WO2009143412A2 - Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides - Google Patents

Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides Download PDF

Info

Publication number
WO2009143412A2
WO2009143412A2 PCT/US2009/044953 US2009044953W WO2009143412A2 WO 2009143412 A2 WO2009143412 A2 WO 2009143412A2 US 2009044953 W US2009044953 W US 2009044953W WO 2009143412 A2 WO2009143412 A2 WO 2009143412A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
oligonucleotide
occurrence
oligonucleotides
independently
Prior art date
Application number
PCT/US2009/044953
Other languages
French (fr)
Other versions
WO2009143412A3 (en
Inventor
Hong Zhao
Prasanna Reddy
Original Assignee
Enzon Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Enzon Pharmaceuticals, Inc. filed Critical Enzon Pharmaceuticals, Inc.
Priority to US12/994,266 priority Critical patent/US20110105413A1/en
Priority to EP09751640.5A priority patent/EP2288261A4/en
Priority to CA2723263A priority patent/CA2723263A1/en
Priority to JP2011510720A priority patent/JP2011520983A/en
Publication of WO2009143412A2 publication Critical patent/WO2009143412A2/en
Publication of WO2009143412A3 publication Critical patent/WO2009143412A3/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/10Vectors comprising a non-peptidic targeting moiety

Definitions

  • Gene -based therapy is a powerful tool in the treatment of disease because a therapeutic gene can selectively modulate gene expression associated with disease and minimize side effects which may incur when other therapeutic approaches are used.
  • LNA locked Nucleic Acid
  • Each LNA monomer contains a methylene bridge between the 2'-oxygen and 4'-carbon of the ribose sugar. This fixes the LNA residue in a favorable RNA-like conformation and enables LNA oligonucleotides to have higher affinity, specificity, and resistance against degradation compared with other art-known oligonucleotides. It has been shown that LNA oligonucleotide inhibits target gene expression in vitro (at sub- nanomolar level).
  • LNA oligonucletides have improved therapeutic activity compared to other art-known nucleic acids, it is still needed to further improve the pharmacokinetic profile and fast clearance from circulation and limited activity in vivo of LNA oligonucleotides. There continues to be a need to provide improved systems and methods for the delivery of LNA oligonucleotides as well as other art-known nucleic acid molecules. The present invention addresses this need.
  • the present invention provides new polymeric delivery systems containing an intracellularly releasable linker.
  • methods of delivering oligonucleotides to tumor cells in a mammal include administering to the mammal having tumor cells a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein
  • R] is a substantially non-antigenic water-soluble polymer; each Z
  • Yi in each occurrence, is independently S or O: Y 2 , in each occurrence, is independently NRj 3 ; R,,, in each occurrence, is the same or a different oligonucleotide; each of Li -4, in each occurrence, is the same or a different bifunctional linker;
  • R t is a folic acid
  • R c in each occurrence, is the same or a different diagnostic agent; each Of R 3-7 is independently selected from among hydrogen, Ci ⁇ alkyls, C 2 . 6 alkenyl, C 2 . 6 alkynyl, C 3 . 19 branched alkyl, C ⁇ cycloalkyl, and C 1 . 6 alkoxy; R
  • R] 2 in each occurrence, is independently selected from among hydrogen, hydroxyl, Ci ⁇ alkyls, C 2 - 6 alkenyl, Ci ⁇ alkynyl, C 3-I ? branched alkyl, C 3 . 8 cycloalkyl, and Ci -6 alkoxy; each of (a) and (d) is independently 0, 1 , 2 or 3; each of (al ) and (a2) is independently 0, 1 , 2 or 3; each (b) is independently 0, 1 , 2, or 3; each (c) is independently 0, 1, 2 or 3; each (e) is independently 0 or 1 ; each (g) is independently 0 or 1 ; and (in) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero and further provided that one or more of Z
  • the present invention provides a method of inhibiting a gene expression in a mammal having prostate or cervical cancer cells.
  • the compound of Formula (I) employed in the method described herein is:
  • PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; Rb is
  • Oligo is an oligonucleotide of from about 8 to 30 nucleotides.
  • methods of inhibiting a gene expression in a mammal for the treatment of various diseases i.e. prostate or cervical cancer.
  • the present invention also provides methods of making the compounds described herein.
  • the releasable PEG-linker technology provides a method for in vivo administration of therapeutic oligonucleotides including LNA. This selective delivery technology allows enhanced therapeutic efficacy of LNA and decrease in toxicity.
  • the releasable delivery systems described herein allow for modulating of the pharmacokinetic properties of oligonucleotides.
  • the release site of therapeutic oligonucleotides from the polymeric conjugates can be selectively targeted via EPR effect and a targeting group such as a folic acid.
  • the oligonucleotides such as LNA attached to the polymers described herein can be released at a targeted area, such as cancer cells, thus allowing the artisan to achieve a desired bioavailability of therapeutic oligonucleotides at a targeted area.
  • the site of release of the oligonucleotides can be modified, i.e., to release oligonucleotides in different compartments of the cells.
  • the polymeric delivery systems described herein allow sufficient amounts of the therapeutic oligonucleotides including LNA to be selectively available at the desired target area, e.g., cytoplasm, micropinosome and endosome.
  • the spatial modifications can be advantageous for treatment of disease.
  • the methods described herein provide an approach for the delivery and improved efficacy of oligonucleotides (e.g., LNA oligonucleotide, siRNA) in vivo.
  • a further advantage of the present invention is that the conjugates described herein allow cellular uptake and specific mRNA downregulation in cancer cells in the absence of transfection agents. This is a significant advantage over prior art technologies and thus significantly simplifies treatment regimens, i.e., the in vivo administration of oligonucleotide drugs. This technology can be applied to the in vivo administration of therapeutic oligonucleotides.
  • the polymeric compounds are stable under buffer conditions and the oligonucleotides or other therapeutic agents are not prematurely excreted from the body.
  • the term "residue” shall be understood to mean that portion of a compound to which it refers, i.e., PEG, oligonucleotide, etc. that remains after it has undergone a substitution reaction with another compound.
  • the term "polymeric residue” or “PEG residue” shall each be understood to mean that portion of the polymer or PEG which remains after it has undergone a reaction with other compounds, moieties, etc.
  • the term "alkyl " as used herein refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
  • the te ⁇ n “alkyl” also includes alkyl-thio-alkyl, alkoxyalkyl, cycloalkylalkyl, heterocycloalkyl, Ci -6 hydrocarbonyl, groups.
  • the alkyl group has 1 to 12 carbons.
  • the alkyl group can be substituted or unsubstituted.
  • the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, Ci ⁇ hydrocarbonyl, aryl, and amino groups.
  • substituted refers to adding or replacing one or more atoms contained within a functional group or compound with one of the moieties from the group of halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, Ci. 6 hydrocarbonyl, aryl, and amino groups.
  • alkenyl 1 refers to groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkenyl group has about 2 to 12 carbons. More preferably, it is a lower alkenyl of from about 2 to 7 carbons, yet more preferably about 2 to 4 carbons.
  • the alkenyl group can be substituted or unsubstituted.
  • the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxy], mercapto, hydroxy, cyano, alkylsilyl, cycloalkyi, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C
  • alkynyl refers to groups containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkynyl group has about 2 to 12 carbons. More preferably, it is a lower alkynyl of from about 2 to 7 carbons, yet more preferably about 2 to 4 carbons.
  • the alkynyl group can be substituted or unsubstituted.
  • the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxy!, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyi, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C
  • alkynyl include propargyl, propyne, and 3-hexyne.
  • aryl refers to an aromatic hydrocarbon ring system containing at least one aromatic ring.
  • the aromatic ring can optionally be fused or otherwise attached to other aromatic hydrocarbon rings or non-aromatic hydrocarbon rings.
  • aryl groups include, for example, phenyl, naphthyl, 1 ,2,3,4-tetrahydronaphthalene and biphenyl.
  • Preferred examples of aryl groups include phenyl and naphthyl.
  • cycloalkyi refers to a C 3 . 8 cyclic hydrocarbon.
  • examples of cycloalkyi include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • 'cycloalkenyF refers to a C 3 .8 cyclic hydrocarbon containing at least one carbon-carbon double bond.
  • Examples of cycloalkenyl include cyclopentenyl, cyclopentadienyl, cyclohexenyl, 1,3-cyclohexadienyl, cycloheptenyl, cycloheptatrienyl, and cyclooctenyl.
  • cycloalkylalkyl refers to an alkyl group substituted with a C 3 .8 cycloalky] group.
  • examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
  • alkoxy refers to an alkyl group of the indicated number of carbon atoms attached to the parent molecular moiety through an oxygen bridge.
  • alkoxy groups include, for example, methoxy, ethoxy, propoxy and isopropoxy.
  • An ''alkylaryl “ group as used herein refers to an aryl group substituted with an alkyl group.
  • An ; 'aralkyl “ group as used herein refers to an alkyl group substituted with an aryl group.
  • alkoxyalkyl refers to an alkyl group substituted with an alkoxy group.
  • alkyl-thio-alkyl refers to an alkyl-S-alkyl thioether, for example, methylthiomethyl or mcthylthioethyl.
  • '"amino refers to a nitrogen containing group as is known in the art derived from ammonia by the replacement of one or more hydrogen radicals by organic radicals.
  • acylamino v and alkylamino refer to specific N-substituted organic radicals with acyl and alky] substituent groups, respectively.
  • alkylcarbonyl refers to a carbonyl group substituted with alkyl group.
  • halogen or halo as used herein refer to fluorine, chlorine, bromine, and iodine.
  • heterocycloalkyl refers to a non-aromatic ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur.
  • the heterocycloalkyl ring can be optionally fused to or otherwise attached to other heterocycloalkyl rings and/or non-aromatic hydrocarbon rings.
  • Preferred heterocycloalkyl groups have from 3 to 7 members. Examples of heterocycloalkyl groups include, for example, piperazine, rnorpholine, piperidine, tetrahydrofuran, pyrrolidine, and pyrazole.
  • Preferred heterocycloalkyl groups include piperidinyl, piperazinyl, morpholinyl, and pyrolidinyl.
  • heteroaryl refers to an aromatic ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur.
  • the heteroaryl ring can be fused or otherwise attached to one or more heteroaryl rings, aromatic or non-aromatic hydrocarbon rings or heterocycloalkyl rings.
  • heteroaryl groups include, for example, pyridine, furan, thiophene, 5,6,7, 8-tetrahydroisoquinoline and pyrimidine.
  • heteroaryl groups include thienyl, benzothienyl, pyridyl, quinolyl, pyrazinyl, pyrimidyl, imidazolyl, benzimidazolyl, furanyl, benzofuranyl, thiazolyl, benzothiazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, benzisothiazolyl, triazolyl, tetrazolyl, pyrrolyl, indolyl, pyrazolyl, and benzopyrazolyl.
  • substituted alkyls include carboxyalkyls, aminoalkyls, dialkylaminos, hydroxyalkyls and mercaptoalkyls; substituted alkenyls include carboxyalkenyls, aminoalkenyls, dialkenylaminos, hydroxyalkenyls and mercaptoalkenyls; substituted alkynyls include carboxyalkynyls, aminoalkynyls, dialkynylaminos, hydroxyalkynyls and mercaptoalkynyls; substituted cycloalkyls include moieties such as 4-chlorocyclohexyl; aryls include moieties such as napthyl; substituted aryls include moieties such as 3-bromo phenyl; aralkyls include moieties such as tolyl; heteroalkyls include
  • positive integer shall be understood to include an integer equal to or greater than 1 and as will be understood by those of ordinary skill to be within the realm of reasonableness by the artisan of ordinary skill, i.e., preferably from 1 to about 10, more preferably 1 or 2 in some embodiments.
  • nucleic acid' or “nucleotide” apply to deoxyribonucleic acid ("DNA”) and ribonucleic acid, (“RNA'”), whether single-stranded or double-stranded, unless otherwise specified, and any chemical modifications thereof.
  • the term "'linked” shall be understood to include covalent (preferably) or noncovalent attachment of one group to another, i.e., as a result of a chemical reaction.
  • therapeutic oligonucleotide * ' refers to an oligonucleotide used as a pharmaceutical or diagnostic agent.
  • modulation of gene expression shall be understood as broadly including down-regulation or up-regulation of any types of genes, preferably associated with cancer and inflammation, compared to a gene expression observed in the absence of the treatment with the compounds described herein, regardless of the route of administration.
  • '"inhibition of gene expression of a target gene shall be understood to mean that mRNA expression or protein translated are reduced or attenuated when compared to that observed in the absence of the treatment with the compound described herein.
  • Suitable assays include, e.g., examination of protein or mRNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELlSA, immunoprecipitation, enzyme function, as well as phenotypic assays known to those of skill in the art.
  • the treated conditions can be confirmed by, for example, decrease in mRNA levels in cells, preferably cancer cells or tissues.
  • successful inhibition or treatment shall be deemed to occur when the desired response is obtained.
  • successful inhibition or treatment can be defined by obtaining e.g., 10% or higher (i.e., 20% 30%, 40%) downregulation of genes associated with tumor growth inhibition.
  • successful treatment can be defined by obtaining at least 20% or preferably 30%, more preferably 40 % or higher (i.e., 50% or 80%) decrease in oncogene mRNA levels in cancer cells or tissues, including other clinical markers contemplated by the artisan in the field, when compared to that observed in the absence of the treatment with the compound described herein.
  • compositions comprising an enzyme refers to one or more molecules of that enzyme. It is also to be understood that this invention is not limited to the particular configurations, process steps, and materials disclosed herein as such configurations, process steps, and materials may vary somewhat.
  • FIG. 1 schematically illustrates synthesis of compound 1 described in Example 7.
  • FIG. 2 schematically illustrates synthesis of compound 10 described in Examples 8-13.
  • FIG. 3 schematically illustrates synthesis of compound 18 described in Examples 14-18.
  • FIG. 4 schematically illustrates synthesis of compound 25 described in Examples 19-23.
  • FIG. 5 schematically illustrates synthesis of compound 30 described in Examples 25-27.
  • FIG. 6 schematically illustrates synthesis of compound 35 described in Examples 28-29.
  • FIG. 7 shows cellular uptake of PEG-LNA conjugates described in Examples 32.
  • FlG. 8 shows receptor-specific cellular uptake of PEG-LNA conjugates described in Example 32.
  • FIG. 9 shows in vitro efficacy of PEG-LNA conjugates described in Example 33.
  • FlG. 10 shows in vivo efficacy and biodistribution of Folate-PEG-LNA conjugates described in Example 34.
  • FIG. 1 1 shows biodistribution of PEG-LNA conjugates described in Example 35.
  • FIG. 12 shows in vivo efficacy of PEG-LNA conjugates described in Example 36.
  • FIG. 13 shows in vivo efficacy of PEG-LNA conjugates described in Example 37.
  • multi-arm PEG e.g., four-arm PEG
  • PEG multi-arm PEG
  • oligonucleotides to tumor cells in a mammal in need thereof.
  • the method includes administering to the mammal having tumor cells a compound of Formula (I): or a pharmaceutically acceptable salt thereof, wherein Ri is a substantially non-antigenic water-soluble polymer; each Z
  • Y ⁇ in each occurrence, is independently S or O, preferably O;
  • Y 2 in each occurrence, is independently NR 13 , preferably NH;
  • R a in each occurrence, is the same or a different oligonucleotide; each of L M . in each occurrence, is the same or a different bifunctional linker;
  • R h in each occurrence, is a folic acid
  • R c in each occurrence, is the same or a different diagnostic agent
  • each of R3.7 is independently selected from among hydrogen, CV 6 alkyls, C 2 . f i alkenyl, C ? . 6 alkynyl. CM? branched alkyl, C3.X cycloalkyl, and C
  • each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably 0; each of (a 1 ) and (a2) is independently zero, 1 , 2, or 3, and preferably 1 ; each (b) is independently zero, 1 , 2, or 3, and preferably 0; each (c) is independently zero, 1, 2, or 3, and preferably 1 ; each (e) is independently zero or one, and preferably 0; each (g) is independently zero or one, and preferably 1 ; and
  • (m) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero and provided that one or more of Z
  • compounds of Fo ⁇ nula (I) are provided: or a pharmaceutically acceptable salt thereof, wherein R
  • Yi in each occurrence, is independently S or O, preferably O;
  • Yi in each occurrence, is independently NR 13 , preferably NH;
  • R a in each occurrence, is the same or a different oligonucleotide; each Of Li -4 , in each occurrence, is the same or a different bifunctional linker;
  • R h in each occurrence, is a folic acid
  • R c in each occurrence, is the same or a different diagnostic agent; each of R3/7 is independently selected from among hydrogen, Ci- 6 alkyls, C 2-6 alkenyl, C 2 . 6 alkynyl, C 3 . 1 1> branched alkyl, C ⁇ .g cycloalkyl, and C
  • R 1 in each occurrence, is independently selected from among hydrogen, Ci -C alkyls, C 2 . & alkenyl, C 2 - 6 alkynyl, C 3 .19 branched alkyl, arid C 3 . 8 cycloalkyl;
  • R ⁇ 2 in each occurrence, is independently selected from among hydrogen, hydroxyl, C ⁇ 6 alkyls, C 2 -6 alkenyl, C 2 -6 alkynyl, C.vj ⁇ > branched alkyl, C 3 .8 cycloalkyl and C ⁇ - 6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably O; each of (al ) and (a2) is independently zero, 1, 2, or 3, and preferably 1 ; each (b) is independently zero, 1 , 2, or 3, and preferably O; each (c) is independently zero, 1 , 2, or 3, and preferably 1 ; each (e) is independently zero or one, arid preferably O; each (g) is independently zero or one, and preferably 1 ; and
  • (m) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero, that one or more of Zi contain an oligonucleotide, and that one or more of Zi contain a folic acid.
  • contains an oligonucleotide and the remaining Z
  • (m) refers to the number of polymer a ⁇ ns.
  • Each polymer arm includes a linear polymer such as polyethylene glycol.
  • (m) equals to from about 2 to about 32.
  • (m) is 32 when R
  • bisPEG is employed in the polymeric compounds described herein.
  • the polymeric compounds can preferably include up to 32 polymer arms, i.e., 4, 8, 16 or 32.
  • the polymeric compounds can include four to eight polymer arms, where (m) can be from 4 to 8 (e.g., 4, 6 or 8).
  • the polymeric portion includes four polymer arms, where (m) is 4.
  • Li is the same or different when (a) is equal to or greater than 2.
  • L 2 is the same or different when (d) is equal to or greater than 2.
  • L 4 is the same or different when (al ) or (a2) is equal to or greater than 2.
  • C(R 4 )(Rs) is the same or different when (b) is equal to or greater than 2.
  • C(Ro)(R 7 ) is the same or different when (c) is equal to or greater than 2.
  • the tumor cells are prostate or cervical cancer cells.
  • the present invention provides a method of inhibiting a gene expression in mammalian cells or tissues. The method includes administering an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof to a mammal in need thereof.
  • the methods described herein are carried out using a compound of Formula (I * ): wherein
  • (m 1 ) is a positive integer from about 1 to about 8 (e.g., 1 , 2, 3, 4, 5, 6, 7, 8);
  • (m2) is zero or a positive integer from about 1 to about 7 (e.g. 0, 1 , 2, 3, 4, 5, 6, 7); and the sum of (m l ) and (m2) is an integer from about 2 to about 8 (e.g., 2, 4, 6, 8).
  • ) contain an oligonucleotide.
  • (m2) is zero and (ml ) is 4 or 8.
  • all (Zi) are the same or different
  • contain a folic acid.
  • one or more Z ⁇ are In this aspect, one Z
  • the compounds described herein include an optional diagnostic agent.
  • each Ri 2 is the same or different groups selected from among H, NH 2 , OH, CO 2 H, C,. 6 alkoxy, C 6 alkyl, and preferably OH.
  • each of R3.7 is the same or different group selected from among hydrogen, methyl, ethyl and isopropyl.
  • R3.7 are all hydrogen
  • (b), (d) and (e) are zero and (c) is 1.
  • the compounds of Formula (I) employed in the method described herein include Zi having the fo ⁇ nula:
  • (a) is 0 or 1 ;
  • (in) is 1 , 2, 4, 8, 16 or 32;
  • Ri 2 in each occurrence, is independently selected from among hydroxyl, Ci. 6 alkyls, Ci-r, alkenyl, CY ⁇ alkynyl, C 3 . 1 i) branched alkyl, and C
  • the compounds described herein have the formula:
  • includes a folic acid.
  • one Zi includes a diagnostic agent.
  • Ri includes a polyalkylene oxide.
  • Ri has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20, 000 to about 45,000 daltons.
  • the methods described herein are conducted with the compounds having the formula:
  • Ri 2 in each occurrence, is independently selected from among hydroxyl, C ⁇ _ 6 alkyls, C 2 -b alkenyl, C 2 . ⁇ alkynyl, C3-m branched alkyl, and Ci -6 alkoxy;
  • (n) is a positive integer and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; and all other variables are the same as defined above.
  • all Z groups include an oligonucleotide.
  • one Z includes an oligonucleotide and remaining one or more Z groups (e.g., 1 , 2, 3, 4, 5, 6 or 7 Z groups) include a targeting agent such as folic acid.
  • one Z includes an oligonucleotide, another Z includes a diagnostic agent, and remaining one or more (e.g., 2, 3, 4, 5, 6) Z include a folic acid.
  • R i is a substantially non-antigenic water-soluble polymer; each Z
  • Y i in each occurrence, is independently S or O; Yi, in each occurrence, is independently NRi 3 ; Rj, in each occurrence, is the same or a different oligonucleotide; each of L M , in each occurrence, is the same or a different bi functional linker; Rb. in each occurrence, is a folic acid; R c , in each occurrence, is the same or a different diagnostic agent; each of R3.7 is independently selected from among hydrogen, C
  • R 1 J in each occurrence, is independently selected from among hydrogen, Cj. ⁇ alkyls, C1. 6 alkenyl, Ci-is alkynyl, C 3 . 1 9 branched alkyl, and CYs cycloalkyl;
  • R1 2 in each occurrence, is independently selected from among hydrogen, hydroxyl, Cu, alkyls, C2-6 alkenyl, Cj.*, alkynyl, Cv 1 0 branched alkyl, C 3 .X cycloalkyl and C ⁇ - 6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3; each of (al ) and (a2) is independently zero, 1 , 2, or 3; each (b) is independently zero, 1 , 2, or 3; each (c) is independently zero, 1 , 2, or 3; each (e) is independently zero or one; each (g) is independently zero or one; and
  • (111) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero, that one or more of Z
  • variables are the same as defined in Fomula (1).
  • Polymers employed in the compounds described herein are preferably water soluble polymers and substantially non-antigenic such as polyalkylene oxides (PAO's).
  • the compounds described herein include a linear, terminally branched or multi-armed polyalkylene oxide.
  • the polyalkylene oxide includes polyethylene glycol and polypropylene glycol.
  • the polyalkylene oxide has the total number average molecular weight of from about 2,000 to about 100,000 daltons, preferably from about 5,000 to about 60,000 daltons.
  • the polyalkylene oxide can be more preferably from about 5,000 to about 25,000 or from about 20,000 to about 45,000 daltons.
  • the compounds described herein include the polyalkylene oxide having the total number average molecular weight of from about 30,000 to about 45,000 daltons.
  • the polymeric portion has the total number average molecular weight of about 40,000 daltons.
  • the polyalkylcne oxide includes polyethylene glycols and polypropylene glycols. More preferably, the polyalkylene oxide includes polyethylene glycol (PEG).
  • PEG is generally represented by the structure: where (n) represents the degree of polymerization for the polymer, and is dependent on the molecular weight of the polymer.
  • PEG residue portion of the compounds described herein can be selected from among:
  • Y 71 and Y7 3 are independently O, S, SO, SO 2 , NR7 3 or a bond;
  • Y 72 is O, S, or NR 74 , preferably O;
  • R7 1 , R 72 , R7 3 and R7 4 are independently selected from the same moieties which can be used for R 3 ;
  • (a71 ), (a72), and (b71) are independently zero or a positive integer (e.g. O. 1 , 2, 3), and preferably 1 ; and (n) is an integer from about 10 to about 2300.
  • the polymers useful in the compounds described herein include multi-arm PEG-OH or "star-PEG " ' products such as those described in NOF Corp. Drug Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference.
  • the polymers can be converted into suitably activated forms, using the activation techniques described in U.S. Patent Nos. 5,122,614 or 5,808,096.
  • a PEG can be of the formula:
  • (u') is an integer from about 4 to about 455.
  • the degree of polymerization for the polymer (u') is from about 28 to about 341 to provide polymers having the total number average molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from about 1 14 to about 239 to provide polymers having the total number average molecular weight of from about 20,000 Da to about 42,000 Da.
  • (u') represents the number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer. In one particular embodiment of the invention, (u") is about 227 to provide the polymeric portion having the total number average molecular weight of about 40,000 Da.
  • all 4 of the PEG arms are converted to suitable activating groups, for facilitating attachment to oligonucleotides or folic acids.
  • suitable activating groups for facilitating attachment to oligonucleotides or folic acids.
  • the polymeric substances included herein are preferably water-soluble at room temperature.
  • a non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
  • substantially or effectively non-antigenic means all materials understood in the art as being nontoxic and not eliciting an appreciable immunogenic response in mammals.
  • polymers having terminal carboxylic acid groups can be employed in the polymeric delivery systems described herein. Methods of preparing polymers having terminal carboxylic acids in high purity are described in U.S. Patent Application Publication No. 2007/0173615, the contents of which are incorporated herein by reference. The methods include first preparing a tertiary alkyl ester of a polyalkylene oxide followed by conversion to the carboxylic acid derivative thereof.
  • the first step of the preparation of the PAO carboxylic acids of the process includes forming an intermediate such as /-butyl ester of polyalkylene oxide carboxylic acid.
  • This intermediate is formed by reacting a PAO with a /-butyl haloacetate in the presence of a base such as potassium /-butoxide.
  • a base such as potassium /-butoxide.
  • polymers having terminal amine groups can be employed to make the compounds described herein.
  • the methods of preparing polymers containing terminal amines in high purity are described in U.S. Patent Application Publication Nos. 2008/0249260 and 2007/0078219, the contents of each of which are incorporated by reference.
  • polymers having azides react with a phosphine-based reducing agent such as triphenylphosphine or an alkali metal borohydride reducing agent such as NaBH 4 .
  • polymers including leaving groups react with protected amine salts such as potassium salt of methyl-tert-butyl imidodicarbonate (KNMeBoc) or the potassium salt of di-tert-butyl iinidodicarbonate (KNBoC 2 ) followed by deprotecting the protected amine group.
  • protected amine salts such as potassium salt of methyl-tert-butyl imidodicarbonate (KNMeBoc) or the potassium salt of di-tert-butyl iinidodicarbonate (KNBoC 2 ) followed by deprotecting the protected amine group.
  • KNMeBoc methyl-tert-butyl imidodicarbonate
  • KNBoC 2 di-tert-butyl iinidodicarbonate
  • Bifunctional linkers include amino acids, amino acid derivatives, and peptides.
  • the amino acids can be among naturally occurring and non-naturally occurring amino acids.
  • Derivatives and analogs of the naturally occurring amino acids, as well as various art-known non-naturally occurring amino acids (D or L), hydrophobic or non-hydrophobic, are also contemplated to be within the scope of the invention.
  • a suitable non-limiting list of the non-natural occurring amino acids includes 2-aininoadipic acid, 3-aminoadipic acid, beta-alanine, beta-aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, piperidinic acid,
  • 6-aminocaproic acid 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2- aminopimelic acid, 2,4-aminobutyric acid, desmosine, 2,2-diaminopinielic acid, 2,3-diaminopropi ⁇ nic acid, N-elhylglycine, N-ethylasparagine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, alloisoleucine, N-inethylglycine, sarcosine, N-methyl- isolcucine, 6-N-methyllysine, N-methylvaline, norvaline, norleucine, and ornithine.
  • Some amino acid residues are selected from glycine, alanine, methionine or sarcosine, and more preferably, glycine.
  • L M are the same or different groups selected from among:
  • R 2 I -2 (J are the same or different groups selected from among hydrogen, C
  • (t) and (O are independently zero or a positive integer, preferably zero or an integer from about 1 to about 12, more preferably an integer from about 1 to about 8, and most preferably 1 or 2; and (v) and (v " ) are independently zero or 1.
  • L ( . 4 of Fomula (I) and Formula (Ia) include the same or different groups selected from among:
  • bifunctional linkers when values for bifunctional linkers are positive integers equal to or greater than 2, the same or different bifunctional linkers can be employed. In one embodiment containing two or more bifunctional linkers, where (a), (al ), and (a2) are equal to or greater than 2, the bifunctional linkers can be the same or different.
  • a further aspect of the invention provides the compounds optionally prepared with a diagnostic tag linked to the polymeric delivery system described herein, wherein the tag is selected for diagnostic or imaging purposes.
  • the compounds described herein can be labeled such as biotinylated compounds, fluorescent compounds (e.g., FAM), radiolab led compounds.
  • a suitable tag is prepared by linking any suitable moiety, e.g., an amino acid residue, to any art-standard emitting isotope, radio-opaque label, magnetic resonance label, or other non-radioactive isotopic labels suitable for magnetic resonance imaging, fluorescence-type labels, labels exhibiting visible colors and/or capable of fluorescing under ultraviolet, infrared or electrochemical stimulation, to allow for imaging tumor tissue during surgical procedures, and so forth.
  • the diagnostic tag is incorporated into and/or linked to a conjugated therapeutic moiety, allowing for monitoring of the distribution of a therapeutic biologically active material within an animal or human patient.
  • the tagged compounds are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels.
  • radioisotope labels include 1 jl Iodine, 1 25 lodine, ""Technetium and/or ' ' 'indium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo.
  • there are a number of art-known methods of linking peptide to Tc-99m including, simply by way of example, those shown by U.S. Patent Nos. 5,328,679; 5,888,474; 5,997,844; and 5,997,845, incorporated by reference herein.
  • the compounds described herein can be used for delivering nucleic acids (oligonucleotides) into cells or tissues.
  • nucleic acid or “nucleotide” apply to deoxyribonucleic acid ("DNA”).
  • An “oligonucleotide” is generally a relatively short polynucleotide, e.g., ranging in size from about 2 to about 200 nucleotides, or more preferably from about 8 to about 30 nucleotides in length.
  • the oligonucleotides according to the invention are generally synthetic nucleic acids, and are single stranded, unless otherwise specified.
  • polynucleotide and '"polynucleic acid
  • oligonucleotides are not limited to a single species of oligonucleotide but, instead, are designed to work with a wide variety of such moieties, it being understood that linkers can attach to one or more of the 3'- or 5'- terminals, usually PO 4 or SO 4 groups of a nucleotide.
  • the nucleic acids molecules contemplated can include a phosphorothioate internucleotide linkage modification, sugar modification, nucleic acid base modification and/or phosphate backbone modification.
  • the oligonucleotides can contain natural phosphorodiester backbone or phosphorothioate backbone or any other modified backbone analogs such as LNA (Locked Nucleic Acid), PNA (nucleic acid with peptide backbone), CpG oligomers, and the like, such as those disclosed at Tides 2002, Oligonucleotide and Peptide Technology Conferences, May 6-8, 2002, Las Vegas, NV and Oligonucleotide & Peptide Technologies, 18th & 19th
  • oligonucleotides contemplated by the invention include, for example, the addition to or substitution of selected nucleotides with functional groups or moieties that permit covalent linkage of an oligonucleotide to a desirable polymer, and/or the addition or substitution of functional moieties that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and functionality to an oligonucleotide.
  • Such modifications include, but are not limited to, 2'-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodouracil, backbone modifications, methylations, base-pairing combinations such as the isobases isocytidine and isoguanidine, and analogous combinations.
  • Oligonucleotides contemplated within the scope of the present invention can also include 3' and/or 5' cap structure.
  • cap structure' shall be understood to mean chemical modifications, which have been incorporated at either te ⁇ ninus of the oligonucleotide.
  • the cap can be present at the 5'-te ⁇ ninus (5'-cap) or at the 3'-terminus (3'-cap) or can be present on both terminus.
  • a non-limiting examples of the 5'-cap includes inverted abasic residue (moiety), 4',5'-methylene nucleotide; 1 -(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide; 1 ,5-anhydrohexitol nucleotide; L-nucleotides; alpha- nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide, 3'-3'-inverted nucleotide moiety; 3'-3'-invcrted abasic moiety;
  • the 3'-ca ⁇ can includes, for example, 4',5'-methylene nucleotide; l -(beta-D-erythrofuranosyl) nucleotide: 4'-thio nucleotide, carbocyclic nucleotide; 5'-amino-alkyl phosphate; l ,3-diamino-2-propyl phosphate,
  • nucleoside analogs has the structure:
  • anti sense refers Io nucleotide sequences which are complementary to a specific DNA or RNA sequence that encodes a gene product or -that encodes a control sequence.
  • antisense strand' is used in reference to a nucleic acid strand that is complementary to the "'sense" strand.
  • the sense strand of a DNA molecule is the strand that encodes polypeptides and/or other gene products.
  • the sense strand serves as a template for synthesis of a messenger RNA ("mRNA' " ) transcript (an antisense strand) which, in turn, directs synthesis of any encoded gene product.
  • Antisense nucleic acid molecules may be produced by any art-known methods, including synthesis by Iigating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated.
  • the designations "negative " or (-) are also art-known to refer to the antisense strand, and "positive " or (+) are also art-known to refer to the sense strand.
  • complementary shall be understood to mean that a nucleic acid sequence forms hydrogen bond(s) with another nucleic acid sequence.
  • a percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule which can form hydrogen bonds, i.e., Watson-Crick base pairing, with a second nucleic acid sequence, i.e.. 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary.
  • Perfectly complementary means that all the contiguous residues of a nucleic acid sequence form hydrogen bonds with the same number of contiguous residues in a second nucleic acid sequence.
  • the choice for conjugation is an oligonucleotide (or "polynucleotide " ') and after conjugation, the target is referred to as a residue of an oligonucleotide.
  • the oligonucleotides can be selected from among any of the known oligonucleotides and oligodeoxynucleotides with phosphorodiester backbones or phosphorothioate backbones.
  • oligonucleotides or oligonucloetide derivatives useful in the compounds described herein can include from about 8 to about 1000 nucleic acids, and preferably relatively short polynucleotides, e.g., ranging in size preferably from about 8 to about 30 nucleotides in length (e.g., about 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30).
  • oligonucleotides and oligodeoxynucleotides with natural phosphorodiester backbone or phosphorothioate backbone or any other modified backbone analogues include:
  • LNA Locked Nucleic Acid
  • PNA nucleic acid with peptide backbone
  • short interfering RNA siRNA
  • miRNA microRNA
  • nucleic acid with peptide backbone PNA
  • PMO phosphorodiamidate morpholino oligonucleotides
  • tricyclo-DNA decoy ODN (double stranded oligonucleotide); catalytic RNA sequence (RNAi); ribozymes; aptamers; aptamers; aptamers (L-conformational oligonucleotides);
  • oligonucleotides can include any suitable art-known nucleotide analogs and derivatives, including those listed by Table 1 , below: TABLE 1. Representative Nucleotide Analogs And Derivatives
  • the oligonucleotide is involved in targeted tumor cells or downregulating a protein implicated in the resistance of tumor cells to anticancer therapeutics.
  • a protein implicated in the resistance of tumor cells to anticancer therapeutics for example, any art-known cellular proteins such as BCL-2 for downregulation by antisense oligonucleotides, for cancer therapy, can be used for the present invention. See U.S. Patent Application No.
  • oligonucleotides include antisense HIF- l ⁇ oligonucleotides, antisense ErbB3 oligonucleotides, antisense survivin oligonucleotides and ⁇ -catenine oligonucleotides.
  • the oligonucleotides useful in the method described herein include phosphorothioate backbone and LNA.
  • the oligonucleotide useful in the method described herein includes antisense bcl-2 oligonucleotides, antisense HIF- l ⁇ oligonucleotides, antisense survivin oligonucleotides, and antisense Erb ⁇ 3 oligonucleotides.
  • the oligonucleotide can be, for example, an oligonucleotide that has the same or substantially similar nucleotide sequence as does Genasense (a/k/a oblimersen sodium, produced by Genta Inc., Berkeley Heights, NJ).
  • Genasense is an 18-nier phosphorothioate antisense oligonucleotide, TCTCCCAGCGTGCGCCAT (SEQ ID NO: 4), that is complementary to the first six codons of the initiating sequence of the human bcl-2 mRNA (human bcl-2 mRNA is art-known, and is described, e.g., as SF.Q ID NO: 19 in U.S. Patent No. 6,414,134, incorporated by reference herein).
  • the U.S. Food and Drug Administration (FDA) gave Genasense Orphan Drug status in August 2000.
  • Preferred embodiments include: Preferred embodiments contemplated include: (i) antisense Survivin LNA oligonucleotide (SEQ ID NO: 1 ) mC 3 -T s - m C 3 -A s- a s -t s -c s -c s -a s -t s -g s -g s - m C 3 -A s -G s -c ; where the upper case letter represents LNA. the "s" represents a phosphorothioate backbone; (ii) antisense Bc!2 siRNA:
  • antisense HIFl ⁇ LNA oligonucleotide (SEQ ID NO: 5) where the upper case letter represents LNA and the "s " represents phosphorothioate backbone.
  • antisense ErbB3 LNA oligonucleotide (SEQ ID NO: 6) where the upper case letter represents LNA and the "s” represents phosphorothioate backbone.
  • LNA includes 2"-O, 4 % -C methylene bicyclonucleotide as shown below:
  • the oligonucleotide comprises SEQ ID NO: 1 , SEQ ID NOs 2 and 3, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6.
  • the oligonucleotides prior to the conjugation to the polymeric systems described herein include (CHT)* sulfhydryl linkers (thio oligonucleotides) at 5 " or 3" end of the oligonucleotides, where (w) in this aspect is a positive integer of from about 1 to about 10 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and preferably 4, 5, 6 or 7.
  • the thio oligonucleotides have the structure SH-(CH 2 X v - Oligonucleotide.
  • the compounds described herein can include oligonucleotides modified with hindered ester-containing (CH 2 ) tt sulfhydryl linkers. Prior to the attachment, the oligonucleotide has the structure:
  • (w) is a positive integer from about 1 to about 10, (e.g., 3, 4, 5, 6). See WO 2008/034] 19, the contents of which are incorporated by reference.
  • the polymeric compounds can release the oligonucleotides without thiol tail.
  • 5' end of the sense strand of siRNA is modified.
  • siRNA employed in the polymeric conjugates is modified with a 5'-C O -SH.
  • One particular embodiment of the present invention employs Bcl2-siRNA having the sequence of SENSE 5'-SH-C 6 -GCAUGCGGCCUCUGUUUGAdTdT-3'
  • modified oligonucleotides include:
  • the methods of preparing compounds described herein include reacting an activated polymer with an oligonucleotide modified with a SH group.
  • Activated polymers useful in the methods described herein include a polymer containing a pyridyl disulfide group at the distal end. The methods provide a polymeric conjugate where the biologically active moiety is bonded to the polymer through -S-S- bond.
  • methods of preparing polymeric compounds described herein include: reacting a polymeric compound of Formula (111): with an olignucleotide modified with a sulfhydryl group-containing moiety under conditions sufficient to form a compound of Formula (1): wherein
  • Ri is a substantially non-antigenic water-soluble polymer; each Z
  • Yi in each occurrence, is independently S or O, preferably O; Y2, in each occurrence, is independently NRn, preferably NH; R 100, in each occurrence is the same or different and selected from among H, a leaving group, an activating group, and
  • each of L M in each occurrence, is the same or a different bifunctional linker;
  • R b in each occurrence, is a folic acid;
  • R c in each occurrence, is the same or a different diagnostic agent;
  • each of R3.7 is independently selected from among hydrogen, C
  • each of Rjj-i 1 is an electron-withdrawing group such as substituted amido, acyl, azido, carboxy, alkyloxycarbonyl, cyano, and nitro, and preferably Rs is nitro, and RQ, R IO and Rn are hydrogen;
  • R 13 in each occurrence, is independently selected from the group consisting of hydrogen, C
  • Ri 2 in each occurrence, is independently selected from the group consisting of hydrogen, hydroxy], C
  • each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably 0; each of (al) and (a2) is independently zero, 1 , 2, or 3, and preferably 1 ; each (b) is independently zero, 1, 2, or 3, and preferably 0; each (c) is independently zero, 1 , 2, or 3, and preferably 1 ; each (e) is independently zero or one, and preferably 0; each (g) is independently zero or one, and preferably 1 ; and
  • (m 1 1 ) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero and provided that one or more of Zi i contain
  • the reactions are carried out in an inert solvent such as methylene chloride, chloroform, DMF or mixtures thereof.
  • the reactions can be preferably conducted in the presence of a base, such as dimethylaminopyridine (DMAP), diisopropylethylamine, pyridine, tri ethyl amine, etc. to neutralize any acids generated.
  • DMAP dimethylaminopyridine
  • the reactions can be carried out at a temperature from about 0 0 C up to about 22 0 C (room temperature). See detailed description in WO/2009/025669, the contents of which are incorporated herein by reference.
  • Oligo is an oligonucleotide, preferably an oligonucleotide modified with a C.i-Q, alkyl (C 6 alkyl);
  • PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; (al ) is one; is zero or one; and ' all other variables are the same as defined above.
  • compounds prepared by the method described herein include
  • Oligo is an oligonucleotide
  • R b is
  • PEG is polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons.
  • compounds prepared by the methods described herein include:
  • Oligo is an oligonucleotide, preferably an oligonucleotide modified with a C 3 -C 6 alkyl (e.g., C 6 alkyl); and
  • PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons.
  • the multi-arm PEG is shown as "PEG".
  • One arm, up to seven arms of the eight-arm PEG (or up to three arms of the four-arm PEG) can be conjugated with a folic acid.
  • the compounds include therapeutic oligonucleotides such as antisense ErbB3 oligonucleotides and antisense Survivin oligonucleotides.
  • the compounds include a C(,-tail modified antisense LNA as follows:
  • the average molecular weight of the polymeric portion is about 40,000 daltons.
  • One aspect of the present invention provides methods of introducing or delivering an oligonucleotide into a mammalian cell.
  • the method according to the present invention includes contacting a cell with a compound of Formula (I) described herein.
  • the present invention is useful for introducing oligonucleotides to a mammal having tumor cells.
  • the compounds described herein can be administered to a mammal, preferably human.
  • the present invention preferably provides methods of inhibiting or downregulating (or modulating) a gene expression in mammalian cells or tissues.
  • the downregulation or inhibition of gene expression can be achieved in vivo and/or in vitro.
  • the methods include contacting human cells or tissues with compounds of Formula (I) described herein.
  • successful inhibition or down-regulation of gene expression such as in mRNA or protein levels shall be deemed to occur when at least about 10%, preferably at least about 20% or higher is realized when measured in vivo or in vitro, when compared to that observed in the absence of the treatment with the compound described herein.
  • inhibiting or “down-regulating” shall be understood to mean that the expression of a target gene, or level of RNAs or equivalent RNAs encoding one or more protein subunits, or activity of one or more protein subunits, such as ErbB3, is reduced below that observed in the absence of the treatment with the conjugates described herein.
  • gene expression of a target gene is inhibited in prostate or cervical cancer cells or tissues, for example, prostate or cervical cancer cells.
  • the cancer cells or tissues can be from one or more of the following: solid tumors, lymphomas, small cell lung cancer, acute lymphocytic leukemia (ALL), pancreatic cancer, glioblastoma, ovarian cancer, gastric cancer, breast cancer, colorectal cancer, ovarian cancer and brain tumors, etc.
  • the compounds according to the methods described herein include, for example, antiscnse bcl-2 oligonucleotides, antisense HIF-I ⁇ oligonucleotides, antisense Survivin oligonucleotides, and antisense Erb ⁇ 3 oligonucleotides.
  • the administering step is via the blood stream of the mammal.
  • a further aspect of the present invention provides methods of treatment for various medical conditions in mammals.
  • the methods include administering, to the mammal in need of such treatment, an effective amount of a pharmaceutical composition containing a compound of Formula (I).
  • the polymeric conjugate compounds are useful for, among other things, treating diseases including, but not limited to, cancer, inflammatory disease, and autoimmune disease.
  • a useful target gene includes, but is not limited to, oncogenes, pro- angiogenesis pathway genes, pro-cell proliferation pathway genes, viral infectious agent genes, and pro-inflammatory pathway genes.
  • methods of treating a patient having a malignancy or cancer comprising administering an effective amount of a pharmaceutical composition containing the compound of Formula (I) to a patient in need thereof.
  • the cancer being treated can be one or more of the following: solid tumors, lymphomas, small cell lung cancer, acute lymphocytic leukemia (ALL), pancreatic cancer, glioblastoma, ovarian cancer, gastric cancers, colorectal cancer, etc.
  • compositions are useful for treating neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mammals by downregulating gene expression of a target gene.
  • Any oligonucleotide, etc. which has therapeutic effects in the unconjugated state can be used in its conjugated fo ⁇ n, made as described herein.
  • the methods described herein include administering polynucleotides (oligonucleotides), preferably antisense oligonucleotides to mammalian cells.
  • the methods include delivering an effective amount of a conjugate prepared as described herein to the condition being treated will depend upon the polynucleotides efficacy for such conditions.
  • the method would include delivering a polymer conjugate containing the oligonucleotides to the cells having susceptibility to the native oligonucleotides.
  • the delivery can be made in vivo as part of a suitable pharmaceutical composition or directly to the cells in an ex vivo environment.
  • the polymeric conjugates including oligonucleotides SEQ ID NO. 1 , SEQ ID NOs: 2 and 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6) can be used.
  • the present invention provides methods of inhibiting the growth or proliferation of cancer cells in vivo or in vitro.
  • the methods include contacting cancer cells with the compounds described herein.
  • the present invention provides methods of modulating apoptosis in cancer cells by administering the compounds described herein to a mammal in need thereof.
  • the methods include introducing the oligonucleotide (antisense LNA) conjugates described herein to cancer cells to reduce survivin expression in the cancer cells or tissues, wherein the antisense oligonucleotide binds to niRNA expressed from the survivin gene and reduces survivin gene expression.
  • oligonucleotide antisense LNA
  • the methods include introducing the compounds described herein to tumor cells to reduce gene expression such as ErbB3 and contacting the tumor cells with an amount of at least one chemotherapeutic agent sufficient to kill a portion of the tumor cells.
  • the portion of tumor cells killed can be greater than the portion which would have been killed by the same amount of the chemotherapeutic agent in the absence of the compounds described herein.
  • a chemotherapeutic agent can be used in combination, simultaneously or sequentially, with the methods employing the compounds described herein.
  • the compounds described herein can be administered concurrently with the chemotherapeutic agent, prior to, or after the administration of the chemotherapeutic agent.
  • the compounds described herein can be administered during or after treatment of the chemotherapeutic agent.
  • compositions including the compounds described herein may be formulated in conjunction with one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically.
  • Proper formulation is dependent upon the route of administration chosen, i.e., whether local or systemic treatment is treated. Parenteral routes are preferred in many aspects of the invention.
  • compositions containing the compounds of Formula (1) described herein may be oral, pulmonary, topical including epidermal, transdermal, ophthalmic and to mucous membranes including vaginal and rectal delivery or parenteral including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion.
  • the compounds containing therapeutic oligonucleotides is administered IV, IP or as a bolus injection.
  • the compounds described herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as physiological saline buffer or polar solvents including, without limitation, a pyrrolidone or dimethylsulfoxide.
  • physiologically compatible buffers such as physiological saline buffer or polar solvents including, without limitation, a pyrrolidone or dimethylsulfoxide.
  • the compounds may also be fo ⁇ nulated for parenteral administration, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers.
  • Useful compositions include, without limitation, suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain adjuncts such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical compositions for parenteral administration include aqueous solutions of a water soluble form, such as. without limitation, a salt (preferred) of the active compound. Additionally, suspensions of the active compounds may be prepared in a lipophilic vehicle.
  • Suitable lipophilic vehicles include fatty oils such as sesame oil, synthetic fatty acid esters such as ethyl oleate and triglycerides, or materials such as liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers and/or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • the compounds can be fo ⁇ nulated by combining the active compounds with pharmaceutically acceptable carriers well-known in the art.
  • Such carriers enable the compounds of the invention to be fo ⁇ nulated as tablets, pills, lozenges, dragees, capsules, liquids, gels, syrups, pastes, slurries, solutions, suspensions, concentrated solutions and suspensions for diluting in the drinking water of a patient, premixes for dilution in the feed of a patient, and the like, for oral ingestion by a patient.
  • Pha ⁇ naceutical preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding other suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Useful excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol, cellulose preparations such as, for example, maize starch, wheat starch, rice starch and potato starch and other materials such as gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methylcellulose, sodium carboxymethylceilulose, and/or polyvinylpyrrolidone (PVP).
  • fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol
  • cellulose preparations such as, for example, maize starch, wheat starch, rice starch and potato starch and other materials such as gelatin, gum traga
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid.
  • a salt such as sodium alginate may also be used.
  • the compounds of the present invention can conveniently be delivered in the form of an aerosol spray using a pressurized pack or a nebulizer ' and a suitable propellant.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as depot preparations. Such long acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection.
  • a compound of this invention may be formulated for this route of administration with suitable polymeric or hydrophobic materials (for instance, in an emulsion with a pharmacologically acceptable oil), with ion exchange resins, or as a sparingly soluble derivative such as, without limitation, a sparingly soluble salt.
  • conjugates may be delivered using a sustained-release system, such as semi -permeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • the therapeutically effective amount can be estimated initially from in vitro assays. Then, the dosage can be formulated for use in animal models so as to achieve a circulating concentration range that includes the effective dosage. Such information can then be used to more accurately determine dosages useful in patients.
  • the amount of the composition, e.g., used as a prodrug, that is administered will depend upon the parent molecule included therein (i.e., efficacy of an unconjugated oligonucleotide). Generally, the amount of prodrug used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various prodrug compounds will vary somewhat depending upon the parent compound (oligonucleotides such as LNA), rate of in vivo hydrolysis, molecular weight of the polymer, etc. In addition, the dosage, of course, can vary depending upon the dosage form and route of administration.
  • the oligonucleotide conjugates described herein can be administered in amounts ranging from about 1 mg/kg/week to about 1 g/kg/week, preferably from about 1 to about 500 mg/kg/week and more preferably from I to about 100 mg/kg/week (i.e., from about 2 to about 60 mg/kg/week).
  • the range set forth above is illustrative and those skilled in the art will determine the optimal dosing of the prodrug selected based on clinical experience and the treatment indication.
  • the exact formulation, route of administration and dosage can be selected by the individual physician in view of the patient's condition.
  • toxicity and therapeutic efficacy of the compounds described herein can be dete ⁇ nined by standard pharmaceutical procedures in cell cultures or experimental animals using methods well-known in the art.
  • dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per subject per day).
  • the amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • the treatment of the present invention includes administering the oligonucleotide conjugates described herein in an amount of from about 2 to about 50 mg/kg/dose, such as from about 5 to about 30 mg/kg/dose to a mammal.
  • the delivery of the oligonucleotide conjugates described herein includes contacting a concentration of oligonucleotides of from about 0.1 to about 1000 nM, preferably from about 10 to about 1000 nM with tumor cells or tissues in vivo or in vitro.
  • compositions may be administered once daily or divided into multiple doses which can be given as part of a multi-week treatment protocol.
  • the precise dose will depend on the stage and severity of the condition, the susceptibility of the tumor to the polyrner-prodrug composition, and the individual characteristics of the patient being treated, as will be appreciated by one of ordinary skill in the art.
  • the dosage amount mentioned is based on the amount of oligonucleotide molecule rather than the amount of polymeric conjugate administered. It is contemplated that the treatment will be given for one or more days until the desired clinical result is obtained.
  • the exact amount, frequency and period of administration of the compound of the present invention will vary, of course, depending upon the sex, age and medical condition of the patent as well as the severity of the disease as determined by the attending clinician.
  • Still further aspects include combining the compound of the present invention described herein with other anticancer therapies for synergistic or additive benefit.
  • DCM dichloromethane
  • DIEA N,N-Diisopropylethylaamine
  • LNA Locked Nucleic Acid
  • MEM Modified Eagle's Medium
  • TEAA tetraethylammonium acetate
  • TFA trifluoroacetic acid
  • RT-qPCR reverse transcription-quantitative polymerase chain reaction
  • the reaction mixtures and the purity of intermediates and final products are monitored by a Beckman Coulter System Gold ® HPLC instrument. It employs a ZORB AX ® 300SB C8 reversed phase column ( 150 * 4.6 mm) or a Phenomenex Jupiter ® 300A Cl 8 reversed phase column ( 15O x 4,6 mm) with a 168 Diode Array UV Detector, using a gradient of 10-90 % of acetonitrile in 0.05 % TFA at a flow rate of 1 lnL/rninute or a gradient of 25-35 % acetonitrile in 50 mM TEAA buffer at a flow rate of 1 mL/min ⁇ te.
  • the anion exchange chromatography was run on AKTA explorer I OOA from GE healthcare (Amersham Biosciences) using Poros 50HQ strong anion exchange resin from Applied Biosystems packed in an AP-Empty glass column from Waters. Desalting was achieved by using HiPrep 26/10 desalting columns from Amersham Biosciences.
  • Example 3 General mRNA Down-Regulation Procedure. The cells were maintained in complete medium (F- 12K or DMEM, supplemented with
  • RNA was prepared using RNAqueous Kit ® (Ambion) following the manufacturer's instruction. The RNA concentrations were determined by OD260 nm using Nanodrop.
  • the reaction was conducted in a PCR the ⁇ nocycler at 25 0 C for 10 minutes, 37 0 C for 120 minutes, 85 0 C for 5 seconds and then stored at 4 0 C.
  • Real-time PCR was conducted with the program of 50 °C-2 minutes, 95 0 C-IO minutes, and 95 0 C-15 seconds / 60 0 C-I minute for 40 cycles. For each qPCR reaction, 1 ⁇ L of cDNA was used in a final volume of 30 ⁇ L.
  • H eterobi functional amino acid PEG (compound 2) is coupled with NHS in the presence of EDC to provide an NHS ester (compound 3).
  • Example 14 Preparation of Compound 12.
  • Compound 11 bis SC-PEG 1 0.35 mmol
  • DMF/DCM 25 mL/45 mL
  • DlEA 3 equivalent for each compound 5
  • the suspension is stirred at room temperature for 5 hours.
  • the reaction mixture is evaporated under vacuum and then precipitated with DCM/Et 2 O at 0 0 C.
  • the solid is filtered and then is dissolved in 80 mL of DCM.
  • Example 21 Preparation of Compound 22 ((NHExtend) r PEG-Cys-SS-NPyS).
  • Example 22 Preparation of Compound 23 ((Folate-NHExtend) 3 -PEG-Cys-SS-NPyS). A solution of compound 1 in DMSO was added to a solution of compound 22 in DCM.
  • Example 23 Preparation of Compound 25 ((Folate-NHExtendb- ⁇ PEG-Cys-SS-C ⁇ -Oligo).
  • (Folate) 3 - 20K PEG-NPyS (compound 23, 120 mg, 5.50 ⁇ mol) was dissolved in pH 6.5 phosphate buffer (3-4 mL), covered in foil and purged with nitrogen gas for 10 minutes.
  • oligonucleotides compound 24, e.g. antisense ErbB3 LNA oligonucleotide, 6.0 mg, 1.10 ⁇ mol
  • the resulting orange yellow mixture was stirred for -2 hours at ambient temperature during which time the solution became deeper yellow in color.
  • the solution was filtered using a 0.45 ⁇ m syringe filter and loaded on a Poros HQ, strong anion exchange column ( 10 cm x 1.0 cm, bed volume - 8 mL) which was pre-equilibrated with 20 mM Tris-HCI buffer, pH 7.0 (buffer A). The column was washed with 3-4 column volumes of buffer A to remove the excess PEG linker. The product was eluted by slow incremental gradient of 1 M NaCl in 20 mM Tris-HCI buffer, pH 7.0 (buffer B).
  • Example 24 Preparation of Compound 26 ((FoIate-NHExtend) 3 - 40K PEG-Cys-SS-C 6 -OIigo- FAM).
  • the solution was filtered using a 0.45 ⁇ m syringe filter and loaded on a Poros HQ, strong anion exchange column (10 cm x 1.0 cm, bed volume - 8 raL) which was pre-equilibrated with 20 mM Tris-HCl buffer, pH 7.0 (buffer A). The column was washed with 3-4 column volumes of buffer A to remove the excess PEG linker. The product was eluted by slow incremental gradient of 1 M NaCl in 20 mM Tris- HCl buffer, pH 7.0 (buffer B).
  • Example 25 Preparation of Compound 28.
  • Example 27 Preparation of Compound 31 (Folate)2-TAMRA- 20K PEG-Cys-SS-C 6 -Oligo.
  • Example 29 Preparation of Compound 35. Different sizes of activated PEG polymer were used to make PEG-LNA conjugates.
  • compound 33 was reacted with oligonucleotides (compound 34) using the conditions described in Example 6. The description of each compound is provided in FIG. 6.
  • Oligonucleotides (compound 34, 400 mg, 0.074 mmol) were added to a solution of compound 33 (505 nig, 0.012 mmol) in 25 mL of pH 6.5, 100 mM Sodium Phosphate. The reaction was stirred at room temperature under nitrogen for 5 hours. The reaction mixture was purified on Source 15RPC column. The column was equilibrated with 100 mM TEAA. Then the reaction mixture was loaded. The column was eluted with IM NaCl, water, and 50% CH3CN. Compound 35 was collected and lyophilized. Yield 150mg, MALDI confirms the molecular weight of 62,590.
  • Examples 31-37 demonstrate improved tumor delivery of oligonucleotides as well as improved antisense knockdown of targeted tumor mRNA using the releasably linked PEG molecule having the formula: wherein
  • PEG is a polyethylene glycol
  • R b is
  • Oligo is uniformly 5'-(CH 2 ) 6 -anti-survivin LNA (SEQ ID NO: 1, referred to as "LNAl”) or 5'-(CH 2 ) 6 -anti-erbB3 LNA (SEQ ID NO: 6, referred to "LNA2”), and the total molecular weight of the polymeric portion of the compound containing PEG is about 40,000 daltons, 20,000 daltons, 10,000 daltons or 5,000 daltons.
  • LNAl 5'-(CH 2 ) 6 -anti-survivin LNA
  • LNA2 5'-(CH 2 ) 6 -anti-erbB3 LNA
  • the compounds include Folate- 40K PEG-Cys-SS-LNA2 (compound 101), Folate- 5K PEG-Cys-SS-LNA2 (compound 1O) 3 401 TEG-CyS-SS-LNAl (compound 103), 10K PEG- Cys-SS-LNAl, (compound 104) and 40K PEG-Cys-S S-LN A2 (compound 105).
  • KB human cervical carcinoma cell line KB cells were plated overnight at 37 0 C. The cells were incubated with 100 nM of Folate-PEG-Cys-SS-LNA2-FAM conjugates (compounds 101 and 10 labeled with FAM) with or without 1 ⁇ M of free folate at 37 0 C for 24 hours. The amount of the PEG-Cys-SS-LNA2-FAM conjugate was based on the amount of LNA2, not the amount of polymeric conjugate. The cells were washed and the samples were observed under fluorescence microscope and confocal microscope.
  • FIG. 7 The results of the cellular uptake of the Folate-PEG-Cys-SS-LNA conjugate are shown in FIG. 7.
  • KB cells were also exposed to Folate- 5K PEG-Cys-SS-LNA2-FAM (compound 10 labeled with FAM) with or without 100 nM of free folate at 37 0 C for 4 hours.
  • the cells were washed and analyzed by FACS for the specificity of binding. The results are shown in FIG. 8.
  • FIG. 7(a) The fluorescence microscope images (FIG. 7(a)) and FACS analysis (FIG. 8) showed that the binding of the folate-PEG conjugate to the folate receptor and subsequent internalization into KB cells was blocked in the presence of free folate. The binding of the folate-PEG conjugate to the folate receptor in KB cells was specific.
  • the efficacy of Folate-PEG-LNA conjugates was evaluated in KB human cervical cancer xenografted mice.
  • Athymic nude Balb/c mice bearing KB tumor were treated with a dose of 35 mg/kg of naked LN A2 or Folate- ⁇ PEG-Cys- SS-LN A2, 40K PEG-LNA2, Folate- 5K PEG-LNA2, or 5K PEG- LNA2 conjugate at q3dx4 for 12 days.
  • the amount of the PEG conjugates administered was based on the amount of LNA2, not the amount of polymeric conjugate administered.
  • Tumor and liver samples were isolated and analyzed by using qRT-PCR for ErbB3 mRNA down-regulation.
  • the results are as shown in FIGS. 10(A) and (B).
  • the folate-PEG conjugates significantly inhibited expression of ErbB3 mRNA compared to naked LN A2 in the tumor tissues.
  • 4OK Folate-PEG-LNA conjugates downregulated target mRNA compared to 5K Folate-PEG-LNA conjugates.
  • the results showed that PEGylation increased accumulation of LNA oligonucleotide in tumor and the folate-PEG conjugates enhanced mRNA downregulation as compared to naked LNA oligonucleotides.
  • A549 (human lung adenocarcinoma epithelial cell line) cells were implanted sc. in athymic nude mice. When tumor reached the average volume of 75 mm 3 , the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LNAl , 40K PEG-Cys- SS LNAl (compound 103, 10 mg/kg equivalent dose of LNA l ) or IOK PEG-Cys-SS-LNAl (compound 104, 10 mg/kg equivalent dose of LNAl ). Plasma samples were collected at 2 and 4 hours time points following the treatment.
  • Tumor tissues were collected from the sacrificed animals at the various time points (2, 4, 12, 24, 48 and 72 hours) following the treatment. Concentrations of equivalent-LNA I oligonucleotides in tumor or plasma samples were measured by an ELISA hybridization assay. Results are shown in FIG. 1 l (A) and (B).
  • mice treated with the PEG-Cys-SS-LNA conjugate had >50 rimes concentration of circulating LNA oligonucleotides in plasma at 2 hours and 4 hours following the treatment, as compared to naked LNA oligonucleotides.
  • the PEG-Cys-SS-LNA conjugates had higher plasma concentrations and longer circulating times compared to naked LNA oligonucleotides.
  • mice treated with the PEG conjugate had 3-fold higher accumulation of LNA oligonucleotides in rumor at 24 hours, as compared to naked LNA oligonucleotides.
  • the results also indicated that the 40K PEG conjugates had >3.5 times tumor accumulation at 12 hours and maintained > 1.5 times accumulation up to 72 hours compared to the 10K PEG conjugates.
  • higher molecular weight PEG (40KDa) conjugates has greater tumor accumulation than lower MW PEG (1 OKDa) PEG conjugates.
  • Example 36 In vivo Efficacy of PEG-Cys-SS-LNA conjugate in KB Xenografted Mice Model
  • mice The efficacy of PEG-LNA conjugates was evaluated in KJB human cervical cancer xenografted mice.
  • KB cells epidermal carcinoma cell line
  • the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LNA2 or 40K PEG-Cys-SS LN A2 (compound 105, 10 mg/kg equivalent dose of LNA2) at q3d x4.
  • Tumor and liver samples were collected 24 hours after the last dose. ErbB3 mRNA downregulation in the samples was measured by using qRT-PCR. The results are shown in FIG. 12.
  • mice 15PC3 human prostate cancer xenografted mice.
  • 15PC3 cells human prostate cancer cell line
  • the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LN A2 or 40K PEG-Cys-SS LN A2 (compound 105, 10 mg/kg equivalent dose of LN A2) at q3d x4.
  • Tumor and liver samples were collected 24 hours after the last dose. ErbB3 mRNA downregulation in the samples was measured by using qRT-PCR. The results are shown in FIG. 13.
  • mice treated with the PEG conjugate inhibited ErbB3 mRNA expression 2 fold more than the mice treated with naked LNA. Additionally, the PEG conjugates inhibited 83% ErbB3 mRNA expression in liver as compared to 73% by naked LNA oligonucleotides.
  • the invention advantageously provides improved methods employing PEG conjugates for the delivery of oligonucleotides to tumor cells in a mammal that greatly increase circulation time, enhance the accumulation of oligonucleotides in tumors in vivo, while also achieving enhanced downregulation of oncogene mRNA expression in tumors compared to corresponding naked anrisense constructs.
  • PEG is a polyethylene glycol
  • Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons;
  • PEG is a polyethylene glycol
  • Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons; and (b) administering the compound or the salt thereof to a mammal having tumor cells, wherein said administration reduces the expression of the preselected gene by the tumor cells.
  • the inhibition of expression of the preselected gene may be as a result of antisense targeting of an mRNA molecule thereby reducing or eliminating translation of the mRNA to a polypeptide.
  • the administration may be by the blood stream of the mammal, for example, by intravenous (i.v.) injection.
  • the oligonucleotides may comprise LNA.
  • the oligonucleotide includes -5 " -(CH 2 )6-antisense-Survivin LNA oligonucleotide or -5'- (CH 2 ) 6 -antisense-ErbB3 LNA oligonucleotide.

Abstract

The present invention provides polymeric prodrugs including an intracellular releasable disulfide linker for the delivery of oligonucleotides. Methods of making the compounds as well as methods of delivering nucleic acids to tumor cells in a mammal using the same are also provided.

Description

POLYMERIC SYSTEMS CONTAINING INTRACELLULAR RELEASABLE DISULFIDE LINKER FOR THE DELIVERY OF OLIGONUCLEOTIDES
CROSS-REFERENCE TO RELATED APPLICATION This application claims the benefit of priority from U.S. Provisional Patent Application
Serial Nos. 61/055,950 filed May 23, 2008, 61/055,869 filed May 23, 2008, 61/106,578 filed October 19, 2008, and 61/106,576 filed October 19, 2008, the contents of each of which are incorporated herein by reference.
BACKGROUND OF THE INVENTION
Gene -based therapy is a powerful tool in the treatment of disease because a therapeutic gene can selectively modulate gene expression associated with disease and minimize side effects which may incur when other therapeutic approaches are used.
A therapy based on locked Nucleic Acid (LNA) antisense oligonucleotide, a new generation of RNA antagonist, has been proposed. Each LNA monomer contains a methylene bridge between the 2'-oxygen and 4'-carbon of the ribose sugar. This fixes the LNA residue in a favorable RNA-like conformation and enables LNA oligonucleotides to have higher affinity, specificity, and resistance against degradation compared with other art-known oligonucleotides. It has been shown that LNA oligonucleotide inhibits target gene expression in vitro (at sub- nanomolar level). While LNA oligonucletides have improved therapeutic activity compared to other art-known nucleic acids, it is still needed to further improve the pharmacokinetic profile and fast clearance from circulation and limited activity in vivo of LNA oligonucleotides. There continues to be a need to provide improved systems and methods for the delivery of LNA oligonucleotides as well as other art-known nucleic acid molecules. The present invention addresses this need.
SUMMARY OF THE INVENTION
In order to overcome the above problems and improve the technology for the delivery of oligonucleotides, the present invention provides new polymeric delivery systems containing an intracellularly releasable linker. In one aspect of the present invention, there are provided methods of delivering oligonucleotides to tumor cells in a mammal. The methods include administering to the mammal having tumor cells a compound of Formula (I):
Figure imgf000003_0002
or a pharmaceutically acceptable salt thereof, wherein
R] is a substantially non-antigenic water-soluble polymer; each Z| is the same or different and selected from among:
Figure imgf000003_0001
Yi, in each occurrence, is independently S or O: Y2, in each occurrence, is independently NRj3; R,,, in each occurrence, is the same or a different oligonucleotide; each of Li -4, in each occurrence, is the same or a different bifunctional linker;
Rt,, in each occurrence, is a folic acid;
Rc, in each occurrence, is the same or a different diagnostic agent; each Of R3-7 is independently selected from among hydrogen, Ci^ alkyls, C2.6 alkenyl, C2. 6 alkynyl, C3.19 branched alkyl, C^ cycloalkyl, and C1.6 alkoxy; R |3, in each occurrence, is independently selected from among hydrogen, Ci.<j alkyls, C2-6 alkenyl, C^ alkynyl, Cj.iς branched alkyl, and C3.8 cycloalkyl;
R]2, in each occurrence, is independently selected from among hydrogen, hydroxyl, Ci^ alkyls, C2-6 alkenyl, Ci^ alkynyl, C3-I? branched alkyl, C3.8 cycloalkyl, and Ci-6 alkoxy; each of (a) and (d) is independently 0, 1 , 2 or 3; each of (al ) and (a2) is independently 0, 1 , 2 or 3; each (b) is independently 0, 1 , 2, or 3; each (c) is independently 0, 1, 2 or 3; each (e) is independently 0 or 1 ; each (g) is independently 0 or 1 ; and (in) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero and further provided that one or more of Z| contain an oligonucleotide.
In another aspect, the present invention provides a method of inhibiting a gene expression in a mammal having prostate or cervical cancer cells. In one embodiment, the compound of Formula (I) employed in the method described herein is:
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof, wherein
PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; Rb is
Figure imgf000005_0001
Oligo is an oligonucleotide of from about 8 to 30 nucleotides. In a further aspect of the invention, there are provided methods of inhibiting a gene expression in a mammal for the treatment of various diseases (i.e. prostate or cervical cancer). The present invention also provides methods of making the compounds described herein.
One advantage of the polymeric transport systems described herein is that the releasable PEG-linker technology provides a method for in vivo administration of therapeutic oligonucleotides including LNA. This selective delivery technology allows enhanced therapeutic efficacy of LNA and decrease in toxicity. Another advantage is that the releasable delivery systems described herein allow for modulating of the pharmacokinetic properties of oligonucleotides. The release site of therapeutic oligonucleotides from the polymeric conjugates can be selectively targeted via EPR effect and a targeting group such as a folic acid. The oligonucleotides such as LNA attached to the polymers described herein can be released at a targeted area, such as cancer cells, thus allowing the artisan to achieve a desired bioavailability of therapeutic oligonucleotides at a targeted area. In addition, the site of release of the oligonucleotides can be modified, i.e., to release oligonucleotides in different compartments of the cells. Thus, the polymeric delivery systems described herein allow sufficient amounts of the therapeutic oligonucleotides including LNA to be selectively available at the desired target area, e.g., cytoplasm, micropinosome and endosome. The spatial modifications can be advantageous for treatment of disease. The methods described herein provide an approach for the delivery and improved efficacy of oligonucleotides (e.g., LNA oligonucleotide, siRNA) in vivo.
A further advantage of the present invention is that the conjugates described herein allow cellular uptake and specific mRNA downregulation in cancer cells in the absence of transfection agents. This is a significant advantage over prior art technologies and thus significantly simplifies treatment regimens, i.e., the in vivo administration of oligonucleotide drugs. This technology can be applied to the in vivo administration of therapeutic oligonucleotides. The polymeric compounds are stable under buffer conditions and the oligonucleotides or other therapeutic agents are not prematurely excreted from the body.
Further advantages will be apparent from the following description and drawings.
For purposes of the present invention, the term "residue" shall be understood to mean that portion of a compound to which it refers, i.e., PEG, oligonucleotide, etc. that remains after it has undergone a substitution reaction with another compound.
For purposes of the present invention, the term "polymeric residue" or "PEG residue" shall each be understood to mean that portion of the polymer or PEG which remains after it has undergone a reaction with other compounds, moieties, etc. For purposes of the present invention, the term "alkyl" as used herein refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups. The teπn "alkyl" also includes alkyl-thio-alkyl, alkoxyalkyl, cycloalkylalkyl, heterocycloalkyl, Ci-6 hydrocarbonyl, groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from about 1 to 7 carbons, yet more preferably about 1 to 4 carbons. The alkyl group can be substituted or unsubstituted. When substituted, the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, Ci^ hydrocarbonyl, aryl, and amino groups. For purposes of the present invention, the term "substituted" as used herein refers to adding or replacing one or more atoms contained within a functional group or compound with one of the moieties from the group of halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, Ci.6 hydrocarbonyl, aryl, and amino groups.
The term "alkenyl1" as used herein refers to groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkenyl group has about 2 to 12 carbons. More preferably, it is a lower alkenyl of from about 2 to 7 carbons, yet more preferably about 2 to 4 carbons. The alkenyl group can be substituted or unsubstituted. When substituted, the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxy], mercapto, hydroxy, cyano, alkylsilyl, cycloalkyi, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C|.6 hydrocarbonyl, aryl, and amino groups.
The term "alkynyl" as used herein refers to groups containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups. Preferably, the alkynyl group has about 2 to 12 carbons. More preferably, it is a lower alkynyl of from about 2 to 7 carbons, yet more preferably about 2 to 4 carbons. The alkynyl group can be substituted or unsubstituted. When substituted, the substituted group(s) preferably includes halo, oxy, azido, nitro, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxy!, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyi, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C|.6 hydrocarbony], aryl, and amino groups. Examples of "alkynyl" include propargyl, propyne, and 3-hexyne.
The term "aryl" as used herein refers to an aromatic hydrocarbon ring system containing at least one aromatic ring. The aromatic ring can optionally be fused or otherwise attached to other aromatic hydrocarbon rings or non-aromatic hydrocarbon rings. Examples of aryl groups include, for example, phenyl, naphthyl, 1 ,2,3,4-tetrahydronaphthalene and biphenyl. Preferred examples of aryl groups include phenyl and naphthyl.
The term "cycloalkyi" as used herein refers to a C3.8 cyclic hydrocarbon. Examples of cycloalkyi include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
The term "'cycloalkenyF as used herein refers to a C3.8 cyclic hydrocarbon containing at least one carbon-carbon double bond. Examples of cycloalkenyl include cyclopentenyl, cyclopentadienyl, cyclohexenyl, 1,3-cyclohexadienyl, cycloheptenyl, cycloheptatrienyl, and cyclooctenyl.
The term "cycloalkylalkyl" as used herein refers to an alkyl group substituted with a C3.8 cycloalky] group. Examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
The term '"alkoxy" as used herein refers to an alkyl group of the indicated number of carbon atoms attached to the parent molecular moiety through an oxygen bridge. Examples of alkoxy groups include, for example, methoxy, ethoxy, propoxy and isopropoxy.
An ''alkylaryl" group as used herein refers to an aryl group substituted with an alkyl group.
An ;'aralkyl" group as used herein refers to an alkyl group substituted with an aryl group. The term "alkoxyalkyl" group as used herein refers to an alkyl group substituted with an alkoxy group.
The term "alkyl-thio-alkyl" as used herein refers to an alkyl-S-alkyl thioether, for example, methylthiomethyl or mcthylthioethyl. The term '"amino" as used herein refers to a nitrogen containing group as is known in the art derived from ammonia by the replacement of one or more hydrogen radicals by organic radicals. For example, the terms "acylaminov and "alkylamino" refer to specific N-substituted organic radicals with acyl and alky] substituent groups, respectively.
The term "alkylcarbonyl" as used herein refers to a carbonyl group substituted with alkyl group.
The terms "halogen" or "halo" as used herein refer to fluorine, chlorine, bromine, and iodine.
The term "heterocycloalkyl" as used herein refers to a non-aromatic ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur. The heterocycloalkyl ring can be optionally fused to or otherwise attached to other heterocycloalkyl rings and/or non-aromatic hydrocarbon rings. Preferred heterocycloalkyl groups have from 3 to 7 members. Examples of heterocycloalkyl groups include, for example, piperazine, rnorpholine, piperidine, tetrahydrofuran, pyrrolidine, and pyrazole. Preferred heterocycloalkyl groups include piperidinyl, piperazinyl, morpholinyl, and pyrolidinyl. The term "heteroaryl" as used herein refers to an aromatic ring system containing at least one heteroatom selected from nitrogen, oxygen, and sulfur. The heteroaryl ring can be fused or otherwise attached to one or more heteroaryl rings, aromatic or non-aromatic hydrocarbon rings or heterocycloalkyl rings. Examples of heteroaryl groups include, for example, pyridine, furan, thiophene, 5,6,7, 8-tetrahydroisoquinoline and pyrimidine. Preferred examples of heteroaryl groups include thienyl, benzothienyl, pyridyl, quinolyl, pyrazinyl, pyrimidyl, imidazolyl, benzimidazolyl, furanyl, benzofuranyl, thiazolyl, benzothiazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, benzisothiazolyl, triazolyl, tetrazolyl, pyrrolyl, indolyl, pyrazolyl, and benzopyrazolyl.
The term "heteroatom" as used herein refers to nitrogen, oxygen, and sulfur. In some embodiments, substituted alkyls include carboxyalkyls, aminoalkyls, dialkylaminos, hydroxyalkyls and mercaptoalkyls; substituted alkenyls include carboxyalkenyls, aminoalkenyls, dialkenylaminos, hydroxyalkenyls and mercaptoalkenyls; substituted alkynyls include carboxyalkynyls, aminoalkynyls, dialkynylaminos, hydroxyalkynyls and mercaptoalkynyls; substituted cycloalkyls include moieties such as 4-chlorocyclohexyl; aryls include moieties such as napthyl; substituted aryls include moieties such as 3-bromo phenyl; aralkyls include moieties such as tolyl; heteroalkyls include moieties such as ethylthiophene; substituted heteroalkyls include moieties such as 3-methoxy-thiophene; alkoxy includes moieties such as methoxy; and phenoxy includes moieties such as 3-nitrophenoxy. Halo shall be understood to include fluoro, chloro, iodo and bromo.
For puiposes of the present invention, "positive integer" shall be understood to include an integer equal to or greater than 1 and as will be understood by those of ordinary skill to be within the realm of reasonableness by the artisan of ordinary skill, i.e., preferably from 1 to about 10, more preferably 1 or 2 in some embodiments.
For purposes of the present invention, the terms, "nucleic acid'" or "nucleotide" apply to deoxyribonucleic acid ("DNA") and ribonucleic acid, ("RNA'"), whether single-stranded or double-stranded, unless otherwise specified, and any chemical modifications thereof.
For purposes of the present invention, the term "'linked" shall be understood to include covalent (preferably) or noncovalent attachment of one group to another, i.e., as a result of a chemical reaction.
The terms "effective amounts" and "sufficient amounts"' for purposes of the present invention shall mean an amount which achieves a desired effect or therapeutic effect as such effect is understood by those of ordinary skill in the art.
For purposes of the present invention, the term "therapeutic oligonucleotide*' refers to an oligonucleotide used as a pharmaceutical or diagnostic agent.
For purposes of the present invention, "modulation of gene expression" shall be understood as broadly including down-regulation or up-regulation of any types of genes, preferably associated with cancer and inflammation, compared to a gene expression observed in the absence of the treatment with the compounds described herein, regardless of the route of administration.
For purposes of the present invention, '"inhibition of gene expression" of a target gene shall be understood to mean that mRNA expression or protein translated are reduced or attenuated when compared to that observed in the absence of the treatment with the compound described herein. Suitable assays include, e.g., examination of protein or mRNA levels using techniques known to those of skill in the art such as dot blots, northern blots, in situ hybridization, ELlSA, immunoprecipitation, enzyme function, as well as phenotypic assays known to those of skill in the art. The treated conditions can be confirmed by, for example, decrease in mRNA levels in cells, preferably cancer cells or tissues.
Broadly speaking, successful inhibition or treatment shall be deemed to occur when the desired response is obtained. For example, successful inhibition or treatment can be defined by obtaining e.g., 10% or higher (i.e., 20% 30%, 40%) downregulation of genes associated with tumor growth inhibition. Alternatively, successful treatment can be defined by obtaining at least 20% or preferably 30%, more preferably 40 % or higher (i.e., 50% or 80%) decrease in oncogene mRNA levels in cancer cells or tissues, including other clinical markers contemplated by the artisan in the field, when compared to that observed in the absence of the treatment with the compound described herein.
Further, the use of singular terms for convenience in description is in no way intended to be so limiting. Thus, for example, reference to a composition comprising an enzyme refers to one or more molecules of that enzyme. It is also to be understood that this invention is not limited to the particular configurations, process steps, and materials disclosed herein as such configurations, process steps, and materials may vary somewhat.
It is also to be understood that the terminology employed herein is used for the purpose of describing particular embodiments only and is not intended to be limiting, since the scope of the present invention will be limited by the appended claims and equivalents thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 schematically illustrates synthesis of compound 1 described in Example 7. FIG. 2 schematically illustrates synthesis of compound 10 described in Examples 8-13.
FIG. 3 schematically illustrates synthesis of compound 18 described in Examples 14-18.
FIG. 4 schematically illustrates synthesis of compound 25 described in Examples 19-23.
FIG. 5 schematically illustrates synthesis of compound 30 described in Examples 25-27.
FIG. 6 schematically illustrates synthesis of compound 35 described in Examples 28-29. FIG. 7 shows cellular uptake of PEG-LNA conjugates described in Examples 32. FlG. 8 shows receptor-specific cellular uptake of PEG-LNA conjugates described in Example 32.
FIG. 9 shows in vitro efficacy of PEG-LNA conjugates described in Example 33.
FlG. 10 shows in vivo efficacy and biodistribution of Folate-PEG-LNA conjugates described in Example 34.
FIG. 1 1 shows biodistribution of PEG-LNA conjugates described in Example 35.
FIG. 12 shows in vivo efficacy of PEG-LNA conjugates described in Example 36.
FIG. 13 shows in vivo efficacy of PEG-LNA conjugates described in Example 37.
For ease of the description and not limitation, multi-arm PEG (e.g., four-arm PEG) is described as "PEG" in the figures.
DETAILED DESCRIPTION OF THE INVENTION A. Overview
In one aspect of the present invention, there are provided methods of delivering oligonucleotides to tumor cells in a mammal in need thereof. The method includes administering to the mammal having tumor cells a compound of Formula (I):
Figure imgf000011_0002
or a pharmaceutically acceptable salt thereof, wherein Ri is a substantially non-antigenic water-soluble polymer; each Z| is the same or different and selected from among
Figure imgf000011_0001
Yι, in each occurrence, is independently S or O, preferably O; Y2, in each occurrence, is independently NR13, preferably NH;
Ra, in each occurrence, is the same or a different oligonucleotide; each of LM. in each occurrence, is the same or a different bifunctional linker;
Rh, in each occurrence, is a folic acid; Rc, in each occurrence, is the same or a different diagnostic agent; each of R3.7 is independently selected from among hydrogen, CV6 alkyls, C2.fi alkenyl, C?. 6 alkynyl. CM? branched alkyl, C3.X cycloalkyl, and C|.& alkoxy;
Ri3, in each occurrence, is independently selected from among hydrogen, Ci_<, alkyls, C2.6 alkenyl, C2-6 alkynyl, C3.19 branched alkyl, and C^ cycloalkyl; Ri2, in each occurrence, is independently selected from among hydrogen, hydroxyl, CK, alkyls, Cz-β alkenyl, C2.6 alkynyl, C3.1Q branched alkyl, C3-s cycloalkyl and C1.6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably 0; each of (a 1 ) and (a2) is independently zero, 1 , 2, or 3, and preferably 1 ; each (b) is independently zero, 1 , 2, or 3, and preferably 0; each (c) is independently zero, 1, 2, or 3, and preferably 1 ; each (e) is independently zero or one, and preferably 0; each (g) is independently zero or one, and preferably 1 ; and
(m) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero and provided that one or more of Z| contain an oligonucleotide.
In another aspect, compounds of Foπnula (I) are provided:
Figure imgf000012_0001
or a pharmaceutically acceptable salt thereof, wherein R| is a substantially non-antigenic water-soluble polymer; each Z| is the same or different and selected from among
Figure imgf000013_0001
Yi, in each occurrence, is independently S or O, preferably O;
Yi, in each occurrence, is independently NR13, preferably NH;
Ra, in each occurrence, is the same or a different oligonucleotide; each Of Li-4, in each occurrence, is the same or a different bifunctional linker;
Rh, in each occurrence, is a folic acid;
Rc, in each occurrence, is the same or a different diagnostic agent; each of R3/7 is independently selected from among hydrogen, Ci-6 alkyls, C2-6 alkenyl, C2. 6 alkynyl, C3.11> branched alkyl, C^.g cycloalkyl, and C|.fi alkoxy;
R 13, in each occurrence, is independently selected from among hydrogen, Ci-C alkyls, C2.& alkenyl, C2-6 alkynyl, C3.19 branched alkyl, arid C3.8 cycloalkyl;
R ι2, in each occurrence, is independently selected from among hydrogen, hydroxyl, C^6 alkyls, C2-6 alkenyl, C2-6 alkynyl, C.vj<> branched alkyl, C3.8 cycloalkyl and Cι-6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably O; each of (al ) and (a2) is independently zero, 1, 2, or 3, and preferably 1 ; each (b) is independently zero, 1 , 2, or 3, and preferably O; each (c) is independently zero, 1 , 2, or 3, and preferably 1 ; each (e) is independently zero or one, arid preferably O; each (g) is independently zero or one, and preferably 1 ; and
(m) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero, that one or more of Zi contain an oligonucleotide, and that one or more of Zi contain a folic acid. In one embodiment, one Z| contains an oligonucleotide and the remaining Z| contains a folic acid.
For purposes of the present invention, (m) refers to the number of polymer aπns. Each polymer arm includes a linear polymer such as polyethylene glycol. Preferably, (m) equals to from about 2 to about 32. For example, (m) is 32 when R| has 32 linear polymer amis. When (m) is 2, bisPEG is employed in the polymeric compounds described herein. Thus, the polymeric compounds can preferably include up to 32 polymer arms, i.e., 4, 8, 16 or 32. In one embodiment, the polymeric compounds can include four to eight polymer arms, where (m) can be from 4 to 8 (e.g., 4, 6 or 8). Preferably, the polymeric portion includes four polymer arms, where (m) is 4.
For purposes of the present invention, Li is the same or different when (a) is equal to or greater than 2.
For purposes of the present invention, L2 is the same or different when (d) is equal to or greater than 2. For purposes of the present invention, L4 is the same or different when (al ) or (a2) is equal to or greater than 2.
For purposes of the present invention, C(R4)(Rs) is the same or different when (b) is equal to or greater than 2.
For puiposes of the present invention, C(Ro)(R7) is the same or different when (c) is equal to or greater than 2.
In one embodiment, the tumor cells are prostate or cervical cancer cells. In another aspect, the present invention provides a method of inhibiting a gene expression in mammalian cells or tissues. The method includes administering an effective amount of the compound of Formula (I) or a pharmaceutically acceptable salt thereof to a mammal in need thereof.
In one embodiment, the methods described herein are carried out using a compound of Formula (I*):
Figure imgf000015_0001
wherein
(m 1 ) is a positive integer from about 1 to about 8 (e.g., 1 , 2, 3, 4, 5, 6, 7, 8); (m2) is zero or a positive integer from about 1 to about 7 (e.g. 0, 1 , 2, 3, 4, 5, 6, 7); and the sum of (m l ) and (m2) is an integer from about 2 to about 8 (e.g., 2, 4, 6, 8). In one particular embodiment, all (Z|) contain an oligonucleotide. In this aspect, (m2) is zero and (ml ) is 4 or 8. Alternatively, all (Zi) are the same or different
Figure imgf000015_0002
• In another particular embodiment, one or more of Z| contain a folic acid. Alternatively, one or more Z\ are
Figure imgf000015_0003
In this aspect, one Z| includes an oligonucleotide and each of the remaining Zi includes a folic acid, when (m) is greater than 2.
In a further embodiment, the compounds described herein include an optional diagnostic agent.
In another embodiment, each Ri2 is the same or different groups selected from among H, NH2, OH, CO2H, C,.6 alkoxy, C6 alkyl, and preferably OH.
In yet another embodiment, each of R3.7 is the same or different group selected from among hydrogen, methyl, ethyl and isopropyl. Preferably, R3.7 are all hydrogen In one preferred embodiment, (b), (d) and (e) are zero and (c) is 1. In one preferred embodiment, the compounds of Formula (I) employed in the method described herein include Zi having the foπnula:
Figure imgf000016_0001
wherein, (a) is 0 or 1 ;
(in) is 1 , 2, 4, 8, 16 or 32;
Ri2, in each occurrence, is independently selected from among hydroxyl, Ci.6 alkyls, Ci-r, alkenyl, CYή alkynyl, C3.1i) branched alkyl, and C|_6 alkoxy; and all other variables are the same as defined above. Alternatively, the compounds described herein have the formula:
Figure imgf000016_0002
wherein (a) is 0 or 1. In this respect, (mj) is zero and all of Zi are
Figure imgf000016_0003
(mi) is I , or one Zi is
Figure imgf000016_0004
wherein each of remaining Z| includes a folic acid. In a further embodiment, one Zi includes a diagnostic agent.
In another aspect, Ri includes a polyalkylene oxide. Preferably, Ri has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20, 000 to about 45,000 daltons. In one preferred aspect of the present invention, the methods described herein are conducted with the compounds having the formula:
Figure imgf000017_0001
wherein each Z is independently
Figure imgf000017_0002
wherein (a) is O or 1 . Ri2, in each occurrence, is independently selected from among hydroxyl, Cι_6 alkyls, C2-b alkenyl, C2.ή alkynyl, C3-m branched alkyl, and Ci-6 alkoxy; (n) is a positive integer and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; and all other variables are the same as defined above.
In this respect, all Z groups include an oligonucleotide. Alternatively, one Z includes an oligonucleotide and remaining one or more Z groups (e.g., 1 , 2, 3, 4, 5, 6 or 7 Z groups) include a targeting agent such as folic acid. In a further embodiment, one Z includes an oligonucleotide, another Z includes a diagnostic agent, and remaining one or more (e.g., 2, 3, 4, 5, 6) Z include a folic acid.
In another aspect of the present invention, there are provided compounds of Formula (Ia):
Figure imgf000018_0002
or a pharmaceutically acceptable salt thereof, wherein
R i is a substantially non-antigenic water-soluble polymer; each Z| is the same or different and selected from among
Figure imgf000018_0001
Y i, in each occurrence, is independently S or O; Yi, in each occurrence, is independently NRi 3; Rj, in each occurrence, is the same or a different oligonucleotide; each of LM, in each occurrence, is the same or a different bi functional linker; Rb. in each occurrence, is a folic acid; Rc, in each occurrence, is the same or a different diagnostic agent; each of R3.7 is independently selected from among hydrogen, C|-f) alkyls, C2.6 alkenyl, C2. ft alkynyl, C3.19 branched alkyl, CYs cycloalkyl, and C|.f, alkoxy;
R1J, in each occurrence, is independently selected from among hydrogen, Cj.ή alkyls, C1.6 alkenyl, Ci-is alkynyl, C3.19 branched alkyl, and CYs cycloalkyl; R12, in each occurrence, is independently selected from among hydrogen, hydroxyl, Cu, alkyls, C2-6 alkenyl, Cj.*, alkynyl, Cv 10 branched alkyl, C3.X cycloalkyl and Cι-6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3; each of (al ) and (a2) is independently zero, 1 , 2, or 3; each (b) is independently zero, 1 , 2, or 3; each (c) is independently zero, 1 , 2, or 3; each (e) is independently zero or one; each (g) is independently zero or one; and
(111) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero, that one or more of Z| contain an oligonucleotide, and that one or more of Zi contain a folic acid.
In certain embodiments, variables are the same as defined in Fomula (1).
B. Substantially Non-antigenic Water-soluble Polymers
Polymers employed in the compounds described herein are preferably water soluble polymers and substantially non-antigenic such as polyalkylene oxides (PAO's).
In one aspect of the invention, the compounds described herein include a linear, terminally branched or multi-armed polyalkylene oxide. In some embodiments of the invention, the polyalkylene oxide includes polyethylene glycol and polypropylene glycol.
The polyalkylene oxide has the total number average molecular weight of from about 2,000 to about 100,000 daltons, preferably from about 5,000 to about 60,000 daltons. The polyalkylene oxide can be more preferably from about 5,000 to about 25,000 or from about 20,000 to about 45,000 daltons. In some particular embodiments, the compounds described herein include the polyalkylene oxide having the total number average molecular weight of from about 30,000 to about 45,000 daltons. In one particular embodiment, the polymeric portion has the total number average molecular weight of about 40,000 daltons. The polyalkylcne oxide includes polyethylene glycols and polypropylene glycols. More preferably, the polyalkylene oxide includes polyethylene glycol (PEG). PEG is generally represented by the structure:
Figure imgf000020_0003
where (n) represents the degree of polymerization for the polymer, and is dependent on the molecular weight of the polymer. Alternatively, the polyethylene glycol (PEG) residue portion of the compounds described herein can be selected from among:
Figure imgf000020_0002
wherein:
Y71 and Y73 are independently O, S, SO, SO2, NR73 or a bond; Y72 is O, S, or NR74, preferably O;
R71, R72, R73 and R74 are independently selected from the same moieties which can be used for R3;
(a71 ), (a72), and (b71) are independently zero or a positive integer (e.g. O. 1 , 2, 3), and preferably 1 ; and (n) is an integer from about 10 to about 2300.
In one preferred aspect, the polymers useful in the compounds described herein include multi-arm PEG-OH or "star-PEG"' products such as those described in NOF Corp. Drug Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference. The polymers can be converted into suitably activated forms, using the activation techniques described in U.S. Patent Nos. 5,122,614 or 5,808,096. Specifically, such a PEG can be of the formula:
Figure imgf000020_0001
or
Figure imgf000021_0001
wherein:
(u') is an integer from about 4 to about 455.
In one preferred embodiment, the degree of polymerization for the polymer (u') is from about 28 to about 341 to provide polymers having the total number average molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from about 1 14 to about 239 to provide polymers having the total number average molecular weight of from about 20,000 Da to about 42,000 Da. (u') represents the number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer. In one particular embodiment of the invention, (u") is about 227 to provide the polymeric portion having the total number average molecular weight of about 40,000 Da.
In some preferred embodiments, all 4 of the PEG arms are converted to suitable activating groups, for facilitating attachment to oligonucleotides or folic acids. Such compounds prior to conversion include:
Figure imgf000021_0002
and
Figure imgf000021_0003
The polymeric substances included herein are preferably water-soluble at room temperature. A non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
For purposes of the present invention, "substantially or effectively non-antigenic" means all materials understood in the art as being nontoxic and not eliciting an appreciable immunogenic response in mammals. In some aspects, polymers having terminal carboxylic acid groups can be employed in the polymeric delivery systems described herein. Methods of preparing polymers having terminal carboxylic acids in high purity are described in U.S. Patent Application Publication No. 2007/0173615, the contents of which are incorporated herein by reference. The methods include first preparing a tertiary alkyl ester of a polyalkylene oxide followed by conversion to the carboxylic acid derivative thereof. The first step of the preparation of the PAO carboxylic acids of the process includes forming an intermediate such as /-butyl ester of polyalkylene oxide carboxylic acid. This intermediate is formed by reacting a PAO with a /-butyl haloacetate in the presence of a base such as potassium /-butoxide. Once the /-butyl ester intermediate has been formed, the carboxylic acid derivative of the polyalkylene oxide can be readily provided in purities exceeding 92%, preferably exceeding 97%, more preferably exceeding 99% and most preferably exceeding 99.5% purity.
In alternative aspects, polymers having terminal amine groups can be employed to make the compounds described herein. The methods of preparing polymers containing terminal amines in high purity are described in U.S. Patent Application Publication Nos. 2008/0249260 and 2007/0078219, the contents of each of which are incorporated by reference. For example, polymers having azides react with a phosphine-based reducing agent such as triphenylphosphine or an alkali metal borohydride reducing agent such as NaBH4. Alternatively, polymers including leaving groups react with protected amine salts such as potassium salt of methyl-tert-butyl imidodicarbonate (KNMeBoc) or the potassium salt of di-tert-butyl iinidodicarbonate (KNBoC2) followed by deprotecting the protected amine group. The purity of the polymers containing the terminal amines formed by these processes is greater than about 95% and preferably greater than 99%.
C. Bifunctioiial Linkers Bifunctional linkers include amino acids, amino acid derivatives, and peptides. The amino acids can be among naturally occurring and non-naturally occurring amino acids. Derivatives and analogs of the naturally occurring amino acids, as well as various art-known non-naturally occurring amino acids (D or L), hydrophobic or non-hydrophobic, are also contemplated to be within the scope of the invention. A suitable non-limiting list of the non- naturally occurring amino acids includes 2-aininoadipic acid, 3-aminoadipic acid, beta-alanine, beta-aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, piperidinic acid,
6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2- aminopimelic acid, 2,4-aminobutyric acid, desmosine, 2,2-diaminopinielic acid, 2,3-diaminopropiυnic acid, N-elhylglycine, N-ethylasparagine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, alloisoleucine, N-inethylglycine, sarcosine, N-methyl- isolcucine, 6-N-methyllysine, N-methylvaline, norvaline, norleucine, and ornithine. Some amino acid residues are selected from glycine, alanine, methionine or sarcosine, and more preferably, glycine.
In an alternative aspect of the present invention, LM are the same or different groups selected from among:
Figure imgf000024_0001
Figure imgf000025_0001
wherein:
R2I-2(J are the same or different groups selected from among hydrogen, C|.6 alkyls, CY12 branched alkyls, C3.8 cycloalkyls, C |.ή substituted alkyls, C6-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls. C|.ή heteroalkyls, substituted C|-6 heteroalkyls, CVδ alkoxy, phenoxy and C|.6heteroalkoxy;
(t) and (O are independently zero or a positive integer, preferably zero or an integer from about 1 to about 12, more preferably an integer from about 1 to about 8, and most preferably 1 or 2; and (v) and (v") are independently zero or 1.
In some preferred embodiments, L(.4 of Fomula (I) and Formula (Ia) (more preferably, L4 of Formula (Ia)) include the same or different groups selected from among:
Figure imgf000025_0002
Figure imgf000026_0001
wherein (r) and (r" ) are independently zero or 1 ; and (s) and (s") are independently 1 , 2, or 3. Both (r) and (r") are not zero simultaneously.
For purposes of the present invention, when values for bifunctional linkers are positive integers equal to or greater than 2, the same or different bifunctional linkers can be employed. In one embodiment containing two or more bifunctional linkers, where (a), (al ), and (a2) are equal to or greater than 2, the bifunctional linkers can be the same or different.
D. Diagnostic Agents A further aspect of the invention provides the compounds optionally prepared with a diagnostic tag linked to the polymeric delivery system described herein, wherein the tag is selected for diagnostic or imaging purposes.
The compounds described herein can be labeled such as biotinylated compounds, fluorescent compounds (e.g., FAM), radiolab led compounds. A suitable tag is prepared by linking any suitable moiety, e.g., an amino acid residue, to any art-standard emitting isotope, radio-opaque label, magnetic resonance label, or other non-radioactive isotopic labels suitable for magnetic resonance imaging, fluorescence-type labels, labels exhibiting visible colors and/or capable of fluorescing under ultraviolet, infrared or electrochemical stimulation, to allow for imaging tumor tissue during surgical procedures, and so forth. Optionally, the diagnostic tag is incorporated into and/or linked to a conjugated therapeutic moiety, allowing for monitoring of the distribution of a therapeutic biologically active material within an animal or human patient.
The tagged compounds are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels. Simply by way of example, these include 1 jl Iodine, 125lodine, ""Technetium and/or ' ' 'indium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo. For instance, there are a number of art-known methods of linking peptide to Tc-99m, including, simply by way of example, those shown by U.S. Patent Nos. 5,328,679; 5,888,474; 5,997,844; and 5,997,845, incorporated by reference herein.
E. Oligonucleotides
The compounds described herein can be used for delivering nucleic acids (oligonucleotides) into cells or tissues.
In order to more fully appreciate the scope of the present invention, the following terms are defined. The artisan will appreciate that the terms. "nucleic acid" or "nucleotide" apply to deoxyribonucleic acid ("DNA"). ribonucleic acid, ("RNA) whether single-stranded or double- stranded, unless otherwise specified, and any chemical modifications thereof. An "oligonucleotide" is generally a relatively short polynucleotide, e.g., ranging in size from about 2 to about 200 nucleotides, or more preferably from about 8 to about 30 nucleotides in length. The oligonucleotides according to the invention are generally synthetic nucleic acids, and are single stranded, unless otherwise specified. The terms, "polynucleotide" and '"polynucleic acid" may also be used synonymously herein. The oligonucleotides (analogs) are not limited to a single species of oligonucleotide but, instead, are designed to work with a wide variety of such moieties, it being understood that linkers can attach to one or more of the 3'- or 5'- terminals, usually PO4 or SO4 groups of a nucleotide. The nucleic acids molecules contemplated can include a phosphorothioate internucleotide linkage modification, sugar modification, nucleic acid base modification and/or phosphate backbone modification. The oligonucleotides can contain natural phosphorodiester backbone or phosphorothioate backbone or any other modified backbone analogs such as LNA (Locked Nucleic Acid), PNA (nucleic acid with peptide backbone), CpG oligomers, and the like, such as those disclosed at Tides 2002, Oligonucleotide and Peptide Technology Conferences, May 6-8, 2002, Las Vegas, NV and Oligonucleotide & Peptide Technologies, 18th & 19th
November 2003, Hamburg, Germany, the contents of which are incorporated herein by reference. Modifications to the oligonucleotides contemplated by the invention include, for example, the addition to or substitution of selected nucleotides with functional groups or moieties that permit covalent linkage of an oligonucleotide to a desirable polymer, and/or the addition or substitution of functional moieties that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and functionality to an oligonucleotide. Such modifications include, but are not limited to, 2'-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodouracil, backbone modifications, methylations, base-pairing combinations such as the isobases isocytidine and isoguanidine, and analogous combinations. Oligonucleotides contemplated within the scope of the present invention can also include 3' and/or 5' cap structure.
For purposes of the present invention, "cap structure'" shall be understood to mean chemical modifications, which have been incorporated at either teπninus of the oligonucleotide. The cap can be present at the 5'-teπninus (5'-cap) or at the 3'-terminus (3'-cap) or can be present on both terminus. A non-limiting examples of the 5'-cap includes inverted abasic residue (moiety), 4',5'-methylene nucleotide; 1 -(beta-D-erythrofuranosyl) nucleotide, 4'-thio nucleotide, carbocyclic nucleotide; 1 ,5-anhydrohexitol nucleotide; L-nucleotides; alpha- nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3',4'-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide, 3'-3'-inverted nucleotide moiety; 3'-3'-invcrted abasic moiety;
3'-2'-inverted nucleotide moiety; 3'-2'-inverted abasic moiety; 1 ,4-butanediol phosphate;
3'-phosphoramidate; hexylphosphate; aminohexyl phosphate; 3'-phosphate;
3'-phosphorothioate; phosphorodithioate; or bridging ov non-bridging methylphosphonate moiety. Details are described in WO 97/26270, incorporated by reference herein. The 3'-caρ can includes, for example, 4',5'-methylene nucleotide; l -(beta-D-erythrofuranosyl) nucleotide: 4'-thio nucleotide, carbocyclic nucleotide; 5'-amino-alkyl phosphate; l ,3-diamino-2-propyl phosphate,
3-aminopropyl phosphate; 6-aminohexyl phosphate; 1 ,2-aminododecyl phosphate; hydroxypropyl phosphate; 1 ,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo-pentofuranosyl nucleotide; acyclic
3',4'-seco nucleotide; 3,4-dihydroxybutyI nucleotide; 3,5-dihydroxypentyl nucleotide,
5'-5'-inverted nucleotide moiety; 5'-5'-invei1ed abasic moiety; 5'-phosphoramidate;
5'-phosphorothioate; 1 ,4-butanediol phosphate; 5 '-amino; bridging and/or non-bridging
5'-phosphoramidate, phosphorothioate and/or phosphorodithioate, bridging or non bridging methylphosphonate and 5'-mercapto moieties. See also Beaucage and Iyer, 1993, Tetrahedron
49, 1925; the contents of which are incorporated by reference herein. A non-limiting list of nucleoside analogs has the structure:
Figure imgf000029_0001
Figure imgf000030_0001
See more examples of nucleoside analogs described in Freier & Altmann; Nucl Acid Res., 1997, 25, 4429-4443 and Uhlmann; Cun: Opinion in Drug Development, 2000, 3(2), 293-213, the contents of each of which are incorporated herein by reference.
The term "anti sense,'" as used herein, refers Io nucleotide sequences which are complementary to a specific DNA or RNA sequence that encodes a gene product or -that encodes a control sequence. The term "antisense strand'" is used in reference to a nucleic acid strand that is complementary to the "'sense" strand. In the normal operation of cellular metabolism, the sense strand of a DNA molecule is the strand that encodes polypeptides and/or other gene products. The sense strand serves as a template for synthesis of a messenger RNA ("mRNA'") transcript (an antisense strand) which, in turn, directs synthesis of any encoded gene product. Antisense nucleic acid molecules may be produced by any art-known methods, including synthesis by Iigating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated. The designations "negative" or (-) are also art-known to refer to the antisense strand, and "positive" or (+) are also art-known to refer to the sense strand.
For purposes of the present invention, "complementary" shall be understood to mean that a nucleic acid sequence forms hydrogen bond(s) with another nucleic acid sequence. A percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule which can form hydrogen bonds, i.e., Watson-Crick base pairing, with a second nucleic acid sequence, i.e.. 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary. "Perfectly complementary" means that all the contiguous residues of a nucleic acid sequence form hydrogen bonds with the same number of contiguous residues in a second nucleic acid sequence.
In one embodiment, the choice for conjugation is an oligonucleotide (or "polynucleotide"') and after conjugation, the target is referred to as a residue of an oligonucleotide. The oligonucleotides can be selected from among any of the known oligonucleotides and oligodeoxynucleotides with phosphorodiester backbones or phosphorothioate backbones.
The oligonucleotides or oligonucloetide derivatives useful in the compounds described herein can include from about 8 to about 1000 nucleic acids, and preferably relatively short polynucleotides, e.g., ranging in size preferably from about 8 to about 30 nucleotides in length (e.g., about 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30).
In one aspect of useful nucleic acids used in the method described herein, oligonucleotides and oligodeoxynucleotides with natural phosphorodiester backbone or phosphorothioate backbone or any other modified backbone analogues include:
LNA (Locked Nucleic Acid); PNA (nucleic acid with peptide backbone); short interfering RNA (siRNA); microRNA (miRNA); nucleic acid with peptide backbone (PNA); phosphorodiamidate morpholino oligonucleotides (PMO); tricyclo-DNA: decoy ODN (double stranded oligonucleotide); catalytic RNA sequence (RNAi); ribozymes; aptamers; spiegelmers (L-conformational oligonucleotides);
CpG oligomers, and the like, such as those disclosed at:
Tides 2002, Oligonucleotide and Peptide Technology Conferences, May 6-8, 2002, Las Vegas, NV and Oligonucleotide & Peptide Technologies, 18th & 19th November 2003, Hamburg, Germany, the contents of which are incorporated herein by reference.
In another aspect of the nucleic acids used in the method described herein, oligonucleotides can include any suitable art-known nucleotide analogs and derivatives, including those listed by Table 1 , below: TABLE 1. Representative Nucleotide Analogs And Derivatives
Figure imgf000032_0001
Figure imgf000033_0001
Preferably, the oligonucleotide is involved in targeted tumor cells or downregulating a protein implicated in the resistance of tumor cells to anticancer therapeutics. For example, any art-known cellular proteins such as BCL-2 for downregulation by antisense oligonucleotides, for cancer therapy, can be used for the present invention. See U.S. Patent Application No.
10/822,205 filed April 9, 2004, the contents of which are incoiporated by reference herein. A non-limiting list of therapeutic oligonucleotides includes antisense HIF- l α oligonucleotides, antisense ErbB3 oligonucleotides, antisense survivin oligonucleotides and β-catenine oligonucleotides. Preferably, the oligonucleotides useful in the method described herein include phosphorothioate backbone and LNA.
In one embodiment, the oligonucleotide useful in the method described herein includes antisense bcl-2 oligonucleotides, antisense HIF- lα oligonucleotides, antisense survivin oligonucleotides, and antisense Erbβ3 oligonucleotides. In one preferred embodiment, the oligonucleotide can be, for example, an oligonucleotide that has the same or substantially similar nucleotide sequence as does Genasense (a/k/a oblimersen sodium, produced by Genta Inc., Berkeley Heights, NJ). Genasense is an 18-nier phosphorothioate antisense oligonucleotide, TCTCCCAGCGTGCGCCAT (SEQ ID NO: 4), that is complementary to the first six codons of the initiating sequence of the human bcl-2 mRNA (human bcl-2 mRNA is art-known, and is described, e.g., as SF.Q ID NO: 19 in U.S. Patent No. 6,414,134, incorporated by reference herein). The U.S. Food and Drug Administration (FDA) gave Genasense Orphan Drug status in August 2000. Preferred embodiments include: Preferred embodiments contemplated include: (i) antisense Survivin LNA oligonucleotide (SEQ ID NO: 1 ) mC3-Ts-mC3-As-as-ts-cs-cs-as-ts-gs-gs-mC3-As-Gs-c; where the upper case letter represents LNA. the "s" represents a phosphorothioate backbone; (ii) antisense Bc!2 siRNA:
SENSE 5"- gcaugcggccucuguuugadTdT-31 (SEQ ID NO: 2) ANTISENSE 3'- dTdTcguacgccggagacaaacu-5' (SEQ ID NO: 3) where dT represents DNA; (iii) Genasense (phosphorothioate antisense oligonucleotide): (SEQ ID NO: 4) ts-Cg-tg-Cs.Cs-Cs-as-gs-Cs-gs-tg-gs-Cs-gs-Cs-Cs-Cs-as-t where the lower case letter represents DNA and "s" represents phosphorothioate backbone;
(iv) antisense HIFl α LNA oligonucleotide (SEQ ID NO: 5)
Figure imgf000034_0002
where the upper case letter represents LNA and the "s" represents phosphorothioate backbone. (v) antisense ErbB3 LNA oligonucleotide (SEQ ID NO: 6)
Figure imgf000034_0003
where the upper case letter represents LNA and the "s" represents phosphorothioate backbone. LNA includes 2"-O, 4%-C methylene bicyclonucleotide as shown below:
Figure imgf000034_0001
See detailed description of Survivin LNA disclosed in U.S. Patent Application Publication Serial Nos. 2006/0154888 and 2005/0014712, the contents of each of which is incorporated herein by reference. See detailed description of HIF-lα LNA disclosed in U.S. Patent Application Publication Nos. 2004/0096848, and 2006/0252721 , the contents of each of which are incorporated herein by reference in its entirety. See also WO2008/1 13832, the contents of which are incorporated herein by reference in its entirety.
In one particular embodiment, the oligonucleotide comprises SEQ ID NO: 1 , SEQ ID NOs 2 and 3, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6. The oligonucleotides prior to the conjugation to the polymeric systems described herein include (CHT)* sulfhydryl linkers (thio oligonucleotides) at 5" or 3" end of the oligonucleotides, where (w) in this aspect is a positive integer of from about 1 to about 10 (1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and preferably 4, 5, 6 or 7. The thio oligonucleotides have the structure SH-(CH2Xv- Oligonucleotide. The compounds described herein can include oligonucleotides modified with hindered ester-containing (CH2)tt sulfhydryl linkers. Prior to the attachment, the oligonucleotide has the structure:
Figure imgf000035_0001
wherein (w) is a positive integer from about 1 to about 10, (e.g., 3, 4, 5, 6). See WO 2008/034] 19, the contents of which are incorporated by reference. The polymeric compounds can release the oligonucleotides without thiol tail.
In one particular embodiment, 5' end of the sense strand of siRNA is modified. For example, siRNA employed in the polymeric conjugates is modified with a 5'-CO-SH. One particular embodiment of the present invention employs Bcl2-siRNA having the sequence of SENSE 5'-SH-C6-GCAUGCGGCCUCUGUUUGAdTdT-3'
ANTISENSE 3'- dTdTCGUACGCCGGAGACAAACU-51. Examples of the modified oligonucleotides include:
Figure imgf000035_0002
Figure imgf000036_0001
F. Synthesis of the Polymeric Delivery Systems
Generally, the methods of preparing compounds described herein include reacting an activated polymer with an oligonucleotide modified with a SH group. Activated polymers useful in the methods described herein include a polymer containing a pyridyl disulfide group at the distal end. The methods provide a polymeric conjugate where the biologically active moiety is bonded to the polymer through -S-S- bond.
In one aspect of the invention, methods of preparing polymeric compounds described herein include: reacting a polymeric compound of Formula (111):
Figure imgf000036_0003
with an olignucleotide modified with a sulfhydryl group-containing moiety under conditions sufficient to form a compound of Formula (1):
Figure imgf000036_0004
wherein
Ri is a substantially non-antigenic water-soluble polymer; each Z| i is the same or different and selected from among
Figure imgf000036_0002
Figure imgf000037_0001
Yi, in each occurrence, is independently S or O, preferably O; Y2, in each occurrence, is independently NRn, preferably NH; R 100, in each occurrence is the same or different and selected from among H, a leaving group, an activating group, and
Figure imgf000037_0002
each of LM, in each occurrence, is the same or a different bifunctional linker; Rb, in each occurrence, is a folic acid; Rc, in each occurrence, is the same or a different diagnostic agent; each of R3.7 is independently selected from among hydrogen, C|.ή alkyls, C2-6 alkenyl, C2. ή alkyny], C3.19 branched alkyl, C3.8 cycloalkyl, and C|.ft alkoxy; each of Rjj-i 1 is an electron-withdrawing group such as substituted amido, acyl, azido, carboxy, alkyloxycarbonyl, cyano, and nitro, and preferably Rs is nitro, and RQ, RIO and Rn are hydrogen;
R 13, in each occurrence, is independently selected from the group consisting of hydrogen, C|.f, alkyls, C2-6 alkenyl, C2^, alkynyl, C3.19 branched alkyl, and C3.8 cycloalkyl;
Ri2, in each occurrence, is independently selected from the group consisting of hydrogen, hydroxy], C|.& alkyls, C:-G alkenyl, C2-O alkynyl, C3.19 branched alkyl, d.s cycloalkyl and Ci .6 alkoxy; each of (a) and (d) is independently zero, 1 , 2, or 3, and preferably 0; each of (al) and (a2) is independently zero, 1 , 2, or 3, and preferably 1 ; each (b) is independently zero, 1, 2, or 3, and preferably 0; each (c) is independently zero, 1 , 2, or 3, and preferably 1 ; each (e) is independently zero or one, and preferably 0; each (g) is independently zero or one, and preferably 1 ; and
(m 1 1 ) is a positive integer from about 2 to about 32 (e.g., 2, 4, 6, 8, 16, 32), provided that (a) and (g) are not simultaneously zero and provided that one or more of Zi i contain
Figure imgf000038_0001
Preferably, the reactions are carried out in an inert solvent such as methylene chloride, chloroform, DMF or mixtures thereof. The reactions can be preferably conducted in the presence of a base, such as dimethylaminopyridine (DMAP), diisopropylethylamine, pyridine, tri ethyl amine, etc. to neutralize any acids generated. The reactions can be carried out at a temperature from about 0 0C up to about 22 0C (room temperature). See detailed description in WO/2009/025669, the contents of which are incorporated herein by reference.
G. Compounds of Formula (I)
Some particular embodiments prepared by the methods described herein have the structure:
Figure imgf000038_0002
Figure imgf000039_0001
Figure imgf000040_0001
wherein:
Oligo is an oligonucleotide, preferably an oligonucleotide modified with a C.i-Q, alkyl (C6 alkyl); PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; (al ) is one; is zero or one;
Figure imgf000040_0003
and ' all other variables are the same as defined above.
For example, compounds prepared by the method described herein include
Figure imgf000040_0002
Figure imgf000041_0001
Oligo is an oligonucleotide;
Rb is
Figure imgf000041_0002
PEG is polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons.
In a further embodiment, compounds prepared by the methods described herein include:
Figure imgf000041_0003
Figure imgf000042_0001
Figure imgf000043_0001
wherein
Oligo is an oligonucleotide, preferably an oligonucleotide modified with a C3-C6 alkyl (e.g., C6 alkyl); and
PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons. For ease of the description and not limitation, the multi-arm PEG is shown as "PEG". One arm, up to seven arms of the eight-arm PEG (or up to three arms of the four-arm PEG) can be conjugated with a folic acid.
Preferably, the compounds include therapeutic oligonucleotides such as antisense ErbB3 oligonucleotides and antisense Survivin oligonucleotides. For example, the compounds include a C(,-tail modified antisense LNA as follows:
-5'-(CH2)6-TsAsGsCsCsTsGsTs CsAsCsTsTsCsTsCs-3' or
-5'-(CH2)6-GsCsTsGsCsCsAsTsGsGsAsTsTsGsAsG -3\ wherein "s*1 represents a phosphorothioate linkage and the first three nucleotides in 5' and 3" terminal are LNA.
Preferably, the average molecular weight of the polymeric portion is about 40,000 daltons.
H. METHODS OF TREATMENT One aspect of the present invention provides methods of introducing or delivering an oligonucleotide into a mammalian cell. The method according to the present invention includes contacting a cell with a compound of Formula (I) described herein.
The present invention is useful for introducing oligonucleotides to a mammal having tumor cells. The compounds described herein can be administered to a mammal, preferably human. According to the present invention, the present invention preferably provides methods of inhibiting or downregulating (or modulating) a gene expression in mammalian cells or tissues. The downregulation or inhibition of gene expression can be achieved in vivo and/or in vitro. The methods include contacting human cells or tissues with compounds of Formula (I) described herein. Once the contacting has occurred, successful inhibition or down-regulation of gene expression such as in mRNA or protein levels shall be deemed to occur when at least about 10%, preferably at least about 20% or higher is realized when measured in vivo or in vitro, when compared to that observed in the absence of the treatment with the compound described herein.
For purposes of the present invention, "inhibiting" or "down-regulating" shall be understood to mean that the expression of a target gene, or level of RNAs or equivalent RNAs encoding one or more protein subunits, or activity of one or more protein subunits, such as ErbB3, is reduced below that observed in the absence of the treatment with the conjugates described herein.
Preferably, gene expression of a target gene is inhibited in prostate or cervical cancer cells or tissues, for example, prostate or cervical cancer cells.
In a further embodiment, the cancer cells or tissues can be from one or more of the following: solid tumors, lymphomas, small cell lung cancer, acute lymphocytic leukemia (ALL), pancreatic cancer, glioblastoma, ovarian cancer, gastric cancer, breast cancer, colorectal cancer, ovarian cancer and brain tumors, etc. In one particular embodiment, the compounds according to the methods described herein include, for example, antiscnse bcl-2 oligonucleotides, antisense HIF-I α oligonucleotides, antisense Survivin oligonucleotides, and antisense Erbβ3 oligonucleotides.
Preferably, the administering step is via the blood stream of the mammal.
A further aspect of the present invention provides methods of treatment for various medical conditions in mammals. The methods include administering, to the mammal in need of such treatment, an effective amount of a pharmaceutical composition containing a compound of Formula (I). The polymeric conjugate compounds are useful for, among other things, treating diseases including, but not limited to, cancer, inflammatory disease, and autoimmune disease.
In this aspect, a useful target gene includes, but is not limited to, oncogenes, pro- angiogenesis pathway genes, pro-cell proliferation pathway genes, viral infectious agent genes, and pro-inflammatory pathway genes. In yet a further aspect, there are also provided methods of treating a patient having a malignancy or cancer, comprising administering an effective amount of a pharmaceutical composition containing the compound of Formula (I) to a patient in need thereof. In alternative aspects, the cancer being treated can be one or more of the following: solid tumors, lymphomas, small cell lung cancer, acute lymphocytic leukemia (ALL), pancreatic cancer, glioblastoma, ovarian cancer, gastric cancers, colorectal cancer, etc. The compositions are useful for treating neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mammals by downregulating gene expression of a target gene. Any oligonucleotide, etc. which has therapeutic effects in the unconjugated state can be used in its conjugated foπn, made as described herein.
In one particular embodiment, the methods described herein include administering polynucleotides (oligonucleotides), preferably antisense oligonucleotides to mammalian cells. The methods include delivering an effective amount of a conjugate prepared as described herein to the condition being treated will depend upon the polynucleotides efficacy for such conditions.
For example, if the unconjugated oligonucleotides (for example antisense ErbB3 oligonucleotides, antisense Survivin oligonucleotides) has efficacy against certain cancer or neoplastic cells, the method would include delivering a polymer conjugate containing the oligonucleotides to the cells having susceptibility to the native oligonucleotides. The delivery can be made in vivo as part of a suitable pharmaceutical composition or directly to the cells in an ex vivo environment. In one particular treatment, the polymeric conjugates including oligonucleotides (SEQ ID NO. 1 , SEQ ID NOs: 2 and 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6) can be used.
In yet another aspect, the present invention provides methods of inhibiting the growth or proliferation of cancer cells in vivo or in vitro. The methods include contacting cancer cells with the compounds described herein. Alternatively, the present invention provides methods of modulating apoptosis in cancer cells by administering the compounds described herein to a mammal in need thereof.
In yet another aspect, there are also provided methods of increasing the sensitivity of cancer cells or tissues to chemotherapeutic agents in vivo or in vitro. In one particular aspect, the methods include introducing the oligonucleotide (antisense LNA) conjugates described herein to cancer cells to reduce survivin expression in the cancer cells or tissues, wherein the antisense oligonucleotide binds to niRNA expressed from the survivin gene and reduces survivin gene expression.
In yet another aspect, there are provided methods of killing tumor cells in vivo or in vitro. The methods include introducing the compounds described herein to tumor cells to reduce gene expression such as ErbB3 and contacting the tumor cells with an amount of at least one chemotherapeutic agent sufficient to kill a portion of the tumor cells. Thus, the portion of tumor cells killed can be greater than the portion which would have been killed by the same amount of the chemotherapeutic agent in the absence of the compounds described herein. In a further aspect of the invention, a chemotherapeutic agent can be used in combination, simultaneously or sequentially, with the methods employing the compounds described herein. The compounds described herein can be administered concurrently with the chemotherapeutic agent, prior to, or after the administration of the chemotherapeutic agent. Thus, the compounds described herein can be administered during or after treatment of the chemotherapeutic agent.
I. Pharmaceutical Compositions/Formulations
Pharmaceutical compositions including the compounds described herein may be formulated in conjunction with one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen, i.e., whether local or systemic treatment is treated. Parenteral routes are preferred in many aspects of the invention.
Administration of pharmaceutical compositions containing the compounds of Formula (1) described herein may be oral, pulmonary, topical including epidermal, transdermal, ophthalmic and to mucous membranes including vaginal and rectal delivery or parenteral including intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion. In one embodiment, the compounds containing therapeutic oligonucleotides is administered IV, IP or as a bolus injection.
For injection, including, without limitation, intravenous, intramuscular and subcutaneous injection, the compounds described herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as physiological saline buffer or polar solvents including, without limitation, a pyrrolidone or dimethylsulfoxide.
The compounds may also be foπnulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers. Useful compositions include, without limitation, suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain adjuncts such as suspending, stabilizing and/or dispersing agents. Pharmaceutical compositions for parenteral administration include aqueous solutions of a water soluble form, such as. without limitation, a salt (preferred) of the active compound. Additionally, suspensions of the active compounds may be prepared in a lipophilic vehicle. Suitable lipophilic vehicles include fatty oils such as sesame oil, synthetic fatty acid esters such as ethyl oleate and triglycerides, or materials such as liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers and/or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
For oral administration, the compounds can be foπnulated by combining the active compounds with pharmaceutically acceptable carriers well-known in the art. Such carriers enable the compounds of the invention to be foπnulated as tablets, pills, lozenges, dragees, capsules, liquids, gels, syrups, pastes, slurries, solutions, suspensions, concentrated solutions and suspensions for diluting in the drinking water of a patient, premixes for dilution in the feed of a patient, and the like, for oral ingestion by a patient. Phaπnaceutical preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding other suitable auxiliaries if desired, to obtain tablets or dragee cores. Useful excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol, cellulose preparations such as, for example, maize starch, wheat starch, rice starch and potato starch and other materials such as gelatin, gum tragacanth, methyl cellulose, hydroxypropyl methylcellulose, sodium carboxymethylceilulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid. A salt such as sodium alginate may also be used. For administration by inhalation, the compounds of the present invention can conveniently be delivered in the form of an aerosol spray using a pressurized pack or a nebulizer ' and a suitable propellant.
The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described previously, the compounds may also be formulated as depot preparations. Such long acting formulations may be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. A compound of this invention may be formulated for this route of administration with suitable polymeric or hydrophobic materials (for instance, in an emulsion with a pharmacologically acceptable oil), with ion exchange resins, or as a sparingly soluble derivative such as, without limitation, a sparingly soluble salt.
Other delivery systems such as liposomes and emulsions can also be used. Additionally, the conjugates may be delivered using a sustained-release system, such as semi -permeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by those skilled in the art.
J. Dosages
Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the disclosure herein.
For any conjugate used in the methods of the invention, the therapeutically effective amount can be estimated initially from in vitro assays. Then, the dosage can be formulated for use in animal models so as to achieve a circulating concentration range that includes the effective dosage. Such information can then be used to more accurately determine dosages useful in patients.
The amount of the composition, e.g., used as a prodrug, that is administered will depend upon the parent molecule included therein (i.e., efficacy of an unconjugated oligonucleotide). Generally, the amount of prodrug used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various prodrug compounds will vary somewhat depending upon the parent compound (oligonucleotides such as LNA), rate of in vivo hydrolysis, molecular weight of the polymer, etc. In addition, the dosage, of course, can vary depending upon the dosage form and route of administration. In general, however, the oligonucleotide conjugates described herein can be administered in amounts ranging from about 1 mg/kg/week to about 1 g/kg/week, preferably from about 1 to about 500 mg/kg/week and more preferably from I to about 100 mg/kg/week (i.e., from about 2 to about 60 mg/kg/week). The range set forth above is illustrative and those skilled in the art will determine the optimal dosing of the prodrug selected based on clinical experience and the treatment indication. Moreover, the exact formulation, route of administration and dosage can be selected by the individual physician in view of the patient's condition. Additionally, toxicity and therapeutic efficacy of the compounds described herein can be deteπnined by standard pharmaceutical procedures in cell cultures or experimental animals using methods well-known in the art.
Alternatively, dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per subject per day). The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient. In one embodiment, the treatment of the present invention includes administering the oligonucleotide conjugates described herein in an amount of from about 2 to about 50 mg/kg/dose, such as from about 5 to about 30 mg/kg/dose to a mammal.
Alternatively, the delivery of the oligonucleotide conjugates described herein includes contacting a concentration of oligonucleotides of from about 0.1 to about 1000 nM, preferably from about 10 to about 1000 nM with tumor cells or tissues in vivo or in vitro.
The compositions may be administered once daily or divided into multiple doses which can be given as part of a multi-week treatment protocol. The precise dose will depend on the stage and severity of the condition, the susceptibility of the tumor to the polyrner-prodrug composition, and the individual characteristics of the patient being treated, as will be appreciated by one of ordinary skill in the art. In all aspects of the invention where polymeric conjugates are administered, the dosage amount mentioned is based on the amount of oligonucleotide molecule rather than the amount of polymeric conjugate administered. It is contemplated that the treatment will be given for one or more days until the desired clinical result is obtained. The exact amount, frequency and period of administration of the compound of the present invention will vary, of course, depending upon the sex, age and medical condition of the patent as well as the severity of the disease as determined by the attending clinician.
Still further aspects include combining the compound of the present invention described herein with other anticancer therapies for synergistic or additive benefit.
EXAMPLES
The following examples serve to provide further appreciation of the invention but are not meant in any way to restrict the effective scope of the invention. The bold-faced numbers recited in the Examples correspond to those shown in FIGs. 1-6.
Example 1. General Experimentals.
All synthesis reactions are run under an atmosphere of dry nitrogen or argon. Commercial reagents are used without further purification. All PEG compounds were dried in vacuo or by azeotropic distillation from toluene prior to use. 1H NMR spectra were obtained at 300 MHz and 13C NMR spectra at 75.46 MHz using a Varian Mercury300 NMR spectrometer and deuterated chloroform as the solvents unless otherwise specified. Chemical shifts (δ) are reported in parts per million (ppm) downfield from tetramethylsilane (TMS).
Abbreviations are used throughout the examples such as DCM (dichloromethane), DIEA (N,N-Diisopropylethylaamine). LNA (Locked Nucleic Acid), MEM (Modified Eagle's Medium). TEAA (tetraethylammonium acetate), TFA (trifluoroacetic acid), and RT-qPCR (reverse transcription-quantitative polymerase chain reaction).
Example 2. General HPLC Method.
The reaction mixtures and the purity of intermediates and final products are monitored by a Beckman Coulter System Gold® HPLC instrument. It employs a ZORB AX® 300SB C8 reversed phase column ( 150 * 4.6 mm) or a Phenomenex Jupiter® 300A Cl 8 reversed phase column ( 15O x 4,6 mm) with a 168 Diode Array UV Detector, using a gradient of 10-90 % of acetonitrile in 0.05 % TFA at a flow rate of 1 lnL/rninute or a gradient of 25-35 % acetonitrile in 50 mM TEAA buffer at a flow rate of 1 mL/minυte. The anion exchange chromatography was run on AKTA explorer I OOA from GE healthcare (Amersham Biosciences) using Poros 50HQ strong anion exchange resin from Applied Biosystems packed in an AP-Empty glass column from Waters. Desalting was achieved by using HiPrep 26/10 desalting columns from Amersham Biosciences.
Example 3. General mRNA Down-Regulation Procedure. The cells were maintained in complete medium (F- 12K or DMEM, supplemented with
10% FBS). A 12 well plate containing 2.5 * 105 cells in each well was incubated overnight at 37 0C. Cells were washed once with Opti-MEM® and 400 μL of Opti-MEM® was added per each well. Then, a solution of the polymer conjugate containing oligonucleotide was added to each well. The cells were incubated for 4 hours, followed by addition of 600 μL of media per well, and incubation for 24 hours. After 24 hours of treatment, the intracellular mRNA levels of the target gene, such as human survivin, and a housekeeping gene, such as GAPDH were qυantitated by RT-qPCR. The expression levels of mRNA noπnalized to that of GAPDH were compared.
Example 4. General RNA Preparation Procedure.
For the in vitro mRNA down-regulation studies, total RNA was prepared using RNAqueous Kit® (Ambion) following the manufacturer's instruction. The RNA concentrations were determined by OD260 nm using Nanodrop.
Example 5. General RT-qPCR Procedure.
All the reagents were from Applied Biosystems: High Capacity cDNA Reverse Transcription Kit® (4368813), 2Ox PCR master mix (4304437), and TaqMan® Gene Expression Assays kits for human GAPDH and survivin. 2.0 μg of total RNA was used for cDNA synthesis in a final volume of 50 μL. The reaction was conducted in a PCR theπnocycler at 25 0C for 10 minutes, 370C for 120 minutes, 85 0C for 5 seconds and then stored at 4 0C. Real-time PCR was conducted with the program of 50 °C-2 minutes, 95 0C-IO minutes, and 95 0C-15 seconds / 600C-I minute for 40 cycles. For each qPCR reaction, 1 μL of cDNA was used in a final volume of 30 μL.
Example 6. General procedure for PEGylation of Oligonυleotides A solution of activated PEG (0.35mmol, about 10-30 eq) and oligonucleotides (0.03mmol,
~1 eq) in PBS buffer (~5 mL /100 mg PEG, pH 7.4) is stirred at room temperature and the reaction progress is monitored by HPLC. The reaction is diluted in Milli-Q water (25 niL) and purified using a HQ/ 10 Poros strong anion exchange column (e.g. Source 15RPC column equilibrated with 10OmM TEAA before loading, 10 mm * 60 mm, bed volume -6 mL). The fractions are eluted using I M NaCl, water, and 50% CH3CN. The fractions containing pure product are pooled and lyophilized to yield pure PEG-Oligo. MALDl is used to confirm the molecular weight of the product.
Example 7. Preparation of Compound 1 (Folate NHS). Folic acid was coupled with NHS in the presence of DCC to provide an NHS ester
(compound 1).
Example 8. Preparation of Compound 3.
H eterobi functional amino acid PEG (compound 2) is coupled with NHS in the presence of EDC to provide an NHS ester (compound 3).
Example 9. Preparation of Compound 5
A solution of 4 N HCl in dioxane (70 mL) was added to BocCys(Npys)-OH (compound 4, 5 g, 13.32 mmol). The suspension was stirred at room temperature for 3 hours, and then was poured into 700 mL of ethyl ether. The solid was filtered through a course fritted funnel without applying vacuum until the end. The cake was washed with ethyl ether (3 x 50 mL) and then dried under vacuum at room temperature overnight. 1H NMR (300 MHz, CD3OD): δ 8.93 (IH. dd, J = 1.5, 4.7 Hz), 8.66 ( 1 H5 dd, J = 1 .5, 8.20 Hz), 7.59 (1 H, dd, J = 4.7, 8.2 Hz), 4.24 (I H, dd, J = 4.1 , 9.4 Hz), 3.58 (I H, dd, J = 4.1 , 14.9 Hz), 3.36 ( I H, dd, J = 9.4, 15.2 Hz). 13C NMR (75.4 MHz, CDCl3): δ 169.40. 156.27. 154.64. 144.13, 135.246, 123.10, 52.77, 39.27. Example 10. Preparation of Compound 6.
Compound 3 (0.35 rninol) is added to a solution of compound 5 (2 equivalent for each NHS to be substituted) in DMF/DCM (25 mL/45 mL), followed by addition of DIEA (3 equivalent for each compound 5). The suspension is stirred at room temperature for 5 hours. The reaction mixture is evaporated under vacuum and then precipitated with DCM/EbO at 0 0C. The solid is filtered and then is dissolved in 80 mL of DCM. After addition of 20 mL of 0.1 N HCl, the mixture is stirred for 5 minutes, then transferred to a separatory funnel and the organic layer is separated and washed again with 0.1 N HCl (20 mL) and brine (20 mL). The organic layer is dried over MgSO4, filtered and evaporated under vacuum. The residue is precipitated with DCM/Et2O at 0 0C. The solid is filtered and dried in the vacuum oven at 30 0C for at least 2 hours to give compound 14.
Example 1 1. Preparation of Compound 7.
A solution of compound 6 in DCM (10 mL/g of compound 6) is added 16 mL TFA (2.5 mlVg of compound 6) at 0 0C. The reaction mixture is stirred at 0 0C to room temperature for 1 hour. After completion of reaction, the solvent is removed in vacuo and the residue is precipitated from 20 mL/300 mL/g of compound 6 Of DCMZEt2O at 0 0C. Solids are filtered and dried to get compound 7.
Example 12. Preparation of Compound 8.
A solution of compound 1 in DMSO is added to a solution of compound 7 in DCM. The reaction mixture is stirred and the crude product is precipitated in DCM/Ether. The solid is filtered and dried in vacuo to give compound 8.
Example 13. Preparation of Compound 10.
Compound 8 is reacted with oligonucleotides (compound 9, HS-5'-(CH2)ό- Oligonucleotide) in the conditions described in Example 6 to give compound 10.
Example 14. Preparation of Compound 12. Compound 11 (bis SC-PEG1 0.35 mmol) is added to a solution of compound 5 (2 equivalent for each NHS to be substituted) in DMF/DCM (25 mL/45 mL), followed by addition of DlEA (3 equivalent for each compound 5). The suspension is stirred at room temperature for 5 hours. The reaction mixture is evaporated under vacuum and then precipitated with DCM/Et2O at 0 0C. The solid is filtered and then is dissolved in 80 mL of DCM. After addition of 20 mL of 0.1 N HCI, the mixture is stirred for 5 minutes, then transferred to a separatory funnel and the organic layer is separated and washed again with 0.1 N HCl (20 mL) and brine (20 mL). The organic layer is dried over MgSθ4, filtered and evaporated under vacuum. The residue was precipitated with DCMZEt2O at 0 0C. The solid is filtered and dried in the vacuum oven at 30 0C for at least 2 hours to give compound 12.
Example 15. Preparation of Compound 14.
Compound 13 is reacted with compound 12 in the presence of DIEA as the base in DCM to give compound 14.
Example 16. Preparation of Compound 15. Compound 14 is treated with TFA in DCM to give compound 15.
Example 17. Preparation of Compound 16.
A solution of compound 1 in DMSO is added to a solution of compound 15 in DCM. The reaction mixture is stirred and the crude product is precipitated in DCM/Ether. The solid is filtered and dried in vacuo to give compound 16.
Example 18. Preparation of Compound 18.
Compound 16 is reacted with oligonucleotides (compound 17, HS-5'-(CH2)ft- Oligonucleotide) in conditions described in Example 6 to give compound 18.
Example 19. Preparation of Compound 20 ((SC)vPEG-Cys-SS-NPyS).
Compound 19 (4 arm SC-2ϋKPEG, 0.35 mmol) is added to a solution of compound 5 (2 equivalent for each NHS to be substituted) in DMF/DCM (25 mL/45 mL), followed by addition of DIEA (3 equivalent for each compound 5). The suspension is stirred at room temperature for 5 hours. The reaction mixture was evaporated under vacuum and then precipitated with
DCM/Et2O at 0 0C. The solid was filtered and then was dissolved in 80 mL of DCM. After addition of 20 m L of 0.1 N HCl, the mixture was stirred for 5 minutes, then transferred to a separatory funnel and the organic layer was separated and washed again with 0.1 N HCl (20 mL) and brine (20 mL). The organic layer was dried over MgSO4, filtered and evaporated under vacuum. The residue was precipitated with DCM/EtiO at 0 0C. The solid is filtered and dried in the vacuum oven at 30 0C for at least 2 hours to give compound 20.
Example 20. Preparation of Compound 21 ((BocNHExfend)rPEG-Cys-SS-NPyS).
Compound 20 was reacted with compound 13 in the presence of DIEA as the base in DCM to give compound 21.
Example 21. Preparation of Compound 22 ((NHExtend)rPEG-Cys-SS-NPyS).
Compound 21 was treated with TFA in DCM to give compound 22.
Example 22. Preparation of Compound 23 ((Folate-NHExtend)3-PEG-Cys-SS-NPyS). A solution of compound 1 in DMSO was added to a solution of compound 22 in DCM.
The reaction mixture is stirred and the crude product is precipitated in DCM/Ether. The solid was filtered and dried in vacuo to give compound 23.
Example 23. Preparation of Compound 25 ((Folate-NHExtendb-^PEG-Cys-SS-Cβ-Oligo). (Folate)3-20KPEG-NPyS (compound 23, 120 mg, 5.50 μmol) was dissolved in pH 6.5 phosphate buffer (3-4 mL), covered in foil and purged with nitrogen gas for 10 minutes. To this solution was added oligonucleotides (compound 24, e.g. antisense ErbB3 LNA oligonucleotide, 6.0 mg, 1.10 μmol) and the resulting orange yellow mixture was stirred for -2 hours at ambient temperature during which time the solution became deeper yellow in color. After this time, the solution was filtered using a 0.45 μm syringe filter and loaded on a Poros HQ, strong anion exchange column ( 10 cm x 1.0 cm, bed volume - 8 mL) which was pre-equilibrated with 20 mM Tris-HCI buffer, pH 7.0 (buffer A). The column was washed with 3-4 column volumes of buffer A to remove the excess PEG linker. The product was eluted by slow incremental gradient of 1 M NaCl in 20 mM Tris-HCI buffer, pH 7.0 (buffer B). The isolated fractions were combined and desalted via reverse-phase chromatography (Source RPC) and the resulting solution was lyophilized to yield the desired PEG-LNA compound as a fluffy yellow solid (4.62 mg, based on oligo, 77.0 %).
Example 24. Preparation of Compound 26 ((FoIate-NHExtend)3-40KPEG-Cys-SS-C6-OIigo- FAM).
(Folate)3-40KPEG-NPyS (0.35 g, 8.33 μmol) was dissolved in pH 6.5 phosphate buffer (5 raL), covered in foil and purged with nitrogen gas for 10 minutes. To this solution was added FAM-modifϊed oligonucleotides (FAM modified compound 24, 10.0 mg, 1.67 μmol) and the resulting orange yellow mixture was stirred for ~3 hours at ambient temperature during which time the solution became deeper yellow in color. After this time, the solution was filtered using a 0.45 μm syringe filter and loaded on a Poros HQ, strong anion exchange column (10 cm x 1.0 cm, bed volume - 8 raL) which was pre-equilibrated with 20 mM Tris-HCl buffer, pH 7.0 (buffer A). The column was washed with 3-4 column volumes of buffer A to remove the excess PEG linker. The product was eluted by slow incremental gradient of 1 M NaCl in 20 mM Tris- HCl buffer, pH 7.0 (buffer B). The isolated fractions were combined and desalted via reverse- phase chromatography (Source RPC) and the resulting solution was lyophilized to yield the desired PEG-LNA compound as a fluffy yellow solid (3.33 mg, based on oligo, 33.3 %).
Example 25. Preparation of Compound 28. Compound 22 is reacted with compound TAMRA-C(=O)-OSu (compound 27) to give compound 28.
Example 26. Preparation of Compound 29.
A solution of compound 1 in DMSO is added to a solution of compound 29 in DCM. The reaction mixture is stirred and the crude product is precipitated in DCM/Ether. The solid is filtered and dried in vacuo to give compound 29.
Example 27. Preparation of Compound 31 (Folate)2-TAMRA-20KPEG-Cys-SS-C6-Oligo.
Compound 29 is reacted with oligonucleotides (compound 30) in conditions described in Example 6 to give compound 31. Example 28. Preparation of Compound 33.
Compound 32 (4 arm SC-20KPEG, 0.35 mmol) was added to a solution of equivalent for each NHS to be substituted) in DMF/DCM (25 niL/45 mL), followed by addition of DIEA (3 equivalent for each compound 5). The suspension is stirred at room temperature for 5 hours. The reaction mixture was evaporated under vacuum and then precipitated with DCM/Et2O at 0 0C. The solid was filtered and then was dissolved in 80 mL of DCM. After addition of 20 mL of 0.1 N HCl, the mixture was stirred for 5 minutes, then transferred to a separatory funnel and the organic layer was separated and washed again with 0.1 N HCl (20 mL) and brine (20 mL), The organic layer was dried over MgSO4, filtered and evaporated under vacuum. The residue is precipitated with DCM/Et2O at 0 0C. The solid is filtered and dried in the vacuum oven at 30 0C for at least 2 hours. 13C NMR (75.4 MHz, CDCl3): d 170.90, 156.66, 155.68, 153.86, 142.41, 133.85, 121.24, 72.96-69.30, 64.08, 53.01, 41.82.
Example 29. Preparation of Compound 35. Different sizes of activated PEG polymer were used to make PEG-LNA conjugates. In general, compound 33 was reacted with oligonucleotides (compound 34) using the conditions described in Example 6. The description of each compound is provided in FIG. 6. Oligonucleotides (compound 34, 400 mg, 0.074 mmol) were added to a solution of compound 33 (505 nig, 0.012 mmol) in 25 mL of pH 6.5, 100 mM Sodium Phosphate. The reaction was stirred at room temperature under nitrogen for 5 hours. The reaction mixture was purified on Source 15RPC column. The column was equilibrated with 100 mM TEAA. Then the reaction mixture was loaded. The column was eluted with IM NaCl, water, and 50% CH3CN. Compound 35 was collected and lyophilized. Yield 150mg, MALDI confirms the molecular weight of 62,590.
Example 30. Compounds of Formula (I)
Examples 31-37 demonstrate improved tumor delivery of oligonucleotides as well as improved antisense knockdown of targeted tumor mRNA using the releasably linked PEG molecule having the formula:
Figure imgf000058_0001
wherein
PEG is a polyethylene glycol;
Rb is
Figure imgf000058_0002
Oligo is uniformly 5'-(CH2)6-anti-survivin LNA (SEQ ID NO: 1, referred to as "LNAl") or 5'-(CH2)6-anti-erbB3 LNA (SEQ ID NO: 6, referred to "LNA2"), and the total molecular weight of the polymeric portion of the compound containing PEG is about 40,000 daltons, 20,000 daltons, 10,000 daltons or 5,000 daltons.
For example, the compounds include Folate-40KPEG-Cys-SS-LNA2 (compound 101), Folate-5KPEG-Cys-SS-LNA2 (compound 1O)3 401TEG-CyS-SS-LNAl (compound 103), 10KPEG- Cys-SS-LNAl, (compound 104) and 40KPEG-Cys-S S-LN A2 (compound 105).
Example 31. In vitro Stability
The PEG-Cys-SS-LNA and Folate-PEG-Cys-SS-LNA conjugates described herein showed good stability in buffers. Example 32. In vitro Cellular Uptake of Folate-PEG-Cys-SS-LNA Conjugate
Cellular uptake of Folate-PEG-Cys-SS-LNA conjugate and specificity o folate receptor were evaluated in KB human cervical carcinoma cell line. KB cells were plated overnight at 37 0C. The cells were incubated with 100 nM of Folate-PEG-Cys-SS-LNA2-FAM conjugates (compounds 101 and 10 labeled with FAM) with or without 1 μM of free folate at 37 0C for 24 hours. The amount of the PEG-Cys-SS-LNA2-FAM conjugate was based on the amount of LNA2, not the amount of polymeric conjugate. The cells were washed and the samples were observed under fluorescence microscope and confocal microscope. The results of the cellular uptake of the Folate-PEG-Cys-SS-LNA conjugate are shown in FIG. 7. KB cells were also exposed to Folate-5KPEG-Cys-SS-LNA2-FAM (compound 10 labeled with FAM) with or without 100 nM of free folate at 37 0C for 4 hours. The cells were washed and analyzed by FACS for the specificity of binding. The results are shown in FIG. 8.
Both fluorescence and confocal microscope studies showed that folate improved cellular uptake of LNA oligonucleotides. The intracellular delivery of the folate-PEG conjugate was comparable to that transfected with lipofectamine. Folic acid enhanced intracellular uptake of oligonucleotides.
The fluorescence microscope images (FIG. 7(a)) and FACS analysis (FIG. 8) showed that the binding of the folate-PEG conjugate to the folate receptor and subsequent internalization into KB cells was blocked in the presence of free folate. The binding of the folate-PEG conjugate to the folate receptor in KB cells was specific.
Example 33. In vitro Efficacy of PEG-Cys-SS-LNA Conjugate in Tumor CeUs
In vitro efficacy of PEG-Cys-SS-LNA conjugates and naked LNA oligonucleotides were performed by using qRT-PCR. 15PC3 human prostate cancer cells were treated with 0.1 nM to 1,000 nM of each of test compounds, naked LNA2 or 40KPEG-Cys-SS-LNA2 (compound 105). The amount of 40KPEG-Cys-SS-LNA2 administered was based on the amount of LNA2, not the amount of polymeric conjugate administered. The cells were collected and analyzed by using qRT-PCR for ErbB3 mRNA downregulation. The results from qRT-PCR were compared to untreated 15PC3 cells with or without lipofectamine. The results are shown in FIG. 9. The results showed that the PEG-Cys-SS-LNA2 conjugate downregulated expression of the target gene ErbB3 mRNA and the efficacy was comparable to naked LNA2. The knockdown of ErbB3 mRNA expression of PEG-Cys-SS-LNA2 conjugate was as potent as equivalent naked LNA2 in 15PC3 cells (IC50 = 5 nM and 8.5 nM, respectively). The PEG attached to the LNA oligonucleotides did not interfere with the potency of the LNA oligonucleotides.
Example 34. In vivo Efficacy of Folate-PEG-Cys-SS-LNA conjugate in KB Xenografted Mice Model
The efficacy of Folate-PEG-LNA conjugates was evaluated in KB human cervical cancer xenografted mice. Athymic nude Balb/c mice bearing KB tumor (epidermoid, human cervical carcinoma cell line) were treated with a dose of 35 mg/kg of naked LN A2 or Folate-^PEG-Cys- SS-LN A2, 40KPEG-LNA2, Folate-5KPEG-LNA2, or 5KPEG- LNA2 conjugate at q3dx4 for 12 days. The amount of the PEG conjugates administered was based on the amount of LNA2, not the amount of polymeric conjugate administered. Tumor and liver samples were isolated and analyzed by using qRT-PCR for ErbB3 mRNA down-regulation.
The results are as shown in FIGS. 10(A) and (B). The folate-PEG conjugates significantly inhibited expression of ErbB3 mRNA compared to naked LN A2 in the tumor tissues. Additionally, 4OK Folate-PEG-LNA conjugates downregulated target mRNA compared to 5K Folate-PEG-LNA conjugates. The results showed that PEGylation increased accumulation of LNA oligonucleotide in tumor and the folate-PEG conjugates enhanced mRNA downregulation as compared to naked LNA oligonucleotides.
Example 35. Biodistribution of PE G-Cy s-S S-LNA in Tumor and Plasma
The circulation of PEG-Cys-SS-LNA conjugates in plasma and retention in tumor was evaluated in A549 human long adenocarcinoma xenografted mice.
A549 (human lung adenocarcinoma epithelial cell line) cells were implanted sc. in athymic nude mice. When tumor reached the average volume of 75 mm3, the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LNAl , 40KPEG-Cys- SS LNAl (compound 103, 10 mg/kg equivalent dose of LNA l ) or IOKPEG-Cys-SS-LNAl (compound 104, 10 mg/kg equivalent dose of LNAl ). Plasma samples were collected at 2 and 4 hours time points following the treatment. Tumor tissues were collected from the sacrificed animals at the various time points (2, 4, 12, 24, 48 and 72 hours) following the treatment. Concentrations of equivalent-LNA I oligonucleotides in tumor or plasma samples were measured by an ELISA hybridization assay. Results are shown in FIG. 1 l (A) and (B).
The results showed that "^PEG-Cys-SS-LNA conjugate had a significantly higher circulation in plasma and accumulation in tumor compared to naked LNA oligonucleotides. The mice treated with the PEG-Cys-SS-LNA conjugate had >50 rimes concentration of circulating LNA oligonucleotides in plasma at 2 hours and 4 hours following the treatment, as compared to naked LNA oligonucleotides. The PEG-Cys-SS-LNA conjugates had higher plasma concentrations and longer circulating times compared to naked LNA oligonucleotides. The mice treated with the PEG conjugate had 3-fold higher accumulation of LNA oligonucleotides in rumor at 24 hours, as compared to naked LNA oligonucleotides. The results also indicated that the 40KPEG conjugates had >3.5 times tumor accumulation at 12 hours and maintained > 1.5 times accumulation up to 72 hours compared to the 10KPEG conjugates. The results indicated that higher molecular weight PEG (40KDa) conjugates has greater tumor accumulation than lower MW PEG (1 OKDa) PEG conjugates.
Example 36. In vivo Efficacy of PEG-Cys-SS-LNA conjugate in KB Xenografted Mice Model
The efficacy of PEG-LNA conjugates was evaluated in KJB human cervical cancer xenografted mice. KB cells (epidermoid, human cervical carcinoma cell line) were implanted sc. in nude mice. When tumor reached the average volume of 75 mm3, the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LNA2 or 40KPEG-Cys-SS LN A2 (compound 105, 10 mg/kg equivalent dose of LNA2) at q3d x4. Tumor and liver samples were collected 24 hours after the last dose. ErbB3 mRNA downregulation in the samples was measured by using qRT-PCR. The results are shown in FIG. 12. The results showed that the PEG-Cys-SS-LNA conjugate significantly inhibited expression of ErbB3 mRNA in tumor compared to naked LNA oligonucleotides. The mice treated with the PEG conjugate inhibited ErbB3 mRNA expression 2 fold more than the mice treated with naked LNA. Additionally, the PEG conjugates inhibited 92% ErbB3 mRNA expression in liver as compared to 88% by naked LNA oligonucleotides. Example 37. Jn vivo Efficacy of PEG-Cys-SS-LNA conjugate in 15PC3 Xenografted Mice Model
The efficacy of PEG-LNA conjugates was evaluated in 15PC3 human prostate cancer xenografted mice. 15PC3 cells (human prostate cancer cell line) were implanted sc. in nude mice. When tumor reached the average volume of 75 mm3, the mice were randomly grouped and injected i.v. with a single dose of 10 mg/kg of naked LN A2 or 40KPEG-Cys-SS LN A2 (compound 105, 10 mg/kg equivalent dose of LN A2) at q3d x4. Tumor and liver samples were collected 24 hours after the last dose. ErbB3 mRNA downregulation in the samples was measured by using qRT-PCR. The results are shown in FIG. 13. The results showed that the PEG-Cys-SS-LNA conjugate significantly inhibited expression of ErbB3 mRNA in tumors compared to naked LNA oligonucleotides. The mice treated with the PEG conjugate inhibited ErbB3 mRNA expression 2 fold more than the mice treated with naked LNA. Additionally, the PEG conjugates inhibited 83% ErbB3 mRNA expression in liver as compared to 73% by naked LNA oligonucleotides.
In view of the above, the invention advantageously provides improved methods employing PEG conjugates for the delivery of oligonucleotides to tumor cells in a mammal that greatly increase circulation time, enhance the accumulation of oligonucleotides in tumors in vivo, while also achieving enhanced downregulation of oncogene mRNA expression in tumors compared to corresponding naked anrisense constructs.
One embodiment of the invention provides an improved method for the delivery of oligonucleotides to tumor cells in a mammal that includes the steps of:
(a) providing a compound having the formula:
Figure imgf000062_0001
Figure imgf000063_0001
or a pharmaceutically acceptable salt thereof, wherein
PEG is a polyethylene glycol; Rh is
Figure imgf000063_0002
Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons; and
(b) administering the compound or the pharmaceutically acceptable salt thereof to a mammal having tumor cells.
A related embodiment of the invention provides an improved method for the in vivo inhibition of tumor gene expression in a mammal that includes the steps of:
(a) providing a compound having the formula:
Figure imgf000063_0003
Figure imgf000064_0001
or a phaπnaceutically acceptable salt thereof, wherein
PEG is a polyethylene glycol; Rb is
Figure imgf000064_0002
Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons; and (b) administering the compound or the salt thereof to a mammal having tumor cells, wherein said administration reduces the expression of the preselected gene by the tumor cells.
The inhibition of expression of the preselected gene may be as a result of antisense targeting of an mRNA molecule thereby reducing or eliminating translation of the mRNA to a polypeptide. In the method embodiments above, the administration may be by the blood stream of the mammal, for example, by intravenous (i.v.) injection. The oligonucleotides may comprise LNA. The oligonucleotide includes -5"-(CH2)6-antisense-Survivin LNA oligonucleotide or -5'- (CH2)6-antisense-ErbB3 LNA oligonucleotide.

Claims

We claim:
1. An improved method of delivering oligonucleotides to tumor cells in a mammal, comprising administering to a mammal having tumor cells a compound of Formula (I):
Figure imgf000065_0001
or a pharmaceutically acceptable salt thereof, wherein
Ri is a substantially non-antigenic water-soluble polymer, each Zi is the same or different and selected from the group consisting of
Figure imgf000065_0002
-(LO81-Rt,; and
-(L»)a2-Rcj
Yi, in each occurrence, is independently S or O;
Y2, in each occurrence, is independently NR13;
R8, in each occurrence, is the same or a different oligonucleotide; each of Li^, in each occurrence, is the same or a different bifunctional linker;
Rb, in each occurrence, is a folic acid;
R0, in each occurrence, is the same or a different diagnostic agent; each of R3-7 is independently selected from the group consisting of hydrogen, Ci-^ alkyls, C2-O alkenyl, C2-6 alkynyl, C3-^ branched alkyl, C3-8 cycloalkyl, and Ci^ alkoxy,
R13, in each occurrence, is independently selected from the group consisting of hydrogen, Ci_6 alkyls, C2-^ alkenyl, C2-6 alkynyl, C3-^ branched alkyl, and C3.8 cycloalkyl;
R12, in each occurrence, is independently selected from the group consisting of hydrogen, hydroxyl, Ci-β alkyls, C2-β alkenyl, C2-6 alkynyl, C3.19 branched alkyl, C3^ cycloalkyl and Ci-6 alkoxy; each of (a) and (d) is independently zero, 1, 2, or 3; each of (al) and (a2) is independently zero, 1, 2, or 3; each (b) is independently zero, 1, 2, or 3; each (c) is independently zero, 1, 2, or 3; each (e) is independently zero or one; each (g) is independently zero or one; and
(m) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero and further provided that one or more of Zi contains an oligonucleotide.
2. The method of claim 1, wherein the compound of Formula (I) has Formula (I'):
Figure imgf000066_0001
wherein
(ml) is a positive integer from about 1 to about 8;
(m2) is zero or a positive integer from about 1 to about 7; and the sum of (ml) and (m2) is an integer from about 2 to about 8.
3. The compound of claim 1, wherein all Zj contain an oligonucleotide.
4. The method of claim 1 , wherein one or more of Zj contains a folic acid.
5. The method of claim 1, wherein Ri2 is OH.
6. The method of claim 1 , wherein R3-7 are all hydrogen.
7. The method of claim 1, wherein (b), (d) and (e) are zero, and (c) is one.
8. The method of claim 1 , wherein Z1 has the formula:
Figure imgf000067_0001
wherein, (a) is O or l;
(m) is an integer from 2 to 8; and (2, 4, 8, 16 or 32);
R12, in each occurrence, is independently selected from the group consisting of hydroxyl, Ci-6 alkyls, C2-6 alkenyl, C2-^ alkynyl, C3.19 branched alkyl, and Cj-6 alkoxy; and all other variables are the same as defined in claim 1.
9. The method of claim 2, wherein the compound of Formula (I) has the formula
Figure imgf000067_0002
wherein (a) is 0 or 1.
10. The method of claim 9, wherein (m2) is zero.
11. The method of claim 1 , wherein (m 1 ) is one.
12. The method of claim 1 , wherein R1 comprises a polyalkylene oxide.
13. The method of claim 12, wherein Rj has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons.
14. A compound of claim 1 selected from the group consisting of:
Figure imgf000067_0003
Figure imgf000068_0001
wherein each Z is independently
Figure imgf000068_0002
wherein (a) is O or l.
Ri2, in each occurrence, is independently selected from the group consisting of hydroxyl, Ci-6 alkyls, C2-6 alkenyl, C2-6 alkynyl, C3-19 branched alkyl, and C1-6 alkoxy,
(n) is a positive integer and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons; and all other variables are the same as defined in claim 1.
15. The method of claim 1 , wherein the oligonucleotide is a single stranded or double stranded oligonucleotide.
16. The method of claim 15, wherein the oligonucleotide is an antisense oligonucleotide.
17. The method of claim 15, wherein the oligonucleotide is selected from the group consisting of deoxynucleotide, ribonucleotide, locked nucleic acids (LNA), short interfering RNA (siRNA), microRNA (miRNA), aptamers, peptide nucleic acid (PNA), phosphorodiamxdate morpholino oligonucleotides (PMO), tricyclo-DNA, double stranded oligonucleotide (decoy ODN), catalytic RNA (RNAi), aptamers, spiegelmers, CpG oligomers and combinations thereof.
18. The method of claim 15, wherein the oligonucleotide has LNA and phosphorothioate linkages.
19. The method of claim 15, wherein the oligonucleotide has from about 8 to about 30 nucleotides.
20. The method of claim 19, wherein the oligonucleotide is selected from the group consisting of antisense bcl-2 oligonucleotides, antisense HIF-Ia oligonucleotides, antisense survivin oligonucleotides and antisense Erbfl3 oligonucleotides.
21. The method of claim 15, wherein the oligonucleotide comprises SEQ ID NO: 1, SEQ ID NOs 2 and 3, SEQ ID NO:3, SEQ BD NO: 4, SEQ ID NO: 5, SEQ ID NO: 5, and SEQ ID NO: 6.
22. The method of claim 1, wherein the compound of Formula (T) is selected from the group consisting of:
Figure imgf000070_0001
Figure imgf000071_0001
wherein: Oligo is an oligonucleotide;
PEG is a polyethylene glycol and the polymeric portion of the compound has the total number average molecular weight of from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons;
(al) is one; and
Figure imgf000071_0002
, wherein (r') is zero or one.
23. The method of claim 1, wherein the tumor cells are prostate or cervical cancer cells.
24. The method of claim 1, wherein the administering step comprises admin blood stream of the mammal.
25. An improved method for delivering oligonucleotides to tumor cells in a mammal, comprising:
(a) providing a compound having the formula:
Figure imgf000072_0001
or a pharmaceutically acceptable salt thereof, wherein
PEG is a polyethylene glycol;
Rb is
Figure imgf000072_0002
Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons; and
(b) administering the compound or the pharmaceutically acceptable salt thereof to a mammal having tumor cells.
26. The method of claim 25, wherein the oligonucleotide comprises LNA.
27. The method of claim 25, wherein Oligo is -5'-(CH2)6-TsAsGsCsCsTsGsTs CsAsCsTsTsCsTsCs-3 ' or -5 '-(CH2)6-GsCsTsGsCsCsAsTsGsGsAsTsTsGsAsG -3 ', wherein the first three nucleotides in 5' and 3' terminal are LNA and "s" represents a phosphorothioate linkage.
28. The method of claim 25, wherein the tumor cells are prostate or cervical cancer cells.
29. An improved method for deliverying oligonucleotides to tumor cells in a mammal, comprising:
(a) providing a compound having the formula:
Figure imgf000073_0001
or a pharmaceutically acceptable salt thereof, wherein
PEG is a polyethylene glycol; Rb is
Figure imgf000074_0001
Oligo is an oligonucleotide of from about 8 to 30 nucleotides, wherein the polymeric portion of the compound has the total number average molecular weight of about 40,000 daltons; and
(b) administering the compound or the salt thereof to a mammal having tumor cells, wherein said administration reduces the expression of the preselected gene by the tumor cells.
30. A method of introducing an oligonucleotide into a cell comprising: contacting a cell with a compound of Formula (I).
31. A method of inhibiting the growth or proliferation of cancer cells comprising: contacting a cancer cell with with a compound of Formula (I).
32. A compound of Formula (Ia):
Figure imgf000074_0003
or a pharmaceutically acceptable salt thereof, wherein
Ri is a substantially non-antigenic water-soluble polymer; each Z] is the same or different and selected from the group among
Figure imgf000074_0002
Yi, in each occurrence, is independently S or O; Y2, in each occurrence, is independently NRi 3; R6, in each occurrence, is the same or a different oligonucleotide; each of Li-4, in each occurrence, is the same or a different bifunctional linker; Rb, in each occurrence, is a folic acid; Rc, in each occurrence, is the same or a different diagnostic agent; each OfR3-7 is independently selected from among hydrogen, Ci-6 alkyls, C2-6 alkenyl, C2- 6 alkynyl, C3-Ig branched alkyl, C$.g cycloalkyl, and Cj-6 alkoxy;
Ri3, in each occurrence, is independently selected from among hydrogen, C1-6 alkyls, Q-^ alkenyl, C2-6 alkynyl, C3-i9 branched alkyl, and C3-8 cycloalkyl; Ri2, in each occurrence, is independently selected from among hydrogen, hydroxyl, Ci-^ alkyls, C2-6 alkenyl, C2-^ alkynyl, C3-19 branched alkyl, C3-S cycloalkyl and Ci-^ alkoxy, each of (a) and (d) is independently zero, 1, 2, or 3; each of (al) and (a2) is independently zero, 1, 2, or 3; each (b) is independently zero, 1, 2, or 3; each (c) is independently zero, 1 , 2, or 3 ; each (e) is independently zero or one; each (g) is independently zero or one; and (m) is a positive integer from about 2 to about 32, provided that (a) and (g) are not simultaneously zero, and further provided that one or more of Zi contain an oligonucleotide, and further provided that one or more of Zi contain a folic acid.
PCT/US2009/044953 2008-05-23 2009-05-22 Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides WO2009143412A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/994,266 US20110105413A1 (en) 2008-05-23 2009-05-22 Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
EP09751640.5A EP2288261A4 (en) 2008-05-23 2009-05-22 Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
CA2723263A CA2723263A1 (en) 2008-05-23 2009-05-22 Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
JP2011510720A JP2011520983A (en) 2008-05-23 2009-05-22 Polymer systems containing intracellular releasable disulfide linkers for oligonucleotide delivery

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US5595008P 2008-05-23 2008-05-23
US5586908P 2008-05-23 2008-05-23
US61/055,950 2008-05-23
US61/055,869 2008-05-23
US10657808P 2008-10-19 2008-10-19
US10657608P 2008-10-19 2008-10-19
US61/106,576 2008-10-19
US61/106,578 2008-10-19

Publications (2)

Publication Number Publication Date
WO2009143412A2 true WO2009143412A2 (en) 2009-11-26
WO2009143412A3 WO2009143412A3 (en) 2010-04-15

Family

ID=41340917

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/044953 WO2009143412A2 (en) 2008-05-23 2009-05-22 Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides

Country Status (6)

Country Link
US (1) US20110105413A1 (en)
EP (1) EP2288261A4 (en)
JP (1) JP2011520983A (en)
CA (1) CA2723263A1 (en)
TW (1) TW201004648A (en)
WO (1) WO2009143412A2 (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2192914A1 (en) * 2007-08-20 2010-06-09 Enzon Pharmaceuticals, Inc. Polymeric linkers containing pyridyl disulfide moieties
WO2015149001A1 (en) 2014-03-27 2015-10-01 The Brigham And Women's Hospital, Inc. Metabolically-activated drug conjugates to overcome resistance in cancer therapy
WO2017148883A1 (en) 2016-03-01 2017-09-08 Ascendis Pharma Bone Diseases A/S Pth prodrugs
WO2018011266A1 (en) 2016-07-13 2018-01-18 Ascendis Pharma A/S Conjugation method for carrier-linked prodrugs
WO2018060314A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Growth Disorders A/S Combination therapy with controlled-release cnp agonists
WO2018060310A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Dosage regimen for a controlled-release pth compound
WO2018060312A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Pth compounds with low peak-to-trough ratios
WO2018060311A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Incremental dose finding in controlled-release pth compounds
WO2018175788A1 (en) 2017-03-22 2018-09-27 Genentech, Inc. Hydrogel cross-linked hyaluronic acid prodrug compositions and methods
WO2019185706A1 (en) 2018-03-28 2019-10-03 Ascendis Pharma A/S Conjugates
WO2019185705A1 (en) 2018-03-28 2019-10-03 Ascendis Pharma A/S Il-2 conjugates
WO2019219896A1 (en) 2018-05-18 2019-11-21 Ascendis Pharma Bone Diseases A/S Starting dose of pth conjugates
WO2020064844A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Treatment of infections
WO2020064847A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Degradable hyaluronic acid hydrogels
WO2020064846A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Novel hydrogel conjugates
WO2020141221A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Conjugates of pattern recognition receptor agonists
WO2020141225A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Minimization of systemic inflammation
WO2020141222A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Sustained local drug levels for innate immune agonists
WO2020141223A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Induction of sustained local inflammation
WO2020165087A1 (en) 2019-02-11 2020-08-20 Ascendis Pharma Bone Diseases A/S Liquid pharmaceutical formulations of pth conjugates
WO2020165081A1 (en) 2019-02-11 2020-08-20 Ascendis Pharma Growth Disorders A/S Dry pharmaceutical formulations of cnp conjugates
WO2020254611A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 conjugates
WO2020254607A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 compounds with localized pd properties
WO2020254617A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 compounds with localized pk properties
WO2020254613A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Controlled-release tyrosine kinase inhibitor compounds with localized pk properties
WO2020254612A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Controlled-release tyrosine kinase inhibitor compounds with localized pd properties
WO2020254609A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Tyrosine kinase inhibitor conjugates
WO2021144249A1 (en) 2020-01-13 2021-07-22 Ascendis Pharma Bone Diseases A/S Hypoparathyroidism treatment
US11154593B2 (en) 2016-01-08 2021-10-26 Ascendis Pharma Growth Disorders A/S CNP prodrugs with large carrier moieties
WO2021224169A1 (en) 2020-05-04 2021-11-11 Ascendis Pharma A/S Hydrogel irradiation
WO2021245130A1 (en) 2020-06-03 2021-12-09 Ascendis Pharma Oncology Division A/S Il-2 sequences and uses thereof
US11224661B2 (en) 2016-01-08 2022-01-18 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with increased NEP stability
WO2022043493A1 (en) 2020-08-28 2022-03-03 Ascendis Pharma Oncology Division A/S Glycosylated il-2 proteins and uses thereof
WO2022064035A1 (en) 2020-09-28 2022-03-31 Ascendis Pharma Bone Diseases A/S Improvement of physical and mental well-being of patients with hypoparathyroidism
US11311604B2 (en) 2016-01-08 2022-04-26 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low NPR-C binding
US11389511B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with reduced side-effects
US11389510B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low initial NPR-B activity
US11413351B2 (en) 2016-01-08 2022-08-16 Ascendis Pharma Growth Disorders A/S CNP prodrugs with carrier attachment at the ring moiety
WO2022207798A1 (en) 2021-04-01 2022-10-06 Ascendis Pharma A/S Use of long-acting growth hormone for treating inflammation-induced diseases
WO2023046732A1 (en) 2021-09-22 2023-03-30 Ascendis Pharma Bone Diseases A/S Long-acting pth compound treatments
WO2023110727A2 (en) 2021-12-13 2023-06-22 Ascendis Pharma Oncology Division A/S Novel cancer treatments with tlr7/8 agonists
WO2023110758A1 (en) 2021-12-13 2023-06-22 Ascendis Pharma Growth Disorders A/S Effective doses of cnp conjugates
WO2023227505A1 (en) 2022-05-23 2023-11-30 Ascendis Pharma Growth Disorders A/S Liquid pharmaceutical formulations of cnp compounds

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITMI20131929A1 (en) * 2013-11-20 2015-05-21 Domenico Terenzio CONJUGATE OF FOLIC ACID AND INDO-3-CARBINOL FOR MEDICAL USE

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE399809T1 (en) * 1998-03-12 2008-07-15 Nektar Therapeutics Al Corp METHOD FOR PRODUCING POLYMER CONJUGATES
ATE452654T1 (en) * 2001-11-09 2010-01-15 Enzon Inc POLYALKYLENE OXIDE CONJUGATES OF THIOL-CONTAINING MEDICATIONS
KR101177340B1 (en) * 2003-01-06 2012-08-30 넥타르 테라퓨틱스 Thiol-selective water-soluble polymer derivatives
WO2004070062A2 (en) * 2003-02-04 2004-08-19 Wyeth Compositions and methods for diagnosing and treating cancers
AU2004230927B9 (en) * 2003-04-13 2011-12-01 Enzon Pharmaceuticals, Inc. Polymeric oligonucleotide prodrugs
ATE494012T1 (en) * 2004-12-21 2011-01-15 Nektar Therapeutics STABILIZED POLYMER THIOL REAGENTS
US7671067B2 (en) * 2006-02-09 2010-03-02 Enzon Pharmaceuticals, Inc. Treatment of non-hodgkin's lymphomas with multi-arm polymeric conjugates of 7-ethyl-10-hydroxycamtothecin
RU2009114154A (en) * 2006-09-15 2010-10-20 Энзон Фармасьютикалз, Инк. (Us) POLYMERIC DRUGS WITH DIRECTED DELIVERY CONTAINING POLYFUNCTIONAL LINKERS
BRPI0716823A2 (en) * 2006-09-15 2015-05-26 Enzon Pharmaceuticals Inc Polymeric assembly containing positively charged portions
US8367065B2 (en) * 2006-09-15 2013-02-05 Enzon Pharmaceuticals, Inc. Targeted polymeric prodrugs containing multifunctional linkers
KR20090108082A (en) * 2007-02-09 2009-10-14 엔존 파마슈티컬즈, 인코포레이티드 Treatment of resistant or refractory cancers with multi-arm polymeric conjugates of 7-ethyl-10-hydroxycamptothecin
BRPI0721948A2 (en) * 2007-08-20 2014-04-08 Enzon Pharmaceuticals Inc POLYMERIC LEADERS CONTAINING FRACTIONS PYRIDYL DISULFECTURE

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2288261A4 *

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2192914A1 (en) * 2007-08-20 2010-06-09 Enzon Pharmaceuticals, Inc. Polymeric linkers containing pyridyl disulfide moieties
EP2192914A4 (en) * 2007-08-20 2014-05-07 Belrose Pharma Inc Polymeric linkers containing pyridyl disulfide moieties
WO2015149001A1 (en) 2014-03-27 2015-10-01 The Brigham And Women's Hospital, Inc. Metabolically-activated drug conjugates to overcome resistance in cancer therapy
US11413351B2 (en) 2016-01-08 2022-08-16 Ascendis Pharma Growth Disorders A/S CNP prodrugs with carrier attachment at the ring moiety
US11224661B2 (en) 2016-01-08 2022-01-18 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with increased NEP stability
US11311604B2 (en) 2016-01-08 2022-04-26 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low NPR-C binding
US11389511B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with reduced side-effects
US11389510B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low initial NPR-B activity
US11154593B2 (en) 2016-01-08 2021-10-26 Ascendis Pharma Growth Disorders A/S CNP prodrugs with large carrier moieties
WO2017148883A1 (en) 2016-03-01 2017-09-08 Ascendis Pharma Bone Diseases A/S Pth prodrugs
US11793861B2 (en) 2016-03-01 2023-10-24 Ascendis Pharma Bone Diseases A/S PTH prodrugs
WO2018011266A1 (en) 2016-07-13 2018-01-18 Ascendis Pharma A/S Conjugation method for carrier-linked prodrugs
US11896671B2 (en) 2016-07-13 2024-02-13 Ascendis Pharma A/S Conjugation method for carrier-linked prodrugs
US11918628B2 (en) 2016-09-29 2024-03-05 Ascendis Pharma Bone Diseases A/S Controlled-release PTH compound
US11759504B2 (en) 2016-09-29 2023-09-19 Ascendis Pharma Bone Diseases A/S PTH compounds with low peak-to-trough ratios
WO2018060314A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Growth Disorders A/S Combination therapy with controlled-release cnp agonists
US11890326B2 (en) 2016-09-29 2024-02-06 Ascendis Pharma Bone Diseases A/S Controlled-release PTH compound
US11857603B2 (en) 2016-09-29 2024-01-02 Ascendis Pharma Bone Diseases A/S PTH compounds with low peak-to-trough ratios
EP4275677A2 (en) 2016-09-29 2023-11-15 Ascendis Pharma Bone Diseases A/S Dosage regimen for a controlled-release pth compound
WO2018060310A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Dosage regimen for a controlled-release pth compound
WO2018060312A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Pth compounds with low peak-to-trough ratios
WO2018060311A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Incremental dose finding in controlled-release pth compounds
US11590207B2 (en) 2016-09-29 2023-02-28 Ascendis Pharma Bone Diseases A/S Dosage regimen for a controlled-release PTH compound
US11564974B2 (en) 2016-09-29 2023-01-31 Ascendis Pharma Growth Disorders A/S Combination therapy with controlled-release CNP agonists
WO2018175788A1 (en) 2017-03-22 2018-09-27 Genentech, Inc. Hydrogel cross-linked hyaluronic acid prodrug compositions and methods
US11642415B2 (en) 2017-03-22 2023-05-09 Ascendis Pharma A/S Hydrogel cross-linked hyaluronic acid prodrug compositions and methods
WO2019185706A1 (en) 2018-03-28 2019-10-03 Ascendis Pharma A/S Conjugates
WO2019185705A1 (en) 2018-03-28 2019-10-03 Ascendis Pharma A/S Il-2 conjugates
WO2019219896A1 (en) 2018-05-18 2019-11-21 Ascendis Pharma Bone Diseases A/S Starting dose of pth conjugates
WO2020064844A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Treatment of infections
WO2020064847A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Degradable hyaluronic acid hydrogels
WO2020064846A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Novel hydrogel conjugates
WO2020141221A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Conjugates of pattern recognition receptor agonists
WO2020141225A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Minimization of systemic inflammation
WO2020141222A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Sustained local drug levels for innate immune agonists
WO2020141223A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Induction of sustained local inflammation
WO2020165081A1 (en) 2019-02-11 2020-08-20 Ascendis Pharma Growth Disorders A/S Dry pharmaceutical formulations of cnp conjugates
WO2020165087A1 (en) 2019-02-11 2020-08-20 Ascendis Pharma Bone Diseases A/S Liquid pharmaceutical formulations of pth conjugates
WO2020254612A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Controlled-release tyrosine kinase inhibitor compounds with localized pd properties
WO2020254613A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Controlled-release tyrosine kinase inhibitor compounds with localized pk properties
WO2020254607A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 compounds with localized pd properties
WO2020254611A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 conjugates
WO2020254617A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 compounds with localized pk properties
WO2020254609A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Tyrosine kinase inhibitor conjugates
WO2021144249A1 (en) 2020-01-13 2021-07-22 Ascendis Pharma Bone Diseases A/S Hypoparathyroidism treatment
WO2021224169A1 (en) 2020-05-04 2021-11-11 Ascendis Pharma A/S Hydrogel irradiation
US11879001B2 (en) 2020-06-03 2024-01-23 Ascendis Pharma Oncology Division A/S Conjugate comprising an IL-2 moiety
WO2021245130A1 (en) 2020-06-03 2021-12-09 Ascendis Pharma Oncology Division A/S Il-2 sequences and uses thereof
WO2022043493A1 (en) 2020-08-28 2022-03-03 Ascendis Pharma Oncology Division A/S Glycosylated il-2 proteins and uses thereof
WO2022064035A1 (en) 2020-09-28 2022-03-31 Ascendis Pharma Bone Diseases A/S Improvement of physical and mental well-being of patients with hypoparathyroidism
WO2022207798A1 (en) 2021-04-01 2022-10-06 Ascendis Pharma A/S Use of long-acting growth hormone for treating inflammation-induced diseases
WO2023046732A1 (en) 2021-09-22 2023-03-30 Ascendis Pharma Bone Diseases A/S Long-acting pth compound treatments
WO2023110758A1 (en) 2021-12-13 2023-06-22 Ascendis Pharma Growth Disorders A/S Effective doses of cnp conjugates
WO2023110727A2 (en) 2021-12-13 2023-06-22 Ascendis Pharma Oncology Division A/S Novel cancer treatments with tlr7/8 agonists
WO2023227505A1 (en) 2022-05-23 2023-11-30 Ascendis Pharma Growth Disorders A/S Liquid pharmaceutical formulations of cnp compounds

Also Published As

Publication number Publication date
CA2723263A1 (en) 2009-11-26
JP2011520983A (en) 2011-07-21
EP2288261A4 (en) 2013-09-25
WO2009143412A3 (en) 2010-04-15
US20110105413A1 (en) 2011-05-05
TW201004648A (en) 2010-02-01
EP2288261A2 (en) 2011-03-02

Similar Documents

Publication Publication Date Title
EP2288261A2 (en) Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
JP6522814B2 (en) Oligomers and oligomer conjugates
JP2012509066A (en) Releasable conjugates for nucleic acid delivery systems
KR20090055623A (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery
US20090202573A1 (en) Polymeric conjugates containing positively-charged moieties
TW201021853A (en) Releasable cationic lipids for nucleic acids delivery systems
KR20090083334A (en) Polyalkylene oxides having hindered ester-based biodegradable linkers
US20100279408A1 (en) Polymeric short interfering rna conjugates
JP4649408B2 (en) Antisense oligonucleotide that inhibits “melanoma inhibitory activity (MIA)”
AU2020398192A1 (en) Peptide docking vehicle for targeted nucleic acid delivery
WO2010120980A1 (en) Methods for inhibiting angiogenesis with multi-arm polymeric conjugates of 7-ethyl-10-hydroxycamptothecin
US8110559B2 (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery
JP7306653B2 (en) Novel cancer therapy using structurally enhanced S-TuD
WO2023198201A1 (en) Aptamer, conjugate, composition, method for preparing same, and use thereof
WO2023198200A1 (en) Conjugate and composition as well as preparation method therefor and use thereof
WO2022172083A2 (en) Targeted nucleic acid therapy for hepatitis b
CN116615246A (en) Multi-conjugates comprising monosubstituted homo-divalent linkers

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09751640

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2723263

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2011510720

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 12994266

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009751640

Country of ref document: EP