AU2007296056A1 - Targeted polymeric prodrugs containing multifunctional linkers - Google Patents

Targeted polymeric prodrugs containing multifunctional linkers Download PDF

Info

Publication number
AU2007296056A1
AU2007296056A1 AU2007296056A AU2007296056A AU2007296056A1 AU 2007296056 A1 AU2007296056 A1 AU 2007296056A1 AU 2007296056 A AU2007296056 A AU 2007296056A AU 2007296056 A AU2007296056 A AU 2007296056A AU 2007296056 A1 AU2007296056 A1 AU 2007296056A1
Authority
AU
Australia
Prior art keywords
cit
val
mpeg
peg
rgd
Prior art date
Legal status (The legal status 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 status listed.)
Granted
Application number
AU2007296056A
Other versions
AU2007296056B2 (en
Inventor
Prasanna Reddy
Maria Belen Rubio
Hong Zhao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Enzon Pharmaceuticals Inc
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
Publication of AU2007296056A1 publication Critical patent/AU2007296056A1/en
Application granted granted Critical
Publication of AU2007296056B2 publication Critical patent/AU2007296056B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C08G65/34Macromolecular compounds obtained by reactions forming an ether link in the main chain of the macromolecule from hydroxy compounds or their metallic derivatives
    • C08G65/48Polymers modified by chemical after-treatment
    • 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
    • C08G73/00Macromolecular compounds obtained by reactions forming a linkage containing nitrogen with or without oxygen or carbon in the main chain of the macromolecule, not provided for in groups C08G12/00 - C08G71/00
    • C08G73/06Polycondensates having nitrogen-containing heterocyclic rings in the main chain of the macromolecule
    • C08G73/10Polyimides; Polyester-imides; Polyamide-imides; Polyamide acids or similar polyimide precursors
    • C08G73/12Unsaturated polyimide precursors
    • C08G73/121Preparatory processes from unsaturated precursors and polyamines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/785Polymers containing nitrogen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6807Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug or compound being a sugar, nucleoside, nucleotide, nucleic acid, e.g. RNA antisense
    • A61K47/6809Antibiotics, e.g. antitumor antibiotics anthracyclins, adriamycin, doxorubicin or daunomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0215Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing natural amino acids, forming a peptide bond via their side chain functional group, e.g. epsilon-Lys, gamma-Glu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06052Val-amino acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06086Dipeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0815Tripeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0819Tripeptides with the first amino acid being acidic
    • 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
    • C08G64/00Macromolecular compounds obtained by reactions forming a carbonic ester link in the main chain of the macromolecule
    • C08G64/20General preparatory processes
    • C08G64/26General preparatory processes using halocarbonates
    • C08G64/28General preparatory processes using halocarbonates and phenols
    • 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
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L71/00Compositions of polyethers obtained by reactions forming an ether link in the main chain; Compositions of derivatives of such polymers
    • C08L71/02Polyalkylene oxides

Description

WO 2008/034124 PCT/US2007/078600 TARGETED POLYMERIC PRODRUGS CONTAINING MULTIFUNCTIONAL LINKERS CROSS-REFERENCE TO RELATED APPLICATION 5 This application claims the benefit of priority from U.S. Provisional Patent Application No. 60/844,943 filed September 15, 2006, the contents of which are incorporated herein by reference. BACKGROUND OF INVENTION 10 It is known that many of small molecular weight anti-cancer agents have significant toxicity. Over the years, there have been various attempts to reduce the toxicity and improve the efficacy of small molecular weight anti-cancer agents. One attempt to redu-ce the toxicity and improve the efficacy of small molecular weight drugs was directed to targeting agent-directed drug delivery systems. For example, the 15 targeting agent such as single-chain antibody, RGD peptides, or folic acid, can direct the delivery of the therapeutic agent and improve its therapeutic effect. At the same time, the targeted delivery of very potent chemotherapeutics would reduce its toxicity. The approach has been validated by FDA's approval of, for example, Wyeth's Mylotarg* gemtuzumab zogamicin, a humanized antibody against CD33 linked to the calicheamicin cytotoxin, for 20 acute myeloid leukemia (AML). Traditionally, the immunoconjugates include three components: a targeting agent like a monoclonal antibody, a cytotoxin and a linker. However, this approach requires that the antibody retain high degree of its binding affinity to its antigen after conjugation to the drug. Furthermore, the antibody-drug 25 conjugates have to be stable in buffers or plasma and not prematurely release the toxin during circulation. They must also be internalized once the antibody binds its antigen on tumor cell surface. Thereafter, the drug molecule has to be released intact inside the targeted tumor cells at a desired speed. Single chain antibodies (SCA's) or single-chain antigen-binding antibodies include the 30 binding domain of full length antibody with only one fourth of the size. However, SCA can bind to antigen specifically with high affinity. A description of the theory and production of single-chain antigen-binding proteins is found, for example, in commonly-assigned U.S. 1 WO 2008/034124 PCT/US2007/078600 Patent Nos. 4,946,778, 5,260,203, 5,455,030 and 5,518,889. The single-chain antigen binding proteins produced under the process recited in the above U.S. patents have binding specificity and affinity substantially similar to that of the corresponding Fab fragment, the content of which are incorporated by reference herein. More recently, commonly-assigned 5 U.S. Patent No. 6,872,393 disclosed polyalkylene oxide-modified single chain polypeptides. The contents of each of the foregoing commonly-assigned patents are incorporated herein by reference. In spite of the attempts and advances, there continues to be a need to provide targeting agent-directed polymeric drag delivery system having desired therapeutic activity and less 10 toxicity. The present invention addresses this need and others. SUMMARY OF THE INVENTION In order to overcome the above problems and improve the technology for polymeric drug delivery, there are provided new and advantageous compounds which employ the use of 15 both targeting agent and PEGylation technologies as well as other improved therapeutic techniques. Therefore, in accordance with one aspect of the invention there are provided compounds of Formula (I):
D
1
(L
1 )a-L 2 -(L3)b-D 2 A wherein: 20 R 1 is a substantially non-antigenic water-soluble polymer; A is a capping group or
D
1
-(L
1
)-L
2 -(L3)b-D 2 each Di is independently selected from among targeting moieties, functional groups and leaving groups, preferably targeting moieties; 25 each D 2 is independently selected from among biologically active moieties, functional groups and leaving groups, preferably biologically active moieties; each L 1 is an independently a permanent linker or a releasable linker, preferably permanent linker; 2 WO 2008/034124 PCT/US2007/078600
L
2 is a multifunctional linker; each L 3 is an independently a permanent linker or a releasable linker, preferably releasable linker; and (a) and (b) are independently zero or a positive integer, preferably zero or 1. 5 In on6 aspect of the invention, the polymer residue is a polyethylene oxide which can be conjugated to both small drug molecules and single chain antibody (SCA) through releasable linkers and permanent linkers through multifunctional linkers. In this way, there are provided compounds to provide SCA-PEG-Linker-Drug delivery platform for targeted delivery of small molecule cytotoxic compounds. Additionally, it is advantageous that the 10 loading of both small molecule and SCA per attaching site are increased over the traditional SCA-drug conjugates. Therefore, there can be provided cost-effective, low-cost, targeted SCA prodrugs. Furthermore, the prodrugs of the present invention provide methods of regulating circulating half-life of the drugs, for example, increasing circulating half-life, when desired, by using proper linkers. 15 One general aspect of the present invention, without limitation, is schematically described in the following structure: Targeting-permanent linker)--multifunctional linker )--(releasable linker-Drug Moiety wherein PEG is attached to both the SCA and the drug such as a cytotoxic agent through a centrally-located multifunctional linker; the linker with the SCA is permanent, while the 20 linker with the cytotoxic agent is releasable and can be designed to be stable in blood, but easily degradable in the presence of a site specific enzyme. In one preferred aspect, the SCA is attached through a permanent linker such as one containing a maleimide group and the cytotoxic agent through a releasable linker such as a peptidyl linker (Val-Cit) which can be specifically degraded by capthesin B. In one preferred 25 embodiment, the cytotoxic agent is SN38. Further aspects of the invention include methods of making the activated polymers containing multifunctional linkers, methods of making conjugates containing the same as well as methods of treatment based on administering effective amounts of conjugates containing a biologically active moiety to a patient (mammal) in need thereof 3 WO 2008/034124 PCT/US2007/078600 One of the advantages of the present invention is that the polymeric delivery systems described herein are stable and thus enhance bioavailability of drugs. Another advantage of the present invention is that the polymeric delivery systems can release drugs intact inside the targeted tumor cells. 5 Yet another advantage is that the release rate of the drugs can be modified to achieve a desired speed. Yet another advantage of the polymeric systems corresponding to the invention is that they are well suited for small molecular weight drugs and oligonucleotides such as antisense, short-interfering RNA (siRNA) or LNA compounds. 10 Other and further advantages will be apparent from the following description. 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. cytotoxin, SN38, permanent linker, multifunctional tinker, releasable linker, etc. that remains after it has undergone a substitution reaction with another compound. 15 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, substituted alkyls include carboxyalkyls, aminoalkyls, dialkylaminos, hydroxyalkyls and mercaptoalkyls; substituted alkenyls include 20 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 25 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, "nucleic acid" or "nucleotide" shall be 30 understood to include deoxyribonucleic acid (DNA), ribonucleic acid (RNA) whether single 4 WO 2008/034124 PCT/US2007/078600 stranded or double-stranded, unless otherwise specified, and any chemical modifications thereof 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 5 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 ease of description and not limitation, it will be understood that the term "biologically active moieties" is interchangeable with "drugs", "cytotoxic agents", "cytotoxins". 10 For ease of description and not limitation, it will be understood that the term "small molecules" are interchangeable with "pharmaceutically active compounds". Unless otherwise defined, for purposes of the present invention: the term "alkyl" shall be understood to include straight, branched, substituted, e.g. halo-, alkoxy-, and nitro- C- 12 alkyls, C 3
-
8 cycloalkyls or substituted cycloalkyls, etc.; 15 the term "substituted" shall be understood to include adding or replacing one or more atoms contained within a functional group or compound with one or more different atoms; the term "substituted alkyls" include carboxyalkyls, aminoalkyls, dialkylaminos, hydroxyalkyls and mercaptoalkyls; the term "substituted cycloalkyls" include moieties such as 4-chlorocyclohexyl; aryls 20 include moieties such as napthyl; substituted aryls include moieties such as 3-bromophenyl; aralkyls include moieties such as toluyl; heteroalkyls include moieties such as ethylthiophene; the term "substituted heteroalkyls" include moieties such as 3-methoxy-thiophene; alkoxy includes moieties such as methoxy; and phenoxy includes moieties such as 3 nitrophenoxy; 25 the term "halo" shall be understood to include fluoro, chloro, iodo and bromo; and the terms "sufficient amounts" and "effective amounts" for purposes of the present invention shall mean an amount which achieves a therapeutic effect as such effect is understood by those of ordinary skill in the art. 30 BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 schematically illustrates methods of synthesis described in Examples 1-5. 5 WO 2008/034124 PCT/US2007/078600 FIG. 2 schematically illustrates methods of synthesis described in Examples 6-7. FIG. 3 schematically illustrates methods of synthesis described in Examples 8-9. FIG. 4 schematically illustrates methods of synthesis described in Examples 10-14. FIG. 5 schematically illustrates methods of synthesis described in Examples 15-17. 5 FIG. 6 schematically illustrates methods of synthesis described in Examples 18-19. FIG. 7 schematically illustrates methods of synthesis described in Examples 20-23. FIG. 8 schematically illustrates methods of synthesis described in Examples 24-25. FIG. 9 schematically illustrates methods of synthesis described in Examples 26-28. FIG. 10 schematically illustrates methods of synthesis described in Examples 29-32. 10 FIG. 11 schematically illustrates methods of synthesis described in Examples 33-35. FIG. 12 schematically illustrates methods of synthesis described in Examples 36-38. FIG. 13 schematically illustrates methods of synthesis described in Examples 39-40. DETAILED DESCRIPTION OF THE INVENTION 15 A. OVERVIEW In one aspect of the present invention, there are provided compounds of Formula (I):
D
1
-(L
1 )a-L 2
-(L
3 )b--D2 A wherein:
R
1 is a substantially non-antigenic water-soluble polymer; 20 A is a capping group or DI-(L)a-L 2 -(L3)b-D 2 each D 1 is independently selected from among targeting moieties, functional groups and leaving groups, preferably targeting moieties; each D 2 is independently selected from among biologically active moieties, functional 25 groups and leaving groups, preferably biologically active moieties; each Li is an independently a permanent linker or a releasable linker, preferably permanent linker;
L
2 is a multifunctional linker; 6 WO 2008/034124 PCT/US2007/078600 each L 3 is an independently a permanent linker or a releasable linker, preferably releasable linker; and (a) and (b) are independently zero or a positive integer, preferably zero or 1. In one preferred aspect of the invention, the targeted polymeric delivery systems 5 include compounds having formula: L3-D2 L3-D2 2 c1 O 2 2 Q A-Rg N A-R 1 -N 00 d1 L1-D 1 (Iha), d2 L1-D 1 (Itb), L3-D2 L3-D2 L3-D2 R2 03 O 4O A-RI-N A-RA-N A-R -N
RN-L
1
-D
1 0 (d5O d3 R'2 (I), d4 L1-D 1 (Ild), L1D1 (ie), "L3-D2 /L3-D2 A-R1-N c'6 0,3D
A--R
1 -N c-6 0'1D 0 0 RC 6 'L -D (G6,c e L 1 D I g 12 ~L3-D2 1 ID A-RgN A-R 1 -N 0-L Dc( S L 1
-D
1 d7 4~4(I~h), 08(Ihi) and 0 R20-L 1
-D
1
A-R
1 -N
S-L
3
-D
3 10 c8 (IIj). wherein
R
2 and R' 2 are independently selected from among hydrogen, C1-6 alkyl, 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, 15 substituted heteroaryl, C 1
-
6 heteroalkyl, substituted C1- 6 heteroalkyl, C 1
.
6 alkoxy, aryloxy, 7 WO 2008/034124 PCT/US2007/078600 Ci- 6 heteroalkoxy, heteroaryloxy, C 2
-
6 alkanoyl, arylcarbonyl, C 2
.
6 alkoxycarbonyl, aryloxycarbonyl,
C
2
-
6 alkanoyloxy, arylcarbonyloxy,
C
2
-
6 substituted alkanoyl, substituted arylcarbonyl, C 2
-
6 substituted alkanoyloxy, substituted aryloxycarbonyl, C 2
-
6 substituted alkanoyloxy and substituted arylcarbonyloxy; 5 (c1), (c2), (e3), (c4), (c5), (c6), (c'6), (c"6), (c7) and (c8) are independently zero or a positive integer, preferably zero or an integer from about 1 to about 10, and more preferably zero, 1 or 2; (dl), (d2), (d3), (d4), (d5) and (d7) are independently zero or a positive integer, preferably zero or an integer from about 1 to about 10, and more preferably zero or an integer 10 from about I to about 4; and all other variables are previously defined. In another aspect, the polymeric systems described herein are either capped on one terminal with a CH 3 group, i.e. mPEG while in other embodiments, bis-activated PEGs are provided such as those corresponding to the formula: 0 0 0 H L-2D-3 H H 3D
A-R
1 -N
N-R
1
-N
15 0 0 0 o 0 0 A-R1-NH 0 0 NH-R 1 -NH 0 L1-D1, D1-L 1 Lj-D1, o 0 0 L3-D2 D2-L3 L3-D2
A-R
1 -NH NH-R§NH L1-Dj D1-L1! L1-D1 0 , 0 0 o 0 0 L3-D2 D2-L3 H L3-D2
A-R
1 -NH
N-R
1 -NH
HN-L
1
-D
1 , D1-L-NH
HN-L
1
-D
1 , 8 WO 2008/034124 PCT/US2007/078600 0 0 0 L,3-D2 D2-L3 L 3
-D
2
A-R
1 -NH NH-R 1 -NH
L
3
-D
2 /-,o 0/\\ F 0
A-R
1 -N 0 N-Rg-N 0 01 SL3-D2 3 2 A-R-NN-R-N O0 6 0 0 L-D, D -iL-I A R 1 NH LD 2
D
2
-L
3 o NH-RgNH & 0 0
L
1 -- D D-Li, L 1
-D
1 A-R-N0 NH-R-NH 5O L-DjL' 00 jD /L3-D2 2L L3D o 0 0 LjDj D1-L1, L-Dj A-R-N N-R 1 -N 5 D-L 1 S L-D o0 0 0 O O R 32-L R R-L3-D
N--R
1 -N 0O O R12 0-L-DD-L1- R 2 0L-D
S-L
3
-D
3 , ad D 3
-L
3 S3 S- L 3
-D
3 Other optional capping groups include H, NH 2 , OH, CO 2 H, Cp 6 r alkoxy and Cp- 6 alkyl. Preferred capping groups include mnethoxy and methyl. 10 in most aspects of the invention, the polymers included herein are generally described as substantially non-antigenic polymers. Within this genus of polymers, polyalkylene oxides 9 WO 2008/034124 PCT/US2007/078600 are preferred and polyethylene glycols (PEG's) are most preferred. For purposes of ease of description rather than limitation, the invention is sometimes described using PEG as the prototypical polymer. It should be understood, however, that the scope of the invention is applicable to a wide variety of polymers which can be linear, substantially linear, branched, 5 etc. In another aspect of the invention, the biological moieties include -NH 2 containing moieties, -OH containing moieties and -SH containing moieties. B. MULTIFUNCTIONAL LINKERS (L2) 10 In view of the structure of the present invention, the multifunctional linker allows attaching (releasable or permanent) 3 or more components, i.e. a targeting agent, a polymer and a biologically active cytotoxic compound like SN38. The artisan can appreciate that other molecules including at least three independent functional groups can also be used. One preferred multifunctional linker can be a residue of an aspartic acid or a lysine. 15 The L 2 having at least three functional site can be selected from among:
F
2 c1 O F 2 c2 2 c3 4 -+N -+N -N O NH-+ dl d2 (1b), d3 2 (Ic), d4 (Id), 0 0-4 c5 -? c6 c -+N R2 R2 c7 O -N e~cO - 04o d5OO 1< (le), c(If, -V (Ig), and 0 R2
O
-- N s- (Ih) wherein 20 R 2 and R'2 are independently selected from among hydrogen, C1-6 alkyl, C 2
-
6 alkenyl,
C
2
-
6 alkynyl, C3 19 branched alkyl, C3.
8 cycloalkyl, C 1
.
6 substituted alkyl, C2- 6 substituted 10 WO 2008/034124 PCT/US2007/078600 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, C 2
-
6 alkanoyl, arylcarbonyl, C 2 -_ alkoxycarbonyl, aryloxycarbonyl, C 2
-
6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted 5 arylcarbonyl, C2- 6 substituted alkanoyloxy, substituted aryloxycarbonyl, C 2
-
6 substituted alkanoyloxy and substituted arylcarbonyloxy; (cl), (c2), (c3), (c4), (c5), (c6), (c'6), (c"6), (c7) and (c8) are previously defined; and (dl), (d2), (d3), (d4), (d5) and (d7) are also previously defined. Preferred embodiments include: 0 O H -- 0 0 0H -- H H-H NN- -N o -1-N O -N N
-
O 9-NH O0/O1 and R s-k C. SUBSTANTIALLY NON-ANTIGENIC WATER-SOLUBLE POLYMERS (R 1 ) 15 The prodrugs of the present invention include a polymer residue R 1 , preferably water soluble and substantially non-antigenic polymers. Suitable examples of such polymers include polyalkylene oxides (PAO) such as polyethylene glycols. Certain preferred polymers are polyethylene glycols such as mPEG. A non-limiting list of such polymers therefore includes polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or 20 polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof. The polymer portion of the compounds described herein has an average molecular weight from about 2,000 to about 100,000 daltons, preferably from about 5,000 to about 11 WO 2008/034124 PCT/US2007/078600 60,000 daltons. In some aspects, the polyalkylene oxide can be from about 5,000 to about 25,000, and preferably from about 12,000 to about 20,000 daltons when proteins or oligonueleotides are attached or alternatively from about 20,000 to about 45,000 daltons, and preferably from about 30,000 to about 40,000 daltons when pharmaceutically active 5 compounds (small molecules such as those having an average molecular weight of less than about 1,500 daltons) are employed in the compounds described herein. Polyethylene glycol (PEG) is generally represented by the structure:
-O-(CH
2
CH
2
O),
where (n) is an integer from about 10 to about 2,300, and is dependent on the number 10 of polymer arms when multi-arm polymers are used. It will be understood that the water soluble polymer will be functionalized for attachment to linkers. Alternatively, the polyethylene glycol (PEG) residue portion of the invention can be represented by the structure:
-Y
7 r(CH 2
CH
2
O).-CH
2
CH
2
Y
7 -, 15 -Y 7 1
-(CH
2
CH
2
O),CH
2 C(=Y72)-Y7r,
-Y
7
"-C(=Y
7 2
)-(CH
2 )a 7
-Y
73
-(CH
2
CH
2 0)CH 2
CH
2
-Y
73
-(CH
2 )a 7 rC(=Y72)-Y7r ,
-Y
7 l-(CR 7 lR72)a 72 -Y73-(CH 2 )b 7 1-O-(CH 2
CH
2 0)(CH2)b71-Y73-(CR71R2)a72-Y71-, -Y7r(CH 2
CH
2
O),-CH
2
CH
2 -,
-Y
7
-(CH
2
CH
2
O).-CH
2 C(=Y72)- , 20 -C(=Y72)-(CH 2 )a 7 rY73-(CH 2
CH
2 0),CH2CH2-Y73-(CH2)a7C(=Y72)- , and -(CR7lR 72 )a 7 2-Y 7 3-(CH 2 )b 7 1-O-(CH 2
CH
2 O)(CH2)b7-Y73-(CR71R72)a72-, wherein:
Y
71 and Y 73 are independently 0, S, SO, S02, NR 73 or a bond;
Y
72 is O, S, or NR 74 ; 25 R 71
.
74 are independently the same moieties which can be used for R2; (a71), (a72), and (b71) 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. As an example, the PEG can be functionalized in the following non-limiting manner: 30 -C(Y 7 4
)-(CH
2 )m-(CH 2
CH
2 O)ir, -C(=Y74)-Y-(CH2)m.-(CH2CH2O)n, 12 WO 2008/034124 PCT/US2007/078600
-C(=Y
7 4 )-NRIi-(CH 2 )m-(CH 2
CH
2 0)ir,
-CR
75
R
76
-(CH
2 )m-(CH 2
CH
2 O)Rc wherein
R
75 and R 76 are independently selected from among of H, C1.
6 alkyls, aryls, substituted 5 aryls, aralkyls, heteroalkyls, substituted heteroalkyls and substituted C 1 _ alkyls; m is zero or is a positive integer, and preferably 1;
Y
74 is O or S; and n represents the degree of polymerization. Although, the prodrugs of the present invention can be formed using any of the 10 substantially non-antigenic polymers described herein, some preferred polyalkylene oxides include:
CH
3 0-PEG-O-(CH 2 )m-CO 2 H; PEG-NHS;
CH
3 0PEG-O-(CH 2 )m-NRjjR12; and 15 CH 3 0-PEG-O-(CH 2
)
2
-S-(CH
2 )m-CO2H; SC-PEG; or any of the art recognized activated PEGs. In particular, polyethylene glycols (PEG's), mono-activated, Ci 4 alkyl-terminated PAO's such as mono-methyl-terminated polyethylene glycols (mPEG's) are preferred when 20 mono- substituted polymers are desired; bis-activated polyethylene oxides are preferred when disubstituted prodrugs are desired. In order to provide the desired linkage, mono- or di-acid activated polymers such as PEG acids or PEG diacids are used. Suitable PAO acids can be synthesized by converting mPEG-OH to an ethyl ester. See also Gehrhardt, H., et al. Polymer Bulletin 18: 487 (1987) 25 and Veronese, F.M., et al., J. Controlled Release 10; 145 (1989). Alternatively, the PAO-acid can be synthesized by converting mPEG-OH into a t-butyl ester. See, for example, commonly assigned U.S. Patent Nos. 5,605,976 and U. S. Patent No. 5,965,566. The disclosures of each of the foregoing are incorporated by reference herein. Branched or U-PEG derivatives are described in U.S. Patents Nos. 5,643,575, 30 5,919,455, 6,113,906 and 6,566,506, the disclosure of each of which is incorporated herein by 13 WO 2008/034124 PCT/US2007/078600 reference. A non-limiting list of such polymers corresponds to polymer systems (i) - (vii) with the following structures: 0 Il mPEG--O-C N CH 2 H Y61 Y62 0 CH NC H mPEG--C
NCH
2 N H (i), 0 H IH r-PEG-N
CH--(Y
6 3CH 2 )wi1C(=0) H r-PEG-N-C o (ii), 0 II H m-PEG-0-C-N (CH2) 4
CH-(Y
63 CH 2 )1C(=) r-PEG--C CN H s 0 (iii), C Il m-PEG-0-C--NH (CH2)w62 N w (CH 2 )w 4 C(=0)--- | (CH2)w63 m-PEG-0-C-N H ) a (iv), 1H r-PEG0-C--N
HC--(Y
63
CH
2 )w 1 C(=0) rn-PEG-- C- N'(H)3 H C (v), and 14 WO 2008/034124 PCT/US2007/078600 0 II m-PEG-C-NH NH (CH2)ws2 HC (Y 63
CH
2 )w 6
IC(=O)
(CH
2 )w 63 m-PEG-C--N H o (vi) wherein: Y61-62 are independently 0, S or NR6i;
Y
63 is 0, NR 62 , S, SO or S9 2 5 (w62), (w63) and (w64) are independently 0 or a positive integer, preferably from about 0 to about 10, more preferably from about I to about 6; (w61) is 0 or 1; mPEG is methoxy PEG wherein PEG is previously defined and a total molecular weight of the polymer 10 portion is from about 2,000 to about 100,000 daltons; and
R
61 and R 6 2 are independently selected from among hydrogen, C 1
-
6 alkyl, C2- 6 alkenyl,
C
2
-
6 alkynyl, C 3
-
19 branched alkyl, C 3
-
8 cycloalkyl, C 1
-
6 substituted alkyl, C2-6 substituted alkenyl, C 2 -5 substituted alkynyl, C 3
-
8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C 1
-
6 heteroalkyl, substituted C1- 6 heteroalkyl, C1-6 alkoxy, aryloxy, 15 C 1 -6 heteroalkoxy, heteroaryloxy, C 2 -6 alkanoyl, arylcarbonyl, C 2
-
6 alkoxycarbonyl, aryloxycarbonyl, C 2
-
6 alkanoyloxy, arylcarbonyloxy, C 2
-
6 substituted alkanoyl, substituted arylcarbonyl, C2- 6 substituted alkanoyloxy, substituted aryloxycarbonyl, C 2
-
6 substituted alkanoyloxy, and substituted and arylcarbonyloxy. In yet another aspect, the polymers include multi-arm PEG-OH or "star-PEG" 20 products such as those described in NOF Corp. Drug Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference. The 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 15 WO 2008/034124 PCT/US2007/078600
H
2 C- O-(CH 2
CH
2 0),H HC-0-(CH 2 CH20),H
CH
2 0
CH
2 HL ---- (CH 2
CH
2 0)nH CH2 CH2 HC- 0-(CH 2
CH
2 0)nH H2C---O- (CH2CH2O)nH wherein: (x) is 0 and a positive integer, i.e. from about 0 to about 28; and (n) is the degree of polymerization. 5 In one particular embodiment of the present invention, the multi-arm PEG has the structure:
H
2 C-O -(CH 2
CH
2 0),H HC-0-(CH2CH 2 0),H CH2 I CH2
HIC---(CH
2
CH
2 0)nH CH2 0 CH2 HC- O-(CH 2
CH
2 0)nH I
H
2 -0-(CH 2 CH20),H 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 10 from 12,000 Da to 40,000 Da. In yet another particular embodiment, the multi-arm PEG has the structure: 16 WO 2008/034124 PCT/US2007/078600 0' 'o H O OH 00 OH OH or
(OCH
2
CH
2 )n-0-1 HO-(CH2CH2O)n
(OCH
2
CH
2 nOH HO.(CH2CH2Q)n 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 5 polymers having a total molecular weight of from about 5,000 Da to about 60,000 Da, and preferably from about 65 to about 270 (12,000-45,000 daltons) to provide polymers having a total molecular weight of 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 10 techniques described in US Patent Nos. 5,122,614 or 5,808,096. Specifically, such PEG can be of the formula: ~(CHHCH2O>Js O CH2CH2'O)~ O'CH2CH2-(OCH 2
CH
2 )u. Star _O-C2CH 0 :C 0,(CH2CH2).'CH2CH2'O_ OC 2H2(OCH2O H2)u7 Star or kO- CH 2
CH
2
-(OCH
2
CH
2 )u.-O O-(CH 2
CH
2 O)u-CH 2 CH-OI VOCH2CH2-(OCH 2
CH
2 )u Multi-arm O CH 2
CH
2 O)u-CH 2 CH2'O~2 wherein: 15 (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. 17 WO 2008/034124 PCT/US2007/078600 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: H3C~- (OCHCH)N 0' (CH2CH2O)..CHCH SO C OCH 2
CH
2 H) H3C(OCHCH)O OH 3 (CH2CH20)C.'CH H3C'-(OCH2CH2)u (CH2CH2O0u*>HCH O 0 (CH2CH2OV).CH2CH2'O H3C'( C 2 H )'N O H3C-(OCH2CH2)u OH 5 HsC- (ocHCH)N- O'CHH)i'CHH2CH2 OH o 0
,(CH
2
CH
2 0).. HO,-~ GO H CH2CH2,OH HOC'(OCH2CH)u''uOcOc H CH2H2-(OHCH)CH 2
H
2 O) HO'-HCH2CH(OCH2CH) CH2CH2OH 0 - Y c 0 (CH2CH2O ) u' 2C2s - ,CH2CH2(OCH2CH2)u.O- O O
H
3
C-(OCH
2
CH
2 )u.- O O-(CH 2
CH
2 0),.-CH 2 CH2-OH 0
H
3
C-(OCH
2
CH
2 )u-O O-CH 2
CH
2 0)u.-CH 3
H
3
C-(OCH
2 CH2)u.-O O-(CH2CH2O).-CH3
H
3
C-(OCH
2 CH2)u*-O 0
CH
2
CH
2 O)u,-CH 2
CH
2 -OH 18 WO 2008/034124 PCT/US2007/078600
H
3 C- (OCH 2 CH).-O O O-(CH 2
CH
2
O)..-CH
2
CH
2 --OH
H
3
C-(OCH
2
CH
2 )u.
0 0
-(CH
2
CH
2 O)u.-CH 2
CH
2 -OH
HO-CH
2
CH
2
-(OCH
2 CH2)u.-0 O-(CH 2
CH
2 0)u.-CH 2
CH
2 -OH 0) H3C-(OCH2CH2)u O 0
(CH
2
CH
2 O),.-CH3
H
3
C-(OCH
2 CH2)u-O
O-(CH
2
CH
2 0)u.-CH 2
CH
2 -OH
HO-CH
2
CH
2
(OCH
2
CH
2 )uK 0 0
CH
2
CH
2 O)u.-CH3
H
3
C-(OCH
2
CH
2 )u. OO O-(CH 2
CH
2 O)1.-CH 2
CH
2 -OH
HO-CH
2
CH
2
-(OCH
2 CH2)u 0 S OCH 2
CH
2 0)u--CH 2 CH2-OH
HO-CH
2
CH
2
-(OCH
2 CH)u- 0 O-(CH 2
CH
2 0)u.-CH 2
CH
2 -OH
H
3
C-(OCH
2 CH2)u O OCH 2
CH
2 0),.-CH 2 CH2-OH 5 and
HO-CH
2
CH
2
-(OCH
2
CH
2 )u.- O O-(CH 2
CH
2
O).-CH
2 CH2-OH
HO-CH
2
CH
2
-(OCH
2 CH2)u 0 0
(CH
2
CH
2 0)u.-CH 2 CH2-OH The polymeric substances included herein are preferably water-soluble at room temperature. A non-limiting list of such polymers include polyalkylene oxide homopolymers 10 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. 19 WO 2008/034124 PCT/US2007/078600 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, hydroxypropylmethacrylamide (HPMA), polyalkylene oxides, and/or copolymers thereof can be used. See also commonly-assigned U.S. Patent No. 6,153,655, the contents of 5 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 10 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 15 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 NaBH 4 . 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 20 (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%. In alternative aspects, polymers having terminal carboxylic acid groups can be employed in the polymeric delivery systems described herein. Methods of preparing 25 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 30 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 20 WO 2008/034124 PCT/US2007/078600 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. 5 D. TARGETING AGENTS (DI) Targeting agents can be attached to the polymeric compounds described herein to guide the conjugates to the target area in vivo. The targeting agents allow biologically active moieties such as pharmaceutically active compounds and oligonucleotides to have therapeutic efficacies at the target area, i.e. tumor site. The targeted delivery in vivo enhances the cellular 10 uptake of these molecules to have better therapeutic efficacies. In certain aspects, some cell penetrating peptides can be replaced with a variety of targeting peptides for targeted delivery to the tumor site. In one aspect of the invention, the targeting moiety, such as a single chain antibody (SCA) or single-chain antigen-binding antibody, monoclonal antibody, cell adhesion peptides .15 such as RGD peptides and Selectin, cell penetrating peptides (CPPs) such as TAT, Penetratin and (Arg) 9 , receptor ligands, targeting carbohydrate molecules or lectins, oligonucleotide, oligonucleotide derivatives such as locked nucleic acid (LNA) and aptamers, or the like, allows cytotoxic drugs to be specifically directed to targeted regions. See Crr Qpin Pharmacol 2006 Oct;6(5):509-1 4 , Cell-penetrating peptides as vectors for peptide, protein 20 and oligonucleotide delivery; see also JPharm Sci. 2006 Sep;95(9):1856-72 Cell adhesion molecules for targeted drug delivery, the contents of each of which are incorporated herein by reference. The targeting moieties can be labeled such as biotinylated compounds, fluorescent compounds, radiolabelled compounds. A suitable tag is prepared by linking any suitable 25 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 30 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. 21 WO 2008/034124 PCT/US2007/078600 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 13 1 Iodine, 12 Iodine, 99 1-Technetium and/or 1 'Indium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo. 5 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. Preferred targeting moieties are single-chain antibodies (SCA's) or single-chain variable fragments of antibodies (sFv). The SCA contains domains of antibodies which can 10 bind or recognize specific molecules of targeting tumor cells. In addition to maintaining an antigen binding site, a PEGylated SCA through linkers can reduce antigenicity and increase the half life of the SCA in the bloodstream. The terms "single chain antibody" (SCA), "single-chain antigen-binding molecule or antibody" or "single-chain Fv" (sFv) are used interchangeably. The single chain antibody has 15 binding affinity for the antigen. Single chain antibody (SCA) or single-chain Fvs can and have been constructed in several ways. A description ofthe theory and production of single chain antigen-binding proteins is found in commonly assigned U.S. Patent Application No. 10/915,069 and U. S. Patent No. 6,824,782, the contents of each of which are incorporated by reference herein. 20 Typically, SCA or Fv domains can be selected among monoclonal antibodies known by their abbreviations in the literature as 26-10, MOPC 315, 741F8, 520C9, McPC 603, D1.3, murine phOx, human phOx, RFL3.8 sTCR, 1A6, Se155-4,18-2-3,4-4-20,7A4-1, B6.2, CC49,3C2,2c, MA-15C5/KlGo, Ox, etc. (see, Huston, J. S. et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); Huston, J. S. et al., SIM News 38(4) (Supp):l1 (1988); McCartney, J. 25 et al., ICSU Short Reports 10:114 (1990); McCartney, J. E. et al., unpublished results (1990); Nedelman, M. A. et al., J. Nuclear Med. 32 (Supp.):1005 (1991); Huston, J. S. et al., In: Molecular Design and Modeling: Concepts and Applications, Part B, edited by J. J. Langone, Methods in Enzymology 203:46-88 (1991); Huston, J. S. et al., In: Advances in the Applications of Monoclonal Antibodies in Clinical Oncology, Epenetos, A. A. (Ed.), London, 30 Chapman & Hall (1993); Bird, R. E. et al., Science 242:423-426 (1988); Bedzyk, W. D. et al., J. Biol. Chem. 265:18615-18620 (1990); Colcher, D. et al., J. Nat. Cancer Inst. 82:1191-1197 22 WO 2008/034124 PCT/US2007/078600 (1990); Gibbs, R. A. et al., Proc. Natl. Acad. Sci. USA 88:4001-4004 (1991); Milenic, D. E. et al., Cancer Research 51:6363-6371 (1991); Pantoliano, M. W. et al., Biochemistry 30:10117-10125 (1991); Chaudhary, V. K. et al., Nature 339:394-397 (1989); Chaudhary, V. K. et al., Proc. Natl. Acad. Sci. USA 87:1066-1070 (1990); Batra, J. K. et al., Biochem. 5 Biophys. Res. Comm. 171:1-6 (1990); Batra, J. K. et al., J. Biol. Chem. 265:15198-15202 (1990); Chaudhary, V. K. et al., Proc. Natl. Acad Sci. USA 87:9491-9494 (1990); Batra, J. K. et al., Mol. Cell. Biol. 11:2200-2205 (1991); Brinkmann, U. et al., Proc. Natl. Acad. Sci. USA 88:8616-8620 (1991); Seetharam, S. et al., J. Biol. Chem. 266:17376-17381 (1991); Brinkmann, U. et al., Proc. Natl. Acad. Sci. USA 89:3075-3079 (1992); Glockshuber, R. et al., 10 Biochemistry 29:1362-1367 (1990); Skerra, A. et al., Bio/Technol. 9:273-278 (1991); Pack, P. et al., Biochemistry 31:1579-1534 (1992); Clackson, T. et al., Nature 352:624-628 (1991); Marks, J. D. et al., J. Mol. Biol. 222:581-597 (1991); Iverson, B. L. et al., Science 249:659 662 (1990); Roberts, V. A. et al., Proc. Nail. Acad. Sci. USA 87:6654-6658 (1990); Condra, J. H. et al., J. Biol. Chem. 265:2292-2295 (1990); Laroche, Y. et al., J. Biol. Chem. 266:16343 15 16349 (1991); Holvoet, P. et al., J. Biol. Chem. 266:19717-19724 (1991); Anand, N. N. et al., J. Biol. Chem. 266:21874-21879 (1991); Fuchs, P. et al., Biol Technol. 9:1369-1372 (1991); Breitling, F. et al., Gene 104:104-153 (1991); Seehaus, T. et al., Gene 114:235-237 (1992); Takkinen, K. et al., Protein Engng. 4:837-841 (1991); Dreher, M. L. et al., J. Immunol. Methods 139:197-205 (1991); Mottez, E. et al., Eur. J. Immunol. 21:467-471 (1991); 20 Traunecker, A. et al., Proc. Natl. Acad. Sci. USA 88:8646-8650 (1991); Traunecker, A. et al., EMBO J. 10:3655-3659 (1991); Hoo, W. F. S. et al., Proc. Natl. Acad. Sci. USA 89:4759 4763 (1993)). Each of the forgoing publications is incorporated herein by reference. Alternatively, the targeting moieties can be oligonucleotides or oligonucleotide derivatives. The oligonucleotides (analogs) are not limited to a single species of 25 oligonucleotide but, instead, are designed to work with a wide variety of such moieties, it being understood that linkers can attach to one or more of the 3'- or 5'- terminals, usually P0 4 or S04 groups of a nucleotide. The oligonucleotides include antisense or complementary oligonucleotides, short interfering RNA (siRNA), locked nucleic acid (LNA), micro RNA (miRNA), peptide nucleic acid (PNA), phosphorodiamidate morpholino oligo (PMO) and 30 aptamer, etc. 23 WO 2008/034124 PCT/US2007/078600 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, PD I and PD 1 peptides, EGF and EGF 5 peptides, RGD peptides, folate, etc. Other optional targeting agents appreciated by artisans in the art can be also employed in the compounds described herein. Preferably, the targeting agents include single chain antibody (SCA), RGD peptides, selectin, TAT, penetratin, Oligo(Arg), preferably (Arg) 9 , folic acid, etc. For purposes of the present invention, the term "TAT" can be understood to mean a portion of trans-activator of 10 transcription activation protein including a peptide sequence of YGRKKRRQRRR, for example. List of the sequences and structures of peptides used in the specification and examples includes: C-TAT: (SEQ ID NO: 1) CYGRKKRRQRRR; C-(Arg) 9 : (SEQ ID NO: 2) CRRRRRRRRR; 15 RGD can be linear or cyclic:
NH
2 HS HN HN O NH HO NH H HN N NH2 HN 0 N NH 2 NH JNH COOH OY COOH- O 000 0 Folic acid 0 OH 0 OH OH N OH H2NH
H
2 N! N Nr .;and 20 (Arg) 9 can include a cysteine for conjugating such as CRRRRRRRRR and TAT can add an additional cysteine at the end of the peptide such as CYGRKKRRQRRRC. 24 WO 2008/034124 PCT/US2007/078600 E. BIOLOGICALLY ACTIVE MOIETIES (Dz) A wide variety of biologically active moieties can be attached to the activated polymers described herein. The biologically active moieties include pharmaceutically active 5 compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides. Any cytotoxic or chemotherapeutic agent, i.e. folic acid, etc. capable of being attached to a polymer, including but not limited to those biologically active moieties are described in commonly assigned U.S. Patent No. 5,965,566, the contents of which are incorporated by 10 reference herein. In one 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. In another aspect, amine- hydroxyl- or thiol-containing biologically active moieties are 15 within the scope of the present invention. The only limitations on the types of the biologically active moieties suitable for inclusion herein is that there is available at least one hydroxyl- or thiol- group 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. 20 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 25 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. In one preferred embodiment, the polymeric transport systems described herein include pharmaceutically active compounds. For purposes of the present invention, it shall be understood to mean that the 30 pharmaceutically active compounds include small molecular weight molecules. Typically, the pharmaceutically active compounds have a molecular weight of less than about 1,500 25 WO 2008/034124 PCT/US2007/078600 daltons. A non-limiting list of such compounds includes DNA topoisomerase I inhibitors such as camptothecin and analogs, taxanes and paclitaxel derivatives, nucleosides including AZT and acyclovir, anthracycline compounds including daunorubicin and doxorubicin, related anti-metabolite compounds including Ara-C (cytosine arabinoside) and gemcitabine, 5 etc. 1. Taxanes and Taxane Derivatives One class of compounds included in the prodrug compositions of the present invention is taxanes. For purposes of the present invention, the term "taxane" includes all compounds 10 within the taxane family of terpenes. Thus, taxol* (paclitaxel), 3'-substituted tert-butoxy carbonyl-amine derivatives (taxotere*) and the like as well as other analogs available from, for example, Sigma Chemical of St. Louis, Missouri, are within the scope of the present invention. These compounds have been found to be effective anti-cancer agents. Numerous 15 studies indicate that the agents have activity against several malignancies. To date, their use has been severely limited by, among other things, their short supply, poor water solubility and hypersensitivity. It is to be understood that other taxanes including the 7-aryl-carbamates and 7-carbazates disclosed in commonly assigned U.S. Patent Nos. 5,622,986 and 5,547,981 can also be included in the prodrugs of the present invention. The contents of the foregoing U.S. 20 patents are incorporated herein by reference. Although the examples describe futer alia paclitaxel for illustrative purposes, it is to be understood that the methods described herein are suitable for all taxanes and related molecules. 25 2. Camptothecin and Related Topoisomerase I Inhibitors Camptothecin is a water-insoluble cytotoxic alkaloid produced by camptoteca accuminata trees indigenous to China and nothapodytes foetida trees indigenous to India. Camptothecin and related compounds and analogs are also known to be potential anticancer or antitumor agents and have been shown to exhibit these activities in vitro and in vivo. 30 Camptothecin and related compounds are also candidates for conversion to the prodrugs of the present invention. See, for example, U.S. Patent No. 5,004,758 and Hawkins, Oncology, 26 WO 2008/034124 PCT/US2007/078600 December 1992, pages 17-23. Camptothecin and related analogues are, for example, Topotecan, Irinotecan (CPT- 11) or SN3S. Additional camptothecin analogs include those reported in the literature including the I 0-hydroxycamptothecins, 11 -hydroxycamptothecins and/or 10,11 -dihydroxycamptothecins, 5 7-and/or 9- alkyl, substituted alkyl, cycloalkyl, alkoxy, alkenyl, aminoalkyl, etc. camptothecins, A-ring substituted camptothecins such as 10,1 1-alkylenedioxycamptothecins, such as those disclosed in U.S. Patent No. 5,646,159, the contents of which are incorporated herein by reference, etc. In one preferred embodiment of the invention, the cytotoxic agent is SN38. 10 3. Oligonucleotides In order to more fully appreciate the scope of the present invention, the following terms are defined. The artisan will appreciate that the terms, "nucleic acid" or "nucleotide" apply to deoxyribonucleic acid ("DNA"), ribonucleic acid, ("RNA) whether single-stranded 15 or double-stranded, unless otherwise specified, and any chemical modifications thereof An "oligonucleotide" is generally a relatively short polynucleotide, e.g., ranging in size from about 2 to about 200 nucleotides, or more preferably from about 10 to about 30 nucleotides in length. The oligonucleotides according to the invention are generally synthetic nucleic acids, and are single stranded, unless otherwise specified. The terms, "polynucleotide" and 20 "polynucleic acid" may also be used synonymously herein. Modifications to the oligonucleotides contemplated in the invention include, for example, the addition to or substitution of selected nucleotides with functional groups or moieties that permit covalent linkage of an oligonucleotide to a desirable polymer, and/or the addition or substitution of functional moieties that incorporate additional charge, 25 polarizability, hydrogen bonding, electrostatic interaction, and functionality to an oligonucleotide. Such modifications include, but are not limited to, 2'-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5 iodouracil, backbone modifications, methylations, base-pairing combinations such as the 30 isobases isocytidine and isoguanidine, and analogous combinations. Oligonucleotide 27 WO 2008/034124 PCT/US2007/078600 modifications can also include 3' and 5' modifications such as capping. A non-limiting list of nucleoside analogs include: O0 B 0B o1 B 0 0 0 0 F 0=P-S O4- Ot voO 0=P-0 Phosphorthioate 2'-O-Methyl 2'-MOE 2'-Fluoro B B B O O B 0 00 04-0 H Nl 2 2'-AP HNA CeNA PNA 0 0B o B B O B 0 /0 0 N P-N _- O=-O- 0 -0 Morpholino OH 3'-Phosphoramidate 2'-F-ANA 2'-(3-hydroxy)propyl 04 0
O=P-BH
3 Boranophosphates See more examples of nueleoside analogues described in Freier & Altmann; NueL Acid Res., 5 1997, 25, 4429-4443 and Uhlmann; Cu. Opinion in Drug Development, 2000, 3(2), 293-213, the contents of each of which are incorporated herein by reference. The term "antisense," as used herein, refers to nucleotide sequences which are complementary to a specific DNA or RNA sequence that encodes a gene product or that 28 WO 2008/034124 PCT/US2007/078600 encodes a control sequence. The term "antisense strand" is used in reference to a nucleic acid strand that is complementary to the "sense" strand. In the normal operation of cellular metabolism, the sense strand of a DNA molecule is the strand that encodes polypeptides and/or other gene products- The sense strand serves as a template for synthesis of a 5 messenger RNA ("mRNA") transcript (an antisense strand) which, in turn, directs synthesis of any encoded gene product. Antisense nucleic acid molecules may be produced by any art known methods, including synthesis by ligating the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to 10 form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated. The designations "negative" or (-) are also art-known to refer to the antisense strand, and "positive" or (+) are also art-known to refer to the sense strand In one preferred embodiment, the choice for conjugation is an oligonucleotide (or 15 "polynucleotide") and after conjugation, the target is referred to as a residue of an oligonucleotide. The oligonucleotides can be selected from among any of the known oligonucleotides and oligodeoxynucleotides with phosphorodiester backbones or phosphorothioate backbones. The oligonucleotides or oligonucloetide derivatives can include from about 10 to 20 about 1000 nucleic acids, and preferably relatively short polynucleotides, e.g., ranging in size from about 2 to about 200 nucleotides, or more preferably from about 10 to about 30 nucleotides in length. Further, oligonucleotides and oligodeoxynucleotides useful according to the invention include, but are not limited to, the following: 25 Oligonucleotides and oligodeoxynucleotides with natural phosphorodiester backbone or phosphorothioate backbone or any other modified backbone analogues; LNA (Locked Nucleic Acid); PNA (nucleic acid with peptide backbone); short interfering RNA (siRNA); 30 microRNA (miRNA); nucleic acid with peptide backbone (PNA); 29 WO 2008/034124 PCT/US2007/078600 phosphorodiamidate morpholino oligonucleotides (PMO); tricyclo-DNA; decoy ODN (double stranded oligonucleotide); catalytic RNA sequence (RNAi); 5 ribozymes; aptamers; spiegelmers (L-conformational oligonucleotides); CpG oligomers, and the like, such as those disclosed at: Tides 2002, Oligonucleotide and Peptide Technology Conferences, May 6-8, 2002, 10 Las Vegas, NV and Oligonucleotide & Peptide Technologies, 18th & 19th November 2003, Hamburg, Germany, the contents of which are incorporated herein by reference. Oligonucleotides according to the invention can also optionally include any suitable art-known nucleotide analogs and derivatives, including those listed by Table 1, below: TABLE 1 Representative Nucleotid Analogs And Derivatives 4-acetyleytidine 5-methoxyaminomethyl-2-thiouridine 5-(carboxyhydroxymethyl)uridine beta, D-mannosylqueuosine 2'-O-methylcytidine 5-methoxycarbonylmethyl-2-thiouidine 5-carboxymethylaminomethyl-2- 5-methoxycarbonylmethyluridine thiouridine 5-carboxymethylaminomethyluridine 5-ethoxyuridine Dihydrouridine 2-methylthio-N6-isopcntenyladenosine 2'-O-methylpseudouridine N-((9-beta-D-ribofaranosyl-2 D-galactosylqueuosine N-((9-beta-D-ribofuranosylpurine-6 2'-O-methylguanosine undine-5-oxyacetic acid-methylester Inosine N6-isopentenyladenosine wybutoxosine 1-methyladenosine pseudoundine 1-methylpseudouridine qucuosme 1-methylguanosine 2-thiocytidine I -methylino sine 5-methyl-2-thiouridine 2,2-dimethylguanosine 2-thiouridme 2-methyladeno sine 4-thiouridine 2-methylguanosine 5-methyluridine 30 WO 2008/034124 PCT/US2007/078600 3-methylcytidine N-((9-beta-D-ribofuranosylpurine-6-yl) carbamoyl)threonine 5-methyleytidine 2'-O-methyl-5-methyluridine N6-methyladenosine 2-0 methyluridine 7-methylguanosine wybutosine 5-methylaminomethyluridine 3-(3-amino-3-carboxy-propyl)udine Locked-adenosine Locked-cytidine Locked-guanosine Locked-thymine Locked-undine Locked-methylcytidine Preferably, the oligonucleotide is involved in targeted tumor cells or downregulating a protein implicated in the resistance of tumor cells to anticancer therapeutics. For example, any art-known cellular proteins such as BCL-2 for downregulation by antisense 5 oligonucleotides, for cancer therapy, can be used for the present invention. See U.S. Patent Application No. 10/822,205 filed April 9, 2004, the contents of which are incorporated by reference herein. A non-limiting list of preferred therapeutic oligonucleotides include antisense HIF- I a oligonucleotides and antisense Survivin oligonucleotides. In one preferred embodiment, the oligonucleotide can be, for example, an 10 oligonucleotide that has the same or substantially similar nucleotide sequence as does Genasense (a/k/a oblimersen sodium, produced by Genta Inc., Berkeley Heights, NJ). Genasense is an 18-mer phosphorothioate antisense oligonucleotide, TCTCCCAGCGTGCGCCAT (SEQ ID NO: 6), that is complementary to the first six codons of the initiating sequence of the human bcl-2 mRNA (human bcl-2 mRNA is art-known, and 15 is described, e.g., as SEQ ID NO: 19 in U.S. Patent No. 6,414,134, incorporated by reference herein). The U.S. Food and Drug Administration (FDA) gave Genasense Orphan Drug status in August 2000. Preferred embodiments include: (i) antisense Survivin LNA (SEQ ID NO: 3) mCs-Ts-mC -As-as-ts-cs-cs-as-ts-gs-gs-mC.-As-Gs-c; 20 where the upper case letter represents LNA, the "s" represents a phosphorothioate backbone; (ii) antisense Bcl2 siRNA: SENSE 5' - GCAUGCGGCCUCUGUUUGAdTdT-3' (SEQ ID NO: 4) ANTISENSE 3 1 - dTdTCGUACGCCGGAGACAAACU- 5' (SEQ ID NO: 5) 25 where dT represents DNA; 31 WO 2008/034124 PCT/US2007/078600 (iii) Genasense (phosphorothioate antisense oligonucleotide): (SEQ ID NO: 6) cts-Cs-ts- 8 s--s-Cs-as-9s-es-gs-ts-gs-Cs9s-Cs-es-Cs-as-t where the lower case letter represents DNA and and "s" represents phosphorothioate backbone; 5 (iv) antisense HIFIa LNA (SEQ ID: 7) 5'- ,TsGsGsc-aagscsastsescsTsGsTsa -3' (SEQ ID NO: 7) where the upper case letter represents LNA and the "s" represents phosphorothioate backbone. LNA includes 2'-O, 4'-C methylene bicyclonucleotide as shown below: B LNA Monomer Ow p-D configuration 01 See detailed description of Survivin LNA disclosed in U.S. Patent Application Serial Nos. 11/272,124, entitled "LNA Oligonucleotides and the Treatmemt of Cancer" and 10/776,934, entitled "Oligomeric Compounds for the Modulation Survivin Expression", the contents of each of which is incorporated herein by reference. See also U.S. Patent Application Serial 15 Nos. 10/407,807, entitled "Oligomeric Compounds for the Modulation HIF-1 Alpha Expression" and 11/271,686, entitled "Potent LNA Oligonucleotides for Inhibition of HIF-1A Expression", the contents of which are also incorporated herein by reference. The oligonucleotides employed in the compounds described herein can be modified with (CH2)w amino linkers at 5' or 3' end of the oligonucleotides, where w in this aspect is a 20 positive integer of preferably from about 1 to about 10, preferably 6. The modified oligonucleotides contemplated in the compounds described herein can be NH-(CH 2 )w Oligonucleotide. In one preferred embodiment, 5' end of the sense strand of siRNA is modified. For example, siRNA employed in the polymeric conjugates is modified with a 5'-C6-NH2. One 25 particular embodiment of the present invention employs Bcl2-siRNA having the sequence of SENSE 5'-(NH 2 -C)GCAUGCGGCCUCUGUUUGAdTdT-3' ANTISENSE 3'- dTdTCGUACGCCGGAGACAAACU-5'. 32 WO 2008/034124 PCT/US2007/078600 In another preferred embodiment, the compounds described herein can include oligonucleotides modified with hindered ester-containing (CH 2 )w amino linkers. See U.S. Provisional Application No. 60/844,942 entitled "Polyalkylene Oxides Having Hindered Ester-Based Biodegradable Linkers", the contents of which are incorporated by reference. 5 The polymeric compounds can release the oligonucleotides without amino tail. For example, the oligonucleotides can have the structure: N H-(CH2)w NH(HO-Oligonucleotide .0 wherein w is a positive integer from about 1 to about 10, preferably about 6. In yet another preferred embodiment, oligonucleotides can be modified with (CH 2 )w 10 sulfhydryl linkers (thio oligonucleotides). The thio oligonucletides can be used for conjugating directly to cysteine of the positively charge peptide or via maleimidyl group. The thio oligonucleotides can have the structure SH-(CH 2 )w-Oligonucleotide. The thio oligonucleotides can also include hindered ester having the structure: SH-(CH ) 0 O-Oligonucleotide 15 Exemplenary of the modified oligonucleotides include: (i) Genasense modified with a C 6 -NH2 tail: 5'- NH2- C 6 - stsestecsagscsgstsgscsgscsast -3' S H2N O -, 0--T-sC-sT-sC-sC-sC-sA-sG-sC.-sG-sT-sG-sC-sG-sC-sC-sA-sT (ii) antisense HIF 1 a LNA modified with a C 6
-NH
2 tail: 20 5'- NH 2
-C
6 - sTGGcsasasgscsastscscsTsGsTsa -3'; (iii) antisense Survivin LNA modified with a C 6
-NH
2 tail: 5'- NH 2 - C 6 - smCT 'CsAsastsescatsgsgs'Cs-sGsc -3'; (iv) antisense Survivin LNA modified with a C 6 -SH tail 5'- HS- C 6 - "CTs"CsAsatcscatgsgsmCAGsc -3'; 25 33 WO 2008/034124 PCT/US2007/078600 (v) Genasense modified with a hindered ester tail 0
H
2 N 0-T-C-T-C-C-C-A-G-C-G-T-G-C-G-C-C-A-T 4. Additional Biologically-Active Moieties 5 In addition to the foregoing molecules, the compounds of the present invention can be prepared using many other compounds. For example, biologically-active compounds such as cis-platin derivatives containing OH groups, floxuridine, podophyllotoxin, and related compounds can be included. Other useful parent compounds include, for example, certain low molecular weight 10 biologically active proteins, enzymes and peptides, including peptido glycans, as well as other anti-tumor agents, cardiovascular agents such as forskolin, anti-neoplastics such as combretastatin, vinblastine, vincristine, doxorubicin, AraC, maytansine, etc. anti-infectives such as vancomycin, etc. anti-fungals such as nystatin or amphoteracin B, anti-inflammatory agents, steroidal agents, and the like. 15 The foregoing is illustrative of the biologically active moieties which are suitable for the prodrugs of the present invention. F. PERMANENT AND RELEASABLE LINKERS: (LI and L 3 ) The L 1 and L 3 linkers include bifunctional linkers. The bifunctional can be permanent 20 or releasable linkers. The bifunctional linkers include amino acids or amino acid derivatives. 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 25 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-diaminopinelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylasparagine, 3-hydroxyproline, 34 WO 2008/034 124 PCT/US2007/078600 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarco sine, N-methyl isolceie, 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. 5 Alternatively, The L, and L 3 can be selected from among: [CQO0)]v(CR 22
R
23 )r-OLC&=O)],' 4[C(=O)]v,(CR 22
R
23 )r-NR 26 [C%=O)]' -[C(=O)]vO(CR R 23 )t[C&'O) iv' 10 -[CQ=O)]O(CR 22
R
23 )tO[CQ=O)]v,', -rc(=O)O(CR 22
R
23 )tNR 2 d[C(=O)],'-, 4[C&0)NRi(CR 22
R
23 )t[Cj0)lv,' K[C(=O)] Nk 2 i(CR22R2)tO[C&=O)]v,'-, -[C{=O)]-,NR 2 1 (CR 22
R
23 )tNR 26 LC(=O)lv-, 15 -[CQ-O)],(CR2R2)tO-KCR2sR29))t'[C(jO)i' -[C(=O)]v(CR22R 3 )tNR 26 -(CR28R29)t'[C(=O)1v,'-, -LCQ=O)]X(CR22R 23 )tS-(CR2sR29)ttC&=O)]v,>
-[C(=O)],O(CR
22
R
23 )tO4(CR 2 sR 29 )t'[CQ=O)]'-, -[CQ=O)1vO(CR 22
R
23 )tNR 2 6 5(CR 2 8R2 9 )t' IC(=O)]I'- , 20 -fCQO0)]vO(CR22R 23 )tS-{CR 2 sR2 9 )t4[C&O)]v'- , -[CQ=O)] ,NR 2 i (CR22R 23 )tO-(CR2sR29)t' [CQ"O)J,' -[C(=O)]vNR 2 K(CR22R4)NR 2 6-(CR2Rs)t'iC&=O)1v'-,
-[C(=O)],NR
2 1(CR 22
R
23 )tS-(CR 2 sR 29 )t'[C&=O)]v' -[CQ"O)Jv(CR22R 23
CR
2 8R 29 )tN R24[C&O)]v'm 25 -rc(=oA1(CR 22
R
23
CR
28
R
29 O)t[C(=O)J,'- , -[C(=O)]vO(CR 22
R
23
CR
2 8R 29 O)tN R 26 [C&-O)],'-, [C(=0)vO(CR 22
R
23
CR
28
R
29 0)t[CQ=O)lv,' 4ECQO)vNR 2 i (CR 22 R23CR 2
R
29 O)tNR 2 6IC&:O)]v'-,
-[C(=O)],NR
2 i (CR22R 23
CR
2 8R29)jC&O)1'-, 30 4[C&O)v(CR22R 23
CR
2 8R 29 O)t(CR24R25)t'W&0=)j, -[CQ=O)]vO(CR 22
R
23
CR
2 gR 29 O)t(CR 24 R25)tf[C(zO)]v' 35 WO 2008/034124 PCT/US2007/078600 -[C(=0)],NR 2 1(CR22R23CR28R290)t(CR24R25)e[C(=O)]v, -[C(=O)],(CR22R23CR 2 8R 2 90)(CR24R25)eO[C(=0)]'m -[C(=0)],(CR 22
R
23 )t(CR 2 4 R2sCR28R290)e[C(=O)11,
-{C(=O)],(CR
22 R23)t(CR 2 4
R
25 CR28R290)NR26[C(=0)],Q-, 5 -[C(=O)],O(CR 2 2 R23CR28R2 9 0)t(CR24R2s)'O[C(=O)Iv
-[C(=O)],O(CR
22
R
23 )t(CR 24
R
2 5CR 2 8R 2 90)t'[C(=O)]v,-, -[C(=O)],(CR22R23)t(CR 24
CR
25 CR28R29O)tNR26[C(=O)],
-[C(=O)]NR
2 1(CR22R 23
CR
2 8R 29 O)t(CR 24 R2)t'O[C(=O)],'-, -[C(=O)],NR2J (CR 22
R
23 )t(CR 2 4 R25CR28R290)t[C(=O)],- , 10 -[C(=O)]vNR 2 1(CR22R 23 )t(CR24R 25 CR28R290)tNR26[C(=O)],, 0 N 0 , H R27
-{C(=O)],O(CR
22
R
23 )t (CR 24
R
2 5)tNR 2 6[C(=0)]v, R27 -[C(=O)]vO(CR 22
R
23 )t
(CR
24
R
25 )tO[C(=0)].
R27 -[C(=O)]vNR 2 1(CR 22
R
23 )t (CR 24
R
2 5)tNR 2 6[C(=O)]v' R27 15 -[C(=O)vNR21
(CR
22
R
23 )t \ / (CR 24
R
25 )tOC(=o)] wherein: R21-29 are independently selected from among hydrogen, C 1
.
6 alkyls, C 3
-
12 branched alkyls, C 3
_
8 cycloalkyls, Ci 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 20 C 1
.
6 heteroalkoxy; 36 WO 2008/034124 PCT/US2007/078600 (t) and (t') are independently zero or a positive integer, preferably zero or an integer from about 1 to about 12, more preferably an integer from about 1 to about 8, and most preferably I or 2; and (v) and (v') are independently zero or 1. 5 Preferably, the bifunctional linkers can be selected from among: -[C(=O)]rNH(CH 2
)
2 CH=N-NHC(=O)-(CH2)2- ,
-[C(=O)]NH(CH
2 )2(CH 2
CH
2
O)
2
(CH
2 )2NH[C(=O)]r -[C(=O)],NH(CH 2
CH
2
)(CH
2
CH
2 0) 2 NH[C(=O)]r-, -{C(=O)]rNH(CH 2 CH2)sNH(CH 2
CH
2 )s,[C(=O)]r 10 -[C(=O)], NH(CH 2
CH
2 )sS(CH 2
CH
2 )s'[C(=O)],, [C(=O)]rNH(CH 2
CH
2
)(CH
2
CH
2 O)[C(=0)]r' [C(=O)]rNH(CH 2
CH
2 )sO(CH 2
CH
2 )s(fC(=O)]r -[C(=O)]rNH(CH 2
CH
2 O)(CH2)NH[C(=0)]r- -[C(=O)]rNH(CH 2
CH
2
O)
2 (CH2)[C(=O)]r'-, 15 -[C(=O)]NH(CH 2
CH
2 0)s(CH2)s'[C(=O)]r,-, -[C(=O)]rNHCH 2
CH
2 NH[C(=O)]r', -[C(=O)]rNH(CH 2
CH
2 )2O[C(=O)]r -, -[C(=O)rNH(CH 2
CH
2 0)[C(=O)lr -C(=O)]rNH(CH 2
CH
2 O)2[C(=O)], 20 -[C(=0)]rNH(CH 2
)
3 [C(=O)]r- , -[C(=O)]rO(CH 2
CH
2
O)
2
(CH
2 )[C(=O)]r--, -[C(=O)]rO(CH 2
)
2
NH(CH
2
)
2 [C(=O)]r', -[C(=O)]rO(CH 2
CH
2 O)2NH[C(=O)]C% -[C(=0)],O(CH 2
)
2 0(CH 2 )2[C(=O) 25 -[C(O)]rO(CH 2
)
2
S(CH
2 )2[C(=O)]r, -[C(=O)},O(CH2CH 2
)NH[C(=O)],
-[C(=O)]rO(CH 2
CH
2 )O[C(=0)1r-, [C(=O)]rO(CH 2
)
3 NH[C(=O)]r'-, -[C(=0)],O(CH 2
)
3 O[C(=O)]r'm 30 -[C(=O)]rO(CH 2
)
3 [C(=0)lr'-,
-[C(=O)],CH
2
NHCH
2 [C(=O)]r', 37 WO 2008/034124 PCT/US2007/078600 -[C(=O)rCH 2
OCH
2 [C(=O)r-, -[C(=0)]CH 2
SCH
2 [C(=O)]r'-,
-[C(=O)]S(CH
2
)
3 [C(=O)]- , -[C(=0)],(CH2)3[C(=O)]r- , 5 -[C(=O)]rOCH 2 \/CH 2 NH[C(=0)]r -[C(=O)]rOCH 2
CH
2 O[C(=O)]r' -{C(=O)]rNHCH 2
CH
2 NH[C(=O)]ra and -[C(=0)]rNHCH 2
CH
2 OC(=0)]r wherein (r) and (r') are independently zero or 1, provided that both (r) and (r') are not 10 simultaneously zero. 1. Releasable Linkers In one preferred embodiment of the invention, the compounds described herein contain a biologically active moiety attached to a releasable linker. One advantage of the 15 invention is that the biologically active moiety can be released in a controlled manner. Among the releasable linkers can be benzyl elimination-based linkers, trialkyl lock based linkers (or trialkyl lock lactonization based), bicine-based linkers, acid labile linkers, lysosomally cleavable peptides and capthepsin B cleavable peptides. Among the acid labile linkers can be disulfide bond, hydrazone-containing linkers and thiopropionate-containing 20 linkers. Alternatively, the releasable linkers are intracellular labile linkers, extracellular linkers and acidic labile linkers. The acidic labile linkers, such as hydrazone linkages, can be hydrolyzed in the acidic lysosome environment. Some suitable releasable linkers are oligopeptides including such as Val-Cit, Ala-Leu-Ala-Leu, Gly-Phe-Leu-Gly and Phe-Lys. 25 One preferred releasable linker is a peptidyl linker (Val-Cit) which can be specifically degraded by capthesin B. Preferably, the L 3 include releasable linker. The preferred releasable linkers include: 38 WO 2008/034124 PCT/US2007/078600 Y1 R31 4 - -L11-- -C---Y12-Ar---- -Y13-C- R32 all -L - bl1 - - e11 lil 11 15 - C-----C--C 0 R34 K36: Ar R37 Y17 R38 || | -C- L13 O0- --- (CR 4 4
R
45 ) -ill1 - - - -C (J x11 1 44 A 4 j L S F I- 42 k l l N 7 A51-(J')>c11---(L14)g11- -C- 1 --- C---(CR 46
R
47 ) m11 - - pl 1 1 R43nl
-
S-
R50
N-N-
5 R51, 0 -Val-Cit-, -Gly-Phe-Leu-Gly-, 39 WO 2008/034124 PCT/US2007/078600 -Ala-Leu-Ala-Leu-, -Phe-Lys-, 0 -FVal-Cit-0-N 0 11 H - -Phe-Lys-C-N \/ HN - -Val-Cit- O-Z 5 0 0, HN -- Phe-Lys-O o 0, -Val-Cit-C(=O)-CH 2 OCH2-C(=O)-, -Val-Cit-C(=0)-CH2SCH2-C(=O)-, and
-NHCH(CH
3 )-C(=O)-NH(CH2)6-C(CH3)2-C(=O) 10 wherein,
Y
11 1 9 are independently 0, S or N 1 4 8 ;
R
3 1 4 8 , R 5 0
.
51 and A 5 1 are independently selected from among hydrogen, C 1
.
6 alkyls, C 3 . 1 2 branched alkyls, C 3 -8 cycloalkyls, C 1
.
6 substituted alkyls, C 3
-
8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, CI 6 heteroalkyls, substituted C 1
-
6 heteroalkyls, C 1
.
6 alkoxy, 15 phenoxy and C 1 .6 heteroalkoxy; Ar is an aryl or heteroaryl moiety; 1
-
15 are independently selected bifunctional spacers; J and J' are independently selected from selected from among moieties actively transported into a target cell, hydrophobic moieties, bifunctional linking moieties and 20 combinations thereof; (c11), (h11), (k11), (111), (m11) and (n11) are independently selected positive integers, preferably 1; (al1), (ell), (g1l), (j11), (o1l) and (ql1) are independently either zero or a positive integers, preferably 1; and 25 (b 11), (x11), (x'l1), (fl1), (il l) and (p11) are independently zero or one. 40 WO 2008/034124 PCT/US2007/078600 Various releasable linkers, benzyl elimination based or trialkyl lock based, are described, for example, in commonly assigned U.S. Patent Nos. 6,180,095, 6,720,306, 5,965,119, 6624,142 and 6,303,569, the contents of each of which are incorporated herein by reference. The bicine-based linkers are also described in commonly assigned U.S. Patent Nos. 5 7,122,189 and 7,087,229 and U.S. Patent Application Nos. 10/557,522, 11/502,108, and 11/011,818, the contents of each of which are incorporated herein by reference. Preferably, the biologically active compounds including oligonucleotides are linked to the polymeric portion of the compounds described herein via acid labile linkers. Without being bound by any theory, the acid labile linkers facilitate release of the oligonucleotides 10 from the parent polymeric compounds within cells and specifically in lysosome, endosome, or macropinosome. 2. Permanent Linkers In preferred aspects of the invention, the targeting moiety such as the SCA is attached 15 to the multifunctional linker through a permanent linker. The permanent linkers are capable of conjugating The targeting moiety and the multifuinctional linker. One preferred permanent linker can be a molecule like a maleimidyl-containing molecule which can provide a thio ether bond. The permanent linkers containing maleimidyl groups can be selected from among: o 0 0 20 0 0 H 0 0 H 0 N H<NX N \-NxN -IN 0 ,0, 0 o 0 0 0 0 N N 0 0 ,0 and In an alternative embodiment, the permanent linkers include structures corresponding to those shown above but instead of maleimidyl group have groups such as vinyl, residues of 25 sulfone, amino, carboxy, mercapto, hydrazide, carbamate and the like instead of maleimidyl. 41 WO 2008/034124 PCT/US2007/078600 G. 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 5 hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, Ci-C 6 alkyloxy, C-C 6 alkanoyloxy, arylcarbonyloxy, ortho-nitrophenoxy, N-hydroxybenzotriazolyl, imidazole, pentafluorophenoxy, 1,3,5-trichlorophenoxy, and 1,3,5-trifluorophenoxy or other suitable leaving groups as will be apparent to those of ordinary skill. 10 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 and a targeting moiety. In some preferred embodiments, functional groups to link the polymeric transport systems to biologically active moieties include maleimidyl, vinyl, residues of sulfone, amino, 15 carboxy, mercapto, hydrazide, carbazate and the like which can be further conjugated to a biologically active group or a targeting group. For purposes of the present invention, when the D 1 or D 2 are functional groups, the L, and L 3 groups are not functional moieties. In one preferred embodiment, the functional group can be maleimidyl and the leaving 20 groups can be selected from among OH, methoxy, tert-butoxy, para-nitrophenoxy and N hydroxysuccinimidyl. H. PREFERRED EMBODIMENTS CORRESPONDING TO FORMULA (I) Some particular embodiments prepared by the methods described herein include: 0 II HNW -Cit-C-
D
2 H mPEG N 0 O 0 0 H ' ' D, 25 42 WO 2008/034124 PCT/US2007/078600 0 0
D
2 -C-Cit-ValNH HN Val-it-C-D2 H H N P N 0 0 0 0 N N N DH 0 H o 0 HN mPEG 0 O 0 ON N 0 H DI o o 0 D- - -- ValNH H C Cit Va HN Val Cit-/- O 'NH H H 0 ONPE N 0 0 0 Nq1NPEG,/,, 0 _t ''-N 0 0.0ND D, H 0 0H 0 Ii 4 ,Val-Cit-C--N D2 HN N mPEG 0 0 00 0 0 D2 /--(7/ - - i - a N - - 2D/ Ho HNH HN 00 N P G N 0 _t -- O -N 0 0 . B 1
,,O-
DlH 0 0 H D 5 o 0 43 WO 2008/034124 PCT/US2007/078600 0 11 HN Val-Cit-C-D 2 H mPEG D 0 t _ N DI 0 0 0
D
2 -C-Cit-Val NH HN Vab-Cit-C-D2 H H D1 O O ~_ PEG D1 N 0 0 0 0 HN -Val--Cit-C\ D2 N mPEG 0 0 0 N D 0 0 0 C-Cit-Val HN Val-Cit-C NN N 0 D DPEG 0 0 N N OO 0 0 HN /Val-Cit--NH O H mPEG 0 0 0 N D 5 0 o 0 D2/ NH- C-- Cit- Val -Val-Cit- -NH / 2 o H H 0 N IN O N PEG N 00
D
1 N 0 0 N D, 0 0 44 WO 2008/034124 PCT/US2007/078600 0 11 H HN Val-Cit-C-D2 mPEG N O N N 0 H D1 0 o 0
D
2 -C-Cit-Val HNVal-Cit-C-D2 H H O0 N PEG N O tN O N N D1 N O H H -D1 N 0 00 H HN 'Val-Cit-C--D2 mPEG N H 0 N0D1 0 D--C-Cit -- VlHN Val-Cit-C--D2 H HNH H02 H O O mPEG O HR51 N-R52 o N S'D1 0 0 0
D
2 -C-CitVal Val-Cit-C-D2 NRH H HN N'R51NH PEG H N' Dj3 N 0 0 N S D1 o-) 0 O 00 45 WO 2008/034124 PCT/US2007/078600 0 H H N X R 5 R 5 / D H 02 N R51wN-R52 mPEG H N o N D1 0 o / -Cit-Val HNVal-Cit D2 IJH H2 NR52, R51N PEG HR 51 '-N R52 2 N-H 0PEG 0 H D S N 0 0 N D1 0 0 0 Va - I -N D2 H HN O / 02 N ItR51,N'R5120 mPEG N H o N S D O 0 0 D2 F / NH-C-Cit--ValN HN'W-Cit---NH \ D2 NOH H o R52N' H N PEG N O HR51 N'R52 0 S N 0 *N S D 0 0 0
II
H NIPhe-Lys-C-D2 H H N R51>N-R52 mPEG 0 H I o N S'D1 50 o 0
D
2 -C-Lys-Phe .Phe-Lys-C-D2 -NH H H HN
R
5 2 'N R 51 N N R51'N'R52 N 0PEG H D1-S Y N 0 0 N S'D1 0 0 46 WO 2008/034124 PCT/US2007/078600 0 H HN Phe-Lys D2 mPG--yN 0 HR51 'N'R52 . mPEG N H~ ,R o N D, 0 O 0 C-Lys-PheNH HN Phe-Lys-C - D/ NH H H I o R R 2 R52N' R51H PEG N H 51 NN 52 D1 S N 0 o N S D1 0 0 0 Phe-Lys-C-NH- D /2 H H Y O N R51'N'R52 O mPEG O H o N S D o and 00 D2/ NH-C-Lys-PheNH HN'Phe-Lys---NHD R5,R1H H - RmN'R H N PEG O N R52 oS NN NO D S, N 0 0 N Dl 0 0 5 wherein mPEG has the formula: CH 3
-O(CH
2
CH
2 O).-; PEG has the formula -O(CH 2
CH
2 0).-; (n) is a positive integer from about 10 to about 2,300; 10 R 51 and R 52 are independently selected from among 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 16 heteroalkyls, substituted C 1
.
6 heteroalkyls, C 14 - alkoxy, C1-6 alkyloxycarbonyl, aryloxycarbonyl, phenoxy and C 1
.
6 heteroalkoxy;
D
1 is a targeting moiety, a functional group or a leaving group; and 15 D 2 is a biologically active moiety, a functional group or a leaving group. Preferred polymeric compounds according to the present invention include: 47 WO 2008/034124 PCT/US2007/078600 0 II HN -Val-Cit-C-SN38 H mPEG 0 O 0 N>~N O H O 0 H 0 O SN38-C-Cit-Val N-Val-Cit-C- SN38 H H O N PEG N O O 04 0 0 Nw N O H OO OHN O II HN Val-Cit-C-SN38 H mPEG 0 0 N - N 0 H/ Ob O 0 SN38-C-Cit-Val'NH HN Val-Cit-C-SN38 H H N PN O PEGN 0H o H H 0 0 H O 0 0 HN Val-Cit H SN38 mPEG 0 O 5 0 48 WO 2008/034124 PCT/US2007/078600 0 0 - /Cit-ValNH HN Val SN38 H H ON NEG 00 0 NN O H O OH O 0 0 HHVal-Cit--N SN38 NH H H - V mPN PEG N O0 0 0 H 11 N0 OH O OO 0 0 SN38- -- i-ai NH -Val-Cit-C-N SN38 HN E0 0 PEG N 0 O0 0 0 H 0 0H 00 HN -Val- it-C SN38 H mPEG 0 0 0 t ,N\ 0 0 0
SN
3 8 C-~Cit-ValNH H H NV-Ct -S3 H NN0 0 0 0 H HN SN38 mnPEG 0 0 0 49 WO 2008/034124 PCT/US2007/078600 O 0 SN38 / -Cit-Val' HH HN Val-Cit-SN38 H H HNVl- - NH ON3 0 0 N PEG N 0 O N O 0 N o 0 0 HN Val-Cit-C-H- SN38 H N mPEG 0 0 N N .0 o 0 N38 N-C-Cit-ValN HVal-Cit-C- S SN38 H HMN 6 0 N PEG N 0 5 N 0 N 0 0 0 HN SSN38 N mPEG H NHBoc o N s o NO 2 , o 0 SN38-C-Cit-Val 'N HHN VlCt -SN38 NHBoc 0 --- PEG o NHBoc N (;NH _,H x ' N 0 0 N S, NO 2 0 o NO 2 , 0 HNaIC tC S N38 mPEG N 0NHBoc o NI S~. N o NO 2 50 WO 2008/034124 PCT/US2007/078600 o 0 -- Cit- Val RHN'Val-Cit C z SN/ SN38 N H H SN38 NNN N o NNHBoC N S NHBoc PEG Vaj-Cit-C-NH-(7) SN38 HN O N ~ NO2c S N 3 8 N - 0Ct - a N HH0a - C t - - NHNS 3 OPEG O
NO
2 N O NO , 0 1 1 Phe-Cit-CNH SN38 SN NG NH~o NN 0< mPEG H O N S O
NO
2 , 50 NO O0 N O2 0 0 Phe-Lys-C S3 HN S3 mPEG N o H NHBoc N 0 0
NO
2 OoNO2N, 11 51 SN8C-y-Pe-N HN.Phe-Lys-C-SN3 mPEG 0 NBO Nq--E Jt Ho H S, Os N,-00N , S 0 0 SN3& LysPhe-'Pe-Ls-CSN3 NN Nc 0S PGNNoN X AE N 0 a NHo 0 N-l-- s0 -Y 0 NN02 51 WO 2008/034124 PCT/US2007/078600 0 0 \ / C Lys-Phe -,Phe-yC SN38 -NH H H HN SN38 S N PEG o NHBoc H Hr " - I --- I N,- 0 0 N S. '
NO
2 0 o NO 2 0 H Phe-Lys-C-NH O SN38 H HN0 mPEG ON NHBoc N O N --- SN ' O NO 2 *-0 0 SN38 / NH-C-Lys-Phe HPhe Lys-C NH O / SN38 0 NH H HMN' 0 N NHBoc N PEG* *-N HNHBoc NA: 0 HH N0N02 N 0 'N0 O S cSEGH
NO
2 OO
NO
2 0 II H HN Val-Cit-C-SN38 N mPEG 0 O
NH
2 O 0 SN38-C-Cit-Val -Val-Cit-C-SN38 H H NH N N PEG N 5
H
2 N 0 0 NH 2 H H LN N OLO-LNA HHNH N mPEG 0 o -INH 2 0 H H 0 LNA-O1 NO-LNA CNH HHHN:) 0 N g-PEG-r 0 [:
H
2 N 0 0 NH 2 52 WO 2008/034124 PCT/US2007/078600 0 II HN Va3-Cit-C-SN38 H mPEG N o H S-SCA o 0 SN38-C-Cit-Val'NHH HN ,Val-Cit-C-SN38 H H SN PEG 0 0 tN N OO NN 0N SCA-S H 0 0 H S-SCA oO 0 II HNXVa--Cit-C-SN38 H mPEG 0 O 0 N N O H S-SCA 0 H0 o 0 SN38-C-Ct-Va NH HN Val-Cit-C-SN38 H H 0 PEG N O N 0 a N SCA-S H 0 H S-SCA o0 0 HNX Val--Cit H SN38 mPEG N O H S-SCA 5~ H 0 5 0 53 WO 2008/034124 PCT/US2007/078600 0 0 C-Cit-ValNH HN-Val--Cit SN38 SN38 H H 0 PEG N 0 N0O N 0 O SCA-S H 0 0 H S-SCA 0 H-Val--Cit-C-N SN3 HN SN38 mPEG N 0 O O 0 0 O H S-SCA o H 0 0 )N-C-Cit--VaI ,Val-Cit-C- N- \ / SN38- N- H 'NH H H HN H SN38 O N P G N 0 00 0 N rPEG -- N 00 0 -Ol N j0 0 N 0 ~ SCA-S H 00 H S-SCA 0 || HN -Va[Cit-C--SN38 H mPEG - yN 0 0 N S-SCA 0 o 0 I I I I SN38-C-Cit-ValNH HN Val-Cit-C-SN38 5 0 0 0 HN -Val-Cit H SN38 mPEG 0 0 0 N S-SCA 0 54 WO 2008/034124 PCT/US2007/078600 0 0 11 11 \ -Cit-Val-NH HN Val-Cit-c SN38 SN38 H H 0 0 N PEG N 0 SCA-S N 0 0 N S-SCA 0 0 0 H HNVal-Cit-C--NH S3 HN / SN38 mPEG -' N 0 00 0 N S-SCA 0 0 0 \N3NH-C--Cit-ValNH HN \Val-Cit-N SN38 SN38 HH H a i HV 0 0 N PEG 0 0 O - N 0 0 N acSCA 0 0 0 S- -It-ValVa-CtNSN38 N mHPEG H 0 Nai NFolio acid 0 0 0 5 O1O1 Ii II SN38-C-Cit Val - CVai-Cit-CSN3 NH H H NH2 ON N 0'S-S 0 0 0 11 HHN ,VltnC SN38 mPEG 0,, t NH 2 0 N -?- ,SSCA 0 55 WO 2008/034124 PCT/US2007/078600 0 0 SN38- -Cit-Val /Val-Cit-C-SN38 NH HHN
NH
2 N PEG H NH 2 SCA-S'S N o 0 N 5 S-SCA 0 0 0 H HN SN38 mPEG N H NH 2 O N &SS-SCA 0 o .0 0O \/ C-Cit- VaI ,aI -Ci-NH HN -Va-Cit- SN38 SN38CH N 0 S mPE1H O NH 2 SN3 P H SSC SCA-S- N a N ( S-SCA O0 0 H H h/ SN3 mPEG N O H NH 2 0 o N FolS-SCA 0 0 0 \/NH-C-Ct-Va 'NH HN VaWCih-C-NH \ / -N3 SNSB-, H H0NS3 0NH.
2 0 ~ N NI- 2 0 IC~S H 0 0 0 0 N mPEG -y to H 0 N-T Folio acid 0 56 WO 2008/034124 PCT/US2007/078600 SN38-C-Lys-Phe NPhe-Lys-C-SN38 NH HN H 0 PEG H Folic acid N 0 0 N Folic acid o 0 0 ll H HN-Phe--Lys-C--SN38 H N mPEG N H NH 2 o N SS-SCA 0 o 0 SN38---Lys-Phe HNPhe-Lys-C-SN38 SN3SNH H HHN Y
NH
2 H PEG N H NH 2
SCA-S-<
3 N O O N3S S-SCA 0 0 0 Phe-Lys- H HN SN38 mPEG N . H NH 2 o N SS-SCA o o 0 \ -Lys-PheNH Phe-Lys- SN38 SN38 H H
NH
2 N PEG N H NH 2 SCA-S's N 0 0 N S'S-SCA 5 0 0 0 HNXPhe-Lys-C-NH H HN SN38 mPEG N H NH 2 0 o N SS-SCA 0 57 WO 2008/034124 PCT/US2007/078600 0 0 SN38 NHC--Lys-Phe Phe Lys---NH SN38 0 NH 2 H N PEG N H NH 2 O SCA-S- N 0 0 S-SCA 0 0 0 II HN Val-Cit-C-SN38 H mPEG N 0 o H S-RGD o 0 SN38-C-Cit-Val H-Val-Cit-- N38 H H 0 PEG N O N N O O N N,,, RGD-S H 0 H S-RGD 0 HN-Val-Cit-C-SN38 H N mPEG 0 0 H O NN o H S-RGD 0 0~ 0 SN38-C-Cit-ValNH HN Val-Cit-C-SN38 H H o PEG N 0 O o 0 N N NN RGD-S H 0 H S-RGD 5a 0 HN Val-Cit- SN38 N mPEG 0 0 N N S-O 0 H H S-RGD 58 WO 2008/034124 PCT/US2007/078600 0 \ -Cit-ValNH HN SN38 SN38 H H 0 0 N PEG N 0 0 0 0 NO>N RGD-S H0 O S-RGD 0 11 Val-Cit-C-N\ HN SN38 mPEG 0 O' N - O '-N 00 H S-RGD SN38 N - H NH H HNVH SN38 N PEG> N O - O- N 0 0N RGD-S H 0 0 O H S-RGD 0 HN Val-Cit-C-SN38 HN N m P E G 0S R 0 t _ N S-RGD 0 o 0 I I I I SN38- C-Cit-Val NH HN Val-Cit-C-SN38 0 Nr-- PEG N 0o RGD-S N 0 0 N S-RGD 5 0 0 HHN Val-Cit-C-E S3 H SN38 mPEG 0 0 0 N S-RGD 0 59 WO 2008/034124 PCT/US2007/078600 0 0 C-Cit-ValNH HHHN Val-Cit-C SN38 SN8H H 0N Y PEG N RGD-S N P 0N S-RGD 0 0 0 11 H ,HNVal-Cit-C--NH S3 H SN38 mPEG 0 O 0 N S-RGD 0 0 0 NH-CCft-Val'N H HN -Val-Cit-C NH 0 0 Nr N E ,I J 00O RGD-S N .0 E N S-RGD O 0 0 Il -Val-Cit-CSN38 H H N PEG H 0 N NH-RGD O 0 0 00 SN38 C Cit-Vali HNC SN38 NPG-" N 0 H H H 0PEGH RGD-NH N 0 0 N NH-RGD 5 00 06 HHN-Vl-itC SN38 mPEG N
NH
2 0 N SSRGD 0 60 WO 2008/034124 PCT/US2007/078600 0 0 11 1 SN38-C-Cit-Val HN Val-Cit-C-SN38 NH HHN
NH
2 0 PEG H NH 2 RGD-S-8 , N, O O N -- ( S-RGD H H N SN38 N H NH2 0 N S S-RGD 0 0 O \-Cit-Val'NH HHN Val-Cit- SN38 HN3 SNH NH2 H N PEG N NH2 RGD-S s N O O N S S-RGD O -( 0 0 C HNVal-Cit-H-NH SN8 HN SN38 H mPEG NH NH2O O N S0 S-RGD O 0 0 0 SN38 NH- I-it-aNH \ /~~lCt HS3 H HNPhe-Lys-C-SN38 H N mPEG 0 H o N NH-RGD o00 61 WO 2008/034124 PCT/US2007/078600 o 0 SN38-C-Lys-Phe Phe-Lys-C-SN38 *NH H H HN H N PEG N o H RGD-NH N 0 0 N NH-RGD 0 0 0 - II H HN Phe-Lys-C-SN38 mPEG N H NH 2 o N SNSRGD 0 o 0 SN38 C-Lys-Phe NPheLsC NH HHHN~~eLsC S3
NH
2 H PEG H NH 2 RGDSS N 0 0 N 0SRGD 0 0 0 P he-Lys-C H HNSN38 N mPEG N H NH 2 o N S-RGD o o 0 SH H HNXe-Lys- SN38 SN38 NH SH3
NH
2 H N PEG 0N O H NH 2 RGD-S-8 N 0 0 N S'S-RGD I0 0 0 HPhe-Lys--C-NH \ / N mPEG H NH 2 0 0 N S S-RGD 0 62 WO 2008/034124 PCT/US2007/078600 o 0 \N3 NH-O--Lys-Phe HNPy -NH SN38 SNS8-- l NH H H NNH0 o NH 2 H N Eo H NH2 RGD-S-S N 0 0 N S S-RGD o 0 H 0 O N 'O-LNA HN H mPEG N 0 H O N Folic Acid 0 o H H 0 N 0 0 N LNA-O NH HN N O-LNA FH ic AcidPEG NYNL Folic Acid Folioc Acid N 0 N oio 0 0 H L N O-LNA HN:: H mPEG 0 H 0 NAiPFolio Acid 0 and H H NH HHHN:: H 0 N 1,-PEG ^rNH Fol io Acid -TrN 0 0 :o-N--,Folic Acid 5 O 0 wherein S-SCA is a single-chain antibody; RGD is 63 WO 2008/034124 PCT/US2007/078600 HS HN NH HN N N NH 2 Hr HN_ NH COOH LNA is locked nucleic acids; Folic acid is a residue of OH N HN N HO N ' ? HNIIl" SOH 5 SN38 is 7-ethyl-10-hydroxycamptothecin; mPEG has the formula: CH 3
-(CH
2
CH
2 O).-; PEG has the formula -O(CH 2
CH
2 O)n-; and (n) is a positive integer from about 10 to about 2,300. 10 . METHODS OF MAKING THE CONJUGATES Generally, the conjugates can be made by sequentially attaching the polymer, cytotoxic agent and targeting moiety to the multifunctional linker. The exact order of addition is not limited to this order and as will be apparent to those of ordinary skill, there are aspects in which the PEG will be first added to the multifunctional linker followed by the addition of 15 the releasably attached cytotoxic drug followed by the addition of the targeting agent like the monoclonal antibody. Details concerning some preferred aspects of this embodiment are provided in the Examples below. In one aspect of the invention, the polymeric compound having a multifimctional linker can be prepared by conjugating a polymeric compound having a OH or a leaving group 20 terminal with a nucleophile containing a cytotoxic agent attached through an optional linker. The artisan can use less amount of the nucleohile compare to the number of the leaving groups on the polymer to form a polymeric intermediate containing both cytotoxic agent and 64 WO 2008/034124 PCT/US2007/078600 leaving groups. This intermediate can further reacted with a targeting moiety to form the polymeric conjugate multisubstituted with cytotoxic agent and targeting agent. Alternatively, the polymer can be activated with different groups to provide different chemical reactivities toward various nucleophilic moieties. For example, different protecting 5 groups such as tert-Bu ester and methyl ester of carboxylic acid terminals can be deprotected selectively and stepwise to provide various degrees of active group to be conjugated with different biologically active agents such as cytotoxic agent and targeting agent. As shown in FIG. 1, maleimidyl group and succinimidyl ester can react selectively with SH or NH 2 containing moieties, respectively. 10 All reactions described herein are standard chemical reactions with necessary steps and conditions known to those of an ordinary skill. The synthetic reactions described herein therefore do not require undue experimentation. The methods of preparing a polymeric conjugate containing multifunctional linkers comprising: 15 reacting a compound of Formula (Ila): Ml-(L 1 )a-L 2 -M2 R1
A
21 (Ilia) with a biologically active moiety-containing compound having Formula (11b): M3-(L3)b-D22 under conditions sufficient to form a compound of Formula (Ilc): Ml-(L 1 )a~L 2 -(L3)b- D 22 1 20 A 22 (IIc) wherein R, is a substantially non-antigenic water-soluble polymer; A21 is a capping group or Ml-(L,),--L2~M2 25 A 22 is a capping group or 65 WO 2008/034124 PCT/US2007/078600
M
1 (L1)a-L 2 -(L3)b-D 22 M 1 is a functional group;
D
22 is a biologically active moiety;
M
2 is OH or a leaving group; 5 M 3 is OH, NH2,or SH;
L
1 is a permanent linker or a releasable linker; L2 is a niultifinctional linker;
L
3 is a permanent linker or a releasable linker; and (a) and (b) are independently zero or a positive integer. 10 The methods further include reacting the compound of Formula (IlIc):
M
1
-(L
1 )a-L 2 -(L3)b-D22 R1
A
22 (IIc) with a nucleophilic moiety-containing moiety having Formula (111d) D21-M4 (Id) 15 under conditions sufficient to fonn a compound of Formula (Ie):
D
2 1-(L1).-L 2 -(L3)b-022
A
23 (Ile) wherein:
A
23 is a capping group or D21-(L1),--L2-~(L3)b-D22 20 M 1 is a functional group;
D
21 is a targeting moiety; and
M
4 is OH, NH 2 , or SH. 66 WO 2008/034124 PCT/US2007/078600 The attachment of the nucleophilic compound such as Formula (11b) to the PEG or other polymer can be done using standard chemical synthetic techniques well known to those of ordinary skill. The activated polymer portion such as SC-PEG, PEG-amine, PEG acids, etc. can be obtained from either commercial sources or synthesized by the artisan without undue 5 experimentation. Attachment of nucleophilic compound such as Formula (11b) 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-i -alkyl-pyridinium halides, (Mukaiyama reagents), 1-(3 10 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, 15 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 0 C up to about 22 0 C (room temperature). Some particular embodiments prepared by the methods described herein include: 20 H. METHODS OF TREATMENT In alternative aspects of the invention, there are also provided methods of treating a mammal, comprising administering an effective amount of a pharmaceutical composition containing a compound of the present invention of Formula (I) to a patient in need thereof. 25 In one preferred aspect of the invention, there are also provided methods of treating a patient having a malignancy or cancer, comprising administering an effective amount of a pharmaceutical composition containing the compound of Formula (I) to a patient in need thereof In some aspects, the cancer being treated can be one or more of the following: solid tumors, lymphomas, small cell lung cancer, acute lymphocytic leukemia (ALL), pancreatic 30 cancer, glioblastoma, ovarian cancer, gastric cancers, etc. The compositions are useful for 67 WO 2008/034124 PCT/US2007/078600 treating neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mammals. Another aspect of the present invention provides methods of treatment for various medical conditions in mammals. Briefly stated, any biologically active moiety which can be 5 attached to the PEG polymer can be administered to a mammal in need of such treatment. Any oligonucleotide, etc. which has therapeutic effects in the unconjugated state can be used in its conjugated form, made as described herein. The amount of the composition that is administered will depend upon the parent molecule included therein. Generally, the amount of prodrug used in the treatment methods is 10 that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various prodrug compounds will vary somewhat depending upon the parent compound, rate of in vivo hydrolysis, molecular weight of the polymer, 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 15 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 20 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 25 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. In a further aspect of the invention, there are provided methods of administering polynucleotides (oligonucleotides), preferably antisense oligonucleotides to mammalian cells. The methods include delivering an effective amount of a conjugate prepared as described 30 herein to the condition being treated will depend upon the polynucleotides efficacy for such conditions. For example, if the unconjugated oligonucleotides (for example antisense BCL2 68 WO 2008/034124 PCT/US2007/078600 oligonucleotides, antisense Survivin oligonucleotides) has efficacy against certain cancer or neoplastic cells, the method would include delivering a polymer conjugate containing the oligonucleotides to the cells having susceptibility to the native oligonucleotides. The delivery can be made in vivo as part of a suitable pharmaceutical composition or directly to the cells in 5 an ex vivo environment. In one particular treatment, the polymeric conjugates including oligonucleotides (SEQ ID NO- 1, SEQ ID NOs: 2 and 3, and SEQ ID NO: 4) can be used. EXAMPLES The following examples serve to provide further appreciation of the invention but are 10 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 Figs. 1. Abbreviations are used throughout the examples such as, DCC (dicyclohexylcarbodiimide), DCM (dichloromethane), DCU (Dicyclohexylurea), DIBA (diisopropylethylamine), DIPC (diisopropylcarbodiimide), DMAP (4-dimethylaminopyridine), DMF (NN'-dimethylformamide), DSC (disuccinimidyl 15 carbonate), EDC (1 -(3 -dimethylaminopropyl)- 3 -ethyl carbodiimide), EEDQ (2-ethoxy-1 ethoxycarbonyl-1,2-dihydroquinoline), IPA (isopropanol), NHS (N-hydroxysuccinimide), NMM (N-methylmorpholine), PEG (polyethylene glycol), SCA-SH (single-chain antibody), SN3 8 (7-ethyl-i 0-hydroxycamptothecin), TBDPS (tert-butyl-dipropylsilyl), and TEA (triethylamine). 20 General Procedures. All reactions are run under an atmosphere of dry nitrogen or argon. Commercial reagents are used without further purification. All PEG compounds are dried in vacuo or by azeotropic distillation from toluene prior to use. 'H NMR spectra were obtained at 300 MHz and 1 3 C NMR spectra at 75.46 MHz using a Varian Mercury300 NMR 25 spectrometer and deuterated chloroform as the solvents unless otherwise specified. Chemical shifts (6) are reported in parts per million (ppm) downfield from tetramethylsilane (TMS). HPLC Method. The reaction mixtures and the purity of intermediates and final products are monitored by a Beckman Coulter System Gold® HPLC instrument. It employs a ZORBAX* 30 300SB C8 reversed phase column (150 x 4.6 mm) or a Phenomenex Jupiter® 300A C18 69 WO 2008/034124 PCT/US2007/078600 reversed phase column (150 x 4.6 mm) with a 168 Diode Array UV Detector, using a gradient of 10-90 % of acetonitrile in 0.05 % trifluoroacetic acid (TFA) at a flow rate of 1 mL/min.) Example 1. N-(Methoxycarbonyl)maleimide (compound 1). 5 Methylchloroformate (4.4 mL, 56.7 mmol, 1 eq) was added to a solution of maleimide (5.5 g, 56.7 mmol, 1 eq) and NMM (6.2 mL, 56.7 mmol, 1 eq) in 200 mL of EtOAc at 0 'C. The suspension was stirred at 0 'C for 30 minutes, filtered and washed with EtOAc. Filtrate and washings were combined and washed with cold water and dried over anhydrous Na 2
SO
4 . After filtration and evaporation under vacuum a pink solid was obtained. Purification by 10 column chromatography on silica gel (Hexane-EtOAc, 1:1, v/v) provided the product (4.8 g, 55%). Example 2. N-Maleoyl-a-(Boc)-L-lysine (compound 2). N-(Methoxycarbonyl)maleimide (315 mg, 2.03 mmol, 1 eq) was added to a solution of Boc 15 L-lysine (500 mg, 2.03 mmol, 1 eq) in 10 mL of saturated aqueous NaHCO 3 at 0 'C. The mixture was stirred vigorously at 0 'C for 40 minutes and at room temperature for an additional hour. After cooling to 0 'C, the solution was acidified to pH 3.0 with concentrated
H
2
SO
4 before extracting with ethyl acetate. The organic layers were combined and dried over anhydrous MgSO 4 . After removing the solvent in vacuo, the residue was purified by column 20 chromatography on silica gel using a mixture of CHCI 3 -MeOH (5:1, v/v) to provide the product (458 mg, 69%): 1 H NMR 6 1.39-1.87 (6 H, m, (CH 2
)
3 ), 1.44 (9H, s, Boc), 3.52 (2H, t, J = 7.2 Hz, NCH 2 ), 4.25-4.30 (1H, m, CH), 5.15 (11H, d, NH), 6.70 (2H, s, maleimide). Example 3. N-Maleoyl-ct-L-lysine (compound 3). 25 A solution of N"-maleoyl-a-(Boc)-L-lysine (172 mg, 0.53 mmol) in 5 mL anhydrous DCM was treated with 10 mL of 2N HC1 in ethyl ether for an hour at room temperature before addition of 10 mL of anhydrous ethyl ether. The resulting solid was filtered to yield 82 mg (59%) of the HCl salt of N-Maleoyl-a-L-lysine: 11 NMR (CD 3 0D) 6 1.36-1.54 (2 H, in,
CH
2 ), 1.65 (2 H, q, J = 7.2 Hz, CH 2 ), 1.83-2.02 (2 H, m, CH 2 ), 1.44 (911, s, Boc), 3.53 (2H, t, 30 J 6.9 Hz, NCH 2 ), 3.95 (1H, t, 6.2 Hz, CH), 6.82 (211, s, maleimide); 1 3 C NMR (CD 3 0D) 6 23.17, 29.03, 31.01, 37.96, 135.26, 172.34, 175.20. 70 WO 2008/034124 PCT/US2007/078600 Example 4. 5KmPEG-amide-Lys(N"-maleoyl)-COOH (compound 4). To a solution of SEmPEGCOONHS (805 mg, 0.16 mmol, 1 eq) in 8 mL of anhydrous DCM was added Ncimaleoyl-a-L-lysine (3, 82 mg, 0.31 i mmol, 2 eq) in 2 mL of DMF followed by 5 DIEA (109 pL, 0.62 mmol, 4 eq). The reaction mixture was stirred at room temperature overnight, filtered and evaporated under vacuum. The residue was first precipitated with DCM/ether and then, recrystallized with CH 3 CN/IPA. 1 3 C NMR (CDCl 3 ): 5 22.53, 28.02, 31.69, 37.32, 51.18, 58.86, 70.11-71.73 (PEG), 133.84, 169.51, 170.38, 172.41. 10 Example 5. 5S mPEG-amide-Lys(N-maleoyl)-NHS (compound 5). 5 KmPEG-Lys(Ne-maleoyl)-COOH (4) and NHS in DCM at 0 'C are treated with DCC. The mixture is stirred at room temperature overnight. The solvent is evaporated under vacuum and the residue is precipitated with DCM/ether and then, recrystallized with CH3CN/TPA. 15 Example 6. sKmPEG-carbamate-Lys(N-maleoyl)-COOH (compound 6). To a solution of sKmPEGCOONHS (5 g, 0.98 mmol, 1 eq) in 45 mL of anhydrous DCM was added N 8 -maleoyl-a-L-lysine (3, 514 mg, 1.95 mmol, 2 eq) in 5 mL of DMF followed by DIEA (0.7 mL, 3.92 mmol, 4 eq). The reaction mixture was stirred at room temperature overnight, filtered and evaporated under vacuum. The residue was first precipitated with 20 DCM/ethyl ether and then, recrystallized with CH 3 CN/IPA to give compound 6: "C NMR 5 22.30, 28.01, 31.89, 37.29, 53.26, 58.89, 64.06, 69.31-71.77 (PEG), 133.87, 155.60, 170.42, 172.82. Example 7. 5 KmPEG-carbamate-Lys(N"-maleoyl)-NHS (compound 7). 25 To a solution of 5mPEG-carbamate-Lys(N'-maleoyl)-COOH (6, 1 g, 0.19 mmol, 1 eq) and NHS (88 mg, 0.76 mmol, 4 eq) in 10 mL of DCM at 0 'C was added DIPC (118 pL, 0:76 mmol, 4 eq). The mixture was stirred at room temperature overnight. The solvents were evaporated under vacuum and the residue was precipitated with DCM/ether and then, recrystallized with CH 3 CN/IPA to give compound 7: "C NMR 5 22.31, 25.90, 28.25, 32.39, 30 37.44, 52.42, 59.30, 64.92, 69.64-72.18 (PEG), 134.25, 155.59, 167.75, 168.55, 170.91. 71 WO 2008/034124 PCT/US2007/078600 Example 8. Compound 8. To a solution of 5'extGS (5 mg, 0.85 ptmol) in PBS buffer (2.5 mL, pH 7.8) was added compound 7 (92 mg, 17 pmol, 20 eq) and the mixture was stirred at room temperature for 2 5 hours. The reaction mixture was diluted to 10 mL with water, filtered and loaded on a Poros HQ, strong anion exchange column (10 mm x 1.5 mm, bed volume ~ 16 mL) which was pre equilibrated with 20 mM Tris-HCl buffer, pH 7.0 (buffer A). The column was washed with 3 4 column volumes of buffer A to remove the excess PEG linker. Then the product was eluted with a gradient of 0 to 100 % 1 M NaCl in 20 mM Tris-HCi buffer, pH 7.0, buffer B in 10 10 minutes, followed by 100 % buffer B for 10 minutes at a flow rate of 10 mL/min. The cluted product was desalted using HiPrep desalting column (50 mL) and lyophilized to give compound 8 (1 mg oligo equivalent, 40% yield). Example 9. Compound 9. 15 To a solution of compound 8 (1 mg oligo equivalent) in PBS buffer (1 mL, pH 7.0) was added peptide cRGDfC (1 mg, 1.7 pmol) and the mixture was stirred at room temperature for 2 hours. The reaction mixture was diluted to 20 mL with water and loaded on a Poros HQ, strong anion exchange column (10 mm x 1.5 mm, bed volume ~ 16 mL) which was pre equilibrated with 20 mM Tris-HC1 buffer, pH 7.0 (buffer A). The column was washed with 3 20 4 column volumes of buffer A to remove the excess PEG linker. Then the product was eluted with a gradient of 0 tolOO % 1 M NaCI in 20 mM Tris-HCl buffer, pH 7.0, buffer B in 10 minutes, followed by 100 % buffer B for 10 minutes at a flow rate of 10 mL/min. The eluted product was desalted using HiPrep desalting column (50 mL) and lyophilized to give compound 9 (0.78 mg oligo equivalent, 78% Yield). 25 Example 10. Compound 10. To a solution of p-amino-benzylalcohol (1 g, 8.12 mmol, 1 eq) in 80 mL of anhydrous THF were added DIEA (1.4 mL, 8.12 mmol, I eq) and Boc 2 O (1.9 mL, 8.12 mmol, 1 eq). The mixture was heated to reflux overnight, then cooled down and evaporated under vacuum. The 30 residue was dissolved in EtOAc. The organic layer was washed with a 0.1 N HCl solution, dried over MgSO 4 , filtered and evaporated under vacuum. The crude product was purified by 72 WO 2008/034124 PCT/US2007/078600 column chromatography on silica gel (Hex-EtOAc, 1:1, v/v) to give 1.85 g of product (quantitative yield): 'H NMR 8 0.1.49 (911, s), 2.17 (1H, s), 4.53 (21H, s), 6.83 (111, s), 7.19 (2H, d, J = 8.5 Hz), 7.28 (2H, d, J 8.2 Hz); 13 C NMR 6 28.28, 64.54, 80.37, 118.49, 127.59, 135.31, 137.46, 152.72. 5 Example 11. Compound 11. To a -20 'C solution of compound 10 (220 mg, 0.98 mmol, 1 eq) and DIEA (188 pL, 1.08 mmol, 1.1 eq) in 8 mL of anhydrous DCM was added dropwise a solution of MsCl (84 pL, 1.08 mmol, 1.1 eq) in 2 mL of anhydrous DCM. The mixture was allowed to warm to rt and 10 then stirred for 1 hour. The reaction mixture was washed with a saturated NaHCO 3 solution, a saturated NH 4 C solution and brine, dried over MgS04, filtered and evaporated under vacuum. The crude compound was used in the next step without further purification. Example 12. Compound 12. 15 To compound 11 (0.98 mmol, 2 eq) dissolved in 15 mL of anhydrous DMF were added SN38 (192 mg, 0.49 mmol, I eq) and K 2
CO
3 (68 mg, 0.49 mmol, 1 eq). The reaction mixture was wanted to 60 'C using a pre-heated oil bath. After 2 h, HPLC showed complete disappearance of SN38 and formation of major product. The reaction mixture was evaporated under vacuum and the residue redissolved in EtOAc and washed with water, saturated NH 4 C1 20 and brine, dried over MgSO 4 , filtered and evaporated under vacuum. The crude product was purified by column chromatography on silica gel (CH 2 C1 2 -EtOAc, 1:1, v/v) to give 140 mg of product (48% yield): 'H NMR (DMSO-d) 8 0.88 (3H, t, J = 7.0 Hz), 1.27 (31, t, J= 7.3 Hz), 1.47 (9H, s), 1.85-1.88 (2H, m), 3.15-3.17 (211, in), 5.25 (211, s), 5.26 (211, s), 5.42 (2H, s), 6.49 (111, s), 7.26 (1H, s), 7.41-7.56 (6H, m), 8.05 (111, d, J = 9.1 Hz), 9.39 (1H, s); 1 3 C NMR 25 (DMSO-d) 5 7.65, 13.31, 22.09, 27.96, 30.11, 49.31, 65.05, 69.47, 72.17, 78.84, 95.74, 103.39, 117.71, 117.92, 122.35, 127.44, 127.99, 128.43, 129.59, 131.07, 139.07, 143.46, 144.06, 145.92, 149.21, 149.69, 152.37, 156.46, 156.82, 172.11. Example 13. Compound 13. 30 A 15% (v/v) TFA in DCM solution was added to compound 12 (535 mg) and the mixture was stirred at room temperature for 1h or until HPLC showed complete disappearance of the 73 WO 2008/034124 PCT/US2007/078600 starting material. Ethyl ether (200 mL) was added and the resulting solid was filtered and washed with more ether (307 mg, 58% yield). Example 14. Compound 14. 5 Compound 13 (48 mg, 0.1 mmol, I eq) and 15 (36 mg, 0.1 mmol, 1 eq) were treated with EEDQ (48 mg, 0.2 mmol, 2 eq). The mixture was stirred in the dark at room temperature overnight. The solvents were removed under vacuum and the resulting solid residue was triturated with 10 mL of ethyl ether. The solid was filtered and purified by column chromatography on silica gel (CHCl 3 :MeOH, 15:1 to 9.1) to give the desired product (8 mg, 10 9% yield): 13 C NMR (DMSO-d) a 7.8, 13.49, 18.11, 18.17, 18.58, 19.13, 19.25, 22.25, 26.51, 26.72, 28.19, 29.59, 30.26, 30.41, 30.50, 49.49, 51.67, 51.80, 52.90, 55.98, 59.26, 59.67, 63.03, 65.21, 69.54, 72.33, 77.91, 78.01, 95.92, 103.63, 118.10, 118.95, 122.52, 127.63, 128.21, 128.58, 131.17, 131.27, 138.53, 143.69, 144.25, 146.10, 149.43, 149.86, 155.15, 155.26, 156.63, 156.97, 158.48, 158.66, 170.39, 171.15, 171.33, 172.15, 172.29. 15 Example 15. Boc-Val-Cit-OH (Compound 15). Boc-Val-NHS (10 g, 31.81 mmol, 1 eq) in 80 mL of DME was added to a solution of L citrulline (5.85 g, 33.40 mmol, 1.05 eq) in 20 mL of THF and NaHCO 3 (2.8g, 33.40 mrnol, 1.05 eq) in 80 mL of water. The mixture was stirred at room temperature overnight. Aqueous 20 citric acid (200 mL of a 15% solution in water) was added and the mixture was extracted with 10% IPA/EtOAc (2 x 200 mL). The suspension was washed with water (2 x 200 mL) and the solvent was evaporated under vacuum. The resulting solid was triturated with 200 mL of ethyl ether and filtered to yield the product (6.08 g, 51% yield): 1 H NMR (CD 3 0D) a 0.92 (3H, d, J = 6.7 Hz), 0.97 (3H, d, J = 6.7 Hz), 1.44 (9H,>s), 1.51-2.06 (5H, m), 3.12 (2H, t, J = 6.7 Hz), 25 3.90 (1H, d, J - 7.0 Hz), 4.37-4.41 (111, in). ' 3 C NMR (CD 3 0D): 6 18.58, 19.83, 27.58, 28.75, 30.03, 32.09, 40.48, 53.33, 61.38, 80.49, 157.75, 162.05, 174.30, 174.69. Example 16. Compound 16 (Boc-Val-Cit-PAB). Boc-Val-Cit (1 g. 2.67 mmol, leg) and p-aminobenzyl alcohol (362 mg, 2.94 mmol, 1.1 eq) in 30 2:1 DCM/MeOH (26 mL/13 mL) were treated with EEDQ (1.32 g, 5.34 mmol, 2 eq). The mixture was stirred in the dark at room temperature overnight. The solvents were removed 74 WO 2008/034124 PCT/US2007/078600 under vacuum and the resulting solid residue was triturated with 10 mL of ethyl ether. The solid was collected by filtration and washed with ethyl ether to yield the product (900 mg, 70%): 11 NMR (CD 3 0D) 5 0.93 (3H, d, J= 7.0 Hz), 0.97 (3H, d, J = 7.0 Hz), 1.44 (9H, s), 1.49-2.06 (5H, in), 3.07-3.29 (2H, in), 3.91 (1H, d, J= 6.7 Hz), 4.50-4.55 (3H, m), 7.29 (2H, 5 d, J = 8.5 Hz), 7.54 (211, d, J = 8.5 Hz), 8.22 (11H, d, J = 7.3 Hz); "C NMR (CD 3 OD) 6 18.64, 19.85, 27.84, 28.75, 30.61, 31.92, 40.28, 54.90, 54.99, 61.73,64.78, 80.62, 121.09, 128.44, 138.47, 138.55, 158.01, 162.11, 172.00, 174.42, 174.52. Example 17. Compound 14 (Boc-Val-Cit-PABE-SN38). 10 To a suspension of Boc-Val-Cit-PAB (214 mg, 0.45 mmol, 1 eq) in 3 mL of CH 3 CN was added DIBA (0.23 mL, 1.35 mmol, 3 eq) followed by dropwise addition of a solution of MsC1 (0.1 mL, 1.35 mmol, 3 eq) in I mL CH 3 CN. After 1 hour, TLC showed no starting material left (CHCl 3 -MeOH, 5:1, v/v). The reaction mixture was evaporated under vacuum and the resulting residue was dissolved in EtOAc. The organic phase was washed with saturated 15 NH 4 Cl, saturated NaHCO 3 and brine; dried over MgSO 4 , filtered and evaporated under vacuum. To the crude residue dissolved in 5 mL of DMF were added SN38 (88 mg, 0.22 mmol, 0.5 eq) and K 2 C0 3 (31 mg, 0.22 eq, 0.5 eq) and the mixture was heated at 60 'C for 3 hours. The product formation was monitored and conformed to product prepared in Example 14 by HPLC. 20 Example 18. Compound 17. Compound 14 in DCM is treated with 2N HCl in ether. After completion of the reaction additional ether is added and the resulting solid is filtered and washed with ether to give Val Cit-PABE-SN38. To a solution of compound 5 in anhydrous DCM is added HCl-Val-Cit 25 PABE-SN38 in anhydrous DMF followed by DIEA. The mixture is stirred at room temperature overnight. The solvent is removed under vacuum and the resulting solid is precipitated with DCM/ether and recrystallized with CH 3 CN/IlPA to give the product. Example 19. Compound 18. 30 To a solution of compound 17 (1 eq) in a mixture of DCM/DMF is added RGDC (2 eq). The reaction mixture is stirred at room temperature overnight and then the solvent is evaporated 75 WO 2008/034124 PCT/US2007/078600 under vacuum. The residue is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product. Example 20. Compound 19. 5 To a solution of 20k4armPEGSC (7 g, 0.35 mmol, I eq) in 60 mL of anhydrous DCM was added Lys(Boc)-OH (690 mg, 2.8 mmol, 2 eq) in 15 mL of DMF followed by DIEA (1 mL, 5.6 mmol, 4 eq). The reaction mixture was stirred at room temperature overnight, filtered and evaporate under vacuum. The residue was first precipitated with DCM/ethyl ether and recrystallized with CH 3 CN/IPA to give compound 19: 1 3 C NMR 8 21.88, 27.97, 29.04, 31.57, 10 39.60, 44.98, 53.00, 63.44, 68.86-70.37 (PEG), 78.04, 155.26, 172.89. Example 21. Compound 20. To a solution of 4arm-PEG-Lys(Boc)-OH in 25 mL of anhydrous DCM was added a 4N HCI solution in dioxane (25 mL). The reaction was stirred at room temperature for 4 hours and 15 then was evaporated under vacuum. The residue was precipitated by addition of ethyl ether. The solid was filtered and washed with ether to give 4arm-PEG-Lys-OH (1.77 g): 'C NMR S 21.97, 26.54, 31.22, 39.63, 45.11, 53.16, 63.66, 69.88-70.51 (PEG), 78.04, 155.59, 172.78. Example 22. Compound 21. 20 A solution of BocCys(NPys)-OH (300 mg, 0.80 mmol, 1 eq) and NHS (97 mg, 0.84 mmol, 1.05 eq) in 10 mL of anhydrous DCM at 0 'C was treated with DCC (173 mg, 0.84 mmol, 1.05 eq). The mixture was allowed to warm to room temperature and stirred overnight. The solid DCU byproduct was filtered off: To the filtrate was added compound 20 (1.7 g, 0.082 mmol, 1 eq) followed by DIEA (114 pL, 0.66 mmol, 8 eq). The reaction mixture was stirred 25 at room temperature overnight and evaporated under vacuum. The residue was first precipitated with DCM/ether and then, recrystallized with CH 3 CN/IPA to give the compound 21 (1.5 g): "C NMR 6 22.32, 25.52, 28.27, 28.83, 31.90, 39.03, 42.45, 45.38, 53.41, 53.58, 63.91, 64.37, 69.30-72.38 (PEG), 80.11, 120.98, 133.94, 142.54, 153.34, 155.51, 155.71, 157.28, 169.98, 173.32. 30 76 WO 2008/034124 PCT/US2007/078600 Example 23. Compound 22. To a solution of compound 21 (1.5 g, 0.07 mmol, 1 eq) and NHS (125 mg, 1.09 mmol, 16 eq) in 15 mL of DCM at 0 'C was added DIPC (168 pL, 1.09 mmol, 16 eq). The mixture was stirred at room temperature overnight. The solvents were evaporated under vacuum and the 5 residue was precipitated with DCMlether and then, recrystallized with CH 3 CN/IPA to give the product (1.3 g): "C NMR 6 22.05, 25.76, 25.93, 28.68, 28.92, 32.01, 38.89, 42.49, 45.79, 52.40, 53.97, 64.78, 69.57-71.77 (PEG), 80.47, 121.32, 134.29, 142.93, 153.75, 155.71, 151.60, 168.18, 169.14, 169.71, 170.56. 10 Example 24. Compound 23. (BocPheLys(Fmoc)-OH). To a solution of BocPheOSu (1.5g, 4.14mmol, lea) in DCM (10.3 mL) and DMF (10.3 mL) was added H-Lys(Fmoc)-OH (1.84g, 4.55 mmol, 1.1eq) followed by DIEA (2.9 mL, 16.56 mmol, 4eq). The resulting reaction mixture was stirred overnight and diluted with ethyl acetate. The solution was washed with water (30 mL x 2) and by brine (30 15 mL x 2). The organic layers were combined, dried over MgSO4 and concentrated in vacuo to yield a yellow residue. The crude residue was then chromatographed over silica gel using CHC13-MeOH (3:1, v/v) to give the product. MS. [M+1]4 616. Example 25. Compound 24. 20 Compound 13 (111 mg, 0.22 mmol, 1 eq) and 16 (208 mg, 0.34 mmol, 1.5 eq) were treated with EEDQ (167 mg, 0.67 mmol, 3 eq). The mixture was stirred in the dark at room temperature overnight. The solvents were removed under vacuum and the resulting solid residue was triturated with 10 mL of ethyl ether. The solid was filtered and purified by column chromatography on silica gel (CH 2 Cl 2 -EtOAc, 1:1 to 1:2, v/v) to give the desired 25 product (20 mg, 8% yield). MS. [M+1]* 1095. Example 26. Compound 25. Compound 24 (8 eq) is treated with 4N HClI in dioxane. The reaction is stirred at room temperature for 4 hours. Ethyl ether is added and the solid is filtered. This solid is dissolved in 30 DMF and added to a solution of compound 22 (leq) in DCM. To the mixture is added DIEA (16 eq) and the reaction is stirred at room temperature overnight. The solvent is removed 77 WO 2008/034124 PCT/US2007/078600 under vacuum and the resulting solid is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product. Example 27. Compound 26. 5 To a solution of compound 25 in DCM is added piperidine. The reaction mixture is stirred for 4 hours. The solvent is removed under vacuum and the resulting solid is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product. Example 28. Compound 27. 10 To a solution of compound 26 (1eq) in a mixture of DCM/DMF is added RGDC (2 eq). The reaction mixture is stirred at room temperature overnight and then the solvent is evaporated under vacuum. The residue is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product. 15 Example 29. FmocVal-Cit-OH (Compound 28). Fmoc-Val-NHS (2.5 g, 5.73 mmol, 1 eq) in 15 mL of DME was added to a solution of L citrulline (1.05 g, 6.01 mmol, 1.05 eq) in 8 mL of THF and NaHCO 3 (505 mg, 6.01 mmol, 1.05 eq) in 15 mL of water. The mixture was stirred at room temperature overnight. Aqueous citric acid (75 mL of a 15% solution in water) was added and the mixture was extracted with 20 10% IPA/EtOAc (2 x 100 mL). The organic layers were washed with water (3 x 100 mL) and the solvent was evaporated under vacuum. The resulting solid was triturated with 100 mL of ethyl ether and filtered to yield the product (1.98 g, 70% yield): 1 H NMR (DMSO-d) 6 0.86 (3H, d, J = 6.7 Hz), 0.90 (3H, d, J = 7.0 Hz), 1.40-1.48 (2H, in), 1.51-1.75 (2H, in), 1.98 (IH, sext, J = 6.8 Hz), 2.95 (2H, q, J = 6.2 Hz), 3.93 (1H, dd, J = 7.3, 8.8 Hz), 4.14-4.29 (4 H, m), 25 5.40 (2H, brs), 5.96 (1H, t, J = 5.6 Hz), 7.32 (211, t, J = 7.5 Hz), 7.39-7.44 (3H, in), 7.75 (2H, t, J= 6.3 Hz), 7.89 (2H, d, J = 7.3 Hz), 8.19 (1H, d, J= 7.3 Hz); 13 C NMR (DMSO-d) 6 18.31, 19.25, 26.75, 28.40, 30.59, 46.68, 51.91, 59.81, 64.92, 65.65, 119.98, 125.30, 126.94, 127.52, 140.55, 143.61, 143.76, 155.88, 158.58, 171.12, 173.25. 30 78 WO 2008/034124 PCT/US2007/078600 Example 30. FmocVal-Cit-PA-B (Compound 29). A solution of compound 28 (1 g, 2.01 mmol, leq) and p-aminobenzyl alcohol (273 mg, 2.21 mmol, 1.1 eq) in a mixture of DCM/MeOH (20 mL/1 0 mL) was treated with EEDQ (996 mg, 4.03 mmol, 2 eq). The mixture was stirred in the dark at room temperature overnight- The 5 solvents were removed under vacuum and the resulting solid residue was triturated with 10 mL of ethyl ether. The solid was collected by filtration and washed with ethyl ether to yield the product (925 mg, 76% yield): 1 H NMR (DMSO-d) 5 0.87 (6H, d, t = 7.5 Hz), 1.36-1.47 (2H, mn), 1.51-1.75 (2H, m), 1.99 (1H, sext, J = 6.7 Hz), 2.90-3.04 (3H, m), 3.93 (1H, dd, J = 7.3, 8.8 Hz), 4.23-4.38 (4 H, m), 4.43 (2H, d, J= 5.3 Hz), 5.13 (11H, t, J= 5.6 Hz), 5.43 (2H, 10 brs), 5.99 (lH, t, J = 5.6 Hz), 7.23 (2H, d, J 8.5 Hz), 7.32 (2H, t, J= 7.3 Hz), 7.39-7.48 (3H, in), 7.54 (2H, d, J =8.5 Hz), 7.74 (2H, t, J= 6.7 Hz), 7.89 (2H, d, J = 7.6 Hz), 8.12 (1H, d, J= 7.3 Hz); 13 C NMR (DMSO-d) 6 18.34, 19.28, 26.84, 29.56, 30.47, 46.67, 53.05, 60.05, 62.55, 65.65, 118.71, 119.98, 125.23, 126.79, 126.94, 127.51, 137.27, 137.36, 140.55, 143.60, 143.73, 155.93, 158.69, 170.17, 171.06. 15 Example 31. Val-Cit-PAB (Compound 30).. To a solution of compound 29 (622 mg, 1.03 mmol) in 10 mL of anhydrous DMF was added piperidine (2 mL). The mixture was stirred at room temperature overnight and then the solvents were evaporated under vacuum. The residue was triturated with DCM and the 20 resulting solid was filtered and washed with DCM to give the product (283 mg, 72% yield): H NMR (DMSO-d) 8 0.79 (311, d, J = 6.7 Hz), 0.89 (3H, d, J = 6.7 Hz), 1.38-1.42 (2H, in), 1.52-1.68 (2H, m), 1.94 (1H, sext, J= 6.3 Hz), 2.89-3.01 (2H, in), 3.05 (1H, d, J= 4.7 Hz), 4.46 (211, s), 5.09 (1H, brs), 5.40 (211, s), 5.98 (111, t, J= 5.3 Hz), 7.23 (2H, d, J= 8.2 Hz), 7.54 (211, d, J= 8.5 Hz), 8.14 (1H, brs), 10.03 (1H, s); 13 C NMR (DMSO-d) 6 16.80, 19.33, 25 26.53, 29.95, 31.10, 52.31, 59.33, 62.35, 118.61, 126.59, 137.06, 137.14, 158.44, 170.06, 173.69. Example 32. Compound 31. To a solution of compound 22 (1.3 g, 0.06 mmol, 1 eq) in 15 mL of anhydrous DCM and 3 30 mL anhydrous DMF was added compound 30 (175 mg, 0.46 mmol, 8 eq). The reaction mixture was stirred overnight at room temperature and then the solvent was evaporated under 79 WO 2008/034124 PCT/US2007/078600 vacuum. The residue was first precipitated with DCM/ether and then, recrystallized with
CH
3 CN/IPA to give the product (1.1 g): 1 3 C NMR (CDCl3/CD 3 0D) 6 18.48, 19.46, 22.85, 26.74, 28.54, 28.94, 29.50, 30.47, 31.81, 39.21, 42.04, 45.79, 53.51, 54.06, 55.32, 59.67, 64.32, 65.04, 69.54-70.99 (PEG), 80.70, 120.23, 121.44, 127.64, 134.31, 137.16, 142.94, 5 153.86, 155.86, 156.85, 160.44, 170.45, 171.02, 171.87, 173.19. Example 33. Compound 32. To a solution compound 31 (1 eq) in a mixture of DCM/DMF (4/1, v/v) is added DSC (16 eq) and the mixture is cooled to 0 'C. Then, pyridine (16 eq) is added and the mixture is gradually 10 warmed to room temperature and stirred overnight. The solvent is evaporated under vacuum and the residue is precipitated with DCM/ethyl ether and then, recrystallized with
CH
3 CN/IPA to give the product. Example 34. Compound 33. 15 To a solution of compound 32 is added doxorbicin. The reaction mixture is stirred at rt overnight. The solvent is evaporated under vacuum and the residue is precipitated with DCM/ethyl ether and then, recrystallized with DMF/IPA to give the product. Example 35. Compound 34. 20 To a solution of compound 33 (leq) in a mixture of DCM/DMF is added RGDC (2 eq). The reaction mixture is stirred at room temperature overnight and then the solvent is evaporated under vacuum. The residue is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product. 25 Example 36. Compound 35. To a solution of compound 19 (6.14 g, 0.307 mmol, 1 eq) and NHS (283 mg, 2.456 mmol, 8 eq) in 31 mL of anhydrous DCM at 0 'C was added DCC (507 mg, 2.456 mmol, 2eq). The reaction mixture was stirred at room temperature overnight, filtered over a pad of celite and evaporated under vacuum. The residue was first precipitated with DCM/ethyl ether and then, 30 recrystallized with CH 3 CN/IPA to give the product (5.69 g, 86%): "C NMR 621.59,25.21, 80 WO 2008/034124 PCT/US2007/078600 28.11, 29.03, 31.42, 39.36, 45.13, 51.80, 64.07, 68.90-70.53 (PEG), 155.15, 155.58, 168.17, 168.54. Example 37. Compound 36 5 To a solution of compound 35 (5.0g, 0.232mmol, leq) in 23 mL each of anhydrous DCM and anhydrous DMF was added HC1.NH 2 Cys(NPys)-OH (0.77g, 2.78mmol, l2eq) followed by DIEA (0.65 mL, 3.71 mmol, 16 eq). The reaction mixture was stirred at room temperature overnight and evaporated under vacuum after filtering through a pad of celite. The residue was first precipitated with DCM/ether and then, recrystallized with CH 3 CN/IPA to give the 10 product (4.88 g, 95%yield): "C NMR 6 22.17, 28.22, 29.27, 31.85, 39.92, 45.22, 51.85, 54.49, 63.98, 69.06-70.62 (PEG), 78.56, 120.87, 132.88, 142.27, 153.44, 155.67, 156.47, 162.14, 170.74, 171.20. Example 38. Compound 37. 15 To a solution of compound 36 (4.38g, 0.197 mmol, I eq) in DCM (44 mL) was added 8.8 mL of TFA and the reaction mixture was allowed to stir overnight at room temperature. After 20 hours the reaction mixture was evaporated under vacuum and the residue was first precipitated with DCM/ether and then, recrystallized with CH 3 CN/IPA to give the product (2.1 g, 49%): 1 3 C NMR 6 21.47, 26.41, 31.64, 39.47, 45.25, 52.32, 54.11, 63.75, 69.12-70.65 20 (PEG), 120.85, 133.46, 142.25, 153.67, 155.64, 156.39, 171.53, 171.79. Example 39. Compound 38. To a solution of folic acid (124mg, 0.28mmol, leq) in 3.1 mL of DMSO at room temperature was added NHS (71mg, 0.616mmol, 2.2eq) followed by addition of DCC (64mg, 0.308 25 mmol, 1.1 eq) and 63 gL of triethylamine. The reaction mixture was stirred overnight in the dark and filtered over a pad of Celite* to obtain a clear yellow solution of FA-NHS which was used as is for the next coupling step with the linker. To a solution of H 2 N-Lys(4arm PEG)CysNPys (37, 381 mg, 0.0175mmol, l eq) in 4 mL each of anhydrous DCM and anhydrous DMF was added the DMSO solution of FA-NHS (prepared as shown above) 30. followed by DIEA (49 pL, 0.28 mmol, 16eq). The reaction mixture was allowed to stir 81 WO 2008/034124 PCT/US2007/078600 overnight in the dark at room temperature. It was then dialysed for 4 days with distilled water. The yellow solution was then lyophilized to obtain 197 mg of the final product. Example 40. Compound 39. 5 To a solution of C6-thio-LNA-survivin (OligoSH) in pH 6.5 phosphate buffer is added compound 38 and the solution is stirred for 1 hour at room temperature. Reaction progress is checked by anion-exchange HPLC. The reaction mixture is filtered through 0.2 micron filter and loaded on Poros anion-exchange column. The product is elated with a gradient using buffer system 20 mM Tris. HCl 2M NaCl at pH 7.0. The product is desalted and lyophilized. 10 82

Claims (29)

1. A compound of the Formula (I) D 1 -(L 1 )a-L2-(L 3 )b-D 2 A wherein: 5 R 1 is a substantially non-antigenic water-soluble polymer; A is a capping group or Dl-(L1)-L 2 -(L 3 )b-D 2 . each Di is independently selected from the group consisting of targeting moieties, functional groups and leaving groups; 10 each D 2 is independently selected from the group consisting of biologically active moieties, functional groups and leaving groups; each L 1 is an independently selected permanent linker or a releasable linker: L 2 is a mutifunctional linker; each L 3 is an independently selected permanent linker or a releasable linker: and 15 (a) and (b) are independently zero or a positive integer.
2. The compound of claim 1, wherein L 2 is selected from the group consisting of: 2 , 2 020 2 3O4 +N 0N . 0N c3-0 c4 O NH- d (a), d2 (Jb), d3 K 2 (Ic), d4 (1d), 0 c5 c6 - N 2 R2 C7 O -N es O - \ O/ (Ie), c"6 d (Ig), and 83 WO 2008/034124 PCT/US2007/078600 0 R2 O--+ c8 (1h) wherein R 2 and R' 2 are independently selected from the group consisting of hydrogen, C 1 _ 6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3 . 1 9 branched alkyl, C 3 . 8 cycloalkyl, C1.6 substituted 5 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 C1.6 heteroalkyl, C 1- 6 alkoxy, aryloxy, C 1 - heteroalkoxy, heteroaryloxy, C 2 -6 alkanoyl, arylearbonyl, C2- 6 alkoxycarbonyl, aryloxycarbonyl, C 2 _ 6 alkanoyloxy, arylearbonyloxy, C 2 - 6 substituted alkanoyl, substituted arylcarbonyl, C2- 6 substituted alkanoyloxy, substituted aryloxycarbonyl, 10 C 2 - 6 substituted alkanoyloxy and substituted arylcarbonyloxy; (cI), (6), (c3), (c4), (c5), (c6), (c'6), (c"6), (7) and (c8) are independently zero or a positive integer; and (d1), (d2), (d3), (d4), (d5) and (47) are independently zero or a positive integer. 15
3. The compound of claim 2, wherein L 2 is selected from the group consisting of: 0 0 H O O O +H - NH H H N -N - 0-N 4-N 0 0 0 0 0 -N 0 ~N0
4-N5 NH 04 1 00/0 and -N N 20 84 WO 2008/034124 PCT/US2007/078600 4. A compound of claim 2, selected from the group consisting of: L3-D2 L3-D2 R2 c1 0 F 2 c2 0 A-RgN A-Rj-N 0 d1 L 1 -D 1 (Ia), d2 L-D 1 (Ib), 05 L3-D2 L3-D2 L3-D2 2 c3 O 4O A-RgN A-R 1 -N A-R-N 0 L(-D1 d5O d3 R'2 (Ile), d4 L 1 -D 1 (Ild), L1-D (Ile), 'L3-D2 ,L3-D2 O O c6 c6 R C R2 f A-R1-N c 6 OL-D1 CO Os c"6a L1-D1 (Io), -6L1-D1 (I1g) L3-D2 R 0 02 7O2 0-L-D A-R-N A-R 1 -2N O-Lj-D1 -R- S L1-D1 5 d7 O-L (Ih) c8 (IIi) and 0 R2 O-L-D 1 A-R 1 -N S-L 3 -D 3 c8 (Ilj).
5. A compound of claim 4, selected from the group consisting of: o 0 0 H L3-D2 D2-L3 H H L3-D2 A-Rg N N-R-N L-D, D 0-L0 L-D1 0o 0 0 L3-D2 D2-L,3 L3-D2 A-R 1 -NH 0 0 NH-R 1 -NH 0 10 LTDj, DrLj LrD1, 85 WO 2008/034124 PCT/US2007/078600 o 0 0 A-RI-NH- L N H- R<NH H 3 o 0 0 o 0 0 L3-D2 2L H 3D A-R 1 -NH NR 1 -NH HN-L 1 -D 1 , D 1 -L 1 -NH -L 1 -D 1 , o 0 0 A-R-NH NH- R-NH L-D2 2L 3D 0lo 0 0i A-R 1 -N N-R-N L -L1, ,I-Li A-R 1 -NH N-R-N 04 o o4 L-D , D -iL-j ALRD2N D 2 -L 3 0A--R1-NH 0' 0 0 A-RI-NH , L3-Li D-L ' lHRNH ,L -D2 0 L-Li-D L8D26 5lD L1-D1,j-D A-- & u( 0 - HR-NH O'LyD20 olLjDjD-L 0 L -D O8O6 WO 2008/034124 PCT/US2007/078600 0 0 R2 O---D2 R2 0--D1 A-R 1 -N A-R 1 -N S-L1-D1, S-Ly-D3, o 0 D 2 -L 3 -0 R2 R2 0-L-D2 N--R 1 -N D1-L1-S S-LrD1, and ~ o 0 D1-L1-0 R2 R2 0-LrD N-R 1 --N D 3 -L 3 -S S-L 3 -D 3 . 5
6. The compound of claim 1, wherein the biologically active moiety is selected from the group consisting of -NH 2 containing moieties, -OH containing moieties and -SH containing moieties.
7. The compound of claim 1, wherein the biologically active moiety is selected from the 10 group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides.
8. The compound of claim 7, wherein the pharmaceutically active compounds are selected from the group consisting of DNA topoisomerase inhibitors, microtubule inhibiting 15 drugs, DNA damaging agents, antimetabolites, nucleoside analogs and anticancer drugs.
9. The compound of claim 1, wherein the biologically active moiety is an oligonucleotide.
10. The compound of claim 9, wherein the oligonucleotide is selected from the group 20 consisting of antisense oligonucleotides, locked nucleic acids (LNA), short interfering RNA (siRNA), microRNA (miRNA), aptamers, peptide nucleic acid (PNA), and phosphorodiamidate morpholino oligonucleotides (PMO).
11. The compound of claim 9, wherein the oligonucleotide is selected from the group 25 consisting of antisens bcl-2 oligonucleotides, antisense HITF-I a oligonucleotides, and antisense Survivin oligonucleotides. 87 WO 2008/034124 PCT/US2007/078600
12. The compound of claim 1, wherein the targeting agent is selected from the group consisting of monoclonal antibodies, single chain antibodies, cell adhesion peptides, cell penetrating peptides, receptor ligands, targeting carbohydrate molecules or lectins and 5 oligonucleotide.
13. The compound of claim 1, wherein the targeting agent is selected from the group consisting of RGD peptide, selectin, TAT, penetratin, (Arg) 9 and folic acid. 10
14. The compound of claim 1, wherein the releasable linker is selected from the group consisting of benzyl elimination-based linkers, trialkyl lock-based linkers, bicine-based linkers, acid-labile linkers, lysosomally cleavable peptides and capthepsin B cleavable peptides.
15 15. The compound of claim 1, wherein the releasable linkers are independently selected from the group consisting of: --- L{1 -- -Y12-Ar- -c--Yl R32 all _ _ b11 - -c11 t{ ll -{ ifil R33 R35 16 C----C- -C 0 q34 R36 Ar gl hl R37 WO 2008/034124 PCT/US2007/078600 Y17 R38 1 1 - t M L13 110-- C -(CR44R 4 5 ), 1 --k1 N - C - - - ( )3 1 42 R40 A 1 -(Z')x .1--(Ll4)ql - -C- L1-- O )- C (CR 4 6 R 4 7 ) m1 p il 0 LR4-3 -S-- R50 N-N 5 -Val-Cit-, -Gly-Phe-Leu-Gly-, -Ala-Leu-Ala-Leu -Phe-Lys-, 0 11 H -QVa1-Cit-C 0 11 H 10 -Phe-Lys-C-N \/ HN - -Val-Cit- OA 0 0 HN - -Phe-Lys o 0, -Val-Cit-C(=O)-CH2OCH2-C(=0)-, -Val-Cit-C(=O)-CH 2 SCH 2 -C(=O)-, and 89 WO 2008/034124 PCT/US2007/078600 -NHCH(CH 3 )-C(=0)-NH(CH 2 ) 6 -C(CH3)2-C(=0)-, wherein Y,-19 are independently 0, S or NR4; R31AS, R 50 , R 1 00 and A 51 are independently selected from the group consisting of 5 hydrogen, C1- 6 alkyls, C3-12 branched alkyls, C 3 - 8 cycloalkyls, Ci- 6 substituted alkyls, C 3 -8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, CI- 6 heteroalkyls, substituted C 1 . 6 heteroalkyls, C 1 . 6 alkoxy, C 16 - alkyloxycarbonyl, phenoxy and C 1 _ 6 heteroalkoxy; Ar is an aryl or heteroaryl moiety; Lii-15 are independently selected bifunctional spacers; 10 Z and Z' are independently selected from selected from the group consisting of moieties actively transported into a target cell, hydrophobic moieties, bifunctional linking moieties and combinations thereof; (ci 1), (hl 1), (k 11), (111), (mll) and (n 11) are independently selected positive integers; (al l), (eli), (g11), (j 11), (o1l) and (qi 1) are independently either zero or a positive 15 integer; and (b11), (x11), (x'12), (fl1), (il l) and (p11) are independently zero or one.
16. The compound of claim 1, wherein the permanent linkers are independently selected from the group consisting of: 0 0 0 o t N H H 20 k /, 0 ,0,C H 0 0 H0 N H V\-N~xs 0 0 O o 0 0 0 0 N NN 0 0 , 0 ,and 0
17. The compound of claim 1, wherein L 1 and L 3 are independently selected from the 25 group consisting of an amino acid, an amino acid derivative, and a peptide. 90 WO 2008/034124 PCT/US2007/078600
18. The compound of claim 1, wherein R 1 comprises a linear, terminally branched or multi-armed polyalkylene oxide. 5
19. The compound of claim 18, wherein the polyalkylene oxide is selected from the group consisting of polyethylene glycol and polypropylene glycol.
20. The compound of claim 18, wherein the polyalkylene oxide is selected from the group consisting of 10 -Y 21 -(CH 2 CH 2 0)X-CH 2 CH 2 Y 2 1 -Y 2 1-(CH 2 CH 2 O).-CH 2 C(=Y22)-Y21 -Y2rC(=Y22)-(CH2)a2-Y23-(CH2CH2O)T-CH2CH2-Y23-(CH2)a2-C( Y22)-Y2r and -Y 2 r(CR51Rs2)a2-Y23-(CH2)b2-0-(CH2CH2O).-(CH2)b2-Y23-(CR51Rs2)a2-Y2r , wherein: 15 Y 2 1 and Y 23 are independently 0, S, SO, S02, NR 53 or a bond; Y 2 2 is 0, S, or NR53; R 51 . 53 are independently selected from the group for R 2 ; (a2) and (b2) are independently zero or a positive integer; and (n) is an integer from about 10 to about 2300. 20
21. The compound of claim 18 wherein the polyalkylene oxide is a polyethylene glycol of the formula, -O-(CH 2 CH 2 O)T wherein (n) is an integer from about 10 to about 2,300. 25
22. The compound of claim 1, wherein R has an average molecular weight from about 2,000 to about 100,000 daltons.
23. The compound of claim 1, wherein Ri has an average molecular weight of from about 5,000 to about 60,000 daltons. 30 91 WO 2008/034124 PCT/US2007/078600
24. The compound of claim 1, wherein R 1 has an average molecular weight from about 5,000 to about 25,000 daltons or from about 20,000 to about 45,000 daltons.
25. A compound of claim 1, selected from the group consisting of: 0 11 H HN ,Val--Cit-C-D2 mPEG N -0 HN00 H N o H 50 o 0 D2C CitH-ValIH HN 2 H H mPE N O C E -- 00 N N 0 PEG 0 O O Cit Val NH HNtV l -Cit H 2 H 2 m PEG N 0a 0 NN O O NN o H D o o 0 </, C-Cit-Val N HN~ \ /-Ctc- D D2 NHH H 0 00 N -^E/,rN 0 _t -N 0 0 N N D- H 0 0 HD 92 WO 2008/034124 PCT/US2007/078600 0 H NVal--Cit-0-N D2 mPEG 0 00 o H O oO 0I 0 D2 -- /] N----Cit-Val HVa-Cit-C- 2 H H \ N PEG N O D1H 0 0 Ho > o00 II HN 2Val-CitC-D2 mPEG 0 0 0N 0 0 0 D2-C-Cit-ValNH HN Val-Cit-C-D2 Ht H P NE P E G N DI N 0 0 t N D o 0 0 O O D -Cit-,Val-NH Ntal Ci H 0N 2 mPEG 0 0 0 ,0 N D 5 0 o 0 C -Cit-Val HN -ct \ / D2 NHH H D2 0 0 N PEG o _ 0 Dj-N 0 0 N D 0 C 93 WO 2008/034124 PCT/US2007/078600 0 HN Val-Cit--NH O H mPEG N 0 0 N D 0 0 0 11I It D2 NH-C-Cit-ValNHH HN -Val-Cit--NH 2 NH H 0 H 0 N PEG N 0 0 D1 N 0 0 N D 0 0 0 II HNVal-Cit-C-D2 HN mPEG 0 O 0 N N 0 H D 0 0 0 D 2 -C-Cit-Val'NH HN ,Val-Cit-C-D2 H H ON PEG N 0 0 0 N0 ~ N N N N D1 H O H O D O0 0 11 H Val-Cit-C-D2 HN mPEG 0 H 0 N-r i 5 0 0 0 D 2 -C-Cit-Va' HN-Val-Cit-C-D2 *NH HHN H N PEG 0 H D -rN 0 0 :o-,N--~ 0 0 94 WO 2008/034124 PCT/US2007/078600 0 11 HH ,Val-Cit-C-D2 HNH N J HR 51 ' N' R 52 mPEG 0 H N o N D, 0 0 0 D 2 -C-Cit-ValN HVal-Cit-C-D 2 R 52 N .R 5 1 NP N R 5 1' R 52 H HN Val--Cit- -' D2 N 0 51'N R52 HmPEG H N N N SN D O O O H-Ct-ValNH HN ValCit- D D2 R52 NR5 1H P HR51'NR522 NPE 0 Hj, E D- r N O O N S D 0 0 O Vl I - D2 H HNX B 2 0NN R 51 N'R52 O mPEHPEH O N 0SN D1 5 0 0 0 D2 /- NH---Cit--ValNH HN Val-Cit--NH O 0R52, -RH PEGN o H R51'NR52 O D1-0 N S.NS 0 0 95 WO 2008/034124 PCT/US2007/078600 0 11 Phl H HN Phe-Lys-C-D 2 mPEG H o N S- D I 0 0 0 D 2 -C-Lys-Phe Phe-L NH H H HN N R 2 R52 N'R 51 N N R 51 'N H PEG H D1-S N 0 0 N S'D1 o 0 0 H HN e-Lys-C D2 mPEG N HR51N'R52 o ND 0 o 0 I I I I C--Lys-Phe Phe--L D2N H Hjt 2N -y- R52'N'R51H N PEG NHR51 N'R52 D1- N 0 0 N 'D1 o 0 0 Phe-Lys-C-NH--, / D2 H H O N R51'N'R52 mPEG O H o N S'D1 5 0 and 0 0 D2 NH-C-Lys-PheNH HNP y N D *HH H R 52 . R 51 O P R51 N R52C 0 - H H - -- E S N 0 0 N S'D1 o 0 wherein mPEG has the formula: CH 3 -O(CH 2 CH 2 O)a-; 10 PEG has the formula -O(CH 2 CH 2 0).-; 96 WO 2008/034124 PCT/US2007/078600 (n) is a positive integer from about 10 to about 2,300; R 51 and R 52 are independently selected from the group consisting of hydrogen, C 1 - alkyls, -C 3 - 1 2 branched alkyls, C 3 - 8 cycloalkyls, C 1 - substituted alkyls, C 3 - 8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C 1 - 6 heteroalkyls, substituted Ci- heteroalkyls, 5 CI. 6 alkoxy, C 1 - alkyloxycarbonyl, aryloxycarbonyl, phenoxy and C 1 - 6 heteroalkoxy; Di is a targeting moiety, a functional group or a leaving group; and D 2 is a biologically active moiety, a functional group or a leaving group. 25. A compound of claim 1, selected from the group consisting of: 0 HN- Val-Cit-C- SN38 H mPEG yN 0 0 O N N O H 10 0 0 [Ili SN38-C-Cit-Val HN -Val-Cit-C- SN38 H H 00 Nf--PEG -- N 00 ON 0 P H 0 OH] o0 0 .0 II HN Val-Cit-C-SN38 H mPEG N 0 N N 0 H 0 o 0 SN38-C-Cit-Val H H Val-Cit-C-SN38 OO N PG N O 00 0 N N tN N O ON H o 0 H 0 97 WO 2008/034124 PCT/US2007/078600 0 H HN _Val-Cit-CS3 H SN38 mPEG N 0 0 O o 0 /-Cit-ValNH HNVal-Cit SN38 H H SN38 ON PG NO 0 PEG 0 0N 0 0 N N N P N O H o 0 O II I HN H C tC-NS3 H- H0H0-< mPEG 0 SN38 C / CCit-ValI N Va -Cit-C- N 3 t NH N mPO PE 0 -_0 0 N 00 HO O 0 0 H 1 mPE 0 N0 r P G 5N 0 0t 0 0 98I WO 2008/034124 PCT/US2007/078600 0 H SN38 mPEG 0 0 0O 0 0 0 z -- Cit-Val'N HNVal-Cit--S3 SN38 NH H HN SN38 0 N PEG N 0 fN 0 0 N oO 0 0 I I HN /Val-Cit--NH O SN38 H 0K mPEG 0 0 0O 0 o 0 - II .1 HNi GPEG 0 0 / N 00 N? *0 0 0 HHN ,VlCtC SN38 50 NO2, SN38-C-Cit-Val'NH HN'-Val-Cit-C-SN38 S' c PEG NHB sN SN0N NO2 O O NO2, 99 WO 2008/034124 PCT/US2007/078600 0 11 HNXVal-Cit H SN38 mPG N It NHBoc N mPEG N O N S O NO 2 O 0 SN38 Cit-Val HN V-/ SN38 NHBoc _! N PEG N o NHBoc H H S -S - J N 0 0 N SN NO 2 O 0 NO 2 0 H HN Val-Cit-C-NH ,\ / SN38 mPEG N ONHBoc N SNU O NH- -- Cit-Val NH HHHN ,Val-Cit-C-NHi() SN38 O N 0 N P NO2 N O NO2 O N H HNPhe-Lys-C mPEG N P N NHBoc H HN 0 S1 N-)- NO 0 0 NO 2 0 10 H 3-CN sPheL-H H 5N38HPh-y-C S3 mPEG 0 Ho NHloc NE : Ho O- N- S0 0 Nl S 0100 WO 2008/034124 PCT/US2007/078600 0 HN e-Lys HjH SN38 mPEG N 0 NHBoc N O N SN O NO 2 0 0 \ -Lys-hH-he Phe-Lys-C SN38 H H H SN38 N NHBoc O N PEG N H NHBoc N IsH "H S s N o 0 N S NO 2 0 0 NO 2 , 0 HNPhe-Lys--NH- SN38 mPEG N NHBoc 0 N -, SNS) O NO 2 0 0 SN38 O /] NH-C--Lys- Phe HN Phe-Lys-C-NH / SN38 /Ha -NH H H NHNo N 0 NHBoc N PEG N H N0 0 N T~s NO 2 O O NO 2 0 11 HN Val-Cit-C-SN38 Ht mPEG N 0 5 0 NH 2 0 0 SN38-C-Cit- Val-- HNVCit-C-SN38 NH HN N PEG 2N H 2 N-- 0 0NH H 0 O N O-LNA H HN mPEG N O 0 NH 2 101 WO 2008/034124 PCT/US2007/078600 0 H H 0 LNA-ON NO 0-LNA NH HHHN H H O PEG 0 H 2 N 0 0 NH 2 0 II HN- Val-Cit-C-SN38 H mPEG 0 O 00 0 N O N o H S-SCA o 0 SN38-C-Cit-ValKNH H H HN3Val-Cit-C-SN38 0 N PEG N 0 N N O O NO SCA-S H 0 0 H S-SCA O 0 0 II HN-Val-Cit-C-SN38 HHN H mPEG 0 O 0 O N N o H S-SCA 0 H0 o 0 SN38-C-Cit-Val NH HN -Val-Cit-C-SN38 H H N N o 0 N> N N NN N SCA-S H a a H S-SCA 5 0 0 0 HN-Val-Ci H SN38 N mPEG 0 o H S-SCA 102 WO 2008/034124 PCT/US2007/078600 o 0 /-Cit-Val -Val-CitSN SN38 H H O N PEG N 0 0 0 NN N SCA-S H 0 0 H S-SCA H HNVal-Cit-C- SN3 mPEG N0 ONN 0 H S-SCA o 0 0 SN38-- N- - i -aNH H H HN Va Cit- SN38 O N PEG -- N O . II A /aI - C t--N 0 0/---- - - HN HVal-Cit-C-SN38 H m P E G C0 0 0 00 o 0 SN38-C--Cit-Va-NH HNVal-Cit-C-SN38 5 O O0 0 H N _ValI- C it -01'l \ H SN38 mPEG N 0 0 N S-SCA 0 103 WO 2008/034124 PCT/US2007/078600 0 0 _-Cit-Val-NH HVal-Cit SN38 SN38 H H C 0 N PEG N 0 o SCA-S N 0 0 N S-SCA 0 0 0 HN Val-Cit--NH SN38 HN N0 mPEG - ' 0 0 N S-SCA 0 0 0 \N38NH-C-Cit-ValN H HN Val -Cit- SN38 SN38 H HV-H H-iC-S3 0 PEG N0H. 0 O0N Noi acN SCA-S- N 0 0 N S-SCA 0 0 0 S C Ci V al - C it- CVSN 3a NH HHHH mPEG H P 0 N r Folic acid 0 0 0 II II SN3-C-Cit -Val -Cit-C-SN3 NH H H SN3 H PEGN H Folio acid N 0 0 N Folio acid 5 0 0 0 10 HHNX V-i--SN38 mPEG N 0 H NH 2 0 N- S SSSCA 0 104 WO 2008/034124 PCT/US2007/078600 o Q SN38--C-Cit-Val Val-Cit-C-SN38 HN HN SN3 NH 2 N4 H N o NH 2 SCA-S- N 0 0 N SS-SCA o 0 0 H HNVal-Cit--NHSNSN3 3 mPEG N O H NH 2 0 N SSSCA 0 0 0 S/ C -Cit-Val, Vl Ct SN38 NH H H HN SN38 NH 2 N PEG N NH 2 H -yH SCA-S "-' N 0 0 N S- S-SCA o 0 0 II H SN38 N Jt mPEG N NH 2 o N FlSi SCA 5 0 100 SN38-C H-C - 'NH-W HN~ / SN38 H Ht SCA-S'S, N, 0 0 N %( SSSCA 0 0 a 11 __H HN~ ,Pe-Lys-C-SN38 mPEG 0 ~t H 0 N -, Folio acid 0 105 WO 2008/034124 PCT/US2007/078600 0 0 SN38-C-Lys-Phe HPhe-Lys-C-SN38 NH H FN H 0 PEG H . . Folio acid qIN 0 0 N Folic acid 0 0 0 II H HN Phe-Lys-C--SN38 H N mPEG N H NH 2 0 N ( S-SCA 0 o 0 SN38- C Lys-Phe HN Phe--Lys-C-SN38 NHH HN NH 2 N PEG N H NH 2 SCA-S-S N 0 0 N S'S-SCA 0 0 0 H .Phe-Lyst 0 \N3 H HNSN38 mPEG N 0 H NH 2 o N S S-SCA o o 0 \ C-Lys-PhePh-sC \/ SN38 NH H H SN38 NH 2 H N PEG N H NH 2 SCA-S- 3 N 0 0 N S'S-SCA 5 0 0 0 II H HNPhe-Lys-C-NH / SN3 NNH0 mPEG H NH2 0 N S S-SCA 0 106 WO 2008/034124 PCT/US2007/078600 0 0 NH--Lys-Phe HPhe-Lys-C-NH SN38 H H N 0 NH 2 H N PEG N 0 NH 2 O SCA-S'S N 0 N S-SCA 0 0 0 11 HN- Val-Cit-C-SN38 H I N mPEG 0 O 0 o H S-RGD o 0 SN38-C-Cit-ValNH HN Val-Ct-C-SN38 H H 0O N P EG N O O N N O N RGD-S H 0 0 H S-RGD 0 11 HN -Val-Cit-C-SN38 H mPEG 0 0 N N H S-RGD 0 H0 0 SN38-C-Cit-Val HVal-Cit-C-SN38 H H 0N PEG N 0 o 0 N N N N RGD-S H o o H S-RGD 5 0 0 HN Val-Cit H SN38 N mPEG 0 O 0 N N o H S-RGD 107 WO 2008/034124 PCT/US2007/078600 0 -- Cit-ValNH HN Val-Cit- SN38 SN38 H H O 0 N PEG N 0 0 N O -- N O O O S R G RGD-SH 0 0 H S-RGD 0 -Va-Cit-C-N \N3 HHN H -O - SN38 mPEG N 0 O O H O S-RGD 00 \ /NC---it-Va VaI-Ct-C-N \/ SN38N V H NHH HN H SN38 0 N N 0 00 0 PE 0 0 0 N__ RGD-S H - 0 0 H S-RGD oO 0 II HN Val-Cit-C-SN38 H mPEG 00 0 S-RGD 0 o 0 SN38--Cit-ValNH HN Val-Cit-C -SN38 H H 00 N PEG-_qN oO0 RGD-S N 0 0 N S-RGD 5 0 0 0 OI HN Val-Cit- SN38 mPEG 0 0 0 l N S-RGD 0 108 WO 2008/034124 PCT/US2007/078600 O 0 11 iii C-Cit-ValNH HN Val-Cit- SN38 SN38 H H 00 N PEG N RGD-S N 0 0 N S-RGD 0 0 0 HHVal-Cit- -NH SN3 HN N N3 mPEG 0 00 0 N S-RGD 0 - 0 0 \ ,NH-C-Cit-VaI HNVal-i\ NC/ SN38 NH- - NH H H HN CSN38 0 0 0 N PEG N S 0 RGD-S- N 0 0 N S-RGD 0 0 0 II H HN Val-Ct-C-SN38 PEG N O H 0 N NH-RGD 0 0 0 SN38-C-Cit-ValNH HN Val-Cit-C-SN38 H N PEG--- H RGD-NH- N 0 0 N NH-RGD 5 0 0 0 II H HN Val-Cit-C-SN38 mPEG N H NH 2 0 N s'S-RGD 0 109 WO 2008/034124 PCT/US2007/078600 0 0 SN38-C-Cit-ValNH HN Val-Cit-C-SN38 N N NH 2 H PEG H NH 2 RGD-S- N 0 0 N SS-RGD 0 0 0 HN Val-Cit H SN38 mPEG N H NH 2 0 N S S-RGD 0 o 0 S C-Cit-ValNH HN Val-Cit-c \ N SN8H H S3 NH 2 H N PEG N H NH 2 RGD-S s N 0 0 N S SRGD o 0 0 Val-Cit--H SN38 N mPEG N H NH 2 0 o N S S-RGD o o 0 SN38 \ NH-C--Cit--ValNH H H HN'-Val-Cit-C-NH SN38 NH2 H N~ PEG N H NH 2 RGD-S-S N 0 0 N S'S-RGD 5 00 0 II H HN y mPEG N H 0 N NH-RGD 0 110 WO 2008/034124 PCT/US2007/078600 0 0 II II SN38- -Lys-PheNH HN Phe-Lys-C-SN38 RGD-NH 0 PEG N NH-RGD o 0 0 II H HN Phe-Lys-C--SN38 mPEG N NH 2 0 N S-RGD 0 0 0 I II SN38-C--Lys-PheNH HN Phe-Lys-C-SN38 H H __H Hjt -Ls--S3 NH 2 H PEG 0 H NH 2 RGD-S-S N 0 0 N "SS-RGD 0 0 0 HPhe-Lys-C H HN SN38 mPEG N H NH 2 o N &SS-RGD o 0 0 -- y - h IIhe SN3 / -- y-N H H HN Phe-Lys- SN38 NH 2 N PEG N H NH 2 RGD-S S N 0 0 N S-RGD 5 0 0 II H Phe-Lys-C-NH \N/ H HNSN38 mPEG N NH 2 0 0 N SN-RGD 0 WO 2008/034124 PCT/US2007/078600 o 0 S8NH--Lys-PheNH HPhe-Lys -- NH S \~~~1 / H---ys-'eHNSN3B SN38- 'NH H H H N NH2 H N PEG N 0 H NH 2 RGD-S-' S N,! O 0 N ,j' S S-RGD 0 H 0 o N O-LNA HN H mPEG N O N Folic Acid 0 H H 0 LNA-O NH HNO N 0-LNA Foi Aci PEG HN Folic Acid 0 O H O N 0-LNA H HNH NN mPEG 0O H o N 0 Folic Acid HH OOan LNA-O N H H0N N-LNA HPE 0 NH_ EG' Folic Acid PEG N Folic Acid 5 0 wherein S-SCA is a single-chain antibody; RGD is 112 WO 2008/034124 PCT/US2007/078600 HS I-N 0; a NH H HNO 0 N NH 2 COOH LNA is locked nucleic acids; Folic acid is a residue of OH N H 2 N N HO N N- -- / - NO N HNI OH 5 mPEG has the formula: CH 3 -O(CH 2 CH 2 O).-; PEG has the formula -O(CH 2 CH 2 0)n-; and (n) is a positive integer from about 10 to about 2,300.
26. A method of preparing a polymeric conjugate containing a multifunctional linker 10 comprising: reacting a compound of Formula (IlIa): M-(L1)a-L 2 ~M 2 A 21 (I1a) with a biologically active moiety-containing compound having Formula (11b): M 3 -(L 3 )b-D 22 15 under conditions sufficient to form a compound of Formula (IIc): Ml-(Ll)a-L 2 -(L3)b-D 2 2 A 22 (IIc) wherein 113 WO 2008/034124 PCT/US2007/078600 R 1 is a substantially non-antigenic water-soluble polymer; A 21 is a capping group or M-(LI)a-L 2 -M 2 A22 is a capping group or 5 MI-(L)aL 2 -(L3)b-D22 M, is a functional group; D 22 is a biologically active moiety; M 2 is OH or a leaving group; M 3 is OH, NH 2 , or SH; 10 L 1 is a permanent linker or a releasable linker; L 2 is a multifunctional tinker; L 3 is a permanent linker or a releasable linker; and (a) and (b) are independently zero or a positive integer. 15
27. The method of claim 26 further comprising: reacting the compound of Formula (IIc): M 1 (L 1 )a-L 2 -(L3)b-D22 A 22 (II1c) with a nucleophilic moiety-containing moiety having Formula (Id) D21-M4 (IId) 20 under conditions sufficient to form a compound of Formula (IIe): D 21 ~(L 1 )a-L2-(L3)b-D22 R, A 23 (IIle) wherein: A 23 is a capping group or 114 WO 2008/034124 PCT/US2007/078600 D 2 1-(Ll)a-L 2 -(L3)b-D 2 2 M 1 is a functional group; D 2 1 is a targeting moiety; M 4 is OH, NH 2 , or SH; and 5 all other variables are the same as defined in claim 27.
28. A method of treating a mammal, comprising administering an effective amount of a compound of Formula (I) to a patient in need thereof. 10
29. A method of administering polynucleotides to mammalian cells, comprising delivering an effective amount of a compound of Formula (I) to a cell requiring such treatment. 115
AU2007296056A 2006-09-15 2007-09-15 Targeted polymeric prodrugs containing multifunctional linkers Expired - Fee Related AU2007296056B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US84494306P 2006-09-15 2006-09-15
US60/844,943 2006-09-15
PCT/US2007/078600 WO2008034124A2 (en) 2006-09-15 2007-09-15 Targeted polymeric prodrugs containing multifunctional linkers

Publications (2)

Publication Number Publication Date
AU2007296056A1 true AU2007296056A1 (en) 2008-03-20
AU2007296056B2 AU2007296056B2 (en) 2012-09-13

Family

ID=39184644

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007296056A Expired - Fee Related AU2007296056B2 (en) 2006-09-15 2007-09-15 Targeted polymeric prodrugs containing multifunctional linkers

Country Status (11)

Country Link
EP (1) EP2073820A4 (en)
JP (1) JP2010503708A (en)
KR (1) KR20090057383A (en)
CN (1) CN101541332A (en)
AU (1) AU2007296056B2 (en)
BR (1) BRPI0716812A2 (en)
CA (1) CA2662981A1 (en)
IL (1) IL197517A0 (en)
MX (1) MX2009002855A (en)
RU (1) RU2009114154A (en)
WO (1) WO2008034124A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11739166B2 (en) 2020-07-02 2023-08-29 Davol Inc. Reactive polysaccharide-based hemostatic agent

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2147116T3 (en) 2007-04-20 2016-08-31 Leadiant Biosciences Ltd Stable recombinant adenosine deaminase
CN102131509B (en) 2008-05-22 2015-01-07 特拉维夫大学拉莫特有限公司 Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US8586019B2 (en) 2008-05-22 2013-11-19 Ramot At Tel-Aviv University Ltd. Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
JP2011520983A (en) * 2008-05-23 2011-07-21 エンゾン ファーマシューティカルズ,インコーポレーテッド Polymer systems containing intracellular releasable disulfide linkers for oligonucleotide delivery
TWI388570B (en) 2008-07-23 2013-03-11 Hanmi Science Co Ltd A polypeptide complex comprising non-peptidyl polymer having three functional ends
US20110305770A1 (en) * 2008-11-17 2011-12-15 Enzon Pharmaceuticals, Inc. Releasable polymeric lipids for nucleic acids delivery system
TW201029668A (en) * 2008-11-17 2010-08-16 Enzon Pharmaceuticals Inc Releasable conjugates for nucleic acids delivery systems
US20130030359A1 (en) * 2010-01-22 2013-01-31 Ascendis Pharma A/S Dipeptide-based prodrug linkers for aromatic amine-containing drugs
CN102869254A (en) * 2010-04-16 2013-01-09 安龙制药公司 Polymeric conjugates of adenine nucleoside analogs
US20130096097A1 (en) 2010-06-11 2013-04-18 Japan Science And Technology Agency Pharmaceutical multimeric particles, and manufacturing method for same
WO2012075087A2 (en) * 2010-11-30 2012-06-07 Board Of Trustees Of The University Of Illinois Silica nanoparticle agent conjugates
CN103045720B (en) * 2011-10-17 2014-08-20 格诺思博生物科技(上海)有限公司 Targeting molecule for detecting pathogenic cell and application thereof
CN104244988A (en) 2012-03-05 2014-12-24 雷蒙特亚特特拉维夫大学有限公司 Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
CN102746316B (en) * 2012-06-26 2015-04-15 济南精合医药科技有限公司 An m-nitroarylmethoxy camptothecin anoxic activation prodrug for antitumor drugs
CN102731518B (en) * 2012-06-26 2014-12-03 济南精合医药科技有限公司 O-nitro aryl methoxycamptothecine anoxic activated prodrug used for antitumor drug
ES2759905T3 (en) * 2013-10-04 2020-05-12 Prolynx Llc SN-38 Slow Release Conjugates
NZ758049A (en) 2013-10-15 2024-03-22 Seagen Inc Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
GB201414098D0 (en) * 2014-08-08 2014-09-24 Illumina Cambridge Ltd Modified nucleotide linkers
WO2016059377A1 (en) 2014-10-14 2016-04-21 Polytherics Limited Process for the conjugation of a peptide or protein with a reagent comprising a leaving group including a portion of peg
CN107073131B (en) * 2014-10-24 2021-05-25 宝力泰锐克斯有限公司 Conjugates and conjugation reagents
JP6570034B2 (en) * 2014-11-26 2019-09-04 日本化薬株式会社 Novel glutamic acid derivatives and uses thereof
CN105641708A (en) * 2014-12-04 2016-06-08 上海中医药大学附属普陀医院 Polypeptide modified poly(oligo(ethylene glycol) methacrylate-co-bufalin) nano preparation, and preparation method thereof
CA2987322C (en) 2015-08-11 2020-02-11 Coherent Biopharma Multi-ligand drug conjugates and uses thereof
JP6412906B2 (en) 2015-11-03 2018-10-24 財團法人工業技術研究院Industrial Technology Research Institute Compound, linker-drug and ligand-drug complex
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
AU2016363013B2 (en) 2015-12-04 2022-03-10 Seagen Inc. Conjugates of quaternized tubulysin compounds
KR20180134351A (en) 2016-03-25 2018-12-18 시애틀 지네틱스, 인크. Process for the preparation of pegylated drug-linkers and intermediates thereof
CN107375288B (en) 2016-05-16 2019-08-23 博瑞生物医药(苏州)股份有限公司 The polymerization target anticancer conjugate of multi-arm
CN106265683A (en) * 2016-09-14 2017-01-04 江南大学 A kind of preparation method with the pathogenic bacterium biomembrane new inhibitor wearing film targeting characteristic
US11135307B2 (en) 2016-11-23 2021-10-05 Mersana Therapeutics, Inc. Peptide-containing linkers for antibody-drug conjugates
JP2020512312A (en) 2017-03-24 2020-04-23 シアトル ジェネティックス, インコーポレイテッド Process for the preparation of glucuronide drug-linker and its intermediates
JP6947909B2 (en) * 2017-04-21 2021-10-13 博瑞生物医薬(蘇州)股▲分▼有限公司Brightgene Bio−Medical Technology Co., Ltd. Multi-arm targeted anti-cancer conjugate
JP7231147B2 (en) * 2017-06-29 2023-03-01 国立大学法人東海国立大学機構 RNA introduction reagent and its use
US11612666B2 (en) * 2017-11-30 2023-03-28 Seagen Inc. Process for the preparation of drug linker compounds
EP3737420A2 (en) 2018-01-08 2020-11-18 Regeneron Pharmaceuticals, Inc. Steroids and antibody-conjugates thereof
US20200360545A1 (en) 2018-01-12 2020-11-19 Prolynx Llc Protocol for minimizing toxicity of combination dosages and imaging agent for verification
KR102141124B1 (en) 2018-01-30 2020-08-04 (주)바이오니아 Double-stranded Oligonucleotide Complex comprising Double― stranded miRNA and Uses thereof
CN110339367A (en) * 2018-04-02 2019-10-18 北京键凯科技股份有限公司 A kind of cell-penetrating peptide-multi-arm polyethylene glycol-drug conjugates and its application with targeting
KR20210084546A (en) * 2018-10-29 2021-07-07 메르사나 테라퓨틱스, 인코포레이티드 Cysteine Engineered Antibody-Drug Conjugates with Peptide-Containing Linkers
SG11202105135QA (en) * 2018-12-17 2021-07-29 Remegen Co A linker for antibody-drug conjugates and its use
CN113995847A (en) * 2020-07-28 2022-02-01 重庆阿普格雷生物科技有限公司 Polyethylene glycol coupled drug, preparation method and application thereof
EP4190360A1 (en) * 2020-07-28 2023-06-07 Chongqing Upgra Biotechnology Co., Ltd. Polyethylene glycol conjugate drug synergist, preparation method therefor, and use thereof
WO2022161452A1 (en) * 2021-01-28 2022-08-04 南京桦冠生物技术有限公司 Conjugate and use thereof
WO2023138682A1 (en) * 2022-01-24 2023-07-27 北京桦冠生物技术有限公司 Conjugate and use thereof

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2626654B2 (en) * 1990-03-31 1997-07-02 科学技術振興事業団 Targeting high molecular weight pharmaceutical compounds and intermediates thereof
US5643575A (en) * 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US7011812B1 (en) * 1996-05-03 2006-03-14 Immunomedics, Inc. Targeted combination immunotherapy of cancer and infectious diseases
US6331289B1 (en) * 1996-10-28 2001-12-18 Nycomed Imaging As Targeted diagnostic/therapeutic agents having more than one different vectors
WO1998049198A1 (en) * 1997-04-30 1998-11-05 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US5965119A (en) * 1997-12-30 1999-10-12 Enzon, Inc. Trialkyl-lock-facilitated polymeric prodrugs of amino-containing bioactive agents
US7060479B2 (en) * 1999-12-08 2006-06-13 Serono Genetics Institute, S.A. Full-length human cDNAs encoding potentially secreted proteins
US6153655A (en) * 1998-04-17 2000-11-28 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
US6251382B1 (en) * 1998-04-17 2001-06-26 Enzon, Inc. Biodegradable high molecular weight polymeric linkers and their conjugates
US6214330B1 (en) * 1998-07-13 2001-04-10 Enzon, Inc. Coumarin and related aromatic-based polymeric prodrugs
US6777387B2 (en) * 2000-03-31 2004-08-17 Enzon Pharmaceuticals, Inc. Terminally-branched polymeric linkers containing extension moieties and polymeric conjugates containing the same
MXPA03007392A (en) * 2001-02-20 2003-12-04 Enzon Inc Terminally-branched polymeric linkers and polymeric conjugates containing the same.
US7091186B2 (en) * 2001-09-24 2006-08-15 Seattle Genetics, Inc. p-Amidobenzylethers in drug delivery agents
CN100475269C (en) * 2002-03-05 2009-04-08 北京键凯科技有限公司 Binding agent of hydrophilic polymer-glutamic acid oligopeptide and medicinal molecular, composition containing said binding agent and use thereof
ITMI20020951A1 (en) * 2002-05-06 2003-11-06 Univ Degli Studi Trieste MULTIFUNCTIONAL DERIVATIVES OF POLYETHYLENGLICLE THEIR PREPARATION AND USE
US7122189B2 (en) * 2002-08-13 2006-10-17 Enzon, Inc. Releasable polymeric conjugates based on aliphatic biodegradable linkers
US7595304B2 (en) * 2003-04-13 2009-09-29 Enzon Pharmaceuticals, Inc. Polymeric oligonucleotide prodrugs
US20040228831A1 (en) * 2003-05-15 2004-11-18 Belinka Benjamin A. Polymeric conjugates for tissue activated drug delivery
GB0411186D0 (en) * 2004-05-19 2004-06-23 Celltech R&D Ltd Biological products
US7365127B2 (en) * 2005-02-04 2008-04-29 Enzon Pharmaceuticals, Inc. Process for the preparation of polymer conjugates
WO2006088248A1 (en) * 2005-02-18 2006-08-24 Nof Corporation Polyoxyalkylene derivative

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11739166B2 (en) 2020-07-02 2023-08-29 Davol Inc. Reactive polysaccharide-based hemostatic agent

Also Published As

Publication number Publication date
AU2007296056B2 (en) 2012-09-13
JP2010503708A (en) 2010-02-04
CN101541332A (en) 2009-09-23
IL197517A0 (en) 2009-12-24
CA2662981A1 (en) 2008-03-20
MX2009002855A (en) 2009-03-30
BRPI0716812A2 (en) 2013-11-05
KR20090057383A (en) 2009-06-05
EP2073820A2 (en) 2009-07-01
WO2008034124A2 (en) 2008-03-20
EP2073820A4 (en) 2014-07-16
WO2008034124A3 (en) 2008-08-07
RU2009114154A (en) 2010-10-20

Similar Documents

Publication Publication Date Title
AU2007296056B2 (en) Targeted polymeric prodrugs containing multifunctional linkers
US8367065B2 (en) Targeted polymeric prodrugs containing multifunctional linkers
US20090202573A1 (en) Polymeric conjugates containing positively-charged moieties
AU2007296054B2 (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery
US20090203706A1 (en) Lysine-based polymeric linkers
KR20100051722A (en) Polymeric linkers containing pyridyl disulfide moieties
CA2723263A1 (en) Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
WO2009009712A1 (en) Polymeric drug delivery system containing a multi-substituted aromatic moiety
US8268318B2 (en) Polyalkylene oxides having hindered ester-based biodegradable linkers
AU2005243417A2 (en) Camptothecins conjugated in position 7 to cyclic peptides as cytostatic agents
US8110559B2 (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery

Legal Events

Date Code Title Description
MK25 Application lapsed reg. 22.2i(2) - failure to pay acceptance fee