" component (e.g., a targeting ligand) that provides added specificity.
As part of a synthetic vector, a polymer of the invention is comprised of monomers or oligomers linked with biodegradable or biocompatible linkages. The nucleic acid binding domain, or "complex forming reagent," is capable of associating with a core nucleic acid complex in a manner that allows assembly of the nucleic acid core complex. The complex forming reagent can be, e.g., a lipid, a synthetic polymer, a natural polymer, a semi-synthetic polymer, a mixture of lipids, a mixture of polymers, a lipid and polymer combination, or a spermine analogue complex. A suitable polymer may contain histidine or an imidazole functional group. WO 01/49324 at, e.g., pages 20-34 disclose suitable DNA binding domains for use in the present invention.
A synthetic vector for use in the present invention preferably provides reduced non-specific interactions, yet effectively can engage in ligand-mediated (i.e., specific) cellular binding. In addition, a synthetic vector for use in the present invention is able to be complexed to one or more therapeutic nucleic acids, which then can be administered to a subject.
In one embodiment, the therapeutic nucleic acid is comprised within a nucleic acid sequence encoding a viral genomic sequence; the entire nucleic acid sequence (i.e. both the viral genomic sequence and therapeutic nucleic acid sequence) is complexed to the DNA binding domain of the synthetic vector. Alternatively, the entire nucleic acid sequence can be cloned into a plasmid to generate multiple copies, then the plasmid can be complexed to the DNA binding domain of the synthetic vector. In either embodiment, the nucleic acid encoding a viral genome can be self-replicative once administered to a subject, thereby providing an ongoing supply of a therapeutic nucleic acid molecule.
A synthetic vector for use in the invention is able to target specific tissues. In the absence of a steric coat, the cationic surface charge of a synthetic vector can act to target a cell. The ability to bind a target cell can be lost when a steric polymer coat is added to the synthetic vector as a hydrophilic polymer domain, such as a hydrophilic polymer domain disclosed herein. Targeting activity of the synthetic vector can be restored by employing a ligand domain, which can effecmate ligand-mediated binding and cellular uptake of the synthetic vector. In one aspect, a ligand may be conjugated to the distal end of the steric polymer in order to mediate binding with one or more cell surface receptors, acidic conditions in the endosome, or reducing conditions within the cytoplasm of cells. Suitable ligands and tissue-targeting molecules are disclosed herein or in references incorporated herein. The synthetic vectors for use in the invention also account for drawbacks associated with other gene delivery vehicles, such as non-specific interactions, which often result from an electrostatic charge differential between a vector and its environment. A synthetic vector, e.g., a condensed cationic reagent-DNA complex, can be made net positive, neutral, or negative depending on the ratio of ' the components in the complex. While electrostatic interactions between a negatively charged cell membrane and a positively charged particle can increase cellular uptake, all cells possess a negative membrane charge. Accordingly, nonspecific interactions can persist in a complex containing a net positive charge.
A hydrophilic polymer domain of a synthetic vector preferably is able to minimize undesirable non-specific interactions by controlling the surface properties of the synthetic vector. The hydrophilic polymer may be selected from the group consisting of poly(ethyleneglycol) ("PEG"), polyoxazoline, polyacetal, hydrolyzed dextran polyacetal polymers, polylactic acid, polyglycolic acid, poly- vinylpyrrolidone, polyvinyl alcohol, polymethyloxazoline, polyethyloxazoline, polyhydroxyethyloxazoline, poly-hydroxypropyloxazoline, and other conventionally known hydrophilic polymers.
A hydrophilic, steric coat can be introduced onto the surface of a synthetic vector by covalently conjugating the polymer to the condensing agent before complexing with therapeutic nucleic acid. This method is preferred over conjugating a hydrophilic, steric polymer to a pre-formed DNA-synthetic vector complex, since chemical reactions carried out after DNA complexing can damage the DNA. Moreover, as the steric barrier is formed, subsequent conjugation reactions are inhibited, which can limit the amount of polymer that can be conjugated to the complex surface.
In one embodiment, any of the foregoing hydrophilic polymers is conjugated to a spermine or spermadine analog, such as those described in Woodle et al, (WO 01/49324, filed December 28, 2000), which, in torn, interacts with a therapeutic nucleic acid molecule. The chosen hydrophilic polymer also may have complexed thereto a ligand or other tissue targeting moiety. According to one method, a hydrophilic polymer is conjugated at random to one or more sites on the nucleic acid binding domain, using either a stable covalent linkage or a linkage that can be cleaved. The grafting density can be varied between 2% and 25% of monomer units. Samples having a single molecular weight of the steric polymer can be used. An alternative steric polymer is polyacetal, derived by oxidation and subsequent reduction of dextran. The polymer is linear, possessing one or two alcohol moieties in place of each hexose ring. Polyacetal has been shown to function as a steric polymer for drug delivery and when conjugated to lipids and polycations.
A steric polymer layer that can block non-specific binding will increase the serum half-life of a synthetic vector, since (i) minimal non-specific interactions render the particles relatively inert, and (ii) the relatively large size allows the synthetic vector to remain in the blood for prolonged periods. Successful construction and use of a steric polymer layer can be observed from blood pharmacokinetics of the complexes following an intravenous administration (e.g., PEG, PEI and DOTAP: Cholesterol complexes).
Therapeutic Methods
The present invention provides methods of administering one or more therapeutic drugs or nucleic acid molecules to a subject, using a vehicle comprised of a polymer of the invention, to bring about a therapeutic benefit to the subject. As used herein, a "therapeutic nucleic acid molecule" or "therapeutic nucleic acid" is any nucleic acid (e.g., DNA or RNA) that, as a nucleic acid or as an expressed nucleic acid or polypeptide, confers a therapeutic benefit to a subject. In the present invention, a therapeutic nucleic acid molecule is administered to a subject as part of, or via, a nucleic acid delivery vehicle. The subject preferably is mammalian such as a mouse, and more preferably is a human being.
Gene delivery vehicles for use in the present invention can be used to stimulate an immune response, which may be protective or therapeutic. Accordingly, the gene delivery vehicles can be used to vaccinate a subject against an antigen.
In this sense, the invention provides methods vaccinating or enhancing a physiological response against a pathogen in a subject. This methodology can entail administering to the subject a first, or priming, dosage of a therapeutic nucleic acid molecule that encodes a therapeutic polypeptide, followed by administering to the subject one or more booster dosages of the nucleic acid molecule.
The administration regimen can vary, depending on, for example, (i) the subject to whom the therapeutic nucleic acid molecule is administered, and (ii) the pathogen that is involved. For instance, a booster dosage of a therapeutic nucleic acid molecule may administered about two weeks after priming, followed by successive booster dosages, which can occur between intervals of constant or increasing duration. It is desirable to administer therapeutic nucleic acid molecules at a periodicity that is appropriate according to the subject's immune response.
In the preceding administration steps, the administered nucleic acid molecule is comprised within a nucleic acid delivery vehicle of the invention. Preferably, expression of the therapeutic nucleic acid molecule in the foregoing steps elicits a humoral and/or cellular response in the subject, causing the subject to exhibit a degree of immunity against the pathogen that is greater than before the therapeutic method is carried out.
The antigen against which the subject exhibits an increased immunity can be the antigen encoded by the therapeutic nucleic acid molecule. Alternatively, the antigen against which the subject exhibits an increased immunity is distinct from, or in addition to, the antigen expressed by the administered nucleic acid molecule. In the latter approach, for instance, the antigen encoded by the therapeutic nucleic acid can act to enhance an immune response against another antigen, e.g., a component of a tumor. The route of administration may vary, depending on the therapeutic application (i.e. preventative or therapeutic vaccine) and the type of disorder to be treated. The nucleic acid delivery vehicle may be injected into the skin, muscle, intravenously, directly to the portal, hepatic vein or bile duct, locally to a tumor or to a joint, or orally.
The invention also contemplates enhanced delivery of a drug or nucleic acid therapeutic agent by employing oral administration. Enhanced delivery can result from protection of the agent by the polymer and binding to target tissues and cells in the gastrointestinal tract.
Nucleic acid refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form. The term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non- naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides. Examples of nucleic acid include, but are not limited to recombinant plasmids and cosmids, vectors that integrate into the host chromosome, episomal vectors, replication-deficient plasmids, mini-plasmids, a recombinant viral genome, a linear nucleic acid fragment, an antisense agent, a linear polynucleotide, a circular polynucleotide, a ribozyme, a cellular promoter, and viral genomes. The nucleic acid can encode one or more proteins, polypeptides or peptides, or multiple copies of peptides, polypeptides and proteins. The nucleic acid can also be an antisense nucleic acid, and may also be an interfering double stranded RNA (RNAi) or an siRNA, or a polynucleotide encoding a double stranded RNA. Examples of suitable double stranded RNA molecules and vectors encoding such molecules are described in, for example, U.S. Patent No. 6,506,559, which is hereby incorporated by reference in its entirety. Examples of nucleic acid analogs include, without limitation, phosphorothioates, phosphorarnidates, methyl phosphonates, chiral methyl phosphonates, 2-0- methyl ribonucleotides, and peptide-nucleic acids (PNAs). In one aspect, a therapeutic nucleic acid molecule(s), which is administered in a gene delivery vehicle, expresses an antigenic polypeptide. Expression of an administered therapeutic nucleic acid may be obtained by repeated administration and with strong expression in antigen presenting cells ("APCs"), e.g., through booster dosages. Nucleic acid expression also may occur by virtue of the self-replicative feature of the gene delivery vehicle of which the therapeutic nucleic acid molecule is comprised.
In yet another embodiment, the invention provides for selective gene expression through use of tissue selective replication of viral nucleic acid. The invention provides for viral vector replication whereby viral vector particles are produced by the target cells and tissues. The viral vector particles produced provide for tissue selective spread and amplification.
Suitable antigens for use in the invention include antigens for infectious agents and for tumor cells that result in the recruitment of antibodies.
An administered therapeutic nucleic acid molecule also may induce an immune response. A response can be achieved to intracellular infectious agents including, for example, tuberculosis, Lyme disease, and others. A response can be achieved by expression of antigen, expression of cytokines, and their combination. The invention also provides for expression of HIV antigens and induction of both a protective and a therapeutic immune response for preventing and treating HIV, respectively.
The invention additionally provides for the expression of antigens, which elicit a humoral and/or a cellular immune response. This heightened immune response can provide protection from a challenge with infectious agents characterized as having the antigen. Preferably, the invention utilizes an adenoviral genomic nucleic acid that (i) expresses an antigen under control of a promoter and (ii) targets an APC.
In one embodiment, the therapeutic nucleic acid encodes a cytokine, which may be expressed with or without an antigen. A cytokine acts to recruit an immune response, which can enhance an immune response to an expressed antigen. Accordingly, cytokine expression can be obtained whereby APCs and other immune response cells are recruited to the vicinity of tumor cells, in which case there is no requirement for co-expression of an antigen by the gene delivery vehicle. Yet, in another embodiment, one or more antigens and cytokines can be co-expressed.
Accordingly, the invention contemplates the use of immunostimulatory cytokines, as well as protein analogues exhibiting biological activity similar to an immunostimulatory cytokine, to vaccinate a subject. Suitable cytokines for use in enhancing an immune response include GM-CSF, IL-1, IL-12, IL-15, interferons, B-40, B-7, tumor necrosis factor (TNF)and others. The invention also contemplates utilizing genes that down-regulate immunosupressant cytokines.
The invention also provides for expression of "recruitment cytokines" at tumors. Expression of cytokines at tumors giving recruitment of immune response cells can initiate a cellular immune response at the tumor site giving recognition and killing of tumor cells at the site of expression and at distal tumor sites. A preferred embodiment of the invention is comprised of an adenoviral genomic nucleic acid, the nucleic acid exhibiting expression of GM-CSF under a tumor-preferential promoter, further comprised of nucleic acid exhibiting rumor- conditional replication to form adenoviral vector particles exhibiting tumor- conditional replication, and yet further comprised by synthetic vector compositions targeting delivery to tumor lesions. Another preferred embodiment of the invention utilizes an adenoviral genomic nucleic acid encoding a cytokine (e.g., GM-CSF) under regulation of a tumor-conditional promoter. This feature would result in enhanced cytokine expression at the site of a tumor. In this embodiment, the adenoviral genomic nucleic acid preferably is administered in conjunction with electroporation to tumor lesions.
A gene delivery vehicle also may be used to treat a disorder characterized by inflammation. In one approach, one or more therapeutic nucleic acid molecules comprised within a nucleic acid delivery vehicle is administered to a subject suffering from a disorder characterized by inflammation, in order to suppress or retard an immune response. Treatable disorders include rheumatoid arthritis, psoriasis, gout and inflammatory bowel disorders.
Suitable therapeutic nucleic acids for use in treating inflammation include nucleic acids that encode an inflammation inhibitory cytokine for use in the present invention include, but are not limited to, IL-IRA, soluble TNF receptor, soluble Fas ligand, and others or nucleic acid such as antisense RNA or DNA and interfering RNA that can inhibit expression of inflammatory cytokines.
The route and site of administration will vary, depending on the disorder and the location of inflammation. The gene delivery vehicle can be administered into a joint; administration thereto can be in conjunction with electroporation.
The nucleic acid delivery vehicles of the invention also can be used to treat a wide variety of diseases and syndromes in a subject, including, but not limited to cancer, cardiovascular diseases, and viral and bacterial infections. Suitable nucleic acids for use in these methods are known in the art, and the present invention provides improved compositions and methods for delivering such nucleic acids to a subject.
For therapeutic applications (cancer): Nucleic acid delivery vehicle containing viral genome, plasmid, antisense RNA, antisense DNA, or interfering RNA may be administered by various routes of administration into a tumor.
Pharmaceutical Compositions
The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. The dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors, including the type, age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, whether a drag delivery system is utilized and whether the compound is administered as part of a drug combination. Thus, the dosage regimen actually employed may vary widely and therefore may deviate from the preferred dosage regimen set forth above.
The compounds of the present invention may be administered orally, parenterally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrastemal injection, or infusion techniques.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Suppositories for rectal administration of the drug can be prepared by mixing the drag with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drag.
Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally by prepared with enteric coatings. Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more therapeutic agents, such as immunomodulators, antiviral agents or anti- infective agents.
The foregoing is merely illustrative of the invention and is not intended to limit the invention to the disclosed compounds. Variations and changes which are obvious to one skilled in the art are intended to be within the scope and natore of the invention which are defined in the appended claims. From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.
All documents referred to herein are specifically incorporated herein by reference in their entireties, including U.S. Provisional Application Nos. 60/352,882, filed February 1, 2002 and 60/358,345, field February 22, 2003, which are incorporated herein by reference in their entirety.

Claims

Claims:
1. A compound represented by the following structure:
Figure imgf000031_0001
wherein, R is H or CH3; and
n is 1-500.
A compound represented by the following structure:
Figure imgf000031_0002
wherein,
Ri is H or CH3;
R2 is H or CH3; R3 is NH2, OH, SH, CN, or N3;
R4 and R5 are each independently H, a hydrocarbon, polyacetal, a ligand conjugated hydrophilic polymer, ligand, or a hydrophilic polymer;
X is a hydrocarbon chain of length 1-10 carbons;
n is 1-50; and
m is 1-100
3. A compound represented by the following structure:
Figure imgf000032_0001
wherein,
n is 1-20;
m is 1-100; and
X is a hydrocarbon of length 1-10 carbons.
4. A compound represented by the following structure:
Figure imgf000033_0001
wherein,
n is 1-100;
m is 1-100; and
X is a hydrocarbon of length 1-10 carbons.
5. A composition comprising at least one nucleic acid molecule and a compound according to any preceding claim.
6. A method of enhancing a physiological response against an antigen in a subject, comprising:
(a) administering to the subject a first dosage of a therapeutic nucleic acid molecule; and
(b) administering to the subject one or more booster dosages of the nucleic acid molecule, wherein, in steps (a) and (b), the administered therapeutic nucleic acid molecule is bound to a compound of any one of claims 1-4,
and wherein the expression of the administered nucleic acid molecule in steps (a) and (b) elicits a humoral and/or cellular response in the subject against the antigen, and wherein after step (b), the subject exhibits a degree of immunity against the antigen that is greater in the subject than before step (a) is carried out.
7. A method according to claim 6, wherein the binding between the nucleic acid molecule and the compound is by electrostatic interaction.
8. A method of reducing inflammation in a subject suffering from a disorder characterized by inflammation, comprising: administering to the subject at, or proximal to, the site of the inflammation a therapeutically effective amount of a nucleic acid molecule that encodes a therapeutic polypeptide, wherein the administered nucleic acid is bound to a compound of any one of claims 1-4, and wherein the expression of the administered nucleic acid alleviates the inflammatory condition.
9. The method of claim 8, wherein the nucleic acid molecule is administered in conjunction with electroporation.
10. The method of claim 8, wherein the disorder is selected from the group consisting of arthritis, gout, and localized bowel inflammatory disorder.
11. A method of treating a disease or syndrome in a subject, comprising administering to said subject an effective amount of a complex according to claim 5.
12. The method according to claim 11, wherein said disease or syndrome is cancer, a tumor, cardiovascular disease, autoimmune disease, bacterial infection, or viral infection.
PCT/US2003/002706 2002-02-01 2003-01-31 Cationic polymers for use in therapeutic agent delivery WO2003066054A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003205380A AU2003205380A1 (en) 2002-02-01 2003-01-31 Cationic polymers for use in therapeutic agent delivery

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US35288202P 2002-02-01 2002-02-01
US60/352,882 2002-02-01
US35834502P 2002-02-22 2002-02-22
US60/358,345 2002-02-22

Publications (1)

Publication Number Publication Date
WO2003066054A1 true WO2003066054A1 (en) 2003-08-14

Family

ID=27737429

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/002706 WO2003066054A1 (en) 2002-02-01 2003-01-31 Cationic polymers for use in therapeutic agent delivery

Country Status (2)

Country Link
AU (1) AU2003205380A1 (en)
WO (1) WO2003066054A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
WO2014071406A1 (en) 2012-11-05 2014-05-08 Pronai Therapeutics, Inc. Methods of using biomarkers for the treatment of cancer by modulation of bcl2|expression
EP3000480A1 (en) 2005-12-01 2016-03-30 ProNAi Therapeutics, Inc. Cancer therapies and pharmaceutical compositions used therein

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5670347A (en) * 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
WO2000032764A1 (en) * 1998-12-02 2000-06-08 I.D.M. Immuno-Designed Molecules New oligomeric conjugates liable to transfer biological molecules into cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5670347A (en) * 1994-05-11 1997-09-23 Amba Biosciences Llc Peptide-mediated gene transfer
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
WO2000032764A1 (en) * 1998-12-02 2000-06-08 I.D.M. Immuno-Designed Molecules New oligomeric conjugates liable to transfer biological molecules into cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ZHOU ET AL.: "Preparation of poly(L-serine ester): a structural analogue of conventional poly(L-serine)", MACROMOLECULES, vol. 23, 1990, pages 3399 - 3406, XP001042094 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3000480A1 (en) 2005-12-01 2016-03-30 ProNAi Therapeutics, Inc. Cancer therapies and pharmaceutical compositions used therein
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
WO2014071406A1 (en) 2012-11-05 2014-05-08 Pronai Therapeutics, Inc. Methods of using biomarkers for the treatment of cancer by modulation of bcl2|expression

Also Published As

Publication number Publication date
AU2003205380A1 (en) 2003-09-02

Similar Documents

Publication Publication Date Title
US20060051315A1 (en) Polymers for delivering peptides and small molecules in vivo
KR100825519B1 (en) A chitosan based polymer conjugate and a method for producing the same
US8592385B2 (en) Polymer micelle complex including nucleic acid
Davis The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic
KR101617790B1 (en) Engineered Tunable Nanoparticles for Delivery of Therapeutics, Diagnostics, and Experimental Compounds and Related Compositions for Therapeutic Use
Wagner Polymers for siRNA delivery: inspired by viruses to be targeted, dynamic, and precise
US6217912B1 (en) Polyester analogue of poly-L-lysine as a soluble, biodegradable gene delivery carrier
US6440743B1 (en) Methods of using polynucleotide compositions
EP2155177A2 (en) Modification of biological targeting groups for the treatment of cancer
EP2197457A2 (en) Therapeutic and vaccine polyelectrolyte nanoparticle compositions
WO2003040399A2 (en) Therapeutic methods for nucleic acid delivery vehicles
JP2002506833A (en) Carrier for in vivo delivery of therapeutic agents
ZA200100286B (en) Actuator with approach pre-stroke and working stroke for operating a tool.
KR101445265B1 (en) Hyaluronic acid-nucleic acid conjugate and composition for nucleic acid delivery containing the same
JP2022502451A (en) Lipid Nanoparticle Formulation Containing Lipidized Cationic Peptide Compounds for Nucleic Acid Delivery
KR20160007529A (en) Polymeric micelle pharmaceutical composition
JP2003530360A (en) Peptide conjugates for drug delivery
US20060014695A1 (en) Hpma-polyamine conjugates and uses therefore
JP6799823B2 (en) Polyion complex of block copolymer containing poly (L-arginine) segment and polyanionic polymer
WO2003066054A1 (en) Cationic polymers for use in therapeutic agent delivery
Tomich et al. Nonviral gene therapy: Peptiplexes
CN117460542A (en) Nanoparticle comprising peptide-lipid conjugate for delivering oligonucleotide into target cell and pharmaceutical composition comprising same
Kuo et al. Intracellular trafficking, metabolism and toxicity of current gene carriers
US20200392289A1 (en) Biodegradable Polymer and Use Thereof
WO2018216793A1 (en) Blood-stable rna-encapsulating micelles

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP