CA2662981A1 - Targeted polymeric prodrugs containing multifunctional linkers - Google Patents

Targeted polymeric prodrugs containing multifunctional linkers Download PDF

Info

Publication number
CA2662981A1
CA2662981A1 CA002662981A CA2662981A CA2662981A1 CA 2662981 A1 CA2662981 A1 CA 2662981A1 CA 002662981 A CA002662981 A CA 002662981A CA 2662981 A CA2662981 A CA 2662981A CA 2662981 A1 CA2662981 A1 CA 2662981A1
Authority
CA
Canada
Prior art keywords
compound
group
substituted
cit
peg
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.)
Abandoned
Application number
CA002662981A
Other languages
French (fr)
Inventor
Hong Zhao
Maria Belen Rubio
Prasanna Reddy
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.)
Belrose Pharma Inc
Original Assignee
Enzon Pharmaceuticals, Inc.
Hong Zhao
Maria Belen Rubio
Prasanna Reddy
Belrose Pharma 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., Hong Zhao, Maria Belen Rubio, Prasanna Reddy, Belrose Pharma Inc. filed Critical Enzon Pharmaceuticals, Inc.
Publication of CA2662981A1 publication Critical patent/CA2662981A1/en
Abandoned legal-status Critical Current

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

Abstract

The present invention provides single chain antibody-directed polymeric prodrugs containing multifunctional linkers. Methods of making the polymeric delivery systems and methods of treating mammals using the same are also disclosed.

Description

TARGETED POLYMERIC PRODRUGS CONTAINING
MULTIFUNCTIONAL LINKERS

CROSS-REFERENCE TO RELATED APPLICATION
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
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 reduce the toxicity and improve the efficacy of small molecular weight drugs was directed to targeting agent-directed drug delivery systems. For example, the 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 cheznotherapeutics 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 calxcheamicin cytotoxin, for 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 conjugates have to be stable in buffers or plasma and not prematurely release the toxin during circulation. They must also be intern.alized 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 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.

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 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 drug delivery system having desired therapeutic activity and less 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 both targeting agent and PEGylation technologies as well as other improved therapeutic techniques. Thcrefore, in accordance with one aspect of the invention there are provided compounds of Formula (I):

D1~(L1)a-L2-(L3)b-D2 Ri A
wherein:
Rl is a substantially non-antigenic water-soluble polymer;
A is a capping group or ~~~(L,)a-L2-(L3)e-pz each D, is independently selected from among targeting moieties, fiinctional groups and leaving groups; preferably targeting moieties;
each D2 is independently selected from among biologically active moieties, functional groups and leaving groups, preferably biologically active moieties;
each L, is an independently a pezxnanent linker or a releasable linker, preferably permanent linker;
L2 is a multifunctional linker;
each L3 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 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 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.
One general aspect of the present invention, without limitation, is schematically described in the following structure:

Targeting perrnanent linker multifunctional linker releasable linker Drug Moiety C~6 wherein PEG is attached to both the SCA and the drug such as a cytotoxic agent thxough a centrally-located multifunctional linker; the linker with the SCA is permanent, while the 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.
Tn one preferred 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 thereo 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.
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.
Other and fuxther 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 linker, releasable linker, etc. that remains after it has undergone a substitution reaction with another compound.
For purposes of the present invention, the terrn "polymeric residue" or "PEG
xesidue"
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 carboxyalkenyls, aminoalkenyls, dialkenylaminos, hydroxyalkenyls and mercaptoalkenyls;
substituted alkynyls include carboxyalkynyls, ax.ninoalkynyls, dialkynylaminos, hydroxyalkynyls and mereaptoalkynyls; substituted cycloalkyls include moieties such as 4-chlorocyclohexyl; aryls include moieties such as napthyl; substituted aryls include moieties such as 3-bromo phenyl; aralkyls include moieties such as tolyl; heteroalkyls include moieties such as ethylthiophene; substituted heteroalkyls include moieties such as 3-methoxy-thiophene; alkoxy includes moieties such as methoxy; and phenoxy includes moieties such as 3-nitrophenoxy. Halo shall be understood to include fluoro, chloro, iodo and brozno_ For purposes of the present invention, "nucleic acid" or "nucleotide" shall be understood to iriclude deoxyribonucleic acid (DNA), ribonucleic acid (RNA) whether single-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 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", "cytotox.izxs".
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- C1_12 alkyls, C3_g cycloalkyls or substituted cycloalkyls, etc.;
the term "substituted" shall be understood to include adding or replacing one or more atoms contained within a functional group or coinpound 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 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;
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.

BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 schematically illustrates methods of synthesis described in Examples 1-5.

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.
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.
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
A. OVERVIEW
In one aspect of the present invention, there are provided compounds of Formula (I):
D1-(L1)a-L2-(L3)b-D2 R~
A -wherein:
R, is a substantially non-antigenic water-soluble polymer;
A is a capping group or I
D1-([-1)a-2'-(L3)b-D2 .

each D, is independently selected from among targeting moieties, functional groups and leaving groups, preferably targeting moieties;
each D2 is independently selected from among biologically active moieties, functional groups and leaving groups, preferably biologically active moieties;
each L, is an independently a permanent linker or a releasable linker, preferably permanent linker;
L2 is a multi?Functionallinker;
each L3 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 include compounds having formula:

R2 c1 O R2 c2 O
A-RI-N O A--RI-N

d~
~
LI-D1 (IIa), d2 L1-Dj (IIb), O

, 'c5 12 cs 0 p A-RI-N
A-Rj N ,a' Rj-N O
O ~
N-Ll-Dl ~d5O
d3 R'2 (IIc), d4 ~-1-D1 (IYd), L~-D1 (Iie), iL3-D2 /L3-D2 O ~
cS ~

A-R1 N c's O' A_R1-N cB O' 1 ~
c"60~L7-Dj (IIf}, c^sO~LlD1 1 (IIg) O
R2 c7 O RZ O-L3-D2 A-Rj-N A-Ri-N
0 {I~}, l S-L~-D~
O--Lj_Dj c8 (IIi) and O
Rz O--Ll -Dj A-Rj-N

c8 (I1j).
wherein R2 and R'2 are independently selected from arnong hydrogen, Q_6 alkyl, C2_6 alkenyl, C2-6 alkynyl, C3_19 branched alkyl, C3_$ cycloalkyl, CI_6 substituted alkyl, C2_6 substituted alkenyl, C2_6 substituted alkynyl, C3-$ substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1_6 heteroalkyl, substituted C1_6 heteroalkyl, CI_6 alkoxy, aryloxy, C1_6heteroalkoxy, heteroaryloxy, C2_6 alkanoyl, arylcarbonyl, C2_fi alkoxycarbonyl, aryloxycarbonyl, C2_6 alkanoyloxy, arylcarbonyloxy, C2_6 substituted alkanoyl, substituted arylcarbonyl, C2_6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2_6 substituted alkanoyloxy and substituted arylcarbonyloxy;
(c1), (c2), (c3), (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 from about 1 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 CH3 group, i..e_ mPEG while in other embodiments, bis-activated PEGs are provided such as those corresponding to the formula:

A-Rj-N N-R,_"N
L1-D, Dj-Lj Lj-Dj O O O
O O O

A-Rj-NH O O NH-Ri-NH O
Li-DI, Dl-L1 Ll-Dl, O O O

A-RI-NH NH-Rj-NH

Lj-Dj DI-LlLI-D, O , 0 0 0 0 0 A--R,-NH N-R1-NH
HN-Lj-Dj, DI-L,-NH HN-Lj-Dj?
p o 0 A-R,-NH NH-R~-NH

Li-DI} Di L, L1-D, --,\o //\-- F-10 A-Rj-N p N-Rj-N

Ll-Dl, Dl-Li Ll-Dl, O
O O
L3-D2 D2-L~ L3-D2 A-RI---N N-Rl-N
\-A O
O /~- O O
Ll-Dl} DI-Li LI-DI, L3-D2 D2-L3 ,L3-D2 A_Rj_NH O L3-D2 D2-L3.0 NH-Rl NH plL3 D2 Ql Ll DI Dj-Li O O`Ll-Dl O O ~
A- -R1_NH p,Ll-DI Dj-Lj.p NH-Ri-NH O~ p' Ll Dl O- Ll-Dl Dj-Li O Ll-Dl p 0 A-Ri-N N-R~-N
S-L, -Dj, S--Ll-Dl, p O O
R2 O-Lj-D1 Dl-L1--0 R2 R2 O-L~-D1 A----RI-N N-R~-N
S-L3-D3, and 3 L3-5 S L3 D3.
Other optional capping groups include H, NH2, OH, COZH, Cz_6 alkoxy and Cz_6 alkyl.
Preferred capping groups include methoxy and methyl.
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 are preferred and polyethylene glycols (PEG's) are most preferrcd. 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, etc.
In another aspect of the invention, the biological moieties include -NHZ
containing moieties, -OH containing moieties and -SH containing moieties.

S. MULTZFUNCTIONAL LINKERS (L2) 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 znultifiinctional linker can be a residue of an aspartic acid or a lysine.

The L2 having at least three functional site can be selected from among: R

c1 O R2 c2 O Z c3 O c4 O
-~ N O -1-N +N ~-N O
"-~ 1 ! s'~
al (Ia), d2 (Ib), d3 Rz (Ic), d4 (Id), O
o ~
c6 os O'Lll`
~-N R2 RZ c7 O
c's O ~-N
O
d5p- ~( O. 0-~-'~ (le), c,6 ~ (I~, a~ (Ig), and O
R2 0+
-~-N
SA.._ c8 (T,) wherein R2 and R'2 are independently selected from among hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkyriyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2_6 substituted alkenyl, C2_6 substituted alkynyl, C3_8 substituted cycloalkyl, aryl, substi.tuted aryl, heteroaryl, substituted heteroaryl, C1_6 heteroalkyl, substituted C1_6heteroalkyl, CI_6 alkoxy, aryloxy, Cz_6heteroalkoxy, heteroaryloxy, C2_6 alkanoyl, arylcarbonyl, C2_b alkoxycarbonyl, aryloxycarbonyl, C2_6 alkanoyloxy, arylcarbonyloxy, C2_6 substituted alkanoyl, substituted arylcarbonyl, C2_6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2_6 substituted alkanoyloxy and substituted arylcarbonyloxy;
(c1), (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:
O
O
O
O +H ~- - ~ O
N N
_~
~ -~ O
Nn HN~, o oo ~o 1 N r O --N O~z ~_NH
~~'N O `-~o~~ o/1 o ~ and RN 7S4_ C. SUBSTANTIALLY NON-ANTIGENIC WATER-SOLUBLE POLYMERS (R1) The prodrugs of the present invention include a polymer residue Ri, 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 polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereo 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 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 oligonucleotides 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 compoun.ds (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-(CH2CH2O)n-where (n) is an integer from about 10 to about 2,300, and is dependent on the number of polymer arms when multi-arm polymers are used. It will be understood that the water-soluble polymer will be functionalized for attachjnent to linkers:
Alternatively, the polyethylene glycol (PEG) residue portion of the invention can be represented by the structure:
-Y71-(CH2CH2O)n-CH2CH2Y2I- , -Y7z-(CH2CH2O)F,-CH2C(-Y72)-Y71- , -Y71-C(=Y72)-(CH2)a2l-Y73-(CH2CHZO)n-CH2CH2-Y23-(CH2)a7z-C(=Y72)-Y7i- , -Y71-(CRnR72)a72-Y73-(CH2)b7i-O-(CH2CH2O)ri (CH2)bn-Y23-(CR7iRI2)a72-y7i- , -Y71-(CH2CH2O)õ-CH2CH2- , -Y71-(CH2CH2O)n-CHzC(=Y72)- , -C(=Y72)-(CH2)a7z-Y73-(CH2CH2O)ri CH2CH2-Y73-(CH2)a71-C(-Y72)- , and -(CR7lRn.)a72-Y73-(CH2)b7i-O-(CH2CH2O).-(CH2)b7i-Y73-(CR7iR72)$72- , wherein:
Y71 and Y73 are independently 0, S, SO, SO2, NR73 or a bond;
Y72 is O, S, or NR74;
R71_74 are independently the same moieties which can be used for R2;
(a7l), (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:
-C(=Y74)-(CH2)õ-(CH2CH2O)n , -C(=Y74)-Y-(CH2),n (CH2CH2O)õ-, -C(=Y74)-NRII-(CH2)m (CHZCH2O),:- , -CR75R76-(CH2)m (CH2CHZO)R_ wherein R75 and R76 are independently selected from among of H, C1_6 alkyls, aryls, substituted aryls, aralkyls, heteroalkyls, substituted heteiroalkyls and substituted C1_6 alkyls;
m is zero or is a positive integer, and preferably 1;
Y74 is O or S; and n represents the degree of polymerization.
Although, the prodrugs of the present invention can be formed usizrg any of the substantially non-antigenic polymers described herein, some preferred polyalkylene oxides in.clude:
CH3O-PEG-O-(CHZ)m-COH;
PEG-NHS;
CH30-PEG-O-(CH2)m NR11R12; and CH3O-PEG-O-(CH2)2-S-(CH2)m CO2H;
SC-PEG; or any of the art recognized activated PEGs.
In particular, polyethylene glycols (PEG's), mono-activated, C1_4 alkyl-terminated PAO's such as mono-methyl-terminated polyethylene glycols (mPEG's) are preferred when 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) 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, 5,919,455, 6,113,906 and 6,566,506, the disclosure of each of which is incorporated herein by reference. A non-limiting list of such polymers corresponds to polymer systems (i) - (vii) with the following structures:

mPEG-O-C~ ____CH2 H Y6i I62 O CHN
S-' mPEG-0-C,,,., 'CH2 H (i), m-PEG-N-C~
CH-.^ (Y63CH2)w61 C(`O)..' H ~
m-PEG-N-C
O (ii), H
m-PEG-O-C-N I., (CH2)4 ,CH-(Y63CH2).63C(=0)-m-PEG-O- m-PEG-0-C-N
li H
0 (iii), m-PEG-O--C-NH
\
( i H2)tv62 I

i C w61 (CH2)w64C(-0) ~C H 2~w63 "
m-PEG-O-C-N
pl H
(iv), m-PEG-0-C-N
( j H2)W62 H i (Y63CH2)w61C(-0)-(CH2~w63 m-PEG-O--C-~-N' H

OI (v), and m-PEG-C-NH

(CH2)w62 HC (Y63CH2)w61 C(=0)-( 1CH2)w63 m=PEG-C-N

O (vi) wherein:
Y61_62 are independently 0, S or NR61;
Y63 is 0, NR62, S, SO or SOZ
(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)is0or1;
mPEG is methoxy PEG
wherein PEG is previously defined and a total molecular weigb.t of the polymer portion is from about 2,000 to about 100,000 daltons; and R61 and R62 are independently selected from among hydrogen, Cr-6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C3-19 branched alkyl, C3_8 cycloalkyl, C1-6 su.bstituted alkyl, C2_6 substituted alkenyl, C2_15 substituted alkynyl, C3_8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1_6 heteroalkyl, substituted C1_6heteroalkyl, C1_6 alkoxy, aryloxy, C1-6 heteroalkoxy, heteroaryloxy, C2_6 alkanoyl, arylcaxbonyl, C2_6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2_6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2_6 substituted alkanoyloxy, and substituted and arylcarbonyloxy.
In yet another aspect, the polymers include rnulti-arm PEG-OH or "star-PEG"
products such as those described in NOF Corp. Drug Delivery System catalog, Ver. 8, April 2006, the disclosure of which is incorporated herein by reference. The 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 H2C- O-(CH2CH2O)õH
HC---O-(CH2CH2O)õH
CHZ

O
E
CHz HC-O-(CH2CH20)nH
x ~
O

HC-`- O-(CH2CHZ0)nH
H2C--- O-(CHzCH2O)nH
wherein:
(x) is 0 and a positive integer, i.e. from about 0 to about 28; and (n) is the degree of polymerization.
In one particular embodiment of the present invention, the multi-arm PEG has the structure:

HZC-O-(CH2CHZ0),H
HC -0-(CHZCHZO)õH

I
O

HC -O-(GHZCHZO)nH

`r a HC- O-(CH2CH20),,H
H2C-O-(CH2CH20),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, from 12,000 Da to 40,000 Da.
In yet another particular embodiment, the multi-ann PEG has the structure:

HO"( O OH

o OH OH
or (OCH2CH2)11-OH

HO-(CH2CH20)n (OCH2CH2)~oH
HIJ(CH2CH20)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 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 an.d is dependent on the molecular weight of the polymer.
The polymers can be converted into a suitably activated polymer, using the activation techniques described in US Patent Nos. 5,122,614 or 5,808,096. Specifically, such PEG can be of the formula:

O (CH2CHZO)~ `CH2CH2_O~
~-O-CHZCHz-(OCHzCH2)~~
O O'-CH CH O _JC ( z z ),,'-CH2CHz_O___ ~_O-CH2CH~,_OCH CH ~O
~ 2 2u' Star Or I'O-CH2CH2 (OCH2CH2)u'O 0---(CH2CH20),,-CH2CH2 O

~'O-CHzCHz-(OCH2CH2)U~~O Multi-arm O~(CHzCH2O)õ'-CH2CH2'O
wherein:
(u') is an integer from about 4 to about 455; and up to 3 terminal portions of the residue is/are capped with a methyl or other lower alkyl.

In some preferred embodiments, all four of the PEG arms can be converted to snitable activating groups, for facilitating attachment to aromatic groups. Such compounds prior to conversion include:

O,(CH2CHZO)~; 'CH2CHZ-OH
H3C-(OCH2CHz)õ~O O
-(CHZCH2O)U'-CH

H3C-(OCH2CH2)11-,(CH2CHZ0)~; 'O CH2CH2-QH
H3C'(OCHZCHz)u'~0 0, (CHZCH2O)U'- CH2CH2-~
'~c H (OCH2CH2)u.

~(CH2CH2O)'i'O CH2CHZ- OH
H3C-(OCHZCH2)u~0 O --Ic O ~(CHZCH2O)õ'-~CH2CHz, OH
HO~CHZCH2-(OCH2CHz)u' O (CH2CH2O)~; ~CHZCH2_ON
HO~CH2CH2-(OCH2CHz)õ~O O
-(CH2CH20),; - CH2CH2~-HO,CH CHz- ~O OH
2 (OCH2CH2)1 H3C-(OCH2CH2)~ -OrO O-(CH2CH2O)~,'-CH2CH2 OH
~
H3C-(OCH2CH2)U ' 'O O, (CH2CH2O)õ.-CH3 H3C-(OCH2CH2)~,-0 O-(CH2CH2O),,.-CH3 H3C-(OCH2CH2)1'- 0 O-(CH2CH2O)1'-CH2CH2-OH

HgC-(OCH2CH2)11'-O rO-,--( O-(CH2CH2O)u'-CH2CH2 OH
H3C-(OCH2CH2)11'- O O-- (CH2CH2O)õ--CH2CH2-OH

HO-CH2CH2--(OCH2CH2)U.-O O-(CH2CH2O),,.-CH2CH2---OH
H3C-(OCH2CH2),, '- O O, (CH2CH2O)õ'-CH3 H3C-(OCH2CH2)U-O rO O-(CH2CH2O),--CH2CH2-OH

HO-CH2CH2--(OCH2CH2)~, ~O O~(CH2CH2O),; -CH3 H3C-(OCH2CH2)u - OrO O-(CH2CH2O)õ -CH2CH2-OH
HO-CH2CH2-(OCH2CH2)UI- 0 O, (CH2CH2O)1=-CH2CH2--OH
HO-CH2CH2 (OCH2CH2)u.-O O-(CH2CH2O)õ.-CH2CH2-OH

H3C-(OCH2CHZ)W~- 0 O-- (CH2CH2O)õ'-CH2CH2-OH
and HO-CH2CH2-(OCH2CH2)U.---0rO~O-(CH2CH20)õ'-CH2CH2-OH
HO-CH2CH2-(OCH2CH2)u'- 0 O, (CH2CH2O),--CH2CH2-OH

The polymeric substances included herein are preferably water-soluble at room temperatu.i-e. A non-limiting list of such polymers include polyalkylene oxide hoxnopolyiners such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.

In a further embodiment, and as an alternative to PAO-based polymers, one or more effectively non-antigenic materials such as dextran, polyvinyl alcohols, carbohydrate-based polymers, hydroxypropyhxxethacrylamide (HPNIA), polyalkylene oxides, and/or copolymers thereof can be used. See also commonly-assigned U.S. Patent No. 6,153,655, the contents of which are incorporated herein by reference. It will be understood by those of ordinary skill that the same type of activation is employed as described here'm as for PAO's such as PEG.
Those of ordinary skill in the art will further realize that the foregoing list is merely illustrative and that all polymeric materials having the qualities described herein are contemplated. For purposes of the present invention, "substantially or effectively non-antigenic" means all materials understood in the art as being nontoxic and not eliciting an appreciable immunogenic response in mammals.
In some aspects, polymers having terminal amine groups can be employed to make the compounds described herein. The methods of preparing polymers containing terminal amines in high purity are described in U.S. Patent Application Nos. 11/508,507 and 11/537,172, the contents of each of which are incorporated by reference. For example, polymers having azides react with phosphine-based reducing agent such as triphenylphosphine or an alkali metal borohydride reducing agent such as NaBH4. Alternatively, polymers including leaving groups react with protected a-tnine salts such as potassium salt of methyl-tert-butyl .imidodicarbonate (KNMeBoc) or the potassium salt of di-tert-butyl imidodicarbonate (KNBoc2) followed by deprotecting the protected amine group. The purity of the polymers containing the terminal amines formed by these processes is greater than about 95% and preferably greater than 99%.
In a.Iternative aspects, polymers having terminal carboxylic acid groups can be employed in the polymeric delivery systems described herein. Methods of preparing polymers having lerminal 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 intennediate such as t-butyl ester of polyalkylene oxide carboxylic acid. This intermediate is formed by reacting a PAO with a t-butyl haloacetate in the presence of a base such as potassium t-butoxide. Once the t-butyl ester interznediate 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.

D. TARGETING AGENTS (Dl) Targeting agents can be attached to the polymeric compounds described herein to guide the conjugates to the target area .i.n 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 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 znolecu.les 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 Curr Op~
Phar.rzzacol. 2006 Oct;6(5):509-14, Cell-penetrating peptides as vectors for peptide, protein 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 moiety, e.g., an amino acid residue, to any art-standard emitting isotope, radio-opaque label, magnetic resonance label, or other non-radioactive isotopic labels suitable for magnetic resonance imaging, fluorescence-type labels, labels exhibiting visible colors and/or capable of fluorescing under ultraviolet, infrared or electrochemical stimulation, to allow for imaging tumor tissue during surgical procedures, and so forth. Optionally, the diagnostic tag is incorporated into and/or linked to a conjugated therapeutic moiety, allowing for monitoring of the distribution of a therapeutic biologically active material within an animal or human patient.

In yet a further aspect of the invention, the inventive tagged conjugates are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels.
Simply by way of example, these include 13'Iodine, 1zsIod.ine, 99iT`Technetium and/or 11lIndium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vrvo.
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 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 anterchangeably. The single chain antibody has 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.
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, MePC 603, D1.3, inurine phOx, human phOx, RFL3.8 sTCR, 1A6, Se155-4,18-2-3;4-4-20,7A4-1, B6.2, CC49,3C2,2c, MA-15C51K12Go, 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):11 (1988);
McCartney, J.
et al., ICSU Short Reports 10:114 (1990); McCartney, J. E. et al., unpublished results (1990);
Nedelu:zan, 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 MoDoclonal Antibodies in Clinical Oncology, Epenetos, A. A.
(Ed.), London, Chapman & Hall (1993); Bird, R. E. et al., Science 242:423-426 (1988); Bedzyk, W. D. et al., J. Biol. Chezn_ 265:18615-18620 (1990); Colcher, D. et al., J. Nat. Cancer Inst. 82:1191-1197 (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.
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);
Gockshuber, R. et al., 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. Natl. 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.
Chern. 266:16343-16349 (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. Imxnunol. 21:467-471 (1991);
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 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 P04 or SO4 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 aptamer, etc.

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 1 and PD 1 peptides, EGF and EGF
peptides, RGD peptides, folate, etc. Other optional targeting agents appreciated by a_Ttisans 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, penctratin, Oligo(Arg), preferably (Arg)g, folic acid, etc. For purposes of the present invention, the term "TAT" can be understood to mean a portion of trans-activator of 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) CYGRK.KRRQRRR ;
C-(Arg)9: (SEQ 1D NO: 2) CRRRRRRRRR ;
RGD can be linear or cyclic:

Hs HN HN
QThy4oNH
NH
O
H
HN O O N NH2 HN O O N~1VH2 N HN ~ ~ HN NH
O O
COOH ~ COOH 0 Y

Folic acid O OH

OH
OH

N~ N\~
~H
H2N N N ; ' and (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.

E. BIOLOGICALLY ACTZ'VE MOIETIES (D2) A wide variety of biologically active moieties can be attached to the activated polymers described herein. The biologically active moieties include pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides.
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 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 thioi-containing biologically -active moieties are 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.
Alternatively, parent compounds suitable for incorporation into the polymeric transport conjugate compounds of the invention, may be active after hydrolytic release from the linked compound, or not active after hydrolytic release but which will become active after undergoing a further chemical process/reaction. For example, an anticancer drug that is delivered to the bloodstream by the polymeric transport system, may remain inactive until entering a cancer or tumor cell, whereupon it is activated by the cancer or tumor cell chemistry, e.g., by an enzymatic reaction unique to that cell.
In one preferred embodiment, the polymeric transport systems described herein include phaaxnaceutically active coznpounds.
For purposes of the present invention, it shall be understood to mean that the pharmaceutically active compounds include small molecular weight molecules.
Typically, the pharmaceutically active compounds have a molecular weight of less than about 1,500 daltons. 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, 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'T includes all compounds within the taxane family of terpenes. Thus, taxol~ (paclitaxel), 3'-substituted tert butoxy-carbonyl-amine derivatives (taxoterJ) 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 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 prodrags of the present invention. The contents of the foregoing U.S.
patents are incorporated herein by reference.
Although the examples describe jzzter alia paclitaxel for illustrative purposes, it is to be understood that the methods described herein are suitable for all taxanes and related molecules.

2. Camptothecin and Related Topoisomerase I Inhibitors Camptothecin is a water-insoluble cytotoxic alkaloid produced by camptoteca accuminata trees indigenous to China and nothapodytes foetlda 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 v.rvo.
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, December 1992, pages 17-23. Camptothecin and related analogues are, for example, Topotecan, Irznotecan (CPT-11) or SN38.
Additional camptothecin analogs include those reported in the literature including the 10-hydroxycamptothecins, 11-hydroxycamptothecins and/or 10, 11 -dihydroxycamptothecins, 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 SN3 8.
3. Oligonucleoti.des In order to more fully appreciate the scope of the present invention, the followi_ng terms are defined. The artisan will appreciate that the terms, "nucleic acid"
or "nucleotide"
apply to deoxyribonucleic acid ("DNA"), ribonucleic acid, ("RNA) whether single-stranded or double-stranded, unless otherwise specified, and any chemical modifications thereof An "oligonucleotide" is generally a relatively short polynucleotide, e.g., ranging in size from about 2 to about 200 nucleotides, or more preferably from about 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 "polynucleic acid" may also be used synonyzuously 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, andJor the addition or substitution of functional moieties that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and functionality to an oligonucleotide. Such modifications include, but are not limited to, 2`-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodouracil, backbone modifications, methylations, base-pairing combinations such as the isobases isocytidine and isoguanidine, and analogous combinations.
Oligonucleotide modifications can also include 3' and 5' modifications such as capping. A non-limiting list of nucleoside analogs include:

o B o B o B o 0 B
o 0 =p-S- p=p-O- 0-p_0- A_Q~ p=P

Phosphorthioate 2'-O-Methyl 2'-MOE 2'-Fluoro B B B
O o B

0 _'O o 0 NHZ
2'-AP HNA CeNA PNA
0 0 B O F$ O B O B
N O~ 0 O O

O P-N I - 04-0- -, N

~ \ O P-0 Morpholino OH 3, 2'-F-ANA Phosphoramidate 2'-(3-hydroxy)Propyl ~
o B

Borarzophosphates See more examples of nucleoside analogues described in Freier & Altmann; NucL
Acid Res., 1997, 25, 4429-4443 and Uhlrnann; Curr. 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 encodes a control sequence. The terxn. "antisense strand" is used in reference to a nucleic acid strand that is complementary to the "sense" strand. In the normal operation of cellular metabolism, the sense strand of a DNA molecule is the strand that encodes polypeptides and/or other gene products_ The sense strand serves as a template for synthesis of a messenger RNA ("inRNA") 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 form duplexes. These duplexes then block either the further transcription or translation. In this manner, mutant phenotypes may be generated. The designatioris "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 "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 oligon.ucloetide derivatives can include from about 10 to 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:
Oligonucleotides and oligodeoxynucleotides with natural phosphorodiester backbone or phosphorothioate backboiae or any other modified backbone analogues;
LNA (Locked Nucleic Acid);
PNA (nucleic acid with peptide backbone);
short interfering RNA (siRNA);
microRNA (miRNA);
nucleic acid with peptide backbone (PNA);

phosphorodiamidate morpholino oligonucleotides (PMO);
tricyclo-DNA;
decoy ODN (double stranded oligonucleotide);
catalytic RNA sequence (RNAi);
ribozymes;
aptamers;
spiegelmers (L-conformational oligonucleotides);
CpG oligomers, and the like, such as those disclosed at:
Tides 2002, Oligonucleotide and Peptide Technology Conferences, May 6-8, 2002, Las Vegas, NV and Oligonuclcotide & Peptide Technologies, 18th & 19th November 2003, Hamburg, Geixnany, 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:

Representative Nucleotide Analogs And Derivatives 4-acetylcytidine 5-methoxyaminomethyl-2-thiouridine 5-(carboxyhydroxymethyl)uridine beta, D-mannosylqueuosine 2'-O-methylcytidine 5-methoxycarbonylmethyl-2-thiouridine 5-carboxymethylaaninomethyl-2- 5-m.ethoxycarbonylmethyluridine thiouridine 5-carboxymethylaminornethyluridine 5-methoxyuridine Dihydrouridine 2-methylthio-N6-isopentenyladenosine 2'-O-znethylpseudouridine N-((9-beta-D-ribofitranosyl-2-methylthiopurine-6-yl) c arb amoyl)threonine D-galactosylqueuosine N-((9-beta-D-ribofi.iranosylpurine-6-yl)N-methylcarbamoyl)threonine 2'-O-methylguanosine uridine-5-oxyacetic acid-methylester Inosine uridine-5-oxyacetic acid N6-isopentenyladenosine butoxosine 1-methyladenosine pseudouridine 1-methylpseudouridine queuosine 1-meth lguanosine 2-thiocytidine 1-methylinosine 5-methyl-2-thiouridine 2,2-diunethylguanosine 2-thiouridine 2-methyladeno sine 4-thiouridine 2-methylguano sine 5-methyluri dine 3-methylcytidine N-((9-beta-D-ribofuranosylpurine-6-yl)-carbamoyl)threonine 5=methylcytidin.e 2'-O-methyl-5-methyluridine N6-methyladenosine 2'--O-methyluridine 7-methylguanosine wybutosine 5-methylaminomethyluridine 3-(3-amino-3-carboxy-propyl)uridine Locked-adenosine Locked-cyEidine Locked-guanosine Locked-thymine Locked-uridine Locked-methylcytidine Preferably, the oligonuclcotide is involved in targeted tumor cells or downregulating a protein implicated in the resistance of tumor cells to anticancer therapeutics. For example, any art-known cellular proteins such as BCL-2 for downregulation by antisense oligonucleotides, for cancer therapy, can be used for the present invention.
See U.S. Patent Application No. 10/822,205 filed Apri19, 2004, the contents of which are incorporated by reference herein. A non-limiting list of preferred therapeutic oligonucleotides include antisense HIF-1a oligonucleotides and antisense Survivin oligonucleotides.
In one preferred embodiment, the oligonucleotide can be, for example, an oligonucleotide that has the same or substantially similar nucleotide sequence as does Genasense (a/k/a oblimersen sodium, produced by Genta Inc., Berkeley Heights, NJ).
Genasense is an 18-na.er phosphorothioate antisense oligonucleotide, TCTCCCAGCGTGCGCCAT (SEQ ID NO: 6), that is complementary to the first six codons of the initiatzng sequence of the human bcl-2 mRNA (human bcl-2 mRNA is art-known, and 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'mCs As-as-ts-Cs-cs-as-ts 9s 9s-mCs-As`Gs-c;
where the upper case letter represents LNA, the "s" represents a phosphorotliioate backbone;
(ii) antisense Bc12 siRNA:
SENSE 5'- GCAUGCGGCCUCUGUUUGAdTdT-3' (SEQ IDNO:4) ANTISENSE 3'- dTdTCGUACGCCGGAGACAAACU- 5' (SEQ ID NO: 5) where dT represents DNA;

(iii) Genasense (phosphorothioate antisense oligonucleotide): (SEQ ID NO: 6) ts-Gs-ts"cs-cs-cs-as-gs cs-9s"ts 9s-Gs-9s-Cs cs Gs-as-t where the lower case letter represents DNA and and "s" represents phosphoi-othioate backbone;

(iv) antisense HIFla LNA (SEQ ID: 7) 5'- TSGGSCaSaSgsC5a5tScScST$GsTsa -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:
~ [) configuration ~ .
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 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), amino linkers at 5' or 3' end of the oligonucleotides, where w in this aspect is a positive integer of preferably from about 1 to about 10, preferably 6. The modified oligonucleotides contemplated in the compounds described herein can be NH-(CH2)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 particular embodiment of the present invention employs Bcl2-siRNA having the sequence of SENSE 5'-(NH2-C6)GCAUGCGGCCUCUGUUUGAdTdT-3' ANTISENSE 3'- dTdTCGUACGCCGGAGACAAACU-5'.

In another preferred embodiment, the compounds described herein can include oligonucleotides modified with hindered ester-containing (CH2)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.
The 5polymeric compounds can release the oligonucleotides without amino tail.
For example, the oligonucl.eotides can have the structure:

NH-(CH2)w O O-Oligonucleotide 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 (CHZ)W
sulfliydryllinkers (thio oligonucleotides). The thio oligonucietides can be used for conjugating directly to cysteine of the positively charge peptide or via maleina.idyl group. The thio oligonucleotides can have the structure SH-(CHz)W-Olzgonucleotide. The thio oliganucleotides can also include hindered ester having the structure:

SH-(CH2)W
0 O-Oligonucleotide Exemplenary of the modified oligonucleotides include:
(i) Genasense modified with a C6-NH2 tail:

5'- NH2- C6- stsestsesCscsasgsesgstsgsCsgsCsesast -3' S
k -l2 N O- P- T-sC-sT-sC-sC-sC-sA-sG-sC-sG-sT-sG -sC-sG-sC-sC-sA-sT
O- -(ii) antisense HiF 1 a LNA modified with a C6-NH2 tail:
5'- NH2-C6- s'TsGsGscSasasg5c5astsescsTSGSTsa -3';
(iii) antisense Survivin LNA modified with a C6-NH2 tail:
5'- NH2- C6- s'nCTsmCsAsastscscsastsgsg'nCsAsC~-Tsc -3' -(iv) antisense Survivin LNA modified with a C6-SH tail 5'- HS- C6- smCsTsmCsAsastsGsesastsgsgsm CSASGsc -3';

(v) Genasense modified with a hindered ester tail O
HZN O T-C-T-C-C-G-A-G-C-G-T-G-C-G-C-C-A-T
4. Additional Biolagically-Active Moieties 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 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-furigals such as nystatin or amphoteracin B,.
anti-inflammatory agents, steroidal agents, and the like.
The foregoing is illustrative of the biologically active moieties which are suitable for the prodrugs of the present invention.

F. PERMANENT AND RELEASABLE LINKERS: (Ll and L3) The L, and L3 linkers include bifiinctional linkers. The bifunctional can be permanent or releasable linkers. The bifunctional linkers include amino acids or amino acid derivatives.
The amino acids can be among naturally occurring and non-naturally occuzring amino acids.
Derivatives and analogs of the naturally occurring amino acids, as well as various art-known non-naturally occurring amino acids (D or L), hydrophobic or non-hydrophobic, are also contemplated to be within the scope of the invention.' A suitable non-limiting list of the non-naturally occurring amino acids includes 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, beta-aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, piperidinic acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2-aminopimelic acid, 2,4-aminobutyric acid, desmosine, 2,2-diaminopimelic acid, 2,3-diaminopropionic acid, N-ethylglycine, N-ethylasparagine, 3=hydroxyproliza.e, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methyl-isoleucine, 6-N-methyl-lysine, N-methylvaline, norvaline, norleucine, and ornithine. Some preferred amino acid residues are selected from glycine, alanine, methionine and sarcosine, and more preferably, glycine.
Alternatively, The Ll and L3 can be selected from among:
-[c(WO)1,(CR22R23)c[C{=0)1, - , -[C(=O)]v(CR22R23)t-O[C(-O)]v>_ -[C(=O)]-,(cR22R23)t-NR26[C(=o)]v'- , -[C(=O)]-,O(CR22R23)t[C(-0)]v'--[C(=O)]O(CR22R23)tO[C(=O)]õ'- , -[C(=O)]r,O(CR22R23)tNRi6[C(=O)], - , -[C(=O)1õNR2z(CR22R23)t[C(-O)].,'- ~
-[C(=O)]~NR21(CR22R23)tO[C(=O)].,'- , -[C(=0)]-,NR21(CR22R23)tNR26[C(=O)]v'- , -[C(=o)],(CR22R23)tO-(CR2sR29)r [C(=0)],'- , -[C(=O)]v(CR22R23)tNR26-(CR2sR29)t'[C(=O)]-,'- ~
-[C(=O)],(CR22R23)tS-(CR28R29)t'[C(=O)],,- a -[C(=O)]õO(CRi2R23)tO-(CR28R29)t [C(=0)]1'--[C(-O)],O(CR22R23)tNR26-(CR2sRzs)t [C(=O)], - , -[C(=0)]vO(CR22R23)tS-(CR28R29)t IC(=O)]v'- ?
-[C(=O)],,NR21(CR22R23)tO-(CR2sR29)t'[C(-O)],,_ , -[C(=O)]vNR21(CRzzRz.3)tNR26-(CR.2sRzs)t [C(=O)]v - , -[C(=O)]vNR2i(CR22R23)1S-(CR2aR29)t [C(=O)]"--[C(=O)],(CRz2R23CR2sR29O)tNR26[C(=O)]" - , -[C(=O)]V(CR22R23CR2sR2g0)t[C(=O)],'- ~
-[C(=O)]vO(CR22R23CR2sR290)tNR26[C(-O)],,- ~
-[C(=O)]-1O(CR22R23CR2sR290)t[C(=O)]v'- , -[C(=O)]õNR21(CR22R23CR2aR-290)tNR26[C(=O)]v - , -[C(=O)]vNR2i(CR22R23CR28R290)t[C(-0)]1'- , -[C(=O)],(CR22R23CR28R290)t(CR24R-25)t [C(-0)], - , -[C(=O)]vO(CR22R23CR2sR29O)t(CR-24R25)t [C(=0)], - ~

-[C(=O)]-,NR2i(CR22R23CR2sR29O)t(CR24R25)ti'[C(=O)]~,'- ~
-[C(=O)],(CR22R23CR2sR290)t(CR24Rzs)t OIC(-0)]v,; , -[C(=0)],(CR22R23)r(CR24R25CR28R290)t'[C(=O)1v,--[C(=O)],(CR22R23)c(CR2aR25CR2sR29O)t NR26[C(=O)]v'- , -[C(=O)]`,O(CR22R23CR2sR29O)c(CR24R2s)x O[C(=O)],,~- , -[C(=O)]õO(CR22R23)c(CR2aR25CR2sR2s0)t'[C(=O)]1'- , -[C(=O)],,O(CR22R23)i(CR24CR25CR28R29O)c'NR26[C(=O)]-,'- , -[C(=O)]~NR2i(CR22R23CR2sR29O)t(CRz4R2s)t O[C(=O)]õ'- , -[C(=O)] õNR2 z (CR22R23)ti(CR24R25CR28R29O)c [C(=O)] , - , -[C(=O],NR21(CR22R23)t(CR24R25CR28R290)c NR26[C(=0)],r - , N O
NN
O H O

-IC(=0)]~O(CR22R23)~ (CRz4R2s)tNR26[C{=0)]V--[C(TO)]VO(CR22R-23)ti (CR24R25)V0[C(=0)]V-OXI-/ -[C(=O)]vNR21(CR22R23)t (CR24R25)t'NR26[C(=O)]v'- and -IC(--O)INR21(CR22R23)t C \:/(CR24R25)t'O[C{=0)]v wherein:
R21_29 are independently selected from among hydrogen, Cz_6 alkyls, C3_z2 branched alkyls, C3_8 cycloalkyls, C1_6 substituted alkyls, C3_8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, Cz_6 heteroalkyls, substitnted Cz_6 heteroalkyls, Cr_6 alkoxy, phenoxy and C1_6heteroalkoxy;

(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.
Preferably, the bifunetional linkers can be selected from among:
-[C(=0)]FNH(CH2)ZCH N-NHC(=O)-(CH2)2- , -[C(=O)]rNH(CH2)2(CH2CH2O)2(CH2)2NH[C(=O)]r' - , -[C(=O)]zNH(CH2CH2)(CH2CH2O)2NH[C(=O)]r'-, -[C(=O)]rNH(CH2CH2)sNH(CH2CH2)s'[C(-O)]r~- , -[C(-O)]r NH(CH2CH2)sS(CH2CH2)$'[C(-O)]T'- , -[C(=O)],NH(CH2CH2)(CH2CH2O)[C(-O)]r,- , -[C(=O)],NH(CH2CHz)SO(CH2CH2)5>[C(=O)]r>- , -[C(=O)]zNH(CHZCH2O)(CHZ)NH[C(=O)]r'- , -[C(=O)]rNH(CH2CH2O)2(CH2)[C(=O)]r'-, -[C(-O)]iNH(CH2CHzO)s(CH2)S'[C(=O)]T>- , -[C(=O)]rNHCH2CH2NH[C(=O)]r'- , -[C(=O)]INH(CH2CH2)2O[C(=O)]x'- , -[C(-0)]rNH(CH2CH2O)[C(=O)]r, -[C(=O)]rNH(CH2CH2O)2[C(=O)]r~- , -[C(=O)]rNH(CH2)3[C(=O)]r-, -[C(=O)]zO(CH2CH2O)2(CH2)[C(=O)]r'- , -[C(=O)]rO(CH2)ZNH(CH2)2[C(-O)]r'- , -[C(=O)]rO(CHZCH2O)zNH[C(-O)]r'- , -[C(=O)]rO(CH2)2O(CHZ)2[C(=O)]r'- , -[C(=O)]rO(CH2)2S(CH2)2[C(=O)]r'- , -[C(=O)]zO(CH2CHZ)NH[C(=O)],'- , -[C(=0)]rO(CH2CH2)O[C(=0)]z'- , -[C(-0)]rO(CH2)3NH[C(=0)]r,- , [C(-O)],,O(CH2)30[C(=O)]r>- , -[C(=O)]xO(CH2)3[C(=0)]r'- ~
-[C(=0)]xCH2NHCH2[C(=0)]r,- , -[C(-0)]rCH2OCH2[C(-0)]r- ~
-[C(=0)]rCH2SCH2[C(-0)]r'- , -[C(=O)]rS(CH2)3[C(-O)]r,- , [C(=O)]z(CH2)3[C(-0)]r- , -[C(=O)]r0CH2 aCH2NH[C(=O)]r' -M=O1OCH2 CH2O[C(=O)1r'-.' -IC(=O)IrNHCH2 CH2NHjC(=0)lr'- and -[C(=O)]rNHCH2 CH20[C(=0)]r'-wherein (r) and (r') are independently zero or 1, provided that both (r) and (r') are not simultaneously zero.

1. Releasable Linkers In one preferred eznbodiment of the invention, the compounds described herein contain a biologically active moiety attached to a releasable linker. One advantage of the 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 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.
One preferred releasable linker is a peptidyl linker (Val-Cit) which can be specifically degraded by capthesin B. Preferably, the L3 include releasable linker.
The preferred releasable linkers include:

y f11~ I R31 Y~14 L11 C Y12--Ar i Y13- C- --al ibli ell e11 f11 L331 R3,571 i~ 16 C I- -R~ C Rss C
o ~ ~
AC g11 h11 I
4Yi R38 i11111 Y'is N C C (J)x11 1-I~ I
A59-(J')x'11 (L14)q11 C ~-98-0 C (CR46R47) m11 p11 /011 n11 +s-S^~

-N-. /~

-Val-Cit-, -Gly-Phe-Leu-Gly-, -Ala-Leu-Ala-Leu-, -Phe-Lys-, _ I~ H
-~-Val-Cit --C-N
O
It H
-~-Ph -Lys-C-N \ / ~, HN ~
$ -J-Val-Cit-~ O O
, HN \ '7/
--Phe-Lys-~
O O, -Val-Cit-C(=0)-CH2OCH2-C(=O)-, -Val-Cit-C(=O)-CH2SCH2-C(=O)-, and -NHCH(CH3)-C(=O)-NH(CH2)6-C (CH3)2-C(=O)-wherein, Y11_z9 are independently 0, S or NR48;
R3148, Rs0-sr and A51 are independently selected from among hydrogen, C1_6 alkyls, C3_ j2 branched alkyls, C3_$ cycloalkyls, C1_6 substituted alkyls, C3_8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1_6 heteroalkyls, substituted Ca_6heteroalkyls, C1_6 alkoxy, phenoxy and C1_6heteroalkoxy;
Ar is an aryl ar'heteroaryl moiety;
T:11_1s are independently selected bifunctional spacers;
J and J' are independently selected from selected from among moieties actively transported irrto a target cell, hydrophobic moieties, bifiinctional linking moieties and combinations thereof;
(c11), (h11), (k11), (111), (m.11) and (n11) are independently selected positive integers, preferably 1;
(a11), (e l l), (g l l), (j 11), (o 11) and (q11) are independently either zero or a positive integers, preferably 1; and (b 11), (xl1), (x'11), (fl 1), (il l) and (p 11) are independently zero or one-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 incozporated herein by reference. The bicine-based linkers are also described in commonly assigned U.S. Patent Nos.
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 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 to tlhe multifunctional linker through a permanent linker. The perm.anenfi linkers are capable of conjugatin~g the targeting moiet,= a,nd thP mõ?tifii-nctional_ 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:

0 -4/--N ~- H~_ ~H N

O O
, > > ~
H O O H O
N~/~/ N N
H
O O

N
O
o , o and o .
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 sulfone, amino, carboxy, xnercapto, hydrazide, carbamate and the like instead of m.aleimidyl.

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-hydroxysuccinimidyl, para-nitrophenoxy, N-hydroxyphtalimide, N-hydroxybenzotriazolyl, imidazole, tosylate, mesylate, tresylate, nosylate, CI-C6 alkyloxy, Cr-C5 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 apparezl.t to those of ordinary skill.
For purposes of the present invention, leaving groups are to be understood as those groups which are capable of reacting with,a nucleophile found on the desired target, i.e. a -biologically active moiety 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, 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 D1 or D2 are fnnetional groups, the La and L3 groups are not funetional moieties.
In one preferred embodiment, the functional group can be maleimidyl and the leaving 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:

HN'_VaI-Cit-C D2 H
mPEG-_'~Y N O O

N

o 11 Dz C-Cit-Va!_NH HN~Va1--Cit-C D2 H H
O O N-f---PEG~N O O
D, H O O H D1 O

I I
HN~VaI-W-Cit-C

mPEG~N O O

O NH D, O , _ II
D C-Cit---VaiNH H H HNiVaI-Cit-C

0 O N-91-~PEG//,,~,N O 0 ' -i0'-'--' D, N H O O H Dl HN~Vaf-Cit-C-H ~(D2 H O \~
O
rr~PEG~N O 0 O
N
O H i D~

0 0 _ D~O ~-C-Cit-Va[~~ H H~N ~VaE-Ci~-C-H ~ ~ D2 // O ~
O O O H~PEG~H O O O
1_tN-'iO`--N N -D H O O H Dt a z fl NN,Va1-Cit-C D2 H
mPEG~N O O
O N D, 11 D2 C---Cit-Val_NH WNiVaI-Cit---C D2 H
O O N_q_-~ PEG--y O O

_ I I
HN~VaI--Cit-C ~ ~

mPEG-^~y N O
O Dj O

_ !I
C-Cit-VaINH HN~-VaI-Cit-C ~D2. H H D2 O O N~PEG~N 'L~N O
Dl N O O Dl .0 0 I I
HN,VaI-Cit-C-NH D2 H
mPEG~N O O O .
O N D, ~ 0 0 0 11 D~0 NH-C-Cit-Val_NH H H HN.VaI-Cit-C-NH DZ
a~\(\
Q -r'~PEG')~ o 0 0 Dj--~N fl 0 N Dt 1]
HN,VaI-Cit-C-D2 H
mPEG~N O 0 0 ~
O H N Dj D2-CW--Cit-Val,NH HN,VaI-Cit-C-D2 H H
O O N_r--~ PEG--YN O O

p1 NH O 0 H DI

HN~,VaI-Cit-C DZ
H
mPEG---~- N O
0 :t,--,N__rD, D2 --C-Cit=Val HN~VaI---Cit-C D2 NH H H
O N~PEGN Ito H
D1~N O 0 N-T--- D, HN_,,VaI-Cit-C D2 H

N
mPEG~N p ` R52 O N--~S~D

11 D2-C-Cit------VaI,NH HNrVaI-Cit---C D2 R52, N' R51 O N~PEG~N jto~~ H F`51~N'1352 D1 SN 0 0 N S` p 1 HN,Va[-Cit-C

mPEG~N p R51 `N R52 p N_ ~ D I
`- I

0 p Q _ C-Cit-Val NH HN~VaI-Cit-C ~ ~
D
D2 R52, N - R51 p N-~, PEG N O R51-N-R52 Z
S H ,D
Dj~ ~N O 0 t,--,N Sl II
_ O
HN,Val-Cit--C-NH~~ D2 N R51-N-R52 O H mPEG~ O
p N_ ),/S,p1 Dz O \/ NH-C-CitJ--VaI NH HN,Va1=Cit-C-NH \/ p D2 H N N R Rs1-NR52 ~\C\O
p R5z- N. R51 p ~PEG~ H
pl" S---Iy N 0 O N-?~-S.Di I I
HNI.Phe-Lys-C D2 mPEG--~-r N O R5'~ - N - R52 H
O NS'D
`I j II {1 Dz-C-Lys-Phe -Phe-Lys-C Dz NH HN
~N S1-NRSz R52, NR51H O N~PEG H
D~ S~N 0 0 N~S, O
li HN~Phe-Lys-C \ /

mPEGN R51-N.R52 N
O
- ~ ~ -C-Lys-Ph ~H H HN'Phe-Lys-C

2 R52- N.R51H O N~PEGN p HR51-N- R52 D1 S~N O O N-I~S.D1 O O
O
11 _ ~Phe---~Lys-C-NH DZ
H HN 0-~-~

mPEG----y Ito O N~S.D

0 and o o _ D2 NH--C-Lys-Ph NH HN' Phe-Lys--C-NH \ ~ O D2 pp ~2~N R51 N N Hj:t Rst-N R52 -~O
p ~PEG~ O H
p1"S,,YN O O N~S,pi O O
wherein mPEG has the formula: CH3-O(CH2CH2O)n ;
PEG has the formula -O(CH2CH2O)õ;
(n) is a positive integer from about 10 to about 2,300;
R51 and R52 are independently selected from among hydrogen, C1-6 alkyls, C3_12 branched alkyls, C3_8 cycloalkyls, C1_6 substituted alkyls, C3_$ substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1_6 heteroalkyls, substituted C1_g heteroalkyls, CI-6 alkoxy, Cz_6 alkyloxycarbonyl, aryloxycarbonyl, phenoxy and C1_6heteroalkoxy;
DI is a targeting moiety, a functional group or a leaving group; and D2 is a biologically active moiety, a functional group or a leaving group.
Preferred polymeric compounds according to the present invention include:

HN~VaI--Cit-C SN38 H
mPEG~N O 0 O N---~iO~
O H
O

li If SN38-C-Cit-Val ~Val-Cit-C SN38 H HN
NH

O N~PEG~N O 0 N N '-'--IZ H

li -HN~VaI-Cit-C-SN38.
H
mP1=G--y O 0 O
IV N
O H

SN38-C-Cit-VaI_NH HN_,VaI-Cit-C-SN38 H H
O O Nlr,,-~ PEG---f N O 0 N
t N /~/~ N N-H O O H /

II
HN,Va[-Cit-C

mP1=G~N O

O H

- p p ~ 11 z C-Cit-Val, NH ~VaI--Cit-C ~ ~

0 O N-rPEG'_-rN O p O~'~ N N~iO~tO H O 0 H

HN~VaI---Cit-C-~ CD~~ SN38 H O ~
O
mPEG~N O 0 O
O H

O , p 0 _ SN38 (' :) ~-C-Cit-Va1_NH HN_VaI-Cit-C-H ~ ~ N38 0 O O N~PEG~N O

N--'-'O--'---N 0 0 N _____\,~'~
<\ ~ i-{ 0 O H /
~O O 0 I l HN-WaI---Cit-C SN38 H

mPE.G"'rN t,,N O
O \

SN38-C-Cit-VaI, NH HN_-VaI-Cit-C SN38 O NPEG_-y p t--_o O O N
N

0 II~
HN~Va{-Cit---C

mPEG O t--_o O N

_ 0 C-Cit-Val_NH HN,VaI-Cit-C

O p N_~, PEG"-~- N O O
N p p Il HN,_VaI-Cit-C-NH (_~ SN38 H O~\(\
mPEG--'-yN t 0 0 O N

0 0 0 _ SN38 ~ ~ NH-C-Cit-VaI, NH HN~,VaI-Cit-C-NH 5N38 ~O H H C_ ,\C\
C C O N~PEG-rN O C C
/ N Q O

~VaI----Cit-C SN38 H HN

mPEG__)~ N. O NHBoc N H O S\S

0 NO2, 11 SN38-C-Cit-Val ,Val-Cit-C--SN38 NH H H HN

N NHBac N~PEGN O NHBoc N~ I
(;~ S~S N 0 0 N S`S

NO2 0 0 NO2 , II -HN~Vai----Cit-C

mP1=G--yN O NHBoc T
S
0 H ~S \ ~

O 0 _ SN38\ ~ C-Cit--Vai_NH H HN~VaI-Cit-C SN38 H
" N NHBoc p N~PEG--,yN O H NHBoc \ I
N O G N__rr),,_,S~S

N02 O O OC/, 1 I
~VaI--Cit-C-NH SN38 H HN G-\(\
mPEG"yN O NHBoc N~ C
C N S`g ~ ~
~
O N02 , SN38 G ~~ NH-C-CitfVal,NH H H HN,VaI Cit-C-NH \ ~ O~ N38 0 ~N NHBoc p N~PBGH NHBoc N~ I C
~/ S.S\)I~ O O N~S S\
NOZ ~O` O NOz O

,Phe-Lys-C SN38 H HN

mPEG-,yN :to,--,N,,'~,S NHBoc N ~
O ` S ~ I.
0 N02a SN38-C-Lys-Phe,NH HN.,Phe--Lys-C-SN38 ~N NHBo~ O N~PEG~N O H NHBoc 1V~ ~
I/ S.S~N O p N~S.S ~
N02 O p NO2 O

Il -",Phe=Lys-C

mPEG--yN to NHBoc N ~
C} N~S.S \ {

o o- _ n n C-Lys-Phe ~Phe-Lys-C \ ~

N NHBo ~ O N~PEG~N o H NHBoc N I
S=S---Iy N 0 0 N__~S, S

_ HN~Phe---Lys-C-NH ~ ~ SN38 H O
mPEG--~yN O NHBoc N~ O H
O N__Jf~S'S\
p NO2 _ a 0 SN38 \ ~ NH-C-Lys-Phe NH HN'Phe-Lys-C-NH \/, O SN38 -\(\
O O ~ N NHBoc p N~PEG~N O NHBoc N'O
5~N O O ~~
S ~

II
,Val-Cit-C SN38 H HN

mPEG~ N Jto-, I I
SN38-C---~Cit-Va[ NH HN,VaI-Cit---C SN38 p N PEG N Ito HzN ~ 0 NH2 ~ O-LNA
HN~\
H
mPEG--)r N O

0 H p p N H 0 LNA-O~ N~CNH ~O-LNA
H H HN/\
p N~PEG~N O

I I
HN,VaI-Cit-CSN38 H
mPEG-~y O 0 N~~p~~
p H S-SCA

SN38-C---Cit--VaI, NH HN,VaI-Cit--CSN38 H H
p PEGN O p O p p I I
HN,VaI-Cit--C--SN38 H
mPEG-~yN O p 0 p H N S-SCA

SN38-C--Ct-VaI, NH HN,VaI-Cit-C--SN38 H H
p p N-r--- PEG 1 N O 0 SCA 5 N~/\ H p p H N S-SCA
p 0 H ~Val-Cit--C ~ ~

mPEG~N O 0 N~~p~~

O , _ C-Cit-VaINH HN -VaI-Cit-C\/ SN38 p N~PEG~N O

SCA-S N/~ \~H 0 0 H N S-SCA
O p 0 ~~ -HN_,VaI-Cit-C----N

mPEG~N O
O

N
O H S-SCA

0 0 _ H-C-Cit VaINH HN~VaI-Cit-C-H \ ~

O O . O N---PEG~N O O 0 O
-'-'O'-N
N-SCAS H O O H S-SCA
O O , II
HN--VaF---Cit-C SN38 H
mPEG~N O 0 11 SN38 C-Cit-Val NH HN_-VaI-Cit-C SN38 H H
O O N)f"PEGN O O
SCA-S N O O t-__N?-S-SCA

HN_-Va!-Cit=C

mPEG~N 0 0 O S-SCA

_ C-Cit-Val HN~Va!-Cit-C

O O PEG~N O O
SGA-S--~ N 0 0 t---7N S-SCA

HN -VaI-Cit-C -NH\/

mPEG O O

SN38 \/ NN-C-Cit-VaINN P " HN-VaI--Cit-C-NH SN38 O O N~PEG--( N t O O

I I
,Val-Cit-C SN38 H HN
mPEG---y N O H
0 N--rFolic acid 11 SN38 C-Cit-VaI_NH HN,VaI-Cit-CSN38 H H
H O N---- PEG~N O H
Foiic acid---- N 0 0 N~Fol9c acid ~Val-Cit-C SN38 H HN

mPEG"-r N O H NH2 0 N-?--:~S, S-SCA

SN38 C-Cit-~-Val NH HNiVaI--Cit-C SN38 H
NH2 H p N,,If,--- PEG"Y N to, H NH2 SCA-S' N 0 O NS`S-SCA
~

If ~Val-Cit-C \ ~

mPEG,-,,rN O H NH2 0 1:t,-, N~S- S SCA
O

C-Cit-Val NH HN~,VaICitC

NH2 N,,r~ PEG---YN O H NH2 SCA-SS_,_~ c N O 0 N S~S-SCA

II -~Val--Cit-C- NH \ ~

mPEG,~y N O H NH2 O N-,(~S~S-SCA

0 _ 0 0 _ SN38 / NH-C---Cit-Val NH H H HN~Va1-Cit-C-NH ~/ gN38 O NH2 H O ~PEG H
SCA-S'S~N 0 0 N S,S-SCA

~Phe-Lys-C SN38 H HN

mPEG~N O H
0 N--TrEolic acid 11 SN38 C-Lys-Phe I.Phe-Lys-C SN38 NH H HN

H O N~PEG~N O H
Folic acidN 0 0 N-ir Fplic acid lf HN~Phe=Lys-C SN38 H
mPEG---y N O H NH2 0 N--(~Sl S-SCA

SN38 C--Lys-Ph NH HN~Phe-Lys-C SN38 H H

NH2 p N~PEG---_rN O H NH2 H SCA-SIS N 0 0 NS,S-SCA
~

II -HN~Phe-Lys--C

mPEG"-r N O H NH2 O N~Sl S-SCA

0 - p 0 SN38 C-Lys -Ph NH H HNPhe-Lys-C SN38 NH2 H p N-f---- PEG----rN O H NH2 SCA-S- S,,,~ r N 0 p N~S,S-SCA

ll ~Phe-Lys-C-NH -\ ~
H HN Ti-SN38 mPEG-'~' N O H NH2 O
0 N~S-S-SCA

_ \ / NH--C--Lys-Ph NH "Phe-Lys-CNH \

O NH2 H O N -ff~--PEG -YN H NH2 O
SCA-S' SN 0 0 S-SCA

HN,VaI-Cit--C SN38 mPEG-~y O O

O H S-RGD
O NO-~-II 1[
SN38 C-Cit-Val_NH HN,VaI-Cit-CSN38 H H
O O N-,,~ Pi=G---f N O O
0~~ ~i0-~
RGD-S tN H O O N N S-RGD

IE
HN,VaI-Cit--C--SN38 H
mPEGN O O
O
O H S-RGD
O

SN38-C-Cit-Val NH HN~VaI-Cit-C-SN.38 H
O PEG~N O
O O

RGD-S N~\H O O H N S-RGD

il HNVal-Cit-C

mPEG-~yN O O
O
N--,~ O--~
O H S-RGD

it C--Cit-VaI.NH HN~,VaI-Cit-C SN38 p NY-PEG'-If N O

N~~p~~N p H~i S RGD

O p HN~VaI-Cit---C----H \ /

O O
mPEG~-N , 0 O O H~~O~
SRGD

H-C-C't---VaLNH H HHNiVal-Cit--G-H \~ SN38 G a p N-~-PEG--rN a RGD-S~^/ \/\H ; N S RGD
o fl , II
HN-,VaI-Cit---C SN38 H
mPEG---YN O O
O N S-RGD

SN38 C-Cit-Val_NH HNVal-Cit-C SN38 H H
O N-a---~PEGN O O
RGD-S N O .0 N S-RGD

_ I I
HN~,VaI-Cit---C \ ~

mPEG~N O O
O N S-RGD

_ CCit-VafNH HN~VaI-----Cit-C \ / SN38 Q p NY-- PEG-'-y N O O

HN,,VaI-Cit-C-NH \ /

mPEG t-_o O
~ N S-RGD

0 o 0 SN38 ~/ NH-C-Cit-VaLNH H H HNiVaI-Cit---CNH () C O N_-PEG"Y N O O O
RGD-S N 0 0 N S-R{''D
//]]

d 0 I I
.,Val-Cit--C SN38 H HN
PEG~N O H
0 N--rNH-RGD

SN38. C-Cit-----VaI_NH HNI-IVaI----Cit-C SN38 H N PEG N

H H RGD
RGD-NH~..~ N ~ 0 N NH

II
,VaI-Cit-C SN38 H HN

mPEG--,-YN O NH2 H 0 N--(~S_ S-RGD

SN38 C-Cit-Val HN,VaI--Cit-CSN38 NH H H
NH2 p N__--- PEG--yN O H NH2 RGD-SIS N 0 O N--(~S,S-RGD
~

I I
HN~VaI-Cit-C

mPEG--y N H
O NHZ

O

SN38 C-Cit----Val NH HNiVaI-Cit-C SN38 H
NH2 H p N-q----PEG--y N H NH2 RG.D-S- S~N 0 O N S, S-RGD

li -~N,VaI-Cit-C-NH

mPEG~N Jto-~ H NH2 O N-Irl~-S-RGD

~ ~
~ ~ NH--C-Cit-Val iVal-Cit C--NH _ C NH2 HC) N-T--- PEG--,rNto-'H NHZ O
RGD-S'S 0 0 N-4--l~S`S-F2GD
~ 0 0 If HN",Phe-Lys-C SN38 H
mPEG~N
~
O --TrNH-RGD

11 SN38 C-Lys-Phe Phe-Lys-C SN38 NH H H HN

H N -Tr----PEG --yN to H
RGD-NH--,,N O O N--r NH-RGD

fI
HN'Phe-Lys-C SN38 H
mPDG--'--r N O H NH2 0 N~S`S=RGD

p O
SN38 C-Lys-Phe HNPhe-Lys--C SN38 NH H H

NH2 O NPEG--'-r N O H NH2 H RGD-S~S N 0 0 NrK~,S~S-RGD

II -H HN'Phe--Lys-C

mPEG--,YN O H NH2 O N~S- S-RGD
O
O _ C-Lys-Ph NM HN~Phe Lys-C \ /

NH2 H N~PEG~'N O H NH2 RGD-S~S 0 0 NS,S-RGD
~ 0 0 0 ~Phe-Lys--C-NH _ ~ ~

mPEG-'t- N O H NH2 O
0 NS, S-RGD

_ 0 NH-C-Lys=Phe NH H HN"Phe-Lys C NH ~ ~ SN38 SN38 \ /
G NH2 H Q N_T-_PEG,-,,rN0 H NH2 RGD~ G
S"J'-N 0 fl N Yl-~- S-RGD

:::~ OWLNA
HN
H
mPEG--~YN H
0 NFQIic Acid p N p NH H H HN O N
LNA-O ~ ::~ "-~ O-LNA
p N~PEG~N O
H ) piIG Acid Fo1iG AGid~N 0 0 N--Ir o H
~ O-LNA
HN
H
mPEG~N H
0 N---rFolic Acid O and W O O N
LNA-O
~NH H H HN::~
p N~Pk*G~N
H Folic Acid Folic Acid~~ 0 O N-ir wherein S-SCA is a singie-chain antibody;
RGD is HS

HN
p NH
H H
H HN NH
N

LNA is locked nucleic acids;
Folic acid is a residue of OH
N
H2N-~' N HO
\N- ~,--~ - O 0 HNIUON

O =
SN38 is 7-ethyl-10-hydroxycamptothecin;
mPEG has the formula: CH3-O(CH2CH2O)n ;
PEG has the formula -O(CH2CH2O)n-; and (n) is a positive integer from about 10 to about 2;300.
I. METHODS OF MAKING THE CONJUGATES
Generally, the conjugates can be made by sequentially attaching the polymer, cytotoxic agent and targeting moiety to the znultifunctional 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 multifunctional linker can be prepared by conjugating a polymeric compound having a OH or a leaving group 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 leaving groups. This intermediate can further reacted with a targeting moiety to form the polymeric conjugate znultisubstituted 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 groups such as tert-Bu ester and methyl ester of carboxylic acid terminals can be deprotected selectively and stepwise to provide various degree.s of active group to be conjugated wzth different biologically active agents such as cytotoxic agent and targeting agent. As shown in FIG. 1, maleimidyl group and succinix.nidyl ester can react selectively with SH or NH2 containing moieties, respectively.
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:
reacting a compound of Formula (IIIa):
Ml_`."(L,)a-L2- M2 Rl A21 (IIIa) with a biologically active m.oiety-containing compound having Formula (IIIb):
M3-(L3)b-D22 under conditions sufficient to form a compound of Formula (IIIc):
M 1-`L 1)a-L2-(L3)b 22 RI
A22 (IIIc) wherein R1 is a substantially non-antigenic water-soluble polymer;
A21 is a capping group or M1-(L,)a-L2-M2 ;
A22 is a capping group or M1-(I-1)a~-2-(L3)b-D22 =

M1 is a functional group;
D22 is a biologically active moiety;
M2 is OH or a leaving group;
M3 is OH, NH2, or SH;
Lz is a permanent linker or a releasable linker;
L2 is a niultifunctional linker;
L3 is a permanent linker or a releasable linker; and (a) and (b) are independently zero or a positive integer.
The methods further include reacting the compound of Formula (IIIc):

M1-(L9 )a-L{2-(L3)b-D22 Rl A22 (IIIc) with a nucleophilic moiety-containing moiety having Formula (IIId) D21-M4 (IIId) under conditions sufficient to orm a compound of Formula (IIIe):

D29-(L1)a-L2-(L3)b-D22 A23 (IIIe) wherein:
A23 is a capping group or D2,-(L1)a-L2-(L3)h-p22 , M, is a functional group;
D21 is a targeting moiety; and M4 is OH, NH2, or SH.

The attachment of the nucleophilic compound such as Formula (IIfb) to the PEG
or other polymer can be done using standard chemical synthetic techniques well known to those of ordin.ary skill. The activated polyinex portion such as SC-PEG, PEG-amine, PEG acids, etc.
can be obtained from either commercial sources or synthesized by the artisan without undue experimentation.
Attachment of nucleophilic compouzxd such as Formula (IIIb) 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-diisopropylcarbodiirnide (DIPC), any suitable dialkyl carbodiimides, 2-halo-l-alkyl-pyridinium halides, (Mukaiyama reagents), 1-(3-dimethylaminopropyl)-3 -ethyl carbodiimide (EDC), propane phosphonic acid cyclic anhydride (PPACA) and phenyl dichlorophosphates, etc. which are available, for example from commercial sources such as Sigma-Aldrich Chemical, or synthesized using lmown teclin:iques_ Preferably, the reactions are carried out in an inert solvent such as methylene chloride, chloroform, DMF or mixtures thereof. The reactions can be preferably conducted in the presence of a base, such as dimethylaminopyridine (DMAP), diisopropylethylamine, pyridine, triethylamine, etc. to neutralize any acids generated. The reactions can be carried out at a temperature from about 0 C up to about 22 C (room temperature). Some particular embodiments prepared by the methods described herein include:

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 Forznula (T) to a patient in need thereof.
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 Fozanula (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 cancer, glioblastoma, ovarian cancer, gastric cancers, etc. The compositions are useful for treating neoplastic disease, reducing tumor burden, preventing metastasis of neoplasms and preventing recurrences of tumor/neoplastic growths in mamznals.
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 attached to the PEG polymer can be administered to a mannnal 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 prodrag used in the treatment methods is that amount which effectively achieves the desired therapeutic result in mammals. Naturally, the dosages of the various prodrug compounds will vary somewhat depending upon the parent compound, rate of in vivo hydrolysis, molecular weight of the polymer, etc.
Those skilled in the art will determi.za.e the optimal dosing of the polymeric transport conjugates selected based on clinical experience and the treatmefit indication. Actual dosages will be apparent to the artisan without undue experimentation.
The compounds of the present invention can be included in one or more suitable pharmaceutical compositions for administration to mammals. The phaxxnaceutical compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral and/or parenteral routes depending upon the needs of the artisan. A solution and/or suspension of the composition may be utilized, for example, as a carrier vehicle for injection or infiltration of the composition by any art known methods, e.g., by intravenous, intramuscular, intraperitoneal, subcutaneous injection and the like. Such administration may also be by infusion into a body space or cavity, as well as by iiahalation 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 m.ammalian cells.
The methods include delivering an effective amount of a conjugate prepared as described herein to the condition being treated will depend upon the polynucleotides efficacy for such conditions. For example, if the unconjugated oligonucleotides (for example antisense BCL2 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 oligoinucleotzdes. The delivery can be made in vivo as part of a suitable pharmaceutical composition or directly to the cells in an ex vivo environment. In one particular treatment, the polymeric conjugates including oligonucieotides. (SEQ 1D 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 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), DTEA (diisopropylethylamine), DIPC
(diisopropylcarbodiimide), DMAP
(4-dimethylaminopyridine), DMF (N,N'-dimethylformatxa.ide), DSC
(disuccinimidyl carbonate), EDC (1 -(3 -dimethylaminopropyl)-3 -ethyl carbodiimide), EEDQ (2-ethoxy-l-ethoxycarbonyl-1,2-dihydroquinoline), IPA (isopropanol), NHS (N-hydroxysuccinimide), NMM (N-methylmorpholine), PEG (polyethylene glycol), SCA-SH (single-chain antibody), SN3 8 (7-ethyl-l0-hydroxycamptothecin), TBDPS (tert-butyl-dipropylsilyl), and TEA
(triethylamin.e).
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 13C NMR spectra at 75.46 MHz using a Varian Mercury300 NMR
spectrometer and deuterated chloroform as the solvents unless otherwise specified. Chemical shifts (S) 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

300SB C8 reversed phase column (150 x 4.6 mm) or a Phenomenex Jupiter 300A

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)male.imide (compound 1).
Methylehloroformate (4.4 mL, 56.7 mmol, 1 eq) was added to a soiution 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 Na2SO4.
After filtration and evaporation under vacuum a pink solid was obtained.
Purification by column chromatography on silica gel (Hexane-EtOAc, 1:1, v/v) provided the product (4.8 g, 55%).

Example 2. N-Maleoyl-a-(Soc)-L-lysine (compound 2).
N-(Methoxycarbonyl)maleimide (315 mg, 2.03 mmol, 1 eq) was added to a solution of Boc-L-lysine (500 mg, 2.03 mmol, 1 eq) in 10 mL of saturated aqueous NaHCO3 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 H2SO4 before extracting with ethyl acetate. The organic layers were combined and dried over anhydrous 1V1gSO4. After removing the solvent izz vacuo, the residue was purified by column chromatography on silica gel using a mixture of CHC13-MeOH (5:1, v/v) to provide the product (458 mg, 69%): 'H NMR S 1.39-1.87 (6 H, m, (CH2)3), 1.44 (9H, s, Boc), 3.52 (2H, t, J- 7.2 Hz, NCH2), 4.25-4.30 (1H, m, CH), 5.15 (1H, d, NH), 6.70 (2H, s, maleimide).
Example 3. N-Maleoyl-ac-L-lysine (compound 3).

A solution of NE-maleoyl-a-(Boc)-L-lysine (172 mg, 0.53 mmol) in 5 mL
anhydrous DCM
was treated with 10 mL of 2N HCl 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 NE-Maleoyl-a-L-lysine: 'H NMR (CD3OD) b 1.36-1.54 (2 H, m, CH2), 1.65 (2 H, q, J = 7.2 Hz, CH2), 1.83-2.02 (2 H, m, CH2), 1.44 (9H; s, Boc), 3.53 (2H, t, J- 6.9 Hz, NCH2), 3.95 (1H, t, 6.2 Hz, CH), 6.82 (2H, s, maleimide); 13C NMR
(CD3OD) S
23.17, 29.03, 31.01, 37.96, 135.26, 172.34, 175.20.

Example 4. $K mPEG-amide-Lys(N-maleoyl)-COOIEI (compound 4).

To a solution of 5KMPEGCOONHS (805 mg, 0.16 mmo1, 1 eq) in 8 mL of anhydrous DCM
was added NE-maleoyl-a-L-lysine (3, 82 mg, 0.31 mmol, 2 eq) in 2 mL of DMF
followed by DIEA (109 L, 0.62 mmol, 4 eq). The reaction znixture was stirred at room temperature overnight, filtered and evaporated under vacuum. The residue was first precipitated with DCMlether and then, recrystallized with CH3CN/IPA. 13C NMR (CDC13): b 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.

Example 5. SKmPEGamide-Lys(NE-maleoyl)~-NHS (compound 5).
"mPEG-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/IPA.

Example 6. sKmPEG-carbamate-Lys(NE-maleoyl)-COOH (compound 6).
To a solution of 5 mPEGCOONHS (5 g, 0.98 mmol, 1 eq) in 45 mL of anhydrous DCM
was added NE-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 DCMlethyl ether and then, recrystallized with CH3CN/IPA to give compound 6: "C
NMR S
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. 5KnPEGcarbamate-Lys(NE-naaleoyl)-NHS (compound 7).

To a solution of 5xmPEG-carbamate-Lys(NE-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 L;
0:76 mmol, 4 eq). The xnixture was stirred at room temperature overnight. The solvents were evaporated under vacuum and the residue was precipitated with DCMlether and then, recrystallized with CH3CN/IPA to give compound 7: "C N114R 622.31, 25.90, 28.25, 32.39, 37.44, 52.42, 59.30, 6492, 69.64-72.18 (PEG), 134.25, 155.59, 167.75, 168.55, 170_91.

Example 8. Compound 8.
To a solution of 5'extGS (5 mg, 0.85 mol) in PBS buffer (2.5 mL, pH 7.8) was added compound 7 (92 mg, 17 mol, 20 eq) and the mixture was stirred at room teraperature for 2 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 rerriove the excess PEG linker_ Then the product was eluted with a gradient of 0 to 100 % 1 M NaCl 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 8 (1 mg oligo equivalent, 40% yield).

Example 9. Compound 9.
To a solution of compound 8 (1 mg oligo equivalent) in PBS buffer (1 mL, pH
7.0) was added peptide cRGDflC (1 mg, 1.7 mol) 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-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 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).
Example 10. Compound 10.
To a solution of p-axnino-benzylalcohol (1 g, 8.12 mmol, I eq) in 80 mL of anhydrous THF
were added DIEA (1.4 mL, 8.12 mmol, I eq) and Boc2O (1.9 mL, 8.12 mmol, 1 eq).
The mixture was heated to reflux overnight, then cooled down and evaporated under vacuum. The residue was dissolved in EtOAc. The organic layer was washed with a 0.1 N HCl solution, dried over MgSO4, filtered and evaporated under vacuum. The crude product was purified by column chromatography on silica gel (Hex-EtOAc, 1:1, v/v) to give 1.85 g of product (quantitative yield): 1H NMR S 0.1.49 (9H, s), 2.17 (1H, s), 4.53 (2H, s), 6_83 (IH, s), 7.19 (2H, d, J= 8.5 Hz), 7.28 (2H, d, J= 8.2 Hz); 13 C NMR b 28.28, 64.54, 80.37, 118.49, 127.59, 135.31, 137.46, 152.72.
Example 11. Compound 11.
To a -20 C solution of compound 10 (220 mg, 0.98 mmol, 1 eq) and DIEA (188 [tL, 1.08 mmol, 1.1 eq) in 8 mL of anhydrous DCM was added dropwise a solution of MsCl (84 L, 1.08 mmol, 1.1 eq) in 2 mL of anhydrous DCM. The mixture was allowed to warm to rt and then stirred for 1 hour. The reaction mixture was washed with a saturated NaHCO3 solution, a saturated NH4C1 solution and brine, dried over MgSO4, filtered and evaporated under vacuum.
The crude compound was used in the next step without fiYrther purification.

Example 12. Compound 12.
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 K2CO3 (68 mg, 0.49 mmol, 1 eq). The reaction mixture was warmed 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 NH4C1 and brine, dried over MgSO4, filtered and evaporated under vacuum. The crude product was purified by column chromatography on silica gel (CH2C12-EtOAc, 1:1, vlv) to give 140 mg of product (48% yield): 'H NMR (DMSO-d6) S 0.88 (3H, t, J = 7.0 Hz), 1.27 (3H, t, J= 7.3 Hz), 1.47 (9H, s), 1.85-1.88 (2H, m), 3_15-3.17 (2H, m), 5.25 (2H, s), 5.26 (2H, s), 5.42 (2H, s), 6.49 (1H, s), 7.26 (1H, s), 7.41-7.56 (6H, m), 8.05 (1H, d, J = 9.1 Hz), 9.39 (1H, s);13C NMR

(DMSO-d6) 8 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..
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 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.
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 (CHC13:MeOH, 15:1 to 9.1) to give the desired product (8 mg, 9% yield): 13 C NMR (DMSO-d6) S 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.
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 NaHCO3 (2.8g, 33.40 mmol, 1.05 eq) in 80 mL of water. The mixture was stirred at room temperature overnight. Aqueous 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 /a yield):1H NMR (CD3OD) S
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, 7= 6.7 Hz), 3.90 (1H, d, J- 7.0 Hz), 4.37-4.41 (1H, m).13C NMR (CD30D): S 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, 1eq) and p-aminobenzyl alcohol (362 mg, 2.94 mmol, 1.1 eq) in 2:1 DCMIMeOH (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 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%): 'H NMR (CD3OD) S 0_93 (311, d, J= 7.0 Hz), 0.97 (3H, d, J = 7.0 Hz), 1.44 (9H, s), 1.49-2.06 (5H, m), 3.07-3.29 (2H, m), 3.91 (1H, d, J= 6.7 Hz), 4.50-4.55 (3H, m), 7.29 (2H, d, J = 8.5 Hz), 7.54 (2H, d, J = 8.5 Hz), 8.22 (1H, d, J = 7_3 Hz);13C NMR
(CD3OD) b 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-Va1-Cit-PABE-SN38).
To a suspension of Boc-Val-Cit-PAB (214 mg, 0.45 mmol, 1 eq) in 3 mL of CH3CN
was added DIEA (0.23 mL, 1.35 minol, 3 eq) followed by dropwise addition of a solution of MsC1 (0.1 mL, 1.35 mmol, 3 eq) in I mL CH3CN. After 1 hour, TLC showed no starting material left (CHC13-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 NH4C1, saturated NaHCO3 and brine; dried over MgSO4, 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 K2C03 (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.
Example 18. Compound 17.
Compound 14 in DCM is treated with 2N HC1 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 HCI-VaI-Cit-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 CH3CN/IPA to give the product.
Example 19. Compound 18.
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 under vacuum. The residue is precipitated with DCM/ethyl ether and recrystallized with DMF/IPA to give the product.

Example 20. Compound 19.
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 evaporated under vacuum. The residue was first precipitated with DCM/ethyl ether and recrystallized with CH3CN/IPA to give compound 19: 13C NMR S 21.88, 27.97, 29.04, 31.57, 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
HCl solution in dioxane (25 mL). The reaction was stirred at room temperature for 4 hours and 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.
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 ovmight. 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 L, 0.66 mmol, 8 eq). The reaction mixture was stirred at room temperature overnight and evaporated under vacuum. The residue was first precipitated with DCM/ether and then, recrystallized with CH3CN/IPA to give the compound 21 (1.5 g): 13C 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, 12098, 133.94, 142.54, 153.34, 155.51, 155.71, 157.28, 169.98, 173.32.

Example 23. Compound 22.
To a solution of campound 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 ~tL, 1.09 mmol, 16 eq). The mixture was stirred at room temperature overnight. The solvents were evaporated under vacuum and the residue was precipitated with DCMlether and then, recrystallized with CH3CN/IPA to give the product (1.3 g): "C NMR b 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, 157.60, 1.68.18, 169.14, 169.71, 170.56.

Example 24. Compound 23.
(BocPheLys(Fmoc)-OH). To a solution of BocPheOSu (1.5g, 4.14mmol; leq) in DCM
(10.3 mL) and DMF (10.3 mL) was added H-Lys(Fmoc)-OH (1.84g, 4.55 mmol, l.leq}
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 mL x 2). The organic layers were combined, dried over MgSO4 and concentrated 1n vacuo to yield a yellow residue. The crude residue was then chromatographed over silica gel using CHCl3-MeOH (3:1, v/v) to give the product. MS. [M+1]+ 616.

Example 25. Compound 24.
Compound 13 (111 mg, 0.22 mmol, 1 eq) and 16 (208 mg, 0.34 mzxzol, 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 (CHZCl2-EtOAc, 1:1 to 1:2, v/v) to give the desired product (20 mg, 8% yield). MS. [M+1]+ 1095.
Example 26. Compound 25.
Compound 24 (8 eq) is treated with 4N HCl 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 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 ovemight_ The solvent is removed under vacuum and the resulting solid is precipitated with DCM/ethyl ether and recrystallized with DMF/Il'A to give the product.

Example 27. Compound 26.
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 DCMlethyl ether and recrystallized with DMF/IPA to give the product.

Example 28. Compound 27.
To a solution of compound 26 (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 under vacuuzxa.. The residue is precipitated with DCM/ethyl ether and recrystallized with DMF/1PA to give the product.

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 NaHCO3 {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 10% IPA/EtO.A.c (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): 1H NMR
(DMSO-d6) S 0.86 (3H, d, J = 6.7 Hz), 0.90 (3H, d, J= 7.0 Hz), 1.40-1.48 (2H, m), 1.51-1.75 (2H, m), 1.98 (1H, 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), 5.40 (211, brs), 5.96 (1H, t, J = 5.6 Hz), 7.32 (2H, t, J= 7.5 Hz), 7.39-7.44 (3H, m), 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-d6) S 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.

Example 30. FmocVal-Cit-PA.S (Compound 29).
A solution of compound 28 (1 g, 2.01 mmol, 1 eq) and p-aminobenzyl alcohol (273 mg, 2.21 mmol, 1.1 eq) in a mixture of DCM/MeOH (20 mL/10 mL) was treated with EEDQ
(996 mg, 4.03 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 collected by filtration and washed with ethyl ether to yield the product (925 mg, 76% yield): 'H NMR (DMSO-d6) S 0.87 (6H, d, t= 7.5 Hz), 1.36-1.47 (2H, m), 1.51-1.75 (2H, m), 1.99 (1H, sext, J = 6.7 Hz), 2.90-3.04 (311, 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 (1H, t, J= 5.6 Hz), 5.43 (2H, brs), 5.99 (1H, 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, m), 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-d6) 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.

Example 31. Va1-Cit-PAL' (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 ovezvight and then the solvents were evaporated under vacuuzn. The residue was triturated with DCM
and the resulting solid was filtered and washed with DCM to give the product (283 mg, 72% yield):
1H NMR (DMSO-d6) fi 0.79 (3H, d, J= 6.7 Hz), 0.89 (3H, d, J = 6.7 Hz), 1-38-1.42 (2H, m), 1.52-1.68 (2H, m), 1.94 (1H, sext, J = 6.3 Hz), 2.89-3.01 (2H, m), 3.05 (1H, d, J 4.7 Hz), 4.46 (211, s), 5.09 (1H, brs), 5_40 (2H, s), 5.98 (1H, t, J- 5.3 Hz), 7.23 (211, 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-d6) 6 16.80, 19.33, 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 mL anhydrous DMF was added compound 30 (175 mg, 0.46 mmol, 8 eq). The reaction mixture was stirred overrnight at room temperature and then the solvent was evaporated under vacuum. The residue was first precipitated with DCM/ether and then, recrystallized with CH3CN/IPA to give the product (1.1 g): 13C NMR (CDCI3/CD34D) 618.48, 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, 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 iiaixture is cooled to 0 C. Then, pyridine (16 eq) is added and the mixture is gradually warmed to room temperature and stirred overnight. The solvent is evaporated under vacuum and the residue is precipitated with DCMlethyl ether and then, recrystallized with CH3CN/IPA to give the product.

Example 34. Compound 33.
To a solution of compound 32 is added doxorubicin. The reaction mixture is stirred at rt overn.ight. 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.
To a solution of compound 33 (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 DCMlethyl ether and recrystallized with DMF/1PA to give the product.

Example 36. Compouu.d 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, recrystallized with CH3CN/IPA to give the product (5.69 g, 86%): "C NN1R
b'21.59, 25.21, 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 To a solution of compound 35 (5.0g, 0.232mmol, leq) in 23 mL each of anhydrous DCM and anhydrous DMF was added HC1.NH2Cys(NPys)-OH (0.77g, 2.78mmol, 12eq) 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 CH3CN/IPA
to give the product (4.88 g, 95%yield): r3C NMR S 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.
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 ovezmight at room telnperature. After 20 hours the reaction mixture was evaporated under vacuum and the residue was first precipitated with DCM/ether and then, recrystallized with CH3CN/IPA to give the product (2.1 g, 49%): 13C NMR S 21.47, 26.41, 31.64, 39.47, 45.25, 52.32, 54.11, 63.75, 69.12-70.65 (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 mucnol, 1.1 eq) and 63 L 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 H2N-Lys(4arm-PEG)CysNPys (37, 3 81 mg, 0.0175mmo1,.1 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 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.
To a solution of C6-thio-LNA-survivin (OligoSH) in pH 6.5 phosphate buffer is added compoun.d 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 eluted with a gradient using buffer system 20 mM Tris. I-IC12M NaCI atpH 7Ø The product is desalted and lyophilized.

Claims (29)

1. A compound of the Formula (I) wherein:
R1 is a substantially non-antigenic water-soluble polymer;
A is a capping group or each D1 is independently selected from the group consisting of targeting moieties, functional groups and leaving groups;
each D2 is independently selected from the group consisting of biologically active moieties, functional groups and leaving groups;
each L1 is an independently selected permanent linker or a releasable linker:
L2 is a multifunctional linker;
each L3 is an independently selected permanent linker or a releasable linker:
and (a) and (b) are independently zero or a positive integer.
2. The compound of claim 1, wherein L2 is selected from the group consisting of:
and wherein R2 and R'2 are independently selected from the group consisting of hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-19 branched alkyl, C3-8 cycloalkyl, C1-6 substituted alkyl, C2-6 substituted alkenyl, C2-6 substituted alkynyl, C3-8 substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, C1-6 heteroalkyl, substituted C1-6 heteroalkyl, C1-6 alkoxy, aryloxy, C1-6 heteroalkoxy, heteroaryloxy, C2-6 alkanoyl, arylcarbonyl, C2-6 alkoxycarbonyl, aryloxycarbonyl, C2-6 alkanoyloxy, arylcarbonyloxy, C2-6 substituted alkanoyl, substituted arylcarbonyl, C2-6 substituted alkanoyloxy, substituted aryloxycarbonyl, C2-6 substituted alkanoyloxy and substituted arylcarbonyloxy;
(c1), (c2), (c3), (c4), (c5), (c6), (c'6), (c"6), (c7) and (c8) are independently zero or a positive integer; and (d1), (d2), (d3), (d4), (d5) and (d7) are independently zero or a positive integer.
3. The compound of claim 2, wherein L2 is selected from the group consisting of
4. A compound of claim 2, selected from the group consisting of:
5. A compound of claim 4, selected from the group consisting of:
6. The compound of claim 1, wherein the biologically active moiety is selected from the group consisting of -NH2 containing moieties, -OH containing moieties and -SH
containing moieties.
7. The compound of claim 1, wherein the biologically active moiety is selected from the group consisting of pharmaceutically active compounds, enzymes, proteins, oligonucleotides, antibodies, monoclonal antibodies, single chain antibodies and peptides:
8. The compound of claim 7, wherein the pharmaceutically active compounds are selected from the group consisting of DNA topoisomerase inhibitors, microtubule inhibiting 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 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 consisting of antisens bc1-2 oligonucleotides, antisense HIF-1a oligonucleotides, and antisense Survivin oligonucleotides.
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 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.
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. The compound of claim 1, wherein the releasable linkers are independently selected from the group consisting of:

-Val-Cit-, -Gly-Phe-Leu-Gly-, -Ala-Leu-Ala-Leu--Phe-Lys-, -Val-Cit-C(=O)-CH2OCH2-C(=O)-, -Val-Cit-C(=O)-CH2SCH2-C(=O)-, and -NHCH(CH3)-C(=O)-NH(CH2)6-C(CH3)2-C(=O)-, wherein Y11-19 are independently O, S or NR 48 R31-48, R50, R100 and A51 are independently selected from the group consisting of hydrogen, C1-6 alkyls, C3-12 branched alkyls, C3-8 cycloalkyls, C1-6 substituted alkyls, C3-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1-6 heteroalkyls, substituted C1-6heteroalkyls, C1-6 alkoxy, C1-6 alkyloxycarbonyl, phenoxy and C1-6heteroalkoxy;

Ar is an aryl or heteroaryl moiety;
L11-15 are independently selected bifunctional spacers;
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;
(c11), (h11), (k11), (l11), (m11) and (n11) are independently selected positive integers;
(a11), (e11), (g11), (j11), (o11) and (q11) are independently either zero or a positive integer; and (b11), (x11), (x'12), (f11), (i11) 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:

17. The compound of claim 1, wherein L1 and L3 are independently selected from the group consisting of an amino acid, an amino acid derivative, and a peptide.
18. The compound of claim 1, wherein R1 comprises a linear, terminally branched or multi-armed polyalkylene oxide.
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 -Y21-(CH2CH2O)n-CH2CH2Y21- , -Y21-(CH2CH2O)n-CH2C(=Y22)-Y21-, -Y21-C(=Y22)-(CH2)a2-Y23-(CH2CH2O)n-CH2CH2-Y23-(CH2)a2-C(=Y22)-Y21- and -Y21-(CR51R52)a2-Y23-(CH2)b2-O-(CH2CH2O)n-(CH2)b2-Y23-(CR51R52)a2-Y21-wherein:
Y21 and Y23 are independently O, S, SO, SO2, NR53 or a bond;
Y22 is O, S, or NR53;
R51-53 are independently selected from the group for R2;
(a2) and (b2) are independently zero or a positive integer; and (n) is an integer from about 10 to about 2300.
21. The compound of claim 18 wherein the polyalkylene oxide is a polyethylene glycol of the formula, -O-(CH2CH20)n-wherein (n) is an integer from about 10 to about 2,300.
22. The compound of claim 1, wherein R1 has an average molecular weight from about 2,000 to about 100,000 daltons.
23. The compound of claim 1, wherein R1 has an average molecular weight of from about 5,000 to about 60,000 daltons.
24. The compound of claim 1, wherein R1 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:

wherein mPEG has the formula: CH3-O(CH2CH2O)n-;
PEG has the formula -O(CH2CH2O)n-;

(n) is a positive integer from about 10 to about 2,300;
R51 and R52 are independently selected from the group consisting of hydrogen, C1-6 alkyls, -C3-12 branched alkyls, C3-8 cycloalkyls, C1-6 substituted alkyls, C3-8 substituted cyloalkyls, aryls, substituted aryls, aralkyls, C1-6 heteroalkyls, substituted C1-6heteroalkyls, C1-6 alkoxy, C1-6 alkyloxycarbonyl, aryloxycarbonyl, phenoxy and C1-6 heteroalkoxy;
D1 is a targeting moiety, a functional group or a leaving group; and D2 is a biologically active moiety, a functional group or a leaving group.

25. A compound of claim 1, selected from the group consisting of:

wherein S-SCA is a single-chain antibody;
RGD is LNA is locked nucleic acids;
Folic acid is a residue of mPEG has the formula: CH3-O(CH2CH2O)n-;
PEG has the formula -O(CH2CH2O)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 comprising:
reacting a compound of Formula (IIIa):

with a biologically active moiety-containing compound having Formula (IIIb):

M3-(L3)b-D22 under conditions sufficient to form a compound of Formula (IIIc):

wherein R1 is a substantially non-antigenic water-soluble polymer;
A21 is a capping group or A22 is a capping group or M1 is a functional group;
D22 is a biologically active moiety;
M2 is OH or a leaving group;
M3 is OH, NH2, or SH;
L1 is a permanent linker or a releasable linker;
L2 is a multifunctional linker;
L3 is a permanent linker or a releasable linker; and (a) and (b) are independently zero or a positive integer.
27. The method of claim 26 further comprising:
reacting the compound of Formula (IIIc):

with a nucleophilic moiety-containing moiety having Formula (IIId) D21-M4 (IIId) under conditions sufficient to form a compound of Formula (IIIe):

wherein:
A23 is a capping group or M1 is a functional group;
D21 is a targeting moiety;
M4 is OH, NH2, or SH; and 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.
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.
CA002662981A 2006-09-15 2007-09-15 Targeted polymeric prodrugs containing multifunctional linkers Abandoned CA2662981A1 (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 (1)

Publication Number Publication Date
CA2662981A1 true CA2662981A1 (en) 2008-03-20

Family

ID=39184644

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002662981A Abandoned CA2662981A1 (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
EP3717016A4 (en) * 2017-11-30 2021-09-01 Seagen Inc. Process for the preparation of drug linker compounds

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2569940T3 (en) 2007-04-20 2016-05-13 Sigma-Tau Rare Disease Ltd Stable recombinant adenosine deaminase
WO2009141823A2 (en) 2008-05-22 2009-11-26 Ramot At Tel Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
CN102105156B (en) * 2008-05-22 2016-05-25 特拉维夫大学拉莫特有限公司 Be connected with therapeutic activity agent and Angiogenesis targeting moiety polymer Novel conjugates and in treatment the purposes in angiogenesis-associated diseases
CA2723263A1 (en) * 2008-05-23 2009-11-26 Enzon Pharmaceuticals, Inc. Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
RU2483081C2 (en) 2008-07-23 2013-05-27 Ханми Сайенс Ко.,Лтд.,Kr Polypeptide complex containing non-peptidyl polymer, having three functional ends
TW201029668A (en) * 2008-11-17 2010-08-16 Enzon Pharmaceuticals Inc Releasable conjugates for nucleic acids delivery systems
JP2012509366A (en) * 2008-11-17 2012-04-19 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 Releasable polymeric lipids for nucleic acid delivery systems
EP2525829A1 (en) * 2010-01-22 2012-11-28 Ascendis Pharma A/S Dipeptide-based prodrug linkers for aromatic amine-containing drugs
US20130018010A1 (en) * 2010-04-16 2013-01-17 Enzon Pharmaceuticals, Inc. Polymeric conjugates of adenine nucleoside analogs
WO2011155501A1 (en) 2010-06-11 2011-12-15 独立行政法人科学技術振興機構 Pharmaceutical multimeric particles, and manufacturing method for same
EP2646818A4 (en) * 2010-11-30 2016-09-21 Univ 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
EP2822598A4 (en) 2012-03-05 2016-04-13 Univ Ramot Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
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
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
ES2759905T3 (en) * 2013-10-04 2020-05-12 Prolynx Llc SN-38 Slow Release Conjugates
KR102645430B1 (en) 2013-10-15 2024-03-11 씨젠 인크. Pegylated drug-linkers for improved ligand-drug conjugate pharmacokinetics
GB201414098D0 (en) 2014-08-08 2014-09-24 Illumina Cambridge Ltd Modified nucleotide linkers
KR20170071485A (en) 2014-10-14 2017-06-23 폴리테릭스 리미티드 Process for the conjugation of a peptide or protein with a reagent comprising a leaving group including a portion of peg
EP3220956B1 (en) * 2014-10-24 2023-08-09 Abzena (UK) Limited Conjugates and conjugating 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
ES2877409T3 (en) * 2015-08-11 2021-11-16 Coherent Biopharma I Ltd Multiligand drug conjugates and their uses
TWI618697B (en) 2015-11-03 2018-03-21 財團法人工業技術研究院 Compounds, linker-drugs and ligand-drug conjugates
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
CA3006000A1 (en) 2015-12-04 2017-06-08 Seattle Genetics, Inc. Conjugates of quaternized tubulysin compounds
KR102626498B1 (en) 2016-03-25 2024-01-19 씨젠 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
ES2859473T3 (en) * 2017-04-21 2021-10-04 Bright Gene Bio Medical Tech Co Ltd Multi-arm Targeted Anticancer 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
WO2019136487A2 (en) 2018-01-08 2019-07-11 Regeneron Pharmaceuticals, Inc. Steroids and antibody-conjugates thereof
AU2019208024A1 (en) 2018-01-12 2020-08-13 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
US20220047713A1 (en) * 2018-04-02 2022-02-17 Jenkem Technology Co., Ltd. (Beijing) Cell-penetrating peptide-multiarm pol yethylene glycol-drug conjugate having targeting property and application thereof
BR112021008012A2 (en) * 2018-10-29 2021-11-03 Mersana Therapeutics Inc Engineered cysteine antibody-drug conjugates with peptide-containing linkers
WO2020125546A1 (en) * 2018-12-17 2020-06-25 荣昌生物制药(烟台)有限公司 Connector for use in antibody medicament conjugate and applications of connector
US11739166B2 (en) 2020-07-02 2023-08-29 Davol Inc. Reactive polysaccharide-based hemostatic agent
US20240115713A1 (en) * 2020-07-28 2024-04-11 Chongqing Upgra Biotechnology Co., Ltd. Polyethylene glycol conjugate drug, and preparation method therefor and use 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
AU2022213743A1 (en) * 2021-01-28 2023-06-15 Nanjing Chempion Biotechnology Co., Ltd. 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
CA2288994C (en) * 1997-04-30 2011-07-05 Enzon, Inc. Polyalkylene oxide-modified single chain polypeptides
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
US6251382B1 (en) * 1998-04-17 2001-06-26 Enzon, Inc. Biodegradable high molecular weight polymeric linkers and their conjugates
US6153655A (en) * 1998-04-17 2000-11-28 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
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
DK1362053T3 (en) * 2001-02-20 2008-03-10 Enzon Inc Terminally branched polymeric linkers and polymeric conjugates containing these
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
NZ542687A (en) * 2003-04-13 2010-02-26 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 (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3717016A4 (en) * 2017-11-30 2021-09-01 Seagen Inc. Process for the preparation of drug linker compounds
US11612666B2 (en) 2017-11-30 2023-03-28 Seagen Inc. Process for the preparation of drug linker compounds

Also Published As

Publication number Publication date
WO2008034124A2 (en) 2008-03-20
JP2010503708A (en) 2010-02-04
CN101541332A (en) 2009-09-23
IL197517A0 (en) 2009-12-24
WO2008034124A3 (en) 2008-08-07
BRPI0716812A2 (en) 2013-11-05
KR20090057383A (en) 2009-06-05
RU2009114154A (en) 2010-10-20
EP2073820A2 (en) 2009-07-01
AU2007296056A1 (en) 2008-03-20
EP2073820A4 (en) 2014-07-16
MX2009002855A (en) 2009-03-30
AU2007296056B2 (en) 2012-09-13

Similar Documents

Publication Publication Date Title
CA2662981A1 (en) Targeted polymeric prodrugs containing multifunctional linkers
US8367065B2 (en) Targeted polymeric prodrugs containing multifunctional linkers
US20090202573A1 (en) Polymeric conjugates containing positively-charged moieties
US20090203706A1 (en) Lysine-based polymeric linkers
CN109316605B (en) Folate receptor binding ligand-drug conjugates
US20090016985A1 (en) Polymeric drug delivery system containing a multi-substituted aromatic moiety
CA2723263A1 (en) Polymeric systems containing intracellular releasable disulfide linker for the delivery of oligonucleotides
KR20100051722A (en) Polymeric linkers containing pyridyl disulfide moieties
US20110230420A1 (en) Releasable conjugates for nucleic acids delivery systems
KR20090055623A (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery
EP1753776B1 (en) Camptothecins conjugated in position 7 to cyclic peptides as cytostatic agents
US8268318B2 (en) Polyalkylene oxides having hindered ester-based biodegradable linkers
US8110559B2 (en) Hindered ester-based biodegradable linkers for oligonucleotide delivery

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued

Effective date: 20150915