WO2009009712A1 - Polymeric drug delivery system containing a multi-substituted aromatic moiety - Google Patents

Polymeric drug delivery system containing a multi-substituted aromatic moiety Download PDF

Info

Publication number
WO2009009712A1
WO2009009712A1 PCT/US2008/069732 US2008069732W WO2009009712A1 WO 2009009712 A1 WO2009009712 A1 WO 2009009712A1 US 2008069732 W US2008069732 W US 2008069732W WO 2009009712 A1 WO2009009712 A1 WO 2009009712A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
group
compound
independently
zero
Prior art date
Application number
PCT/US2008/069732
Other languages
French (fr)
Inventor
Hong Zhao
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 CA2693616A priority Critical patent/CA2693616A1/en
Priority to EP08781660.9A priority patent/EP2175878A4/en
Priority to JP2010516258A priority patent/JP2010533202A/en
Publication of WO2009009712A1 publication Critical patent/WO2009009712A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/331Polymers modified by chemical after-treatment with organic compounds containing oxygen
    • C08G65/332Polymers modified by chemical after-treatment with organic compounds containing oxygen containing carboxyl groups, or halides, or esters thereof
    • C08G65/3324Polymers modified by chemical after-treatment with organic compounds containing oxygen containing carboxyl groups, or halides, or esters thereof cyclic
    • C08G65/3326Polymers modified by chemical after-treatment with organic compounds containing oxygen containing carboxyl groups, or halides, or esters thereof cyclic aromatic
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/321Polymers modified by chemical after-treatment with inorganic compounds
    • C08G65/326Polymers modified by chemical after-treatment with inorganic compounds containing sulfur
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/33331Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing imide group
    • C08G65/33337Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing imide group cyclic
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/333Polymers modified by chemical after-treatment with organic compounds containing nitrogen
    • C08G65/33379Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing nitro group
    • C08G65/33386Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing nitro group cyclic
    • C08G65/33389Polymers modified by chemical after-treatment with organic compounds containing nitrogen containing nitro group cyclic aromatic
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G65/00Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule
    • C08G65/02Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from cyclic ethers by opening of the heterocyclic ring
    • C08G65/32Polymers modified by chemical after-treatment
    • C08G65/329Polymers modified by chemical after-treatment with organic compounds
    • C08G65/337Polymers modified by chemical after-treatment with organic compounds containing other elements
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L2203/00Applications
    • C08L2203/02Applications for biomedical use

Definitions

  • the present invention relates to drug delivery systems.
  • the invention relates to polymeric-based drug delivery system having a multi-substituted aromatic moiety.
  • the aromatic moiety is conjugated to targeting groups and biologically active moieties such as therapeutic agents, enzymes, proteins and the like.
  • Medicinal agents are quite often insoluble in aqueous fluid or rapidly degraded in vivo.
  • alkaloids are often difficult to be solubilized and proteins are often prematurely degraded upon administration into the body.
  • One of the attempts to solve the obstacles is to include such medicinal agents as part of a soluble transport system.
  • Such transport systems can include permanent conjugate-based systems or prodrugs.
  • polymeric transport systems can improve the solubility and stability of medicinal agents.
  • Multifunctional therapeutics such as proteins can be employed in permanent co ⁇ jugate- based transport systems including polymers. Proteins employed in such systems maintain biological activities to achieve therapeutic effects. Examples of polymeric conjugates of proteins are described in U.S. Patent No. 4,179,337, the disclosure of which is incorporated herein by reference. Most of permanent conjugate systems include aliphatic linking moieties between polymers and amino-containing biologically active moieties. On the other hand, prodrugs are often biologically inert or substantially inactive forms of a parent or active drug. Among many factors which influence the rate of release of the parent drug, i.e. the rate of hydrolysis, the release rate is especially modified by the linkages joining the parent drug to the rest of the prodrug system.
  • Prodrugs including polymers can improve the circulating half-life of the drug.
  • the prodrug linkages can modify in vivo hydrolysis rate at a rate which eventually generates sufficient amounts of the parent drug after administration thereby providing improved control of the pharmacokinetics of therapeutic moieties like small molecule drugs and the like.
  • A is a capping group
  • R 1 is a substantially non-antigenic water-soluble polymer
  • X 1 and X' 1 are independently O, S, SO, SO 2 , NR 6 or a bond
  • Ar and Ar are independently an aryl or heteroaryl moiety:
  • Y 1 and Y' 1 are independently O, S, or NR 6 ;
  • L 1 and L' 1 are independently selected bifunctional linkers;
  • D 1 and D' 1 are independently selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties;
  • R 2-5 , R' 2-5 , and R 6 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C 1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C 1-6 alkylthio, C 1-6 alkyls, C 2-6 alkenyl, C 2-6 alkynyl, C 3-19 branched alkyl, C 3-8 cycloalkyl, C 1-6 substituted alkyl, C 2-6 substituted alkenyl, C 2-6 substituted alkynyl, C 3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C 1-6 heteroalkyl, substituted C 1-6 heteroalkyl, C 1-6 alkoxy, aryloxy, C 1-6 heteroalkoxy, heteroaryloxy
  • (p), (p'), (r) and (r') are independently zero or a positive integer;
  • (q 1 ), (q' 1 ), (q 2 ), (q' 2 ), (q 3 ), (q' 3 ), (q 4 ) and (q' 4 ) are independently zero or one;
  • Q 1-4 and Q' 1-4 are independently selected from among the same moieties which can be used for R 2 or each can be:
  • R 7 and R 8 are independently selected from the same group as that which defines
  • Y 2 is O, S or NR 6 ;
  • L 3 is a bifuncional linker; (z) is zero or one;
  • (w) is zero or a positive integer
  • D 3 is selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties such as medicinal agents including small molecular weight compounds, provided that the sum of (q 1 ) + (q 2 ) + (q 3 ) + (q 4 ) is not zero and that at least one of Q 1-4 and Q' 1-4 is
  • D 3 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
  • the aromatic portion of the systems is conjugated to at least one target group and at least one biologically active moiety.
  • R 1 includes a linear, branched or multi-armed poly(ethylene glycol) residue with molecular weight of from about 5,000 to about 60,000.
  • (p) is zero or one and (1) is 0, 1 or 2.
  • R 2-5 , R' 2-5 , R 7 and R 8 are selected from among hydrogen, methyl and ethyl, and each is more preferably hydrogen.
  • the polymeric delivery systems described herein include a novel linker which can form a permanent bond such as amide or carbamate bond between polymers and biologically active moieties.
  • the polymeric systems are based on an aromatic structure which is built as part of the PEG backbone permanently and activated as PEG acid ester such as NHS ester.
  • PEG acid ester such as NHS ester.
  • the activated forms can react with an amino group to form the amide bond.
  • the polymeric delivery systems include a releasable bond between the polymers and biologically active moieties. These polymeric systems can release the parent compound in vivo upon chemical hydrolysis or enzymatic metabolism.
  • polymeric delivery systems have improved stability.
  • hydrophobic microenvironment around the covalent linkage between polymers and a moiety such as functional groups, biologically active moieties and targeting groups, protects the covalent linkage from exposure to basic aqueous medium or enzymes which can modify the covalent linkage, thereby stabilizing the covalent linkage.
  • the stability of the polymeric systems also allows long-term storage prior to attaching to targeting groups or biologically active moieties.
  • Another advantage of the polymeric systems described herein allows attaching a second agent. Substitution can be easily arranged on the aromatic ring so that artisans in the art can attach a second drug to have a synergistic effect for therapy or a targeting group for selectively targeted delivery. A further advantage is that the aromatic moiety allows the polymeric systems to become
  • Yet another advantage is that multiple steps previously required to attach a second agent can be avoided.
  • certain bifunctional groups can be directly attached to a second agent and therefore eliminate steps for activating the polymeric systems.
  • the term "residue” shall be understood to mean that portion of a compound, to which it refers, i.e. PEG, etc. that remains after it has undergone a substitution reaction with another compound.
  • a biologically active moiety and "a residue of a biologically active moiety” shall be understood to mean that portion of a biologically active compound which remains after the biologically active compound has undergone a substitution reaction in which the transport carrier portion has been attached.
  • 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.
  • alkyl refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
  • alkyl also includes alkyl-thio-alkyl, alkoxyalkyl, cycloalkylalkyl, heterocycloalkyl, C 1-6 hydrocarbonyl, groups.
  • 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.
  • the substituted group(s) preferably include halo, oxy, azido, nitio, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C 1-6 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. C 1-6 hydrocarbonyl, aryl, and amino groups.
  • alkenyl 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 include 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, C 1-6 hydrocarbonyl, aryl, and amino groups.
  • 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 include 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, C 1-6 hydrocarbonyl, aryl, and amino groups.
  • 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.
  • cycloalkyl refers to a C 3-8 cyclic hydrocarbon.
  • examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • cycloalkenyl 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 alklyl group substituted with a C 3-8 cycloalkyl group.
  • examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
  • alkoxy refers to an alkyl group of 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 refers to an aryl group substituted with an alkyl group.
  • an "aralkyl” group refers to an alkyl group substituted with an aryl group.
  • alkoxyalkyl refers to an alkyl group substituted with an alkloxy group.
  • alkyl-thio-alkyl refers to an alkyl-S- alkyl thioether, for example, methylthiomethyl or methylthioethyl.
  • 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 and “alkylamino” refer to specific N-substituted organic radicals with acyl and alkyl 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, morpholine, 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.
  • 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.
  • 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.
  • bond shall be understood to mean that an atom is absent and moieties adjacent to the group designated as “bond” are linked directly.
  • the pharmaceutically active compounds include small molecular weight molecules.
  • the pharmaceutically active compounds have a molecular weight of less than about 1,500 daltons and optionally derivarized with amine-, hydroxyl- or thiol- containing moieties to provide a reactive site for conjugation with polymer.
  • successful treatment i.e. tumor growth inhibition or inhibition of inflammation
  • successful treatment can be defined by obtaining e.g., 10% or higher (i.e. 20% 30%, 40%) down regulation or up-regulation of genes associated with cancer or inflammation.
  • FIG. 1 schematically illustrates methods of synthesis described in Examples 1-7.
  • A is a capping group
  • R 1 is a substantially non-antigenic water-soluble polymer
  • X 1 and X' 1 are independently O, S, SO, SO 2 , NR 6 or a bond; Ar and Ar' are independently an aryl or heteroaryl moiety; Y 1 and Y' 1 are independently O, S, or NR 6 , and preferably Y 1 and Y' 1 are O; L 1 and L' 1 are independently selected bifunctional linkers; D 1 and D' 1 are independently selected from among hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties such as medicinal agents including small molecular weight compounds; R 2-5 , R' 2-5 , and R 6 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C 1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C 1-6 alkylthio, C 1-6 alkyls, C 2
  • (p), (p'), (r) and (r') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and most preferably 0, 1 or 2;
  • (q 1 ), (q' 1 ), (q 2 ), (q' 2 ), to (q' 3 ), (q 4 ) and (q' 4 ) are independently zero or one; (s) and (s') are independently zero or a positive integer, preferably from about 0 to about
  • Q 1-4 and Q' 1-4 are independently selected from among the same moieties which can be used for R 2 or each can be:
  • R 7 and R 8 are independently selected from among the same moieties which can be used for R 2 ;
  • Y 2 is O, S or NR 6 , and preferably Y 2 is O; L 3 is a bifuncional linker; (z) is zero or one;
  • (w) is zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), more preferably zero, 1 or 2, and yet more preferably (z) is 0 and (w) is 1 ; and D 3 is selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties provided that the sum of (q 1 ) + (q 2 ) + (q 3 ) + (q 4 ) is not zero (preferably, the sum of (q 1 ) + (q 2 ) + (q 3 ) + (q 4 ) or the sum of (q' 1 ) + (q' 2 ) + (q' 3 ) + (q' 4 ) is 1), and that at least one (e.g, 1, 2, 3 or 4, preferably 1 or 2, and more preferably 1) of Q 1-4 and Q' 1-4 is
  • D 3 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
  • the leaving group is preferably selected from among N-hydroxysuccinimidyl, para-nitrophenoxy, ortho-nitrophenoxy and C 1 -C 6 alkyloxy
  • the functional group is preferably selected from among maleimidyl, vinyl, and residues of sulfone.
  • the substituents contemplated for substitution can include, for example, acyl, amino, amido, amidine, ara-alkyl, aryl, azido, alkylmercapto, arylmercapto, carbonyl, carboxylate, cyano, ester, ether, formyl, halogen, heteroaryl, heterocycloalkyl, hydroxy, imino, nitro, thiocarbonyl, thioester, thioacetate, thioformate, alkoxy, phosphoryl, phosphonate, phosphinate, silyl, sulfhydryl, sulfate, sulfonate, sulfamoyl, sulfonamide, and sulfonyl.
  • the compounds described herein include at least one targeting group and at least one biologically active moiety.
  • both (p) and (r) are zero or one.
  • (p) is 0 and (r) is 2.
  • the polymeric portion attached to the phenyl ring would not be in an ortho position in relation to D 1 , D' 1 or D 3 .
  • C(R 2 )(R 3 ) is the same or different when (p) and/or (p') are equal to or greater than 2.
  • C(R 4 )(R 5 ) is the same or different when (r) and/or (r') are equal to or greater than 2.
  • each of L 1 , L' 1 and L 3 are the same or different when (s) and (w) are equal to or greater than 2.
  • the biological moieties include amine containing moieties, hydroxyl containing moieties and thiol containing moieties.
  • A can be selected from among H, NH 2 , OH, CO 2 H, C 1-6 alkoxy, and C 1-6 alkyls.
  • A can be methyl, ethyl, methoxy, ethoxy, H, and OH.
  • A is more preferably methyl or methoxy.
  • compounds described herein have the formula:
  • A is a capping group
  • both (p) and (r) are zero or 1 ; or (p) is zero and (r) is 2. More preferably, (z) is 0 and (w) is 1.
  • One particular embodiment can have the formula:
  • R 7 and R 8 include hydrogen or CH 3 , preferably hydrogen.
  • X 1 and X' 1 include O, NH or a bond.
  • Y 1 and Y' 1 include O.
  • R 2-5 and R' 2-5 include hydrogen or CH 3 , more preferably hydrogen.
  • D 1 is a targeting group, a diagnostic agent or a biologically active moiety.
  • the multi-arm polymer includes at least one targeting group and at least one biologically active moiety.
  • the multi-arm polymeric conjugates containing one or more biologically active moieties are contemplated.
  • 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-aimed polyalkylene oxide.
  • the polyalkylene oxide includes polyethylene glycol and polypropylene glycol.
  • the polyalkylene oxide has an average molecular weight 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 alternatively from about 20,000 to about 45,000 daltons (preferably when small molecular weight compounds having an average molecular weight of less than 1,500 daltons (for example, up to 1,200 daltons) are conjugated to the polymer).
  • the compounds described herein include the polyalkylene oxide having an average molecular weight of from about 12,000 to about 20,000 daltons or from about 30,000 to about 45,000 daltons.
  • polymeric portion has a molecular weight of about 12,000 or 40,000 daltons.
  • the polyalkylene oxide includes polyethylene glycols and polypropylene glycols. More preferably, the polyalkylene oxide includes polyethylene glycol (PEG).
  • PEG is generally represented by the structure:
  • Y 11 and Y 12 are independently O, S, or NR 33 ;
  • R 31-33 are independently the same moieties which can be used for R 2 ;
  • (a') and (b') are independently zero or a positive integer, preferably 0-6 and more preferably 1 ;
  • (n) is an integer from about 10 to about 2300.
  • R 51-52 are polyalkylene oxide
  • Y 11 and Y 51-52 are independently O, S or NR 33 ;
  • X 21 is O, NR 6 , S, SO or SO 2
  • the polymers include multi-arm PEG-OH or "star-PEG" products such as those described in NOF Corp. Drag Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference. See also Shearwater Corporation's 2001 catalog “Polyethylene Glycol and Derivatives for Biomedical Application", the disclosure of which is incorporated herein by reference.
  • the multi-arm polymer conjugates contain four or more polymer arms and preferably four or eight polymer arms.
  • the multi-arm polyethylene glycol (PEG) residue can be any multi-arm polyethylene glycol (PEG) residue.
  • PEG polyethylene glycol
  • (x) is zero and a positive integer, i.e. from about 0 to about 28; and (n) is the degree of polymerization.
  • the multi-arm PEG has the structure:
  • the polymers have a total molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from 20,000 Da to 45,000 Da.
  • the multi-arm PEG has the structure:
  • the degree of polymerization for the multi-arm polymer (n) is from about 28 to about 350 to provide polymers having a total molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from 12,000 Da to 45,000 Da. This represents the number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer.
  • the polymers can be converted into a suitably activated polymer, using the activation techniques described in U.S. Patent Nos. 5,122,614 or 5,808,096.
  • PEG can be of the formula:
  • (u') is an integer from about 4 to about 455: and up to 3 terminal portions of the residue is/are capped with a methyl or other lower alkyl.
  • all four of the PEG arms can be converted to suitable activating groups, for facilitating attachment to aromatic groups.
  • suitable activating groups for facilitating attachment to aromatic groups.
  • 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.
  • PEG polyethylene glycol
  • PAO-based polymers one or more effectively non-antigenic materials such as dextran, polyvinyl alcohols, carbohydrate-based polymers, hydiOxypropylmethacrylamide (HPMA), polyalkylene oxides, and/or copolymers thereof can be used.
  • HPMA hydiOxypropylmethacrylamide
  • polymers having azides react with 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 imidodicarbonate (KNBoC 2 ) 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%.
  • 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 No. 11/328,662, 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 t-butyl ester of polyalkylene oxide carboxylic acid.
  • This intermediate is formed by reacting a PAO with a t-butyl haloacetate in the presence of a base such as potassium t-butoxide.
  • a base such as potassium t-butoxide.
  • Aromatic moieties (Ar) employed in the compounds described herein include a multi- substituted aromatic or heteroaromatic hydrocarbon.
  • Ar/Ar group is aromatic in nature.
  • the ⁇ electrons must be shared within a ""cloud" both above and below the plane of a cyclic molecule.
  • the number of ⁇ electrons must satisfy the H ⁇ ckle rule (4n+2).
  • the aromatic moieties include
  • aromatic moieties include:
  • J is O, S, or NRn
  • E and Z are each independently CR 12 or NR 13
  • R 11 , R 12 and R 13 can be selected from among the same moieties which can be used for R 2 .
  • Isomers of the five and six-membered rings are also contemplated as well as benzo- and dibenzo- rings such as anthracine and napthlene and their related congeners are also contemplated within the scope of the invention.
  • aromatic or heteroaromatic moieties may optionally be substituted with halogen(s) and/or side chains.
  • All structures suitable for Ar moieties of the present invention are capable of allowing the substituents on the aromatic group to be aligned within the same plane. Ortho, meta and para substituted aromatic rings can be used. D. Bifunctional 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-natural amino acids includes 2-aminoadipic 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-diaminopimelic acid, 2,3- diaminopropionic acid, N-ethylglycine, N-ethylasparagine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methyl-isoleucine, 6-N-methyl- lysine, N-methylvaline, norvaline, norleucine, and ornithine
  • L 1 , L 1 ' and L 3 can be selected from among
  • R 21-29 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 3-12 branched alkyls, C 3-8 cycloalkyls, C 1-6 substituted alkyls, C 3-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C 1-6 heteroalkyls, substituted C 1-6 heteroalkyls, C 1-6 alkoxy, phenoxy and C 1-6 heteroalkoxy;
  • (t) and (t') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and yet more preferably 0, 1 or 2; and
  • C(R 24 )(R 25 ) is the same or different when (t) or (t') is equal to or greater than 2.
  • (s1) and (s1') are independently zero or a positive integer, preferably from about 0 to about 4 (e.g., 0, 1, 2, 3, or 4), more preferably 0, 1 or 2, provided that both (r1) and (r1') are not zero simultaneously.
  • L 1 , L' 1 and L 3 include:
  • L 1 , L' 1 and L 3 include structures corresponding to those shown above but having vinyl, residues of sulfone, amino, carboxy, mercapto, hydrazide, carbazate and the like instead of maleimidyl. These bifunctional groups allow a second agent to be directly conjugated and therefore eliminate the need of attaching a functional group for conjugating to a second agent.
  • suitable leaving groups include, without limitations halogen (Br, Cl), activated carbonate, carbonyl imidazole, cyclic imide thione, isocyanate, N-hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, C 1 -C 6 alkyloxy, C 1 -C 6 alkanoyloxy, arylcarbonyloxy, ortho- nitrophenoxy, N-hydroxybenzotriazolyl, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5- trifluorophenoxy or other suitable leaving groups as will be apparent to those of ordinary skill.
  • leaving groups are to be understood as those groups which are capable of reacting with a nucleophile found on the desired target, i.e. a biologically active moiety, a diagnostic agent, a targeting moiety, a bifunctional spacer, intermediate, etc.
  • the targets thus contain a group for displacement, such as OH, NH 2 or SH groups found on proteins, peptides, enzymes, naturally or chemically synthesized therapeutic molecules such as doxorubicin, and spacers such as mono-protected diamines.
  • functional groups to link the polymeric transport systems to biologically active moieties include maleimidyl, vinyl, residues of sulfone, amino, carboxy, mercapto, hydrazide, carbazate and the like which can be further conjugated to a biologically active group.
  • D 1 , D' 1 and D 3 can be selected from among OH, methoxy, tert-butoxy, N-hydroxysuccinimidyl and maleimidyl.
  • the biologically active moieties include pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides-
  • the activated polymer of the invention can further contain a biologically active moiety as D 1 , D' 1 and D 3 which includes amine-, hydroxyl-, or thiol- containing compounds.
  • D 1 , D' 1 and D 3 which includes amine-, hydroxyl-, or thiol- containing compounds.
  • suitable compounds includes organic compounds, enzymes, proteins, polypeptides, antibodies, monoclonal antibodies, single chain antibodies or oligonucleotides, etc.
  • the pharmaceutically active compounds include small molecular weight molecules.
  • the pharmaceutically active compounds have a molecular weight of less than about 1,500 daltons and optionally derivatized with thiol containing moiety to provide reactive site for conjugation with polymer.
  • biologically active moieties include amine-, hydroxyl-, or thiol-containing compounds.
  • suitable compounds includes organic compounds, enzymes, proteins, polypeptides, antibodies, monoclonal antibodies, single chain antibodies or oligonucleotides, etc.
  • Organic compounds include, without limitation, moieties such as camptothecin and analogs (e.g., SN38 and irinotecan), and related topoisomerase I inhibitors, taxanes and paclitaxel derivatives, nucleosides including AZT, anthracycline compounds including daunorubicin, doxorubicin; p-aminoaniline mustard, melphalan, Ara-C (cytosine arabinoside) and related anti-metabolite compounds, e.g., gemcitabine, etc.
  • moieties such as camptothecin and analogs (e.g., SN38 and irinotecan), and related topoisomerase I inhibitors, taxanes and paclitaxel derivatives, nucleosides including AZT, anthracycline compounds including daunorubicin, doxorubicin; p-aminoaniline mustard, melphalan, Ara-C (cytosine arabino
  • biologically active moieties can include cardiovascular agents, anti-neoplastic, anti-infective, anti-fungal such as nystatin and amphotericin B, anti-anxiety agents, gastrointestinal agents, central nervous system-activating agents, analgesic, fertility agents, contraceptive agents, anti-inflammatory agents, steroidal agents, anti-urecemic agents, vasodilating agents, and vasoconstricting agents, etc. It is to be understood that other biologically active materials not specifically mentioned but having suitable amine-, hydroxyl- or thiol-containing groups are also intended and are within the scope of the present invention.
  • the biologically active compounds are suitable for medicinal or diagnostic use in the treatment of animals, e.g., mammals, including humans, for conditions for which such treatment is desired.
  • biologically active moieties suitable for inclusion herein there is available at least one amine-, hydroxyl-, or thiol-containing position which can react and link with a carrier portion and that there is not substantial loss of bioactivity in the form of conjugated to the polymeric delivery systems described herein.
  • parent compounds suitable for incorporation into the polymeric transport conjugate compounds of the invention may be active after hydrolytic release from the linked compound, or not active after hydrolytic release but which will become active after undergoing a further chemical process/reaction.
  • an anticancer drug that is delivered to the bloodstream by the polymeric transport system may remain inactive until entering a cancer or tumor cell, whereupon it is activated by the cancer or tumor cell chemistry, e.g., by an enzymatic reaction unique to that cell.
  • a further aspect of the invention provides the conjugate 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.
  • 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 inventive tagged conjugates are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels.
  • radioisotope labels include 131 Iodine, 125 Iodine, 99m Technetium and/or 111 Indium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo.
  • 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 include targeting groups.
  • the targeting groups include receptor ligands, an antibodies or antibody fragments, single chain antibodies, targeting peptides such as cell adhesion peptides and cell penetrating peptides (CPPs), targeting carbohydrate molecules or lectins.
  • Targeting groups enhance binding or uptake of the compounds described herein a target tissue and cell population.
  • a non-limiting list of targeting groups includes vascular endothelial cell growth factor, FGF2, somatostatin and somatostatin analogs, transferrin, melanotropin, ApoE and ApoE peptides, von Willebrand's Factor and von Willebrand's Factor peptides; adenoviral fiber protein and adenoviral fiber protein peptides; PDl and PDl peptides, EGF and EGF peptides, RGD peptides, folate, etc.
  • Other suitable targeting groups include selectin, TAT, penetratin, and Arg 9 .
  • the targeting groups can be optionally labeled with biotin, fluorescent compounds, radio-labeled compounds by art-known methods.
  • the methods of preparing the compounds described herein include reacting a polymer with an aromatic acid ester to form a polymer-aromatic acid.
  • the method includes reacting a compound of Formula (II) having the structure of:
  • a 1 is a capping group or M 1 -X' 1 -;
  • A is a capping group
  • R 1 is a substantially non-antigenic water-soluble polyinei-
  • M 1 is -OH,-SH Or-NHR 41 ;
  • M 2 is a leaving group;
  • Ar and Ar' are independently an aryl or heteroaryl moiety
  • X 1 and X' 1 are independently O, S, SO, SO 3 , NR 6 , or a bond;
  • Y 1 and Y' 1 are independently O, S, or NR 6 ;
  • L 1 and L' 1 are independently selected bifunctional linkers; D 4 and D' 4 are independently selected from among hydrogen, OH, OR 42 , leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
  • R 2-5 , R' 2-5 , R 6 and R 41 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C 1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C 1-6 alkylthio, C 1-6 alkyls, C 2-6 alkenyl, C 2-6 alkynyl, C 3-19 branched alkyl, C 3-8 cycloalkyl, C 1-6 substituted alkyl, C 2-6 substituted alkenyl, C 2-6 substituted alkynyl, C 3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C 1-6 heteroalkyl, substituted C 1-6 heteroalkyl, C 1-6 alkoxy, aryloxy, C 1-6 heteroalkoxy, heteroary
  • R 42 is C 1-6 alkyl;
  • (p), (p'), (r) and (r') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and most preferably 0, 1 or 2;
  • (s) and (s') are independently zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6) and more preferably zero, 1 or 2;
  • Q 1-4 and Q' 1-4 are independently selected from among the same moieties which can be used for R 2 or each can be:
  • R 7 and R 8 are independently selected from the same as used for R 2 ;
  • Y 2 is O, S or NR 6 ;
  • L 3 is a bifuncional linker
  • (w) is zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), more preferably zero, 1 or 2 (for exzmple, (z) is 0 and (w) is 1 and preferably
  • D 5 is selected from the group consisting of hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties; provided that the sum of (q 1 ) + (q 2 ) + (q 3 ) + (q 4 ) is not zero and that at least one of Q 1-4 and Q' 1-4 is
  • D 5 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
  • the polymeric portion attached to the phenyl ring would not be in an ortho position in relation to D 4 , D' 4 or D 5 .
  • the leaving group M 1 includes halogen (Br, Cl), activated carbonate, carbonyl imidazole, cyclic imide thione, isocyanate, N-hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, C 1 -C 6 alkyloxy, C 1 -C 6 alkanoyloxy, arylcarbonyloxy, ortho-nitrophenoxy, N-hydroxybenzotriazolyl, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5-trifluorophenoxy or other suitable leaving groups that is apparent to those of ordinary skill in the art.
  • the resulting compounds of Formula (IV) can be then deprotected to form a polymer- aromatic acid.
  • the polymer-aromatic acids are further activated with an amine or a hydroxyl containing compound.
  • a bifunctional group can be attached to the aromatic moiety to provide a functional group.
  • the functional groups can be further conjugated to a biologically active moiety, or a targeting moiety.
  • methods can include reacting a polymer containing a leaving group with an aromatic-containing moiety to form a polymer-aromatic acid.
  • the method includes: a) reacting a protected aromatic compound with a substantially non-antigenic polymer compound to form a polymer compound of Formula (Ia): b) converting the nitro moiety on the compound of Formula (Ia) under a suitable reaction condition to form an amine; c) reacting the amine with a suitable bifunctional spacer to form a compound of Formula (Ic):
  • NH-drug is an amine-contaiing biologically active moiety
  • Ab is an antibody such as monoclonal antibody, single chain antibody and active fragments thereof;
  • Polymer is a substantially non-antigenic polymer.
  • Attachment of the bifunctional group to the polymer portion is preferably carried out in the presence of a coupling agent.
  • suitable coupling agents include 1,3- diisopropylcarbodiimide (DIPC), any suitable dialkyl carbodiimides, 2-halo-1-alkyl-pyridinium halides, (Mukaiyama reagents), 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide (EDC), propane phosphonic acid cyclic anhydride (PPACA) and phenyl dichlorophosphates, etc. which are available, for example from commercial sources such as Sigma-Aldrich Chemical, or synthesized using known techniques.
  • DIPC 1,3- diisopropylcarbodiimide
  • EDC 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide
  • PPACA propane phosphonic acid cyclic anhydride
  • phenyl dichlorophosphates etc.
  • 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, triethylamine, etc. to neutralize any acids generated.
  • DMAP dimethylaminopyridine
  • the reactions can be carried out at a temperature from about 0°C up to about 22°C (room temperature).
  • D 2 is hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
  • Ab is an antibody
  • mPEG has the formula CH 3 O(CH 2 CH 2 O) n -;
  • PEG has the formula -O(CH 2 CH 2 O) n - , and
  • (n) is an integer from about 10 to about 2,300.
  • the ''S-Ab represents antibodies, such as monoclonal antibodies, single chain antibodies, and active fragments thereof.
  • (n) is an integer from about 10 to about 2300;
  • D 2 is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides;
  • Ab is an antibody.
  • the compounds prepared by the methods include:
  • (n) is an integer from about 10 to about 2300;
  • D 2 is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides;
  • Ab is an antibody.
  • Another 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 compound described herein.
  • the polymeric conjugate compounds are useful for, among other things, treating diseases which are similar to those which are treated with the parent compound, e.g. enzyme replacement therapy, neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mammals.
  • the amount of the polymeric conjugate that is administered will depend upon the amount of the parent molecule included therein. Generally, the amount of polymeric conjugate used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various polymeric conjugate compounds will vary somewhat depending upon the parent compound, molecular weight of the polymer, rate of in vivo hydrolysis, etc. Those skilled in the art will determine the optimal dosing of the polymeric transport conjugates selected based on clinical experience and the treatment indication. Actual dosages will be apparent to the artisan without undue experimentation.
  • the compounds of the present invention can be included in one or more suitable pharmaceutical compositions for administration to mammals.
  • the pharmaceutical compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral and/or parenteral routes depending upon the needs of the artisan.
  • a solution and/or suspension of the composition may be utilized, for example, as a carrier vehicle for injection or infiltration of the composition by any art known methods, e.g., by intravenous, intramuscular, intraperitoneal, subcutaneous injection and the like.
  • Such administration may also be by infusion into a body space or cavity, as well as by inhalation and/or intranasal routes.
  • the polymeric conjugates are parenterally administered to mammals in need thereof.
  • DMF N,N'-dimethylformamide
  • DSC disuccinimidyl carbonate
  • EDC 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide
  • IPA isopropanol
  • NHS N- hydroxysuccinimide
  • PEG polyethylene glycol
  • SCA-SH single-chain antibody
  • TEA triethylamine
  • a solution of 5.0 g (1.0 mmole) of mPEG 5K -OH (compound 2) in 130ml of toluene is azeotroped for 2 hours, while removing 65ml of toluene/water.
  • This solution is cooled to 25°C, followed by addition of 2.0 ml (2.0 mmole) of 1.0 molar t-BuOK in t-butanol.
  • This solution is stirred for 30 minutes at 25°C, followed by the addition of 30ml of anhydrous DMF.
  • To this reaction mixture is added dropwise, a solution of compound 1 (2.0 mmol) in anhydrous DMF. This solution is added at a rate of 10ml per 20 min.
  • Example 2 Compound 4 Compound 3 is suspended in a mixture of water and THF and is added Na 2 S 2 O 4 . The mixture is stirred overnight at room temperature. The mixture is concentrated in vacuo and the product is extracted with DCM twice. The organic layers are combined and dried over anhydrous Na 2 SO 4 , filtered, and concentrated to a minimum volume. Anhydrous ether is added to the residual solution to precipitate the product, which is collected by vacuum filtration and dried in the vacuum oven at 45 °C to give the product.
  • Example 4 Trifluoroacetic acid (TFA, 2.5 mL) is added to a solution of compound 6 (3.2 mmol) of in 5 mL of methylene chloride followed by stirring at room temperature for 30 minutes. Ether is added until the solid is precipitated. The solid is filtered and washed thoroughly with ether until all the excess TFA is removed. The acid is dried in the vacuum oven at 45 °C.
  • TFA Trifluoroacetic acid
  • PEG linker (compound 8) (0.084 mmol) is added to native (L)-asparaginase (0.00027 mmol) in 3 mL of sodium phosphate buffer (0.1 M, pH 7.8) with gentle stirring. The solution is stirred at 30 °C for 30 minutes. A GPC column (Zorbax GF-450) is used to monitor PEG conjugation. At the end of the reaction (as evidenced by the absence of native enzyme), the mixture is diluted with 12 mL of formulation buffer (0.05 M sodium phosphate, 0.85% sodium chloride, pH 7.3) and diafiltered with a Centriprep concentrator (Amicon) to remove the unreacted PEG. Dialfiltration is continued as needed at 4 °C until no more free PEG is detected by mixing equal amount of filtrate and 0.1% PMA (polymethacrylic acid in 0.1 M HCl) to give the product.
  • PMA polymethacrylic acid in 0.1 M HCl

Abstract

The present invention provides polymeric delivery systems including a multi-substituted aromatic moiety. Methods of making the polymeric delivery systems and methods of treating mammals using the same are also disclosed.

Description

POLYMERIC DRUG DELIVERY SYSTEM CONTAINING A MULTI-SUBSTITUTED AROMATIC MOIETY
CROSS-REFERENCE TO RELATED APPLICATION This application claims the benefit of priority from U.S. Provisional Patent Application
Serial No. 60/949,168 filed July 11 , 2007, the contents of which are incorporated herein by reference.
FIELD OF THE INVENTION The present invention relates to drug delivery systems. In particular, the invention relates to polymeric-based drug delivery system having a multi-substituted aromatic moiety. The aromatic moiety is conjugated to targeting groups and biologically active moieties such as therapeutic agents, enzymes, proteins and the like.
BACKGROUND OF THE INVENTION
Over the years, numerous methods have been proposed for administering biologically- effective materials. Medicinal agents are quite often insoluble in aqueous fluid or rapidly degraded in vivo. For example, alkaloids are often difficult to be solubilized and proteins are often prematurely degraded upon administration into the body. One of the attempts to solve the obstacles is to include such medicinal agents as part of a soluble transport system. Such transport systems can include permanent conjugate-based systems or prodrugs. In particular, polymeric transport systems can improve the solubility and stability of medicinal agents.
Multifunctional therapeutics such as proteins can be employed in permanent coηjugate- based transport systems including polymers. Proteins employed in such systems maintain biological activities to achieve therapeutic effects. Examples of polymeric conjugates of proteins are described in U.S. Patent No. 4,179,337, the disclosure of which is incorporated herein by reference. Most of permanent conjugate systems include aliphatic linking moieties between polymers and amino-containing biologically active moieties. On the other hand, prodrugs are often biologically inert or substantially inactive forms of a parent or active drug. Among many factors which influence the rate of release of the parent drug, i.e. the rate of hydrolysis, the release rate is especially modified by the linkages joining the parent drug to the rest of the prodrug system. Care must thus be taken to avoid the prodrugs from being eliminated through the kidney or reticular endothelial system, etc. before a sufficient amount of hydrolysis occurs to release the parent drug. Prodrugs including polymers can improve the circulating half-life of the drug. The prodrug linkages can modify in vivo hydrolysis rate at a rate which eventually generates sufficient amounts of the parent drug after administration thereby providing improved control of the pharmacokinetics of therapeutic moieties like small molecule drugs and the like. Some examples of polymeric prodrugs are described in commonly-assigned U.S. Patent Nos. 6,180,095 and 6,720,306, the contents of each of which are incorporated herein by reference.
In spite of the attempts and advances, there still continues to be a need to improve polymeric delivery platforms. The present invention addresses this need and others.
SUMMARY OF THE INVENTION
In one aspect of the invention, there are provided compounds of Formula (I):
Figure imgf000003_0001
wherein:
A is a capping group or
Figure imgf000003_0002
R1 is a substantially non-antigenic water-soluble polymer; X1 and X'1 are independently O, S, SO, SO2, NR6 or a bond; Ar and Ar are independently an aryl or heteroaryl moiety: Y1 and Y'1 are independently O, S, or NR6; L1 and L'1 are independently selected bifunctional linkers; D1 and D'1 are independently selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties;
R2-5, R'2-5, and R6 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C1-6 alkylthio, C1-6alkyls, C2-6 alkenyl, C2-6alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6 heteroalkyl, substituted C1-6heteroalkyl, C1-6alkoxy, aryloxy, C1-6 heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy, substituted and arylcarbonyloxy,
(p), (p'), (r) and (r') are independently zero or a positive integer; (q1), (q'1), (q2), (q'2), (q3), (q'3), (q4) and (q'4) are independently zero or one;
(s) and (s') are independently zero or a positive integer;
Q1-4 and Q'1-4 are independently selected from among the same moieties which can be used for R2 or each can be:
;
Figure imgf000004_0001
wherein
R7 and R8 are independently selected from the same group as that which defines
R2;
Y2 is O, S or NR6; L3 is a bifuncional linker; (z) is zero or one;
(w) is zero or a positive integer; and D3 is selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties such as medicinal agents including small molecular weight compounds, provided that the sum of (q1) + (q2) + (q3) + (q4) is not zero and that at least one of Q1-4 and Q'1-4 is
Figure imgf000005_0001
wherein at least one of D3 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
In one preferred aspect of the invention, the aromatic portion of the systems is conjugated to at least one target group and at least one biologically active moiety.
In some preferred aspects, R1 includes a linear, branched or multi-armed poly(ethylene glycol) residue with molecular weight of from about 5,000 to about 60,000. In certain embodiments, (p) is zero or one and (1) is 0, 1 or 2. In one preferred embodiment, R2-5, R'2-5, R7 and R8 are selected from among hydrogen, methyl and ethyl, and each is more preferably hydrogen.
In another aspect of the invention, there are provided methods of preparing the compounds described herein and methods of treatment using the compounds described herein.
The polymeric delivery systems described herein include a novel linker which can form a permanent bond such as amide or carbamate bond between polymers and biologically active moieties. For example, the polymeric systems are based on an aromatic structure which is built as part of the PEG backbone permanently and activated as PEG acid ester such as NHS ester. The activated forms can react with an amino group to form the amide bond.
In another aspect of the invention, the polymeric delivery systems include a releasable bond between the polymers and biologically active moieties. These polymeric systems can release the parent compound in vivo upon chemical hydrolysis or enzymatic metabolism.
One advantage of the aromatic moiety-based polymeric transport systems described herein is that the polymeric delivery systems have improved stability. Without being bound by any theories, the hydrophobic microenvironment around the covalent linkage between polymers and a moiety such as functional groups, biologically active moieties and targeting groups, protects the covalent linkage from exposure to basic aqueous medium or enzymes which can modify the covalent linkage, thereby stabilizing the covalent linkage. The stability of the polymeric systems also allows long-term storage prior to attaching to targeting groups or biologically active moieties.
Another advantage of the polymeric systems described herein allows attaching a second agent. Substitution can be easily arranged on the aromatic ring so that artisans in the art can attach a second drug to have a synergistic effect for therapy or a targeting group for selectively targeted delivery. A further advantage is that the aromatic moiety allows the polymeric systems to become
UV visible. Artisans in the art can easily and efficiently check purity of the polymeric systems and degree of reaction completion. The property, therefore, allows saving costs and time associated with analytical steps during preparation. The property also allows preparing the polymeric systems described herein in high purity and thereby having uniform pharmacokinetic properties.
Yet another advantage is that multiple steps previously required to attach a second agent can be avoided. For example, certain bifunctional groups can be directly attached to a second agent and therefore eliminate steps for activating the polymeric systems.
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, etc. that remains after it has undergone a substitution reaction with another compound.
For purposes of the present invention, the terms "a biologically active moiety" and "a residue of a biologically active moiety" shall be understood to mean that portion of a biologically active compound which remains after the biologically active compound has undergone a substitution reaction in which the transport carrier portion has been attached.
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 term "alkyl" also includes alkyl-thio-alkyl, alkoxyalkyl, cycloalkylalkyl, heterocycloalkyl, C1-6 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 include halo, oxy, azido, nitio, cyano, alkyl, alkoxy, alkyl-thio, alkyl-thio-alkyl, alkoxyalkyl, alkylamino, trihalomethyl, hydroxyl, mercapto, hydroxy, cyano, alkylsilyl, cycloalkyl, cycloalkylalkyl, heterocycloalkyl, heteroaryl, alkenyl, alkynyl, C1-6 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. C1-6 hydrocarbonyl, aryl, and amino groups.
For purposes of the present invention, the term "alkenyl" 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 include 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, C1-6 hydrocarbonyl, aryl, and amino groups.
For purposes of the present invention, the term "alkynyl" 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 include 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, C1-6 hydrocarbonyl, aryl, and amino groups. Examples of "alkynyl" include propargyl, propyne, and 3-hexyne. For purposes of the present invention, the term "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. 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.
For purposes of the present invention, the term "cycloalkyl" refers to a C3-8 cyclic hydrocarbon. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
For purposes of the present invention, the term "cycloalkenyl" 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.
For purposes of the present invention, the term "cycloalkylalkyl" refers to an alklyl group substituted with a C3-8 cycloalkyl group. Examples of cycloalkylalkyl groups include cyclopropylmethyl and cyclopentylethyl.
For purposes of the present invention, the term "alkoxy" as used herein refers to an alkyl group of 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. For purposes of the present invention, an "alkylaryl" group refers to an aryl group substituted with an alkyl group.
For purposes of the present invention, an "aralkyl" group refers to an alkyl group substituted with an aryl group.
For purposes of the present invention, the term "alkoxyalkyl" group refers to an alkyl group substituted with an alkloxy group.
For purposes of the present invention, the term "alkyl-thio-alkyl" refers to an alkyl-S- alkyl thioether, for example, methylthiomethyl or methylthioethyl.
For purposes of the present invention, 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 "acylamino" and "alkylamino" refer to specific N-substituted organic radicals with acyl and alkyl substituent groups, respectively.
For purposes of the present invention, the terra "alkylcarbonyl" refers to a carbonyl group substituted with alkyl group. For purposes of the present invention, the terms "halogen" or "halo" as used herein refer to fluorine, chlorine, bromine, and iodine.
For purposes of the present invention, the term "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, morpholine, piperidine, tetrahydrofuran, pyrrolidine, and pyrazole. Preferred heterocycloalkyl groups include piperidinyl, piperazinyl, morpholinyl, and pyrolidinyl.
For purposes of the present invention, the term "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. 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.
For purposes of the present invention, the term "heteroatom" 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 purposes 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 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.
For purposes of the present invention, the term "bond" shall be understood to mean that an atom is absent and moieties adjacent to the group designated as "bond" are linked directly.
For purposes of the present invention, it shall be understood to mean that the pharmaceutically active compounds include small molecular weight molecules. Typically, the pharmaceutically active compounds have a molecular weight of less than about 1,500 daltons and optionally derivarized with amine-, hydroxyl- or thiol- containing moieties to provide a reactive site for conjugation with polymer.
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.
Broadly speaking, successful treatment (i.e. tumor growth inhibition or inhibition of inflammation) shall be deemed to occur when the desired response is obtained, compared to that observed in the absence of the treatment with the compound described herein. For example, successful treatment, (i.e., tumor growth inhibition or inhibition of inflammation) can be defined by obtaining e.g., 10% or higher (i.e. 20% 30%, 40%) down regulation or up-regulation of genes associated with cancer or inflammation.
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 methods of synthesis described in Examples 1-7.
DETAILED DESCRIPTION OF THE INVENTION A. Overview In one aspect of the present invention, there are provided compounds of Formula (I):
Figure imgf000011_0001
wherein: A is a capping group or
Figure imgf000011_0002
R1 is a substantially non-antigenic water-soluble polymer;
X1 and X'1 are independently O, S, SO, SO2, NR6 or a bond; Ar and Ar' are independently an aryl or heteroaryl moiety; Y1 and Y'1 are independently O, S, or NR6, and preferably Y1 and Y'1 are O; L1 and L'1 are independently selected bifunctional linkers; D1 and D'1 are independently selected from among hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties such as medicinal agents including small molecular weight compounds; R2-5, R'2-5, and R6 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C1-6 alkylthio, C1-6alkyls, C2-6 alkenyl, C2-6alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6, heteroalkyl, substituted C1-6heteroalkyl, C1-6 alkoxy, aryloxy, C1-6 heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy, substituted and arylcarbonyloxy;
(p), (p'), (r) and (r') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and most preferably 0, 1 or 2;
(q1), (q'1), (q2), (q'2), to (q'3), (q4) and (q'4) are independently zero or one; (s) and (s') are independently zero or a positive integer, preferably from about 0 to about
6 (e.g., 0, 1, 2, 3, 4, 5 or 6), more preferably zero, 1 or 2, and yet more preferably zero or 1 ;
Q1-4 and Q'1-4 are independently selected from among the same moieties which can be used for R2 or each can be:
Figure imgf000012_0001
wherein
R7 and R8 are independently selected from among the same moieties which can be used for R2;
Y2 is O, S or NR6, and preferably Y2 is O; L3 is a bifuncional linker; (z) is zero or one;
(w) is zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), more preferably zero, 1 or 2, and yet more preferably (z) is 0 and (w) is 1 ; and D3 is selected from among hydrogen, OH, leaving groups, functional groups, targeting groups and biologically active moieties provided that the sum of (q1) + (q2) + (q3) + (q4) is not zero (preferably, the sum of (q1) + (q2) + (q3) + (q4) or the sum of (q'1) + (q'2) + (q'3) + (q'4) is 1), and that at least one (e.g, 1, 2, 3 or 4, preferably 1 or 2, and more preferably 1) of Q1-4 and Q'1-4 is
Figure imgf000013_0001
wherein at least one of D3 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero. With this respect, the leaving group is preferably selected from among N-hydroxysuccinimidyl, para-nitrophenoxy, ortho-nitrophenoxy and C1-C6 alkyloxy, and the functional group is preferably selected from among maleimidyl, vinyl, and residues of sulfone.
Within those aspects of the invention, the substituents contemplated for substitution, where said moieties corresponding to R2-5, R'2-5, R6, R7 and R8 are indicated as being possibly substituted can include, for example, acyl, amino, amido, amidine, ara-alkyl, aryl, azido, alkylmercapto, arylmercapto, carbonyl, carboxylate, cyano, ester, ether, formyl, halogen, heteroaryl, heterocycloalkyl, hydroxy, imino, nitro, thiocarbonyl, thioester, thioacetate, thioformate, alkoxy, phosphoryl, phosphonate, phosphinate, silyl, sulfhydryl, sulfate, sulfonate, sulfamoyl, sulfonamide, and sulfonyl.
In one preferred aspect, the compounds described herein include at least one targeting group and at least one biologically active moiety.
In some preferred embodiments, both (p) and (r) are zero or one. Alternatively, (p) is 0 and (r) is 2.
In certain aspects of the invention, when (p) is zero, there are preferably a sufficient number of atoms, e.g., more than five or six atoms, present between X1/X' 1 and C(=Y1)/C(=Y'1), so that a releasable cyclic moiety is not formed. For example, when (p) is zero, the polymeric portion attached to the phenyl ring would not be in an ortho position in relation to D1, D'1 or D3. For purposes of the present invention, C(R2)(R3) is the same or different when (p) and/or (p') are equal to or greater than 2. For purposes of the present invention, C(R4)(R5) is the same or different when (r) and/or (r') are equal to or greater than 2.
Forpurposes of the present invention, each of L1, L'1 and L3 are the same or different when (s) and (w) are equal to or greater than 2. In another aspect of the invention, the biological moieties include amine containing moieties, hydroxyl containing moieties and thiol containing moieties.
In yet another aspect, A can be selected from among H, NH2, OH, CO2H, C1-6 alkoxy, and C1-6 alkyls. In some preferred embodiments, A can be methyl, ethyl, methoxy, ethoxy, H, and OH. A is more preferably methyl or methoxy. In one embodiment, compounds described herein have the formula:
Figure imgf000014_0001
wherein A is a capping group or
Figure imgf000014_0002
In certain particular embodiments, compounds described herein have the formula,
Figure imgf000014_0003
or
Figure imgf000015_0001
In one preferred embodiment, the sum of (q1) + (q2) + (q3) + (q4) or (q'1) + (q'2) + (q'3) + (q'4) equals to 1. Preferred polymeric compounds can have the formula:
Figure imgf000015_0002
Figure imgf000015_0003
Figure imgf000015_0004
Figure imgf000016_0001
In a further preferred embodiment, both (p) and (r) are zero or 1 ; or (p) is zero and (r) is 2. More preferably, (z) is 0 and (w) is 1. One particular embodiment can have the formula:
Figure imgf000016_0002
In another embodiment, R7 and R8 include hydrogen or CH3, preferably hydrogen. In yet another embodiment, X1 and X'1 include O, NH or a bond. In yet anther embodiment, Y1 and Y'1 include O.
In yet another preferred embodiment, R2-5 and R'2-5 include hydrogen or CH3, more preferably hydrogen.
In another embodiment, the compounds described herein have the formula:
Figure imgf000017_0001
wherein
Ra is
Figure imgf000017_0002
and at least one of D1 is a targeting group, a diagnostic agent or a biologically active moiety. Preferably the multi-arm polymer includes at least one targeting group and at least one biologically active moiety. The multi-arm polymeric conjugates containing one or more biologically active moieties are contemplated.
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-aimed polyalkylene oxide. In some preferred embodiments, the polyalkylene oxide includes polyethylene glycol and polypropylene glycol.
The polyalkylene oxide has an average molecular weight 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 alternatively from about 20,000 to about 45,000 daltons (preferably when small molecular weight compounds having an average molecular weight of less than 1,500 daltons (for example, up to 1,200 daltons) are conjugated to the polymer). In some particularly preferred embodiments, the compounds described herein include the polyalkylene oxide having an average molecular weight of from about 12,000 to about 20,000 daltons or from about 30,000 to about 45,000 daltons. In one particular embodiment, polymeric portion has a molecular weight of about 12,000 or 40,000 daltons. The polyalkylene oxide includes polyethylene glycols and polypropylene glycols. More preferably, the polyalkylene oxide includes polyethylene glycol (PEG). PEG is generally represented by the structure:
-O-(CH2CH2O)n- 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 invention can be selected from among: -X11-(CH2CH2O)n-CH2CH2X11- , -X11-(CH2CH2O)n-CH2C(=Y11)-X11- ,
-X11-C(=Y11)-(CH2)a-Y12-(CH2CH2O)n-CH2CH2-Y12-(CH2)a-C(=Y11)-X1,- , and -X11-(CR31R32)a-Y12-(CH2)b-O-(CH2CH2O)n-(CH2)b-Y12-(CR31R32)a-X11- , wherein: X11 is O, S, SO, SO2, NR33 or a bond;
Y11 and Y12 are independently O, S, or NR33;
R31-33 are independently the same moieties which can be used for R2;
(a') and (b') are independently zero or a positive integer, preferably 0-6 and more preferably 1 ; and
(n) is an integer from about 10 to about 2300.
Branched or U-PEG derivatives are described in U.S. Patent Nos. 5,643,575, 5,919,455, 6,113,906 and 6,566,506, the disclosure of each of which is incoiporated herein by reference. A non-limiting list of such polymers corresponds to polymer systems (i) - (vii) with the following structures:
Figure imgf000019_0001
Figure imgf000020_0001
wherein: R51-52 are polyalkylene oxide;
Y11 and Y51-52 are independently O, S or NR33; X21 is O, NR6, S, SO or SO2
(c') and (t1') are independently 0 or a positive integer such as 1, 2, 3, 4 and 5; (d') is 0 or l; mPEG is methoxy PEG wherein PEG is previously defined and a total molecular weight of the polymer portion is from about 2,000 to about 100,000 daltons. R6 and R33 are previously defined. In yet another aspect, the polymers include multi-arm PEG-OH or "star-PEG" products such as those described in NOF Corp. Drag Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference. See also Shearwater Corporation's 2001 catalog "Polyethylene Glycol and Derivatives for Biomedical Application", the disclosure of which is incorporated herein by reference. The multi-arm polymer conjugates contain four or more polymer arms and preferably four or eight polymer arms.
For purposes of illustration and not limitation, the multi-arm polyethylene glycol (PEG) residue can be
Figure imgf000021_0001
wherein:
(x) is zero and a positive integer, i.e. from about 0 to about 28; and (n) is the degree of polymerization.
In one particular embodiment of the present invention, the multi-arm PEG has the structure:
Figure imgf000021_0002
wherein (n )is a positive integer. In one preferred embodiment of the invention, the polymers have a total molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from 20,000 Da to 45,000 Da.
In yet another particular embodiment, the multi-arm PEG has the structure:
Figure imgf000022_0003
or
Figure imgf000022_0001
wherein (n) is a positive integer. In one preferred embodiment of the invention, the degree of polymerization for the multi-arm polymer (n) is from about 28 to about 350 to provide polymers having a total molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from 12,000 Da to 45,000 Da. This represents the number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer.
The polymers can be converted into a suitably activated polymer, using the activation techniques described in U.S. Patent Nos. 5,122,614 or 5,808,096. Specifically, such PEG can be of the formula:
Figure imgf000022_0004
or
Figure imgf000022_0002
wherein: (u') is an integer from about 4 to about 455: and up to 3 terminal portions of the residue is/are capped with a methyl or other lower alkyl.
In some preferred embodiments, all four of the PEG arms can be converted to suitable activating groups, for facilitating attachment to aromatic groups. Such compounds prior to conversion include:
Figure imgf000023_0001
Figure imgf000024_0001
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. In a further embodiment, and as an alternative to PAO-based polymers, one or more effectively non-antigenic materials such as dextran, polyvinyl alcohols, carbohydrate-based polymers, hydiOxypropylmethacrylamide (HPMA), polyalkylene oxides, and/or copolymers thereof can be used. See also commonly-assigned U.S. Patent No. 6,153,655, the contents of which are incorporated herein by reference. It will be understood by those of ordinary skill that the same type of activation is employed as described herein as for PAO's such as PEG. Those of ordinary skill in the art will further realize that the foregoing list is merely illustrative and that all polymeric materials having the qualities described herein are contemplated. 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 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 Nos. 11/508,507 and 11/537,172, the contents of each of which are incorporated by reference. For example, polymers having azides react with 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 imidodicarbonate (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%.
In alternative 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 No. 11/328,662, 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 t-butyl ester of polyalkylene oxide carboxylic acid. This intermediate is formed by reacting a PAO with a t-butyl haloacetate in the presence of a base such as potassium t-butoxide. Once the t-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.
C. Aromatic Moieties
Aromatic moieties (Ar) employed in the compounds described herein include a multi- substituted aromatic or heteroaromatic hydrocarbon. A key feature is that the Ar/Ar group is aromatic in nature. Generally, the π electrons must be shared within a ""cloud" both above and below the plane of a cyclic molecule. Furthermore, the number of π electrons must satisfy the Hϋckle rule (4n+2). Those of ordinary skill will realize that a myriad of moieties will satisfy the aromatic requirement of the moiety and thus are suitable for use herein.
In one particular embodiment of the invention, the aromatic moieties include
Figure imgf000026_0001
Other suitable aromatic moieties include:
Figure imgf000026_0002
wherein J is O, S, or NRn; E and Z are each independently CR12 or NR13; and R11, R12 and R13 can be selected from among the same moieties which can be used for R2.
Isomers of the five and six-membered rings are also contemplated as well as benzo- and dibenzo- rings such as anthracine and napthlene and their related congeners are also contemplated within the scope of the invention.
Furthermore, the aromatic or heteroaromatic moieties may optionally be substituted with halogen(s) and/or side chains. All structures suitable for Ar moieties of the present invention are capable of allowing the substituents on the aromatic group to be aligned within the same plane. Ortho, meta and para substituted aromatic rings can be used. D. Bifunctional 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-aminoadipic 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-diaminopimelic acid, 2,3- diaminopropionic acid, N-ethylglycine, N-ethylasparagine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methyl-isoleucine, 6-N-methyl- lysine, N-methylvaline, norvaline, norleucine, and ornithine. Some preferred amino acid residues are selected from glycine, alanine, methionine and sarcosine, and more preferably, glycine.
Alternatively, L1, L1' and L3 can be selected from among
-[C(=O)]v(CR22R23)t[C{=O)]v'- ,
-[C(=O)]v(CR22R23)t-O[C(=O)]v'- ,
-[C(=O)]v(CR22R23)t-NR26[C(=O)]v'- , -[C(=O)]vO(CR22R23)t[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tO[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tNR26[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)t[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)tO[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tNR26[C(=O)]v'- ,
-[C(=O)]v(CR22R23)tO-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23)tNR26-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23)tS-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tO-(CR28R29)t'[C(=O)]v'- , -[C(=O)]vO(CR22R23)tNR26-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tS-(CR28R29)t'[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tO-(CR28R29)t'[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tNR26-(CR28R29)t'[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tS-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23CR28R29O)tNR26[C(=O)]v'- , -[C(=O)]v(CR22R23CR28R29O)t[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR28R29O)tNR26[C(=O)]v'- , -[C(=O)]vO(CR22R23CR28R29O)t[C(=O)]v'- , -[C(=O)]vNR21(CR22R23CR28R29O)tNR26[C(=O)]v'- , -[C(=O)]vNR21(CR22R23CR28R29O)t[C(=O)]v'- , -[C(=O)]v(CR22R23CR28R29O)t(CR24R25)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR28R29O)t(CR24R25)t'[C(=O)]v'- , -[C(=O)]vNR21(CR22R23CR28R29O)t(CR24R25)t'[C(=O)]v'- , -[C(=O)]v(CR22R23CR28R29O)t(CR24R25)t'O[C(=O)]v'- , -[C(=O)]v(CR22R23)t(CR24R25CR28R29O)t'[C(=O)]v'- , -[C(=O)]v(CR22R23)t(CR24R25CR28R29O)t'NR26[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR28R29O)t(CR24R2s)t'O[C(=O)]v'- , -[C(=O)]vO(CR22R23)t(CR24R25CR28R29O)t'[C(=O)]v'- , -[C(=O)]vO(CR22R23)t(CR24CR25CR28R29O)t'NR26[C(=O)]v'- , -[C(=O)]vNR21(CR22R23CR28R29O)t(CR24R25)t'O[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)t(CR24R25CR28R29O)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)t(CR24R25CR28R29O)t'NR26[C(=O)]v'- ,
Figure imgf000028_0001
Figure imgf000029_0001
and
Figure imgf000029_0002
wherein:
R21-29 are independently selected from the group consisting of hydrogen, C1-6 alkyls, C3-12 branched alkyls, C3-8 cycloalkyls, C1-6 substituted alkyls, C3-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1-6 heteroalkyls, substituted C1-6heteroalkyls, C1-6alkoxy, phenoxy and C1-6heteroalkoxy;
(t) and (t') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and yet more preferably 0, 1 or 2; and
(v) and (v') are independently zero or 1.
For purposes of the present invention, C(R24)(R25) is the same or different when (t) or (t') is equal to or greater than 2.
Preferably, L1, L1' and L3 can be selected from among: -[C(=O)]r1NH(CH2)2CH-N-NHC(=O)-(CH2)2- ,
-[C(=O)]r1NH(CH2)2(CH2CH2O)2(CH2)2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)(CH2CH2O)2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1NH(CH2CH2)s1'[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1S(CH2CH2)s1'[C(=O)]r1'- , -[C(=O)]r1NH(CH2CH2)(CH2CH2O)[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1O(CH2CH2)s1'[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)(CH2)NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)2(CH2)[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)s1(CH2)s1'[C(=O)]r1'- , -[C(=O)]r1NHCH2CH2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)2O[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)[C(=O)]r1'- , -[C(=O)]r1NH(CH2CH2O)2[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2)3[C(=O)]r1'- ,
-[C(=O)]r1O(CH2CH2O)2(CH2)[C(=O)]r1'- ,
-[C(=O)]r1O(CH2)2NH(CH2)2[C(=O)]r1'- , -[C(=O)]r1O(CH2CH2O)2NH[C(=O)]r1'- ,
-[C(=O)]r1O(CH2)2O(CH2)2[C(=O)]r1'- ,
-[C(=O)]r1O(CH2)2S(CH2)2[C(=O)]r1'- ,
-[C(=O)]r1O(CH2CH2)NH[C(=O)]r1'- ,
-[C(=O)]r1O(CH2CH2)O[C(=O)]r1'- , -[C(=O)]r1O(CH2)3NH[C(=O)]r1'- ,
-[C(=O)]r1O(CΗ2)3O[C(=O)]r1'- ,
-[C(=O)]r1O(CH2)3[C(=O)]r1'- ,
-[C(=O)]r1CH2NHCH2[C(=O)]r1'- ,
-[C(=O)]rCH2OCH2[C(=O)]r1'- , -[C(=O)]r1CH2SCH2[C(=O)]r1'- ,
-[C(=O)]r1S(CH2)3[C(=O)]r1'- ,
-[C(=O)]r1(CH2)3[C(=O)]r1'- ,
Figure imgf000030_0001
wherein
(r1) and (r1') are independently zero or 1 ; and
(s1) and (s1') are independently zero or a positive integer, preferably from about 0 to about 4 (e.g., 0, 1, 2, 3, or 4), more preferably 0, 1 or 2, provided that both (r1) and (r1') are not zero simultaneously.
In yet further alternative aspects of the invention, L1, L'1 and L3 include:
Figure imgf000031_0001
In a further embodiment and as an alternative, L1, L'1 and L3 include structures corresponding to those shown above but having vinyl, residues of sulfone, amino, carboxy, mercapto, hydrazide, carbazate and the like instead of maleimidyl. These bifunctional groups allow a second agent to be directly conjugated and therefore eliminate the need of attaching a functional group for conjugating to a second agent.
E. D1, D'1 and D3 Groups
1. Leaving Groups and Functional Groups
In some aspects, suitable leaving groups include, without limitations halogen (Br, Cl), activated carbonate, carbonyl imidazole, cyclic imide thione, isocyanate, N-hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, C1-C6 alkyloxy, C1-C6 alkanoyloxy, arylcarbonyloxy, ortho- nitrophenoxy, N-hydroxybenzotriazolyl, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5- trifluorophenoxy or other suitable leaving groups as will be apparent to those of ordinary skill.
For purposes of the present invention, leaving groups are to be understood as those groups which are capable of reacting with a nucleophile found on the desired target, i.e. a biologically active moiety, a diagnostic agent, a targeting moiety, a bifunctional spacer, intermediate, etc. The targets thus contain a group for displacement, such as OH, NH2 or SH groups found on proteins, peptides, enzymes, naturally or chemically synthesized therapeutic molecules such as doxorubicin, and spacers such as mono-protected diamines. In some preferred embodiments, functional groups to link the polymeric transport systems to biologically active moieties include maleimidyl, vinyl, residues of sulfone, amino, carboxy, mercapto, hydrazide, carbazate and the like which can be further conjugated to a biologically active group. In particularly preferred embodiments of the invention, D1, D'1 and D3 can be selected from among OH, methoxy, tert-butoxy, N-hydroxysuccinimidyl and maleimidyl.
2. Biologically Active Moieties
A wide variety of biologically active moieties can be attached to the activated polymers described herein. The biologically active moieties include pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides- In addition, the activated polymer of the invention can further contain a biologically active moiety as D1, D' 1 and D3 which includes amine-, hydroxyl-, or thiol- containing compounds. A non-limiting list of such suitable compounds includes organic compounds, enzymes, proteins, polypeptides, antibodies, monoclonal antibodies, single chain antibodies or oligonucleotides, etc.
For purposes of the present invention, it shall be understood to mean that the pharmaceutically active compounds include small molecular weight molecules. Typically, the pharmaceutically active compounds have a molecular weight of less than about 1,500 daltons and optionally derivatized with thiol containing moiety to provide reactive site for conjugation with polymer.
In some aspects of the invention, biologically active moieties include amine-, hydroxyl-, or thiol-containing compounds. A non-limiting list of such suitable compounds includes organic compounds, enzymes, proteins, polypeptides, antibodies, monoclonal antibodies, single chain antibodies or oligonucleotides, etc. Organic compounds include, without limitation, moieties such as camptothecin and analogs (e.g., SN38 and irinotecan), and related topoisomerase I inhibitors, taxanes and paclitaxel derivatives, nucleosides including AZT, anthracycline compounds including daunorubicin, doxorubicin; p-aminoaniline mustard, melphalan, Ara-C (cytosine arabinoside) and related anti-metabolite compounds, e.g., gemcitabine, etc. Alternatively, biologically active moieties can include cardiovascular agents, anti-neoplastic, anti-infective, anti-fungal such as nystatin and amphotericin B, anti-anxiety agents, gastrointestinal agents, central nervous system-activating agents, analgesic, fertility agents, contraceptive agents, anti-inflammatory agents, steroidal agents, anti-urecemic agents, vasodilating agents, and vasoconstricting agents, etc. It is to be understood that other biologically active materials not specifically mentioned but having suitable amine-, hydroxyl- or thiol-containing groups are also intended and are within the scope of the present invention.
In another aspect of the invention, the biologically active compounds are suitable for medicinal or diagnostic use in the treatment of animals, e.g., mammals, including humans, for conditions for which such treatment is desired.
The only limitations on the types of the biologically active moieties suitable for inclusion herein is that there is available at least one amine-, hydroxyl-, or thiol-containing position which can react and link with a carrier portion and that there is not substantial loss of bioactivity in the form of conjugated to the polymeric delivery systems described herein. Alternatively, parent compounds suitable for incorporation into the polymeric transport conjugate compounds of the invention, may be active after hydrolytic release from the linked compound, or not active after hydrolytic release but which will become active after undergoing a further chemical process/reaction. For example, an anticancer drug that is delivered to the bloodstream by the polymeric transport system, may remain inactive until entering a cancer or tumor cell, whereupon it is activated by the cancer or tumor cell chemistry, e.g., by an enzymatic reaction unique to that cell. A further aspect of the invention provides the conjugate 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. Thus, 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. In yet a further aspect of the invention, the inventive tagged conjugates are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels. Simply by way of example, these include 131Iodine, 125Iodine, 99mTechnetium and/or 111Indium 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.
3. Targeting Groups
In some aspects, the compounds described herein include targeting groups. The targeting groups include receptor ligands, an antibodies or antibody fragments, single chain antibodies, targeting peptides such as cell adhesion peptides and cell penetrating peptides (CPPs), targeting carbohydrate molecules or lectins. Targeting groups enhance binding or uptake of the compounds described herein a target tissue and cell population. For example, a non-limiting list of targeting groups includes vascular endothelial cell growth factor, FGF2, somatostatin and somatostatin analogs, transferrin, melanotropin, ApoE and ApoE peptides, von Willebrand's Factor and von Willebrand's Factor peptides; adenoviral fiber protein and adenoviral fiber protein peptides; PDl and PDl peptides, EGF and EGF peptides, RGD peptides, folate, etc. Other suitable targeting groups include selectin, TAT, penetratin, and Arg9.
In a further aspect of the invention, the targeting groups can be optionally labeled with biotin, fluorescent compounds, radio-labeled compounds by art-known methods.
F. Synthesis of the Polymeric Delivery Systems
Generally, the methods of preparing the compounds described herein include reacting a polymer with an aromatic acid ester to form a polymer-aromatic acid.
In one embodiment, the method includes reacting a compound of Formula (II) having the structure of:
A1 — R1 — X1 — M1
(II) with a compound of Formula (III) having the structure of:
Figure imgf000035_0001
under conditions effective to form the compound of Formula (IV) having the structure of
Figure imgf000035_0002
wherein:
A1 is a capping group or M1-X'1-;
A is a capping group or
Figure imgf000035_0003
R1 is a substantially non-antigenic water-soluble polyinei-;
M1 is -OH,-SH Or-NHR41; M2 is a leaving group;
Ar and Ar' are independently an aryl or heteroaryl moiety;
X1 and X'1 are independently O, S, SO, SO3, NR6, or a bond;
Y1 and Y'1 are independently O, S, or NR6;
L1 and L'1 are independently selected bifunctional linkers; D4 and D'4 are independently selected from among hydrogen, OH, OR42, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
R2-5, R'2-5, R6 and R41 are independently selected from among hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C1-6 alkylthio, C1-6alkyls, C2-6 alkenyl, C2-6alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6 heteroalkyl, substituted C1-6heteroalkyl, C1-6alkoxy, aryloxy, C1-6 heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy, substituted and arylcarbonyloxy;
R42 is C1-6 alkyl; (p), (p'), (r) and (r') are independently zero or a positive integer, preferably from about 0 to about 10 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10), more preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), and most preferably 0, 1 or 2;
(q1), (q'1), (q2), (q'2), (q3), (q'3), (q4) and (q'4) are independently zero or one;
(s) and (s') are independently zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6) and more preferably zero, 1 or 2;
Q1-4 and Q'1-4 are independently selected from among the same moieties which can be used for R2 or each can be:
Figure imgf000036_0001
wherein R7 and R8 are independently selected from the same as used for R2;
Y2 is O, S or NR6;
L3 is a bifuncional linker,
(z) is zero or one;
(w) is zero or a positive integer, preferably from about 0 to about 6 (e.g., 0, 1, 2, 3, 4, 5 or 6), more preferably zero, 1 or 2 (for exzmple, (z) is 0 and (w) is 1 and preferably
(z) is 1 and (w) is 1); and
D5 is selected from the group consisting of hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties; provided that the sum of (q1) + (q2) + (q3) + (q4) is not zero and that at least one of Q1-4 and Q'1-4 is
Figure imgf000037_0001
wherein at least one of D5 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
In certain aspects of the invention, when (p) is zero, there are preferably a sufficient number of atoms, e.g., more than five or six atoms, present between X1/X'1 and C(=Y1)/C(=Y'1), so that a releasable cyclic moiety is not formed. For example, when (p) is zero, the polymeric portion attached to the phenyl ring would not be in an ortho position in relation to D4, D'4 or D5. The leaving group M1 includes halogen (Br, Cl), activated carbonate, carbonyl imidazole, cyclic imide thione, isocyanate, N-hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, C1-C6 alkyloxy, C1-C6 alkanoyloxy, arylcarbonyloxy, ortho-nitrophenoxy, N-hydroxybenzotriazolyl, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5-trifluorophenoxy or other suitable leaving groups that is apparent to those of ordinary skill in the art.
The resulting compounds of Formula (IV) can be then deprotected to form a polymer- aromatic acid. The polymer-aromatic acids are further activated with an amine or a hydroxyl containing compound. Alternatively/additionally, a bifunctional group can be attached to the aromatic moiety to provide a functional group. The functional groups can be further conjugated to a biologically active moiety, or a targeting moiety.
Alternatively, it is also contemplated that methods can include reacting a polymer containing a leaving group with an aromatic-containing moiety to form a polymer-aromatic acid.
In one embodiment, the method includes: a) reacting a protected aromatic compound with a substantially non-antigenic polymer compound to form a polymer compound of Formula (Ia):
Figure imgf000038_0001
b) converting the nitro moiety on the compound of Formula (Ia) under a suitable reaction condition to form an amine; c) reacting the amine with a suitable bifunctional spacer to form a compound of Formula (Ic):
Figure imgf000038_0002
d) deprotecting the ester of the compound of Formula (Ic) to form an acid; e) activating the acid resulting from step (d) and reacting the activated acid with an amine-containing biologically active moiety to form a compound of Formula (Ie):
Figure imgf000038_0003
and f) reacting the compound of Formula (Ie) with a thiol containing moiety to form a compound of Formula (If):
Figure imgf000038_0004
wherein, NH-drug is an amine-contaiing biologically active moiety;
Spacer is selected from the same group as defined for L3; Ab is an antibody such as monoclonal antibody, single chain antibody and active fragments thereof; and
Polymer is a substantially non-antigenic polymer.
Attachment of the bifunctional group to the polymer portion is preferably carried out in the presence of a coupling agent. A non-limiting list of suitable coupling agents include 1,3- diisopropylcarbodiimide (DIPC), any suitable dialkyl carbodiimides, 2-halo-1-alkyl-pyridinium halides, (Mukaiyama reagents), 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide (EDC), propane phosphonic acid cyclic anhydride (PPACA) and phenyl dichlorophosphates, etc. which are available, for example from commercial sources such as Sigma-Aldrich Chemical, or synthesized using known techniques.
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, triethylamine, etc. to neutralize any acids generated. The reactions can be carried out at a temperature from about 0°C up to about 22°C (room temperature).
G. COMPOUNDS OF FORMULA (I)
Some particular embodiments prepared by the methods described herein have the structure:
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
and
Figure imgf000042_0002
wherein: one or more, preferably all, D2 is hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
Ab is an antibody; mPEG has the formula CH3O(CH2CH2O)n-;
PEG has the formula -O(CH2CH2O)n- , and
(n) is an integer from about 10 to about 2,300.
For purposes of the present invention, it will be understood that the ''S-Ab" represents antibodies, such as monoclonal antibodies, single chain antibodies, and active fragments thereof.
For example, a non-limiting list of particular embodiments includes:
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
and
Figure imgf000047_0002
wherein
(n) is an integer from about 10 to about 2300; D2 is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides; and
Ab is an antibody.
Preferably, the compounds prepared by the methods include:
Figure imgf000048_0001
wherein
(n) is an integer from about 10 to about 2300;
D2 is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides; and
Ab is an antibody.
H. METHODS OF TREATMENT
Another 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 compound described herein. The polymeric conjugate compounds are useful for, among other things, treating diseases which are similar to those which are treated with the parent compound, e.g. enzyme replacement therapy, neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mammals.
The amount of the polymeric conjugate that is administered will depend upon the amount of the parent molecule included therein. Generally, the amount of polymeric conjugate used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various polymeric conjugate compounds will vary somewhat depending upon the parent compound, molecular weight of the polymer, rate of in vivo hydrolysis, etc. Those skilled in the art will determine the optimal dosing of the polymeric transport conjugates selected based on clinical experience and the treatment indication. Actual dosages will be apparent to the artisan without undue experimentation.
The compounds of the present invention can be included in one or more suitable pharmaceutical compositions for administration to mammals. The pharmaceutical compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral and/or parenteral routes depending upon the needs of the artisan. A solution and/or suspension of the composition may be utilized, for example, as a carrier vehicle for injection or infiltration of the composition by any art known methods, e.g., by intravenous, intramuscular, intraperitoneal, subcutaneous injection and the like. Such administration may also be by infusion into a body space or cavity, as well as by inhalation and/or intranasal routes. In preferred aspects of the invention, however, the polymeric conjugates are parenterally administered to mammals in need thereof.
EXAMPLES
The following examples serve to provide further appreciation of the invention but are not meant in any way to restrict the scope of the invention. The bold-faced numbers recited in the Examples correspond to those shown in Fig. 1. Abbreviations are used throughout the examples such as, DCM (dichloromethane), DIEA (diisopropylethylamine), DMAP (4- dimethylaminopyridine). DMF (N,N'-dimethylformamide), DSC (disuccinimidyl carbonate), EDC (1-(3-dimethylaminopropyl)-3-ethyl carbodiimide), IPA (isopropanol), NHS (N- hydroxysuccinimide), PEG (polyethylene glycol), SCA-SH (single-chain antibody), and TEA (triethylamine).
General
All reactions can be conducted under an atmosphere of dry nitrogen. Commercial regents and anhydrous solvents can be used without further purification. NMR spectra can be recorded at a Varian Mercury 300 MHz NMR spectrometer using deuterated solvent indicated. Chemical shifts (δ) are reported in parts per million (ppm) downfield from tetramethylsilane (TMS) and coupling constants (J values) are given in hertz (Hz).
Example 1. Compound 3
A solution of 5.0 g (1.0 mmole) of mPEG5K-OH (compound 2) in 130ml of toluene is azeotroped for 2 hours, while removing 65ml of toluene/water. This solution is cooled to 25°C, followed by addition of 2.0 ml (2.0 mmole) of 1.0 molar t-BuOK in t-butanol. This solution is stirred for 30 minutes at 25°C, followed by the addition of 30ml of anhydrous DMF. To this reaction mixture is added dropwise, a solution of compound 1 (2.0 mmol) in anhydrous DMF. This solution is added at a rate of 10ml per 20 min. During addition of the 4-(bromomethyl)- phenylacetic acid solution, while the pH of the reaction mixture is monitored. When the pH reaches ~8.0, a 1.0mI aliquot of 1.0 molar t-BuOK in t-butanol is added, total volume 7.0ml over 40 minutes. The reaction mixture is then poured into 700mI of ether, and the precipitate is collected by filtration and washed with ether. The solid is dissolved in 70ml of 0.2N HCI solution, and extracted with methylene chloride. The combined methylene chloride extracts are dried over sodium sulfate, filtered, and the solvent partially removed by rotovap. The product is precipitated out with ether, collected, washed with ether, and recrystallized from 12% DMF/1PA to give the product.
Example 2. Compound 4 Compound 3 is suspended in a mixture of water and THF and is added Na2S2O4. The mixture is stirred overnight at room temperature. The mixture is concentrated in vacuo and the product is extracted with DCM twice. The organic layers are combined and dried over anhydrous Na2SO4, filtered, and concentrated to a minimum volume. Anhydrous ether is added to the residual solution to precipitate the product, which is collected by vacuum filtration and dried in the vacuum oven at 45 °C to give the product.
Example 3. Compound 6
A solution of compound 4 (1.72 mmol), compound 5 (0.80g, 6.9 mmol), DIEA (1.3g, 10.3 mmol), and DMAP (50 mg, 0.4 mmol) in 75 ml of dry methylene chloride is cooled to 0°C in an ice bath, followed by addition of EDC hydrochloride (1.66g, 8.6 mmol). This mixture is allowed to warm to room temperature overnight. The solvent is partially removed by rotovap. The product is precipitated with ether, and collected and washed with ether to give the crude product, which is dried in the vacuum oven at 45 °C to give the product.
Example 4. Compound 7 Trifluoroacetic acid (TFA, 2.5 mL) is added to a solution of compound 6 (3.2 mmol) of in 5 mL of methylene chloride followed by stirring at room temperature for 30 minutes. Ether is added until the solid is precipitated. The solid is filtered and washed thoroughly with ether until all the excess TFA is removed. The acid is dried in the vacuum oven at 45 °C.
Example 5. Compound 8
A solution of compound 7 (1.72 mmol), NHS (0.80g, 6.9 mmol), DIEA (1.3g, 10.3 mmol), and DMAP (50 mg, 0.4 mmol) in 75 ml of dry methylene chloride is cooled to 0°C in an ice bath, followed by addition of EDC hydrochloride (1.66g, 8.6 mmol). This mixture is allowed to warm to room temperature overnight. The solvent is partially removed by rotovap. The product is precipitated with ether, and collected and washed with ether to give the crude product, which is dried in the vacuum oven at 45 °C to give the product.
Example 6. Compound 9
PEG linker (compound 8) (0.084 mmol) is added to native (L)-asparaginase (0.00027 mmol) in 3 mL of sodium phosphate buffer (0.1 M, pH 7.8) with gentle stirring. The solution is stirred at 30 °C for 30 minutes. A GPC column (Zorbax GF-450) is used to monitor PEG conjugation. At the end of the reaction (as evidenced by the absence of native enzyme), the mixture is diluted with 12 mL of formulation buffer (0.05 M sodium phosphate, 0.85% sodium chloride, pH 7.3) and diafiltered with a Centriprep concentrator (Amicon) to remove the unreacted PEG. Dialfiltration is continued as needed at 4 °C until no more free PEG is detected by mixing equal amount of filtrate and 0.1% PMA (polymethacrylic acid in 0.1 M HCl) to give the product.
Example 7. Compound 10
Compound 9 (0.084 mmol) is added to SCA-SH (0.00027 mmol) in 3 mL of sodium phosphate buffer (0.1 M, pH 7.8) with gentle stirring. The solution is stirred at 30 °C for 30 minutes. A GPC column (Zorbax GF-450) is used to monitor PEG conjugation. At the end of the reaction (as evidenced by the absence of native enzyme), the mixture is diluted with 12 mL of formulation buffer (0.05 M sodium phosphate, 0.85% sodium chloride, pH 7.3) and diafiltered with a Centriprep concentrator (Amicon) to remove the unreacted PEG reactant. Dialfiltration is continued as needed at 4 °C until no more free PEG was detected by mixing equal amount of filtrate and 0.1 % PMA (polymethacrylic acid in 0.1 M HCl) to give the product.

Claims

We claim:
1. A compound of Formula (I):
Figure imgf000053_0001
wherein:
A is a capping group or
Figure imgf000053_0002
R1 is a substantially non-antigenic water-soluble polymer;
X1 and X'1 are independently O, S, SO, SO2, NR6 or a bond;
Ar and Ar' are independently an aryl or heteroaryl moiety;
Y1 and Y'1 are independently O, S, or NR6;
L1 and L'1 are independently selected bifunctional linkers;
D1 and D'1 are independently selected from the group consisting of hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
R2-5, R'2-5, and R6 are independently selected from the group consisting of hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C1-6 alkylthio, C1-6alkyls, C2-6 alkenyl, C2-6alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6 heteroalkyl, substituted C1-6heteroalkyl, C1-6alkoxy, aryloxy, C1-6heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, Ci-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy, substituted and arylcarbonyloxy;
(p)' (p'), (r) and (r') are independently zero or a positive integer;
(q1), (q'1), (q2), (q'2), (q3), (q'3), (q4) and (q'4) are independently zero or one;
(s) and (s') are independently zero or a positive integer;
Q1-4 and Q'1-4 are independently selected from the same moieties which can be used for R2 or each can be:
Figure imgf000054_0002
wherein
R7 and R8 are independently selected from the same group as that which defines
R2;
Y2 is O, S or NR6; L3 is a bifuncional linker; (z) is zero or one;
(w) is zero or a positive integer; and
D3 is selected from the group consisting of hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties, provided that the sum of (q1) + (q2) + (q3) + (q4) is not zero and that at least one of Q1-4 and Q'1-4 is
Figure imgf000054_0001
wherein at least one of D3 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
2. The compound of claim 1, wherein the leaving group is selected from the group consisting of halogens, activated carbonates, carbonyl imidazole, cyclic imide thione, isocyanate, N-hydroxybenzotriazolyl, N-hydroxyphtalimide, N-hydroxysuccinimidyl, imidazole, mesylate, nosylate, tosylate, tresylate, C1-C6 alkyloxy, C1-C6 alkanoyloxy, arylcarbonyloxy, para- nitrophenoxy, ortho-nitrophenoxy, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5- trifluorophenoxy.
3. The compound of claim 1, wherein the functional group is selected from the group consisting of maleimidyl, vinyl, residues of sulfone, amino, carboxy, mercapto, hydrazide, and carbazate.
4. The compound of claim 1, wherein D1, D'1 and D3 are independently selected from the group consisting of OH, methoxy, tert-butoxy, N-hydroxysuccinimidyl and maleimidyl.
5. The compound of claim 1, wherein the biologically active moiety is selected from the group consisting of amine-containing moieties, hydroxyl-containing moieties and thiol- containing moieties.
6. The compound of claim 1, wherein the biologically active moiety is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides.
7. The compound of claim 1, wherein L1, L'1 and L3 are independently selected from the group consisting of:
-[C(=O)]v(CR22R23)t[C(=O)]v'- , -[C(=O)]v(CR22R23)t-O[C(=O)]v'- ,
-[C(=O)]v(CR22Ra3)t-NR26[C(=O)]v'- , -[C(=O)]vO(CR22R23)t[C(=O)]v'- , -[C(=O)]vO(CR22R23)tO[C(=O)]v'- , -[C(=O)]vO(CR22R23)tNR26[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)t[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tO[C(=O)]v'- , -[C(=O)]vNR21(CR22R23)tNR26[C(=O)]v'- , -[C(=O)]v(CR22R23)tO-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23)tNR26-(CR28R29V[C(=O)]v'- ,
-[C(=O)]v(CR22R23)tS-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tO-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tNR26-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)tS-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)tO-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)tNR26-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)tS-(CR28R29)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23CR28R29O)tNR26[C(=O)]v'- ,
-[C(=O)]v(CR22R23CR28R29O)t[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR28R29O)tNR26[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR28R29O)t[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23CR28R29O)tNR26[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23CR28R29O)t[C(=O)]v'- ,
-[C(=O)]v(CR22R23CR28R29O)t(CR24R25)t'[C(=O)]v'- ,
-[C(=O)]vO(CR22R23CR2sR29O)t(CR24R25)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23CR28R29O)t(CR24R25)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23CR28R29O)t(CR24R2S)t'O[C(=O)]v'- ,
-[C(=O)]v(CR22R23)t(CR24R25CR28R29O)t'[C(=O)]v'- ,
-[C(=O)]v(CR22R23)t(CR24R25CR28R29O)t'NR26[C(=O)]v'- ,
.[C(=O)]vO(CR22R23CR28R29O)t(CR24R25)t'O[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)t(CR24R25CR28R29O)r[C(=O)]v'- ,
-[C(=O)]vO(CR22R23)t(CR24CR25CR28R29O)t'NR26[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23CR28R29O)t(CR24R25)t'O[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)t(CR24R25CR28R29O)t'[C(=O)]v'- ,
-[C(=O)]vNR21(CR22R23)t(CR24R25CR28R29O)t'NR26[C(=O)]v'- ,
Figure imgf000056_0001
Figure imgf000057_0001
wherein:
R21-29 are independently selected from the group consisting of hydrogen, C1-6alkyls, C3-12 branched alkyls, C3-8 cycloalkyls, C1-6 substituted alkyls, C3-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1-6 heteroalkyls, substituted C1-6 heteroalkyls, C1-6alkoxy, phenoxy and C1-6heteroalkoxy;
(t) and (t') are independently zero or a positive integer; and
(v) and (v') are independently zero or 1.
8. The compound of claim 1, wherein L1, L'1 and L3 are independently selected from the group consisting of:
-[C(=O)]r1NH(CH2)2CH=N-NHC(=O)-(CH2)2- ,
-[C(=O)]r1NH(CH2)2(CH2CH2O)2(CH2)2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)(CH2CH2O)2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1NH(CH2CH2)s1'[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1S(CH2CH2)s1'[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)(CH2CH2O)[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2)s1O(CH2CH2)s1'[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)(CH)2NH[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)2(CH2)[C(=O)]r1'- ,
-[C(=O)]r1NH(CH2CH2O)s1(CH2)s1'[C(=O)]r1'- , -[C(=O)]r1NHCH2CH2NH[C(=O)]r1'- , -[C(=O)]r1NH(CH2CH2)2O[C(=O)]r1'- , -[C(=O)]r1NH(CH2CH2O)[C(=O)]r1'- , -[C(=O)]r1NH(CH2CH2O)2[C(=O)]r1'- , -[C(=O)]r1NH(CH2)3[C(=O)]r1'- , -[C(=O)]r1O(CH2CH2O)2(CH2)[C(=O)]r1'- , -[C(=O)]r1O(CH2)2NH(CH2)2[C(=O)]r1'- , -[C(=O)]r1O(CH2CH2O)2NH[C(=O)]r1'- , -[C(=O)]r1O(CH2)2O(CH2)2[C(=O)]r1'- , -[C(=O)]r1O(CH2)2S(CH2)2[C(=O)]r1'- , -[C(=O)]r1O(CH2CH2)NH[C(=O)]r1'- , -[C(=O)]r1O(CH2CH2)O[C(=O)]r1'- , -[C(=O)]r1O(CH2)3NH[C(=O)]r1'- , -[C(=O)]r1O(CH2)3O[C(=O)]r1'- , -[C(=O)]r1O(CH2)3[C(=O)]r1'- , -[C(=O)]r1CH2NHCH2[C(=O)]r1'- , -[C(=O)]rCH2OCH2[C(=O)]r1'- , -[C(=O)]r1CH2SCH2[C(=O)]r1'- , -[C(=O)]r1S(CH2)3[C(=O)]r1'- , -[C(=O)]r1(CH2)3[C(=O)]r1'- ,
Figure imgf000058_0001
wherein
(r1) and (r1') are independently zero or 1; and
(s1) and (s1') are independently zero or a positive integer, provided that both (r1) and (r1') are not zero simultaneously.
9. The compound of claim 1, wherein L1, L'1 and L3 are independently selected from the group consisting of amino acids, amino acid derivatives and peptides.
10. The compound of claim 1, wherein L3 is selected from the group consisting of:
Figure imgf000059_0001
11. The compound of claim 1, wherein A is selected from the group consisting of H, NH2, OH, CO2H, C1-6 alkoxy, and C1-6 alkyl.
12. The compound of claim 1, wherein A is methyl or methoxy.
13. The compound of claim 1, wherein Ar and Ar' are independently selected from the group consisting of:
Figure imgf000059_0002
Figure imgf000060_0001
wherein
J is O, S, or NR11;
E and Z are independently CR 12 or NR13; and
R11, R12 and R13 are independently selected from the same group as that which defines R2.
14. The compound of claim 1 having the formula:
Figure imgf000060_0002
wherein A is a capping group or
Figure imgf000060_0003
15. The compound of claim 1, having the formula:
Figure imgf000060_0004
16. The compound of claim 3 , wherein R2-5 and R'2-5 are independently hydiOgen or CH3.
17. The compound of claim 1, wherein R1 comprises a linear, terminally branched or multi- armed polyalkylene oxide.
18. The compound of claim 17, wherein the polyalkylene oxide is selected from the group consisting of polyethylene glycol and polypropylene glycol.
19. The compound of claim 17, wherein the polyalkylene oxide is a polyethylene glycol of the formula, -O-(CH2CH2O)n- wherein (n) is an integer from about 10 to about 2,300.
20. The compound of claim 17, wherein the polyalkylene oxide has an average molecular weight from about 2,000 to about 100,000 Daltons.
21. The compound of claim 17, wherein the polyalkylene oxide residue has an average molecular weight of from about 5,000 to about 60,000 daltons.
22. The compound of claim 17, wherein the polyalkylene oxide has an average molecular weight from about 5,000 to about 25,000 Daltons or from about 20,000 to about 45,000 Daltons.
23. A compound of claim 1, selected from the group consisting of:
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
and
Figure imgf000064_0002
wherein: D2 is hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties; Ab is an antibody; mPEG has the formula CH3O(CH2CH2O)n-; PEG has the formula -O(CH2CH2O)n- , and (n) is an integer from about 10 to about 2,300.
24. The compound of claim 23 selected from the group consisting of:
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
and
Figure imgf000068_0001
25. A method of preparing a substantially non-antigenic polymer compound having a multisubstituted aromatic moiety comprising: reacting a compound of Formula (II):
A1 — R1 — X1 — M1
(II) with a compound of Formula (III):
Figure imgf000068_0002
under conditions effective to form a compound of Formula (IV):
Figure imgf000068_0003
wherein:
A1 is a capping group or M1-X'1-;
A is a capping group or
Figure imgf000068_0004
R1 is a substantially non-antigenic water-soluble polymer; M1 is -OH, -SH Or-NHR41;
M2 is a leaving group;
Ar and Ar' are independently an aryl or heteroaryl moiety;
X1 and X'1 are independently O, S, SO, SO2, NR6 or a bond;
Y1 and Y'1 are independently O, S, or NR6;
L1 and L'1 are independently selected bifunctional linkers;
D4 and D'4 are independently selected from the group consisting of hydrogen, OH, OR42, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties;
R2-5, R'2-5, R6, and R41 are independently selected from the group consisting of hydrogen, amino, substituted amino, azido, carboxy, cyano, halo, hydroxyl, nitro, silyl ether, sulfonyl, mercapto, C1-6 alkylmercapto, arylmercapto, substituted arylmercapto, substituted C1-6 alkylthio, C1-6 alkyls, C2-6 alkenyl, C2-6 alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6 heteroalkyl, substituted C1-6heteroalkyl, C1-6alkoxy, aryloxy, C1-6heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy, substituted and arylcarbonyloxy;
R42 is C1-6 alkyl;
(p), (p'), (r) and (r') are independently zero or a positive integer;
(q1), (q'1), (q2), (q'2), (q3), (q'3), (q4) and (q'4) are independently zero or one;
(s) and (s') are independently zero or a positive integer:
Q1-4 and Q'1-4 are independently selected from the same moieties which can be used for R2 or each can be:
Figure imgf000069_0001
wherein R7 and R8 are independently selected from the same group as that which defines
R2;
Y2 is O, S or NR6;
L3 is a bifuncional linker;
(z) is zero or one;
(w) is zero or a positive integer; and
D5 is selected from the group consisting of hydrogen, OH, leaving groups, functional groups, targeting groups, diagnostic agents and biologically active moieties; provided that the sum of (q1) + (q2) + (q3) + (q4) is not zero and that at least one of Q1-4 and Q'1-4 is
Figure imgf000070_0001
wherein at least one of D5 is a leaving group, a functional group, a targeting group, a diagnostic agent or a biologically active moiety; and provided that (z) is not zero when (w) is zero.
26. A method of treating a mammal, comprising administering an effective amount of a compound of claim 1 to a patient in need thereof, wherein at least one of D1, D'1, and D3 is a biologically active moiety.
PCT/US2008/069732 2007-07-11 2008-07-11 Polymeric drug delivery system containing a multi-substituted aromatic moiety WO2009009712A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2693616A CA2693616A1 (en) 2007-07-11 2008-07-11 Polymeric drug delivery system containing a multi-substituted aromatic moiety
EP08781660.9A EP2175878A4 (en) 2007-07-11 2008-07-11 Polymeric drug delivery system containing a multi-substituted aromatic moiety
JP2010516258A JP2010533202A (en) 2007-07-11 2008-07-11 Polymeric drug delivery system comprising a polysubstituted aromatic moiety

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US94916807P 2007-07-11 2007-07-11
US60/949,168 2007-07-11

Publications (1)

Publication Number Publication Date
WO2009009712A1 true WO2009009712A1 (en) 2009-01-15

Family

ID=40229074

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/069732 WO2009009712A1 (en) 2007-07-11 2008-07-11 Polymeric drug delivery system containing a multi-substituted aromatic moiety

Country Status (6)

Country Link
US (1) US20090016985A1 (en)
EP (1) EP2175878A4 (en)
JP (1) JP2010533202A (en)
CA (1) CA2693616A1 (en)
TW (1) TW200922624A (en)
WO (1) WO2009009712A1 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
WO2018060311A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Incremental dose finding in controlled-release pth compounds
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
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
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
WO2020064844A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Treatment of infections
WO2020141222A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Sustained local drug levels for innate immune agonists
WO2020141225A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Minimization of systemic inflammation
WO2020141223A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Induction of sustained local inflammation
WO2020141221A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Conjugates of pattern recognition receptor agonists
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
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
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
WO2021144249A1 (en) 2020-01-13 2021-07-22 Ascendis Pharma Bone Diseases A/S Hypoparathyroidism treatment
US11103593B2 (en) 2013-10-15 2021-08-31 Seagen Inc. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
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
US11730822B2 (en) 2017-03-24 2023-08-22 Seagen Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
WO2023227505A1 (en) 2022-05-23 2023-11-30 Ascendis Pharma Growth Disorders A/S Liquid pharmaceutical formulations of cnp compounds
US11844839B2 (en) 2016-03-25 2023-12-19 Seagen Inc. Process for the preparation of pegylated drug-linkers and intermediates thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003061577A2 (en) * 2002-01-18 2003-07-31 Biogen Idec Ma Inc. Polyalkylene glycol with moiety for conjugating biologically active compound
US20060009590A1 (en) * 2002-12-31 2006-01-12 Antoni Kozlowski Hydrolytically stable maleimide-terminated polymers

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US5328679A (en) * 1988-04-01 1994-07-12 Immunomedics, Inc. Methods for technetium/rhenium labeling of proteins
US5122614A (en) * 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
JP2997004B2 (en) * 1989-05-26 2000-01-11 住友製薬株式会社 Polyethylene glycol derivative, modified peptide and production thereof
US5229366A (en) * 1990-10-23 1993-07-20 Fuji Photo Film Co., Ltd. Peptide-containing polyethylene glycol derivatives and application thereof
US5645815A (en) * 1991-02-08 1997-07-08 Diatide, Inc. Radiolabled compounds for thrombus imaging
US5997844A (en) * 1991-02-08 1999-12-07 Diatide, Inc. Technetium-99m labeled peptides for imaging
KR930703024A (en) * 1991-02-08 1993-11-29 리챠드 티. 딘 Technetium-99m Labeled Polypeptide for Imaging
US5643575A (en) * 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5919455A (en) * 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
EP0816381B1 (en) * 1995-03-10 2004-01-14 NAKAMURA, Toshikazu Polyethylene glycol modified hepatocyte growth factor (hgf)
US6180095B1 (en) * 1997-12-17 2001-01-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
WO1999030727A1 (en) * 1997-12-17 1999-06-24 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
US6624142B2 (en) * 1997-12-30 2003-09-23 Enzon, Inc. Trimethyl lock based tetrapartate prodrugs
US5965119A (en) * 1997-12-30 1999-10-12 Enzon, Inc. Trialkyl-lock-facilitated polymeric prodrugs of amino-containing bioactive agents
US6153655A (en) * 1998-04-17 2000-11-28 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
CN1434726A (en) * 2000-06-08 2003-08-06 拉卓拉药物公司 Multivalent platform molecules comprising high molecular weight polyethylene oxide
ES2331562T3 (en) * 2001-03-23 2010-01-08 Enzon, Inc. PROFARMACOS OF ANTI-BANERIGIN AGENTS WHO USE SUBSTITUTED AROMATIC ACIDS.
US7309615B2 (en) * 2002-09-27 2007-12-18 Siemens Medical Solutions Diagnostic High quantum yield acridinium compounds and their uses in improving assay sensitivity
US6767881B1 (en) * 2003-03-19 2004-07-27 Ecolab, Inc. Cleaning concentrate
CA2786794C (en) * 2004-03-23 2017-09-12 Complex Biosystems Gmbh Aromatic polymeric cascade prodrug linker reagent
TW200616604A (en) * 2004-08-26 2006-06-01 Nicholas Piramal India Ltd Nitric oxide releasing prodrugs containing bio-cleavable linker
PT2279758E (en) * 2005-06-16 2015-05-27 Nektar Therapeutics Conjugates having a degradable linkage and polymeric reagents useful in preparing such conjugates
EP2099495A2 (en) * 2005-08-04 2009-09-16 Nektar Therapeutics AL, Corporation Conjugates of a g-csf moiety and a polymer
US7301003B2 (en) * 2005-08-26 2007-11-27 Enzon Pharmaceuticals, Inc. Method of preparing polymers having terminal amine groups
US7601798B2 (en) * 2005-10-04 2009-10-13 Enzon Pharmaceuticals, Inc. Methods of preparing polymers having terminal amine groups using protected amine salts
US7989554B2 (en) * 2006-01-10 2011-08-02 Enzon Pharmaceuticals, Inc. Reacting polyalkylene oxide with base, tertiary alkyl haloacetate, then acid to prepare polyalkylene oxide carboxylic acid
US20090017004A1 (en) * 2007-07-11 2009-01-15 Enzon Pharmaceuticals, Inc. Polymeric drug delivery systems containing an aromatic allylic acid

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003061577A2 (en) * 2002-01-18 2003-07-31 Biogen Idec Ma Inc. Polyalkylene glycol with moiety for conjugating biologically active compound
US20060009590A1 (en) * 2002-12-31 2006-01-12 Antoni Kozlowski Hydrolytically stable maleimide-terminated polymers

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2175878A4 *

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11103593B2 (en) 2013-10-15 2021-08-31 Seagen Inc. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
US11413351B2 (en) 2016-01-08 2022-08-16 Ascendis Pharma Growth Disorders A/S CNP prodrugs with carrier attachment at the ring moiety
US11389510B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low initial NPR-B activity
US11389511B2 (en) 2016-01-08 2022-07-19 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with reduced side-effects
US11311604B2 (en) 2016-01-08 2022-04-26 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with low NPR-C binding
US11224661B2 (en) 2016-01-08 2022-01-18 Ascendis Pharma Growth Disorders A/S Controlled-release CNP agonists with increased NEP stability
US11154593B2 (en) 2016-01-08 2021-10-26 Ascendis Pharma Growth Disorders A/S CNP prodrugs with large carrier moieties
US11793861B2 (en) 2016-03-01 2023-10-24 Ascendis Pharma Bone Diseases A/S PTH prodrugs
WO2017148883A1 (en) 2016-03-01 2017-09-08 Ascendis Pharma Bone Diseases A/S Pth prodrugs
US11844839B2 (en) 2016-03-25 2023-12-19 Seagen Inc. Process for the preparation of pegylated drug-linkers and intermediates thereof
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
US11590207B2 (en) 2016-09-29 2023-02-28 Ascendis Pharma Bone Diseases A/S Dosage regimen for a controlled-release PTH compound
EP4275677A2 (en) 2016-09-29 2023-11-15 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
US11759504B2 (en) 2016-09-29 2023-09-19 Ascendis Pharma Bone Diseases A/S PTH compounds with low peak-to-trough ratios
WO2018060310A1 (en) 2016-09-29 2018-04-05 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
WO2018060311A1 (en) 2016-09-29 2018-04-05 Ascendis Pharma Bone Diseases A/S Incremental dose finding in controlled-release pth compounds
US11918628B2 (en) 2016-09-29 2024-03-05 Ascendis Pharma Bone Diseases A/S Controlled-release PTH compound
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
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
US11730822B2 (en) 2017-03-24 2023-08-22 Seagen Inc. Process for the preparation of glucuronide drug-linkers and intermediates thereof
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
WO2020141223A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Induction of sustained local inflammation
WO2020141221A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Conjugates of pattern recognition receptor agonists
WO2020141222A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Sustained local drug levels for innate immune agonists
WO2020141225A1 (en) 2019-01-04 2020-07-09 Ascendis Pharma A/S Minimization of systemic 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
WO2020254617A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Anti-ctla4 compounds with localized pk properties
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
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
WO2020254613A1 (en) 2019-06-21 2020-12-24 Ascendis Pharma Oncology Division A/S Controlled-release tyrosine kinase inhibitor compounds with localized pk properties
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
JP2010533202A (en) 2010-10-21
EP2175878A1 (en) 2010-04-21
CA2693616A1 (en) 2009-01-15
EP2175878A4 (en) 2014-12-03
US20090016985A1 (en) 2009-01-15
TW200922624A (en) 2009-06-01

Similar Documents

Publication Publication Date Title
WO2009009712A1 (en) Polymeric drug delivery system containing a multi-substituted aromatic moiety
US20090203706A1 (en) Lysine-based polymeric linkers
US20100203066A1 (en) Polymeric linkers containing pyridyl disulfide moieties
US20090017004A1 (en) Polymeric drug delivery systems containing an aromatic allylic acid
AU2003262622B2 (en) Releasable polymeric conjugates based on aliphatic biodegradable linkers
AU2003262623B2 (en) Releasable polymeric conjugates based on aliphatic biodegradable linkers
US8268318B2 (en) Polyalkylene oxides having hindered ester-based biodegradable linkers
WO2003097356A9 (en) Camptothecin derivatives and polymeric conjugates thereof
CN101516336A (en) Lysine-based polymeric 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: 08781660

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2693616

Country of ref document: CA

Ref document number: 2010516258

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008781660

Country of ref document: EP