WO1995022991A2 - Block copolymers - Google Patents

Block copolymers Download PDF

Info

Publication number
WO1995022991A2
WO1995022991A2 PCT/GB1995/000418 GB9500418W WO9522991A2 WO 1995022991 A2 WO1995022991 A2 WO 1995022991A2 GB 9500418 W GB9500418 W GB 9500418W WO 9522991 A2 WO9522991 A2 WO 9522991A2
Authority
WO
WIPO (PCT)
Prior art keywords
copolymer
chohch
peptide
mmol
glycine
Prior art date
Application number
PCT/GB1995/000418
Other languages
French (fr)
Other versions
WO1995022991A3 (en
Inventor
Eugene Cooper
Stephen Jones
Colin Pouton
Michael Threadgill
Original Assignee
Nycomed Imaging As
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 Nycomed Imaging As filed Critical Nycomed Imaging As
Priority to MX9603680A priority Critical patent/MX9603680A/en
Priority to AU18170/95A priority patent/AU1817095A/en
Priority to EP95909864A priority patent/EP0748226A1/en
Priority to JP7522220A priority patent/JPH09509443A/en
Publication of WO1995022991A2 publication Critical patent/WO1995022991A2/en
Publication of WO1995022991A3 publication Critical patent/WO1995022991A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino 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
    • A61K47/56Medicinal 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • 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
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • 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
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • BLOCK COPOLYMERS This invention relates to linear block copolymers useful in diagnostic imaging, drug delivery, and as drugs, and in particular to such polymers having
  • polypeptide and polyalkylene oxide moieties in the polymer backbone are polypeptide and polyalkylene oxide moieties in the polymer backbone.
  • British Patent 1,469,472 discloses low molecular weight polyethylene oxide immobilized proteins, said to have low immunogenicity.
  • the invention concerns a linear block copolymer comprising single or repeating units of poly (alkylene oxide) (PAG) linked to units of peptide.
  • PAG poly (alkylene oxide)
  • the copolymer can be tailored to produce water-soluble polymers which are stable in the blood circulation but ultimately will be degraded to allow more facile excretion of low molecular weight PAG derivatives in the urine.
  • the present invention provides a linear block copolymer comprising units of an alkylene oxide linked to units of peptide via a linking group comprising a -CH 2 CHOHCH 2 N(R) - moiety, wherein R is a lower alkyl group, (eg. C 1-6 -alkyl), eg a copolymer comprising units of polyalkyleneoxide linked to
  • polypeptide units via a linker group comprising an amine :epoxide conjugation product.
  • copolymers of the invention have a variety of end uses. In particular they may be used as diagnostic agents, eg. image contrast enhancing agents in
  • the peptide units may have chelating agents coupled thereto (eg. to the peptide side chains) such that the resultant chelating moieties may be metallated with metal species useful diagnostically or
  • drug or pro-drug species may be coupled to the peptide side chains, so that the
  • copolymer acts in effect both as a targetting agent and as a reservoir for release of the drug species.
  • the targetting delivery system effected by the copolymers of the invention is of course also especially useful for delivery of metal species useful in diagnostic imaging of body organs or tissues or as cytotoxic agents.
  • the present invention also provides a pharmaceutical
  • composition comprising a copolymer according to the invention together with at least one physiologically acceptable carrier or excipient.
  • compositions of the present invention may include one or more of the polymers of this invention formulated into compositions together with one or more non-toxic physiologically acceptable carriers, adjuvants or vehicles which are collectively referred to herein as carriers, for parenteral injection, for oral
  • the copolymers of the invention may be produced by a particularly elegant and simple condensation of a bisamine reagent with a bisepoxide reagent, the amine: epoxide condensation yielding the linking group moiety -CH 2 CHOHCH 2 N(R) - mentioned above.
  • a bisamine reagent with a bisepoxide reagent
  • the amine: epoxide condensation yielding the linking group moiety -CH 2 CHOHCH 2 N(R) - mentioned above.
  • polymeric such reagents one incorporating a polyalkylene oxide chain and the other a polypeptide chain, the linear block copolymer structure of the compounds of the invention is produced.
  • the invention provides a process for the preparation of a linear block copolymer according to the invention, said process comprising reating a bisepoxide reagent with a bisamine reagent, one of said reagents incorporating said peptide units and the other incorporating said alkylene oxide units.
  • the linking group in the copolymers according to the invention preferably comprises a -CH 2 -CHOH-CH 2 N(CH 3 ) - moiety, particularly preferably attached at the nitrogen end to an alkylene chain (CH 2 ) P (where p is an integer having a value of from 1 to 6) and optionally attached at the carbon end to a phenyleneoxy moiety.
  • the linking group preferably comprises a moiety
  • the alkylene oxide residues are preferably residues of lower alkylene oxides, eg. C 2-6 , preferably C 2-4 and especially preferably ethylene oxide residues. These will generally be chain end derivatised to link up to the amine :epoxide conjugation product component of the linking group, eg. to link the terminal ether oxygens of the polyalkyleneoxide chain to epoxide-reactive amine groups or to amine-reactive epoxide groups.
  • the nature of the chain-end derivatization is not critical and such bifunctional reagents may be represented by the formulae HNR-(PAG)-NRH or with the PAG representing the polyalkyleneoxide group and the
  • the peptide units again generally occur in a polypeptide chain containing a plurality of amino acid residues.
  • Either synthetic or naturally occurring polypeptide units or fragments may be used and these may in and of themselves provide a therapeutic or targetting moiety or alternatively, if desired, further moieties such as drugs, prodrugs or chelating agents may be conjugated to the peptide side chains.
  • the polypeptide chains may be chain end derivatised to link up to the amine:epoxide conjugation product component of the linking group, eg. to link carbonyl carbon or amine nitrogen termini to epoxides-reactive amines or to amine-reactive epoxides.
  • CO(peptide)NH is generally used to indicate a peptide group showing the terminal carbonyl and amine groups outside the brackets.
  • R is a 1-4 carbon alkyl
  • p is from 1 to 6) are preferred.
  • copolymer compounds can be tailored for
  • altering the peptide composition to provide differing blood pool residence time, enzymatic breakdown rates, and tissue distributions.
  • invention preferably has a molecular weight of at least about 5000 and a metal ion useful as a contrast
  • An imaging metal is defined as a metal useful in x-ray imaging (eg. a metal of atomic number 50 or above) or a metal useful in magnetic resonance imaging (preferably a paramagnetic metal and more preferably a lanthanide metal or transition metal) or a metal useful in fluorescence imaging (preferably a lanthanide metal, most preferably europium).
  • the polymeric chelates of this invention provide effective imaging contrast enhancement of the blood pool within the vascular system for remarkably long periods of time.
  • polymeric compounds are provided having a specificity toward accumulation in different tissues, for example, in tumors and the liver.
  • PAG refers to polyalkylene oxide moieties having a single type of repeating unit or differing (non-repeating) units of alkylene oxide, or a mixture thereof in each PAG.
  • Each alkylene oxide unit in the PAG preferably contains from 2 to about 4 carbons, and is linear or branched.
  • Poly(alkylene oxide) units in the polymer may also differ in length and composition from each other.
  • Exemplary PAG moieties include poly (ethylene oxides), poly(propylene oxides) and poly (butylene oxides).
  • Preferred PAG moieties include poly (ethylene oxides), poly(propylene oxides) and random and block copolymers thereof. Poly(ethylene oxide) containing polymers are particularly preferred when it is desired for the final polymer to possess solubility in water. It is also contemplated that the poly(alkylene oxide) moiety can comprise glycerol poly(alkylene oxide) triethers, polyglycidols, linear, block and graft copolymers of alkylene oxides with compatible comonomers such as poly(proplene oxide-coethylene oxide), or poly(butylene oxide-co-ethylene oxide) and grafted block copolymers. These moieties can be derived from poly (alkylene oxide) moieties which are commercially available or
  • polyalkyleneoxide moieties derived from poly (ethylene oxide) can be represented by the structure:
  • PAG moieties and their reactive derivatives useful in preparing the polymer of the invention, are known in the art.
  • bis (methyl amino) polyethylene glycol and its use as an intermediate in the preparation of block copolymers is known in the art.
  • PAG derivatives may be prepared by known chemical techniques, examples of which are described hereinbelow.
  • peptide refers to an amino acid chain of at least 2 amino acids, wherein each of the amino acids in the peptide may or may not be the same, and may or may not be selected from the 20 naturally occurring L-amino acids.
  • peptide units may contain D-amino acids, artificial amino acids or amino acid derivatives, such as glutamate esters, lysyKe- amino) amides and the like.
  • This definition also includes proteins, enzymes, polypeptides and
  • oligopeptides which are art recognized amino acid chains.
  • preferred peptides include small enzymes less than 100 kD), peptide hormones, peptide recognition domains, peptide drugs, and peptides with known enzymatic breakdown rates.
  • Boc refers to the t-butoxy carbonyl radical commonly used as a blocking agent in solid phase peptide synthesis.
  • Conventional three letter abbreviations for amino acid residues are used throughout the specification.
  • OPFP refers to
  • Bn refers to benzyl
  • CBZ refers to phenylmethoxycarbonyl
  • OTCP refers to 2,4,5-trichlorophenyloxy
  • Troc refers to 2,2,2-trichloroethoxycarbonyl.
  • Copolymerization can occur by reaction of
  • the monomer units of PAG and peptide can be prepared as either bis(oxiranyl)
  • the polymer of the invention has between its PAG and peptide subunits , a linking group.
  • the linking group contains a -CH 2 CHOHCH 2 N(R)- diradical, typically derived from the reaction of an amine and an epoxide. It is preferred that a bisepoxide subunit be reacted with a bisamine subunit.
  • the skilled artisan will appreciate that the recitation used throughout the specification of each type of linking group diradical can be reversed and have the same meaning. Thus the sense of the linking group can be reversed (end for end), with one terminus attached to the PAG moiety, and the other terminus attached to the peptide or vise versa, while its recitation in the specification and the claims is the same.
  • Peptides used to prepare the copolymers of the invention can be prepared by standard procedures known in the art.
  • Useful peptides include those derived from native or recombinant organisms, solid phase peptide synthesis or traditional wet chemistry peptide
  • Protein expression and purification from natural and recombinant sources is in the prior art (cf. Protein Expression and Purification (1990); Harris et al., Protein Purification Methods
  • Linear peptide fragments can be tailored such that they are stable in blood, but are susceptible to lysosomal degradation by commonly occurring proteases.
  • susceptible peptide units are gly-phe-leu-gly, gly-phe-tyr-ala, ala-gly-val-phe, gly-phe-ala-gly, and others known in the art.
  • the prior art describes such olig ⁇ peptides as useful in preparing prodrugs, when the drug is attached to one terminus of the oligopeptide. (See generally "Polymers Containing Enzymatically Degradable Bonds" Makromol. Chem. 184
  • the peptide portion of the polymer can be tailored to recognize (or target) certain cells or functions of cells. Because the polymer can use more than one peptide and thus more than one type of peptide, the polymer can advantageously target more than one type of cell or tissue at once. Judicious choice of peptide allows treatment or targeting of more than one type of cancer cell, for example, or other disease state. This choice is facilitated by the prior art which contains a myriad of known oligopeptides which are antigenic to certain cells. Furthermore, the invention allows such targeting without the cost of raising antibodies to certain cells, harvesting such antibodies, conjugating antibodies to drug and further testing for maintained specificity after conjugation. The invention allows specific targeting to be achieved by short recognition sequences. Cell specific delivery can be achieved by incorporating targeting agents into the polymer.
  • Preferred peptides are those which have a receptor molecule specific to a ligand of interest.
  • a specific binding reaction involving the reagent can be used for the targeting expected.
  • the peptides can be selected from a wide variety of naturally occurring or synthetically prepared materials, including, for example enzymes, proteins, peptide hormones, virus coats, or proteins derived from blood components, tissue and organ components, including haptens,
  • these targeting peptides include: the integrin binding motif RGDS (arg-gly-asp-ser), which is present on many extracellular matrix proteins and can be used to interfere with cell adhesions involved in migration of leukocytes.
  • Other peptide sequences which can be used to deliver the polymer include cationic sequences (ie. rich in lys or arg) which are useful in producing a DNA-binding polymer for use in supression of gene expression, antisense oligomer delivery and the like; peptide hormones such as ⁇ MSH which can be used for targeting to melanoma; and relatively low molecular weight (15-2OkDa) engineered hypervariable antibody binding domains (V H +V L constructs) raised against any target.
  • Such sequences are obtained by synthesis, isolation from cells or bacteriophages or they can also be raised against cells, proteins, or foreign
  • recognition sequences include rabbits, goats, mice, and the like. These and other methods of obtaining
  • the above-described peptide can be an immunoreactive group, which would be found in a living organism or which finds utility in the diagnosis, treatment or genetic engineering of cellular material of living organisms.
  • the peptide has a capacity for interaction with another component which may be found in biological fluids, cells or associated with cells to be treated or imaged, such as, for example tumor cells and the like.
  • Preferred immunoreactive groups therefore include antibodies, or immunoreactive fragments thereof, to tumor-associated antigens.
  • B72.3 antibodies (described in U.S. Patents Nos. 4,522,918 and 4,612,282) which recognize colorectal tumors, 9.2.27 antimelanoma antibodies, D612 antibodies which recognize colorectal tumors, UJ13A antibodies which recognize small cell lung carcinomas, NRLU-10 antibodies which recognize small cell lung carcinomas and colorectal tumors (Pan- carcinoma), 7E11C5 antibodies which recognize prostate-tumors, CC49 antibodies which recognize colorectal tumors, TNT antibodies which recognize necrotic tissue, PR1A3 antibodies, which recognize colon carcinoma, ING-1 antibodies, which are known in the art and are described in International Patent Publication WO-A-90/02569, B174 antibodies which recognize squamous cell carcinomas, B43 antibodies which are reactive with certain lymphomas and leukemias and any other antibody which may be of particular interest.
  • the peptides of the polymer are linear, they can provide functional groups for coupling of diagnostic agents, drugs, or prodrugs or other
  • Functional groups can also be added by reacting or derivatizing functionalizable basic groups (found for example in lysyl or argininyl residues) or acidic groups (as found in aspartate, glutamate, providing free carboxyl groups), or sulfhydryl groups, (e.g. cysteine), hydroxyl groups (such as found in serine) and the like. This coupling is done by reacting or derivatizing functionalizable basic groups (found for example in lysyl or argininyl residues) or acidic groups (as found in aspartate, glutamate, providing free carboxyl groups), or sulfhydryl groups, (e.g. cysteine), hydroxyl groups (such as found in serine) and the like. This coupling is done by reacting or derivatizing functionalizable basic groups (found for example in lysyl or argininyl residues) or acidic groups (as found in aspartate, glutamate, providing
  • Cytotoxic drugs can also be coupled to the polymer to produce prodrugs which are released as a drug to targeted cells or tissues. Such coupling methods are known in the art, see for example; Duncan, P.
  • Drugs contemplated to be useful include any drug which can be covalently attached to the polymer and retains its activity when so attached. It is contemplated that drugs which become active only when liberated from the polymer are also useful, and as such are prodrugs.
  • Drugs which are contemplated to be useful in the polymer include cytotoxic agents, and immunomodulating peptides and proteins as described above.
  • cytotoxic agent any agent able to kill cells, including, chemotherapeutic agents such as cytotoxic drugs and cytotoxic antibiotics, chelated radionuclides and toxins or any agent which initiates or which leads to cell death.
  • chemotherapeutic agents such as cytotoxic drugs and cytotoxic antibiotics, chelated radionuclides and toxins or any agent which initiates or which leads to cell death.
  • cytotoxic agents also includes agents which activate a host's immune response leading to cell death.
  • the cytotoxic agent will be selected with reference to factors, such as the type of disease state, for example the type of cancer tumor and the efficacy of a certain chemotherapy agent for treating the cancer tumor involved, and the like.
  • the cytotoxic agent may be selected from
  • alkylating agents alkylating agents, antimetabolites, natural products useful as cytotoxic drugs, hormones and antagonists and other types of cytotoxic compounds.
  • alkylating agents include the nitrogen mustards (i.e. the 2-chloroethylamines) such as, for example, chloromethine, chlorambucil, melphalan, uramustine, mannomustine, extramustine phosphate, mechlor-thaminoxide, cyclophosphamide, ifosamide and trifosfamide; alkylating agents having a substituted aziridine group such as, for example, tretamine, thiotepa, triaziquone and mitomycin; alkylating agents of the alkyl sulfonate type, such as, for example, busulfan and piposulfan; alkylating N-alkyl-N-nitrosourea derivatives such as, for example,
  • carmustine, lomustine, semustine or streptozotocne alkylating agents of the mitobronitole, decarbazine and procarbazine type
  • platinum complexes such as for example, cisplatin and carboplatin and others.
  • antimetabolites include folic acid
  • derivatives such as, for example, methotrexate, aminopterin and 3'-dichloromethotrexate; pyrimidine derivatives such as, for example, 5-fluorouracil, floxuridine, tegafur, cytarabine, idoxuridine, and flucytosine; purine derivatives such as, for example, mercaptopurine, thioguanine, azathioprine, tiamiprine, vidarabine, pentostatin and puromycin and others.
  • pyrimidine derivatives such as, for example, 5-fluorouracil, floxuridine, tegafur, cytarabine, idoxuridine, and flucytosine
  • purine derivatives such as, for example, mercaptopurine, thioguanine, azathioprine, tiamiprine, vidarabine, pentostatin and puromycin and others.
  • cytotoxic agents examples include for example vinca alkaloids, such as vinblastine and vincristine; epipodophylotoxins such as, for example, etoposide, and teniposide; antibiotics such as, for example, adrimycin, daunomycin,
  • dactinomycin dactinomycin, daunorubicin, doxorubicin, mithramycin, bleomycin and mitomycin
  • enzymes such as, for example, L-asparaginase
  • biological response modifiers such as, for example, alpha-interferon; camptothecin; taxol
  • retinoids such as retinoic acid and the like.
  • hormones and antagonists include adrenocorticoids, such as, for example, prednisone;
  • progestins such as, for example, hydroxyprogesterone acetate, medroxyprogesterone acetate and megestrol acetate; estrogens such as, for example,
  • diethylstilbestrol and ethinyl estradiol diethylstilbestrol and ethinyl estradiol
  • antiestrogens such as for example, tamoxifen
  • androgens such as, for example, testosterone propionate and fluoxymestrone
  • antiandrogens such as, for example, flutamide
  • gonadotropinreleasing hormone analogs such as, for example, leuprolide.
  • miscellaneous cytotoxic agents include anthracenediones such as for example, mitoxantrone;
  • the cytotoxic agent can be ionically associated with the chelating residue.
  • the cytotoxic agent is a radionuclide comprising a radioactive metal ion such as described below associated with a peptide-linked cheating residue.
  • the polymer of the invention can contain one or more of a wide variety of chelating agents.
  • a chelating agent is a compound containing donor atoms that can combine by coordinate bonding with a cation to form a cyclic structure called a chelation complex or chelate.
  • Chelating residues may also be attached via the functionalizable side chains of the peptide via known chemistry. These chelating residues can be coupled to the polymer to produce contrast agents useful in diagnostic imaging or cytotoxic agents when complexed with the appropriate metal .
  • the chelating residue is attached to an available amino acid side chain in the peptide portion of the polymer by a protein reacting group.
  • protein reactive group it is meant any group which can react with any functional groups typically found in proteins, especially an amino acid side chain.
  • Preferred protein reactive groups can be selected from but are not limited to:
  • a group that will react directly with the amine or sulfhydryl groups on an amino acid side chain For example, active halogen containing groups
  • chloromethylphenyl groups and chloroacetyl [Cl-CH 2 CO-] groups activated 2-leaving- group-substituted ethylsulfonyl and ethylcarbonyl groups such as 2-chloroethylsulfonyl and 2-chloroethylcarbonyl; vinylsulfonyl; vinylcarbonyl;
  • epoxy epoxy; isocyanato; isothiocyanato; aldehyde; aziridine; succinimidoxycarbonyl; activated acyl groups such as carboxylic acid halides; mixed anhydrides and the like; and other groups known to be useful in attaching molecules to proteins or crosslinking proteins and the like.
  • the alkyl portions of the protein reactive group can contain from 1 to about 18 carbon atoms or be a lower alkyl group as described for R above.
  • the aryl portions of the protein reactive group can contain from about 6 to about 20 carbon atoms.
  • crosslinking agent A group that can be linked to the amino acid side chain or similar biological molecule, or to the modified peptide as noted in (1) and (2) above by use of a crosslinking agent.
  • Certain useful crosslinking agents such as, for example, difunctional gelatin hardeners, bisisocyanates etc., which become a part of a linking group in the polymer during the crosslinking reaction.
  • Other useful crosslinking agents such as, for example, consumable catalysts, are not present in the final conjugate. Examples of such crosslinking agents are carbodiimide and carbamoylonium crosslinking agents as disclosed in U.S. Patent 4,421,847 and the dication ethers of U.S. Patent 4,877,724.
  • crosslinking agents one of the reactants must have a carboxyl group and the other an amine, alcohol, or sulfhydryl group.
  • the crosslinking agent first reacts selectively with the carboxyl group, then is cleaved during reaction of the "activated" carboxyl group with, for example, an amine to form an amide linkage between the peptide portion of the polymer and metal complexing agents, thus covalently bonding the two moieties.
  • An advantage of this approach is that crosslinking of like molecules, e.g., amino acid side chains with imino acid side chains or complexing agents with complexing agents is avoided, whereas the reaction of difunctional crosslinking agents is less selective.
  • Especially preferred protein reactive groups include amino and isothiocyanato.
  • Preferred chelating agent precursors have anhydride, sulfonylylchloride, alkylsulfate, vinyl sulfate, vinylsulfate, or ester functionalkyl.
  • the chelating residues can be derived from
  • chelating moieties which are selected to contain electron donating atoms which will chelate a metal, by forming coordination bonds therewith.
  • These moieties can be selected from polyphosphates, such as sodium tripolyphosphate and hexametaphosphoric acid;
  • linear, branched or cyclic aminocarboxylic acids such as ethylenediaminetetra-acetic acid, N-(2- hydroxyethyl) ethylenediaminetriacetic acid,
  • nitrilotriacetic acid N,N-di (2-hydroxyethyl)glycine, ethylenebis (hydroxyphenylglycine), diethylenetriamine pentaacetic acid and the N-carboxymethylated
  • 1, 3-diketones such as acetylacetone
  • hydroxycarboxylic acids such as tartaric acid, citric acid, gluconic acid, and 5-sulfosalicylic acid
  • polyamines such as ethylenediamine
  • aminoalcohols such as triethanolamine and N-(2-hydroxyethyl)ethylenediamine
  • aromatic heterocyclic bases such as 2,2'-dipyridyl, 2,2'-diimidazole, dipicoline amino and 1,10-phenanthroline;
  • phenols such as salicylaldehyde
  • aminophenols such as 8-hydroxyquinoline and oxinesulfonic acid
  • oximes such as dimethylglyoxime and
  • tetrapyrroles such as tetraphenylporphin and phthalocyanine
  • sulfur compounds such as toluenedithiol, meso- 2,3-dimercaptosuccinic acid, dimercaptopropanol, thioglycolic acid , potassium ethyl xanthate, sodium diethyldithiocarbamate, dithizone, diethyl
  • ethylenediaminetetra methylenephosphonic acid
  • hydroxyethylidenediphosphonic acid or combinations of two or more of the above agents.
  • Preferred chelating residues contain polycarboxlic acid or carboxylate groups and include elements present in: ethylenediamine-N,N,N',N'-tetraacetic acid (EDTA);
  • EDTA ethylenediamine-N,N,N',N'-tetraacetic acid
  • DTPA N,N,N',N",N"-diethylenetriaminepentaacetic acid
  • 1,4,7,10-tetraazacyclododecane-N,N',N",N"'tetraacetic acid (DOTA); 1,4,7,10-tetraazacyclododecane-N,N',N"-triacetic acid (D03A); 1-oxa-4,7,10-triazacyclo-dodecane-N,N',N"-triacetic acid (OTTA); trans(1,2)-cyclohexanodiethylenetriamine pentaacetic acid (CDTPA);
  • Such chelating compounds including their
  • the chelating residue is made of multiple chelating moieties or subunits, such subunits can be linked together by a linking group. Thus, more than one chelating moiety can be used to make up the
  • chelating residue If more than one chelating moiety is present in the chelating residue, these may be the same or different. Chelating moieties can be linked together using known chemistries. Thus the chelating residue can be one moiety or a "core" of chelating moieties. For example, a core of DTPA residues may be prepared by reacting DTPA dianhydride with a diamine, such as ethylene diamine, to form a "core" of DTPA chelators. Other chelating residues, made up of multiple chelating moieties are well known in the art and are prepared by known chemistries as well.
  • the chelated metal ion M( +a ) preferably represents a
  • paramagnetic metal ion such as an ion of metals of atomic number 21 to 29, 42, 44 and 57 to 71, especially 57 to 71.
  • Ions of the following metals are preferred: Cr, V, Mn, Fe, Co, Ni, Cu, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm and Yb.
  • Cr, V, Mn, Fe, Co, Ni, Cu, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm and Yb are especially preferred.
  • the cytotoxic agent can be a radioactive isotope, preferably a radioactive metal ion isotope.
  • This radioactive metal isotope can be an ion of an isotope of a metal selected, for example, from Sc, Fe, Pb, Ga, Y, Bi, Mn, Cu, Cr, Zn, Ge, Mo, Tc, Ru, In, Sn, Re, Sr, Sm, Lu, Du, Sb, W, Re, Po, Ta and Tl ions.
  • radioisotopes which are also useful in diagnostic imaging applications are
  • radioactive metal ions for this embodiment include 44 Sc, 64 , 67 Cu, 111 In, 212 Pb, 68 Ga, 90 y, 153 Sm, 212 Bi, 99m Tc and 188 Re for this embodiment.
  • the metal content in the polymer can vary from about 0.1 up to about 20% based on the total weight of the polymer.
  • the polymer preferably contains a paramagnetic metal ion in an amount of from 1 to 25%, more preferably 2-20% by weight.
  • the metal content in the polymer can vary from about 0.1 up to about 20% based on the total weight of the polymer.
  • the polymer preferably contains a paramagnetic metal ion in an amount of from 1 to 25%, more preferably 2-20% by weight.
  • the metal content in the polymer can vary from about 0.1 up to about 20% based on the total weight of the polymer.
  • the polymer preferably contains a paramagnetic metal ion in an amount of from 1 to 25%, more preferably 2-20% by weight.
  • radionuclide metal ion is present in roughly the same amounts as for imaging.
  • the PAG moiety in this composition can be capped at the terminus with a capping moiety selected from a hydrogen, hydroxy, alkyl, amino, or alkoxy.
  • Preferred capping groups are hydrogen or hydroxyl groups.
  • capping is done by known chemistry, and precapped prepolymers are available. It is further contemplated that cyclic copolymers can be prepared.
  • the copolymers of this invention can be prepared in water-soluble, water dispersible or water-insoluble forms depending upon the intended application.
  • the copolymer can have a molecular weight ranging from 10,000 to 1 million preferably 11,000 to 80,000. The preferred molecular weight varies according to the application as described below.
  • the polymer can be selectively delivered to specific cells, tissue types, or organs with or without the aid of a targeting agent.
  • a targeting agent such targeted delivery is based on size (hydrodynamic radius) and charge alone.
  • the charge of the polymer can be altered by judicious choice of the aminoacids used in the peptide component of the copolymer to suit the application.
  • the size of the polymer can be chosen by altering the size of PAG or peptide used to prepare the polymer or by altering the degree of polymerization.
  • the size and charge of the polymer in the tissue is a function of the size and charge of the polymer administered.
  • Distribution of the unmetabolized polymer to tissues will be influenced by the nature of the local vascular endothelium in each tissue and the prescence or absence of a lymphatic system.
  • Three general categories of vascular endothelium in each tissue will be influenced by the nature of the local vascular endothelium in each tissue and the prescence or absence of a lymphatic system.
  • endothelium are sinusoidal epithelium, characterized by discontinuity and little or no basement membrane;
  • vascular endothelium fenestrated vascular endothelium
  • continuous vascular endothelium characterized by tight junctions and basement membrane.
  • the lymphatic system is known to recirculate proteins and other molecules which can float freely in the plasma, but escape the circulatory system, exist for a time in tissue and then are
  • the skilled artisan can determine which tissues will be passively targeted by the polymer by approximating the molecular weight or more preferably the hydrodynamic radius of known proteins diffusing through the tissue in a known given period.
  • Tissues such as bone marrow, liver and spleen tissue are characterized by sinusoidal endothelium, (which allows escape of large molecules from the circulating system into the surrounding tissue) thus larger polymer molecules are useful in passively targeting such tissues.
  • Tissues such as those found in the GI tract, kidney glomeruli, and endocrine gland tissue are characterized by fenestrated endothelium
  • Tissues such as muscle and lung tissue are characterized by continuous vascular endothelium (which allows small molecules to escape from the circulatory system into the surrounding tissue), thus smaller polymer molecules are useful in passively targeting these tissues.
  • the hydrodynamic radius of albumin is approximately 37 A, its molecular weight is 67 Kd, and its charge is known. It is known that the average half life for albumin circulation through tissue is
  • the polymer finds utility in imaging and/or treating such inflamed tissues or inflamed tissue sites.
  • a lymphatic system in a tissue will perturb the concentration and increase the half life of macromolecules in a tissue because no convenient mechanism is provided for the scavenging of such macromolecules. Such is the case in growing tumors.
  • cytotoxic agent will result in accumulation of such agent in the growing surface of the tumor.
  • molecular weight and charge of the polymer may be tailored to the specific application based on tissue type, presence or absence of inflammation, tumor and/or vasculature type and presence or absence of a lymphatic system to provide a polymer with the correct characteristics for targeting the desired tissue.
  • acylation of hydroxy- or amino-substituted species to prepare the corresponding esters or amides, respectively; simple aromatic and heterocyclic substitutions or displacements; cleavage of alkyl or benzyl ethers to produce the corresponding alcohols or phenols; and hydrolysis of esters or amides to produce the corresponding acids, alcohols or amines, preparation of anhydrides, acid halides, aldehydes, simple aromatic alkylation and the like as desired can be carried aromatic out.
  • chelating agents and precursors thereof containing reactive functionality including, for example,
  • polycarboxylic acids in dianhydride form, di(sulfonyl chlorides), di(alkyl sulfates), di(vinyl sulfones), diesters, and the like.
  • Such known transformations are also useful in attaching the chelator to the polymer or polymer precursor, and in preparing the polymer itself.
  • obtaining the desired product by some reactions will be better facilitated by blocking or rendering certain functional groups inert. This practice is well recognized in the art, see for example, Theodora
  • the chelating residue containing reactive functionality can be prevented from reacting to form undesired products by suitably blocking the chelating residue precursor which can be contacted with the reactive poly (alkylene oxide) moiety to form the polymer, and then the blocking group can be subsequently removed by techniques known in the art.
  • hydroxy substituents are to be selectively present in the final polymer, they preferably should be temporarily blocked during polymerization, such as by formation of an alkyl ether from the hydroxyl by conventional blocking techniques to minimize formation of undesirable by products.
  • products which contain one or more linkages formed by unblocked reactive precursor groups in the backbone of the polymer are contemplated to be useful.
  • Small proteins or peptides may be incorporated into the polymer by methods as described hereinbelow.
  • An advantage of this chemistry is that the N and C terminus of the peptide can be reversed or randomized in the polymer of the invention, reducing
  • Apep Bis-(oxiranyl)-peptide monomers
  • Apag bis-(alkylamino)-PAG derivative monomers
  • a linking group precursor is added to the PAG monomers at the terminal hydroxy.
  • the reaction of the known linking group precursor with the known PAG moiety forms a (PAG)-linking group precursor radical.
  • the precursor radical is conveniently chosen from
  • aminoalkylamino N-sarcosyl-aminoalkyl-amino, or N-sarcosylaminoalkylamino-N'-carboxy.
  • the peptide monomers conveniently have 4-(oxiranylmethoxy) aryl radicals connected as linking group precursors using carboxy funtionality to attach to the N terminus of the peptide or amino functionality to attach to the C terminus of the peptide, thus forming amide bonds with the N terminus with the C terminus of the peptide monomer with the one end of each linking group precursor, and having an oxirane at the other end of each linking group
  • Apep This oxirane functionalized peptide is referred to as Apep.
  • Apep can be;
  • oxiranyl functionality can be used on PAG derivative monomers while using amino
  • peptide derivative monomers functionality on peptide derivative monomers.
  • Bpep bis(alkylamino)-peptide monomers
  • Bpag bis-(oxiranyl)-PAG monomers
  • the peptide has a linking group precursor radical attached to the C and N termini so as to provide terminal amine functionality.
  • Glycine or sarcosine can be used as the linking group precursor for the N terminus.
  • the C terminus is attached to a -NH(CH 2 ) p NHCOCH 2 NH(R) or
  • linking group precursor radical which is derived from a diamine (wherein p is one to six, R is an alkyl radical, linear or branched, of 1 to about 4 carbons) and glycine or sarcosine.
  • R is an alkyl radical, linear or branched, of 1 to about 4 carbons
  • glycine or sarcosine is attached to the linking group precursor via amide linkages at both the N and C termini.
  • Bpep An example of Bpep is:
  • alkylamine and bis (oxiranyl) functionality may be on either the PAG moiety or the peptide moiety; so long as the polymerization takes place between a peptide and PAG, using the reaction of an amine and an epoxide.
  • suitable chelating agents and precursors thereof may be attached to the polymer or polymer precursor.
  • reactive functionality can be contacted with the reactive amino acid side chain incorporated into the polymer or polymer precursor to form the chelate-polymer or chelate polymer precursor, and then any blocking groups can be subsequently removed by
  • the metallized polymer can be formed by contacting the unmetallized polymer sequentially or simultaneously with one or more sources of metal ions. This can be conveniently accomplished by adding one or more metal ion solutions or one or more metal ion solid salts or metal ion oxides, preferably sequentially, to a
  • the chelated polymer preferably is
  • the copolymer preferably is prepared in a water soluble, for example, an injectable form when used as magnetic resonance contrast agent for blood pool imaging, as a composition intended to be administered intravenously, and the like.
  • a water soluble for example, an injectable form when used as magnetic resonance contrast agent for blood pool imaging, as a composition intended to be administered intravenously, and the like.
  • the preparation of water-soluble compounds of molecular weight 10,000 to 50,000 can be accomplished by known methods by one skilled in the art.
  • copolymer carries an overall charge
  • it will conveniently be used in the form of a salt with a physiologically acceptable counterion, for example an ammonium, substituted ammonium, alkali metal or
  • alkaline earth metal eg. calcium
  • meglumine salts are particularly preferred.
  • the copolymer may be formulated with conventional pharmaceutical or veterinary formulation aids, for example stabilizers, antioxidants, osmolality adjusting agents, buffers, pH adjusting agents, etc. and may be in a form suitable for parenteral or enteral administration, for example injection or infusion or administration directly or after dispersion or dilution with a physiologically tolerable medium, eg. water for injections into a body cavity having an external escape duct, for example the gastrointestinal tract, the bladder or the uterus.
  • a physiologically tolerable medium eg. water for injections into a body cavity having an external escape duct, for example the gastrointestinal tract, the bladder or the uterus.
  • compositions of the present invention may be in conventional pharmaceutical administration forms such as powders, solutions, suspensions, dispersions, etc; however, solutions, suspensions and dispersions in physiologically acceptable carrier media, for example water for injections, will generally be preferred.
  • copolymers according to the invention may therefore be formulated for administration using physiologically acceptable carriers or excipients in a manner fully within the skill of the art.
  • the copolymers optionally with the addition of
  • Suitable additives include, for example, physiologically biocompatible buffers (as for example, tromethamine hydrochloride), additions (eg. 0.01 to 10 mole percent) of chelants (such as, for example, DTPA) or calcium chelate complexes (as for example calcium DTPA, CaNaDTPA-bisamide, or calcium salts) or,
  • additions eg., 1 to 50 mole percent of calcium of sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate and the like).
  • calcium of sodium salts for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate and the like.
  • copolymers are to be formulated in
  • suspension form eg., in water or physiological saline for oral administration, a small amount of soluble chelate may be mixed with one or more of the inactive ingredients traditionally present in oral solutions and/or surfactants and/or aromatics for flavouring.
  • parentral eg., intravenous administration
  • parenterally administrable forms eg., intravenous solutions
  • Suitable vehicles include aqueous vehicles customarily used for administering parenteral solutions such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection and other solutions such as are described in Remington's Pharmaceutical Sciences, 15th ed., Easton: Mack
  • the solutions can contain preservatives, antimicrobial agents, buffers and antioxidants conventionally used for parenteral solutions, excipients and other
  • the copolymer comprises a chelated toxic metal species, eg. a heavy metal ion, it may be
  • a slight excess of a chelating agent eg. as discussed by
  • compositions of the present invention may be varied so as to obtain an amount of active ingredient that is effective to obtain the desired effect for a particular composition and method of administration.
  • the selected dosage level therefore depends upon the desired effect, on the route of administration, on the desired duration of treatment and other commonly considered factors.
  • the dosages of the contrast agent used according to the method of the present invention will vary according to the precise nature of the contrast agent used. Preferably however, the dosage should be kept as low as is consistent with achieving contrast enhanced imaging and volumes minimized for IV drip or bolus injection. In this way, the toxicity potential is minimized.
  • the diagnostic agent of the present invention if in solution, suspension or dispersion form, will generally contain the chelated metal at concentration in the range 1 micromole to 1.5 mole per litre, preferably 0.1 to 700mM.
  • the composition may however be supplied in a more concentrated form for dilution prior to
  • the appropriate dosage will generally range from 0.02 to 3 mmol paramagnetic metal/kg body weight, especially 0.05 to 1.5 mmol/kg, particularly 0.08 to 0.5, more especially 0.1 to 0.4 mmol/kg. It is well within the skill of the average practitioner in this field to determine the optimum dosage for any particular contrast agent for both in vivo or in vitro applications.
  • the dose of the contrast agent should generally be higher and for scintigraphic examination the dose should generally be lower than for MR examination.
  • conventional or sub conventional dosages may be used.
  • the total daily dose of the compounds of this invention administered to a host in single or divided dose may be in amounts, for example, of from about 1 picomol to about 10 millimoles of cytotoxic agent per kilogram of body weight.
  • Dosage unit compositions may contain such amounts or such submultiples thereof as may be used to make up the daily dose. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the body weight, general health, sex, diet, time and route of administration, rates of absorption and excretion, combination with other drugs and the severity of the particular disease being treated.
  • invention provides a method of generating enhanced images of the human or non-human animal body, which method comprises administering to said body a
  • invention provides a method of therapy practised on the human or non-human animal body, which method comprises administering to said body a therapeutically effective copolymer according to the invention, eg. one
  • a drug or prodrug or a therapeutically effective, eg. cytotoxic, chelated metal e.g. a drug or prodrug or a therapeutically effective, eg. cytotoxic, chelated metal.
  • the present invention may include one or more of the polymers of this invention formulated into
  • compositions can be administered to humans and animals either orally, rectally, parenterally
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders and lyophilizates for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous solutions may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders and lyophilizates for reconstitution into sterile injectable solutions or dispersions.
  • nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of
  • compositions may also contain adjuvants such as preserving, wetting, emulsifying, cryoprotecting, and dispensing agents.
  • adjuvants such as preserving, wetting, emulsifying, cryoprotecting, and dispensing agents.
  • Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens,
  • chlorobutanol sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is admixed with at least one inert customary excipient (or
  • a carrier such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glylcerol, (d) disintegrating agents, as for example, agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate, (h)
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol and silicic
  • adsorbents as for example, kaolin and bentonite
  • lubricants as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate or mixtures thereof.
  • the dosage forms may also comprise buffering agents.
  • compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain
  • opacifying agents and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner.
  • embedding compositions which can be used are polymeric substances and waxes.
  • the active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions,
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol,
  • inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, di
  • composition can also include adjuvants, such as wetting agents,
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example,
  • compositions for rectal or vaginal administrations are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
  • Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays and inhalants.
  • the active component is admixed under sterile conditions with a physiologically
  • the present invention also provides the use of the copolymers according to the invention for the manufacture of diagnostic or therapeutic agents for use in methods of image generation or therapy practised on the human or non-human animal body.
  • the present invention provides a process for the preparation of a chelated metal bearing copolymer, said process comprising metallating a chelating moiety containing copolymer according to the invention, eg. by admixing the copolymer in a solvent together with an at least sparingly soluble compound of the metal, for example a chloride, oxide, acetate or carbonate.
  • the present invention provides a process for the
  • N-(N-(1,1-Dimethyl-ethoxycarbonyl)-phenylalanyl)leucine pentafluorophenylester N-(N-(1,1-Dimethylethoxycarbonyl)-phenylalanyl)leucine (23.0 g, 61 mmol) (prepared by a literature method [Anderson, G.W.; McGregor, A.C., t-Butoxycarbonyl amino acids and their use in peptide synthesis, J. Am. Chem. Soc, 1957, 79, 6180-6183]) was stirred with
  • Phenylmethyl 4- (phenylmethoxy)benzoate (48.8 g, 76%) was boiled under reflux with aqueous sodium hydroxide (2 M; 250 mL) and ethanol (250 mL) for 4 hours. The ethanol was evaporated under reduced pressure. Water (1000 mL) was added.
  • the white solid was collected by filtration, warmed to 65°C with aqueous sulphuric acid (2 M; 300 mL) for 1 hour and extracted with warm ethyl acetate. The ethyl acetate solution was dried with anhydrous
  • N-(4-(Phenylmethoxy)benzoyl)-glycine methyl ester 14.75 g, 49.2 mmol
  • methanolic sodium hydroxide (1M) 80 mL
  • the solvent was evaporated under reduced pressure.
  • the residue was dissolved in water and was acidified by addition of aqueous hydrochloric acid.
  • the suspension was extracted with ethyl acetate.
  • the extract was washed with saturated brine and was dried with anhydrous magnesium sulphate.
  • the solvent was evaporated under reduced pressure to give N-(4-(phenylmethoxy)benzoyl)glycine (6.59 g, 47%).
  • Dicyclohexylcarbodiimide (720 mg, 3.5 mmol) was added to N-(4-(phenylmethoxy)benzoyl)glycine (100 g, 3.5 mmol) in dry tetrahydrofuran (100 mL) and the mixture was taken to 0°C.
  • Pentafluorophenol (640 g, 3.5 mmol) was added dropwise and the mixture was stirred for 17 hours at 0°C. The suspension was filtered and the solvent was evaporated from the filtrate under reduced pressure.
  • N-(1,1-Dimethylethoxvcarbonyl)glycine N-(2-(4-phenylmethoxy)phenyl)ethyl)amide.
  • N-(1,1- Dimethylethoxy-carbonyl)glycine 850 mg, 4.85 mmol was stirred with dicyclohexylcarbodiimide (1.00 g, 4.85 mmol) and 2-(4-(phenylmethoxy)phenyl)ethylamine
  • N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanyl)-leucine pentafluorophenyl ester (1.19 g, 2.18 mmol) (Example A, Intermediate A) in tetrahydrofuran (30 mL) was added dropwise to glycine N-(2-(4-(phenylmethoxy)-phenyl)ethyl)amide (620 mg, 2.18 mmol), N,N-diisopropylethylamine (310 mg, 2.4 mmol) and 1-hydroxybenzotriazole (20 mg) in tetrahydrofuran (30 mL) and the mixture was stirred for 16 hours. The solvent was evaporated under reduced pressure.
  • N-(1,1-Dimethylethoxycarbonyl)sarcosine 2,4,5-trichlorophenyl ester N-(1,1-Dimethylethoxycarbonyl)-sarcosine (10.0 g, 53 mmol) was stirred with 2,4,5-trichlorophenol (10.6 g, 53 mmol) and
  • N-(1,1-Dimethylethoxycarbonyl)sarcosine N-(2-aminoethyl)amide N-(1,1-Dimethylethoxycarbonyl)-sarcosine 2,4,5-trichlorophenyl ester (12.7 g, 34.5 mmol) in dichloromethane (50 mL) was added during 30 minutes to ethane-1,2-diamine (20.7 g, 345 mmol) in dichloromethane (150 mL) and the solution was stirred for a further 2 hours. The solution was washed with water and with 10% aqueous sodium carbonate and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure to give N-(1,1-dimethylethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (6.9 g, 86%).
  • Example A Preparation of the peptide portion of Example A 1.
  • N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanylleucyl)glycine N-(2- (4-(phenylmethoxy) phenyl)ethyl)amide (3.89 g, 6.05 mmol) was treated with excess hydrogen chloride in dichloromethane (200 mL) for 3 hours. The solvent and excess reagent were evaporated under reduced pressure.
  • N-(N-Phenylalanylleucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)amide hydrochloride 165 mg, 284 mol was stirred with N,N-diisopropylethyl amine (100 mg, 774 mol), 4-(dimethylamino)pyridine (10 mg) and 1-hydroxybenzotriazole (10 mg) in dry
  • N-(N- (N-(N-(4-(Phenylmethoxy)benzoyl)glycyl)phenyalanyl)-leucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)-amide (444 mg, 547 mol) in ethanol (45 mL) was stirred vigorously with palladium on charcoal (10%; 50 mg) and hydrogen for 12 hours.
  • N-(N-(N-(N-(4-Hydroxybenzoyl)-glycyl)phenylalanyl)leucyl)glycine N-(2-(4-hydroxyphenyl)ethyl)amide (106 mg, 0.131 mol) was suspended in water (12 mL) containing sodium hydroxide (52.3 mg, 1.31 mmol).
  • N-(1,1-Dimethylethoxycarbonyl)leucine 2,4,5-trichlorophenyl ester.
  • N-(1,1-Dimethylethoxycarbonyl)leucine (6.32 g, 15 mmol) was stirred with 2,4, 5-trichlorophenol (3.01 g, 15.2 mmol) and
  • N-(1,1-Dimethylethoxycarbonyl)glycine (6.12 g, 35 mmol) was stirred with 2, 4, 5-trichlorophenol (6.91 g, 35 mmol) and
  • lysine monohydrochloride (9.14 g, 50 mmol) was stirred under reflux with copper (II) carbonate (21.6 g, 75 mmol) in water (180 mL) for 3 hours. The solution was filtered while hot and the filtrate was cooled to 20oC. 2,2,2-Trichloroethyl chloroformate (15.9 g, 75 mmol) and aqueous sodium carbonate (13.3 g, 125 mmol in 40 mL) were added alternately in portions to the filtrate during 30 minutes and the mixture was stirred vigorously at 0oC for 20 hours.
  • N ⁇ -(1,1-Dimethylethoxycarbonyl)-N ⁇ (2,2,2-trichloroethoxycarbonyl)lysine 2,4,5-trichlorophenyl ester N ⁇ -(1,1- Dimethylethoxycarbonyl)-N ⁇ -(2,2 , 2 -trichloroethoxy-carbonyl)lysine (6.32 g, 15 mmol) was stirred with 2, 4, 5-trichlorophenol (2.96 g, 15 mmol) and dicyclohexylcarbodiimide (3.10 g, 15 mmol) in ethyl acetate (100 mL) at 0°C for 20 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was
  • Succinic anhydride (tetrahydrofuran-2,5-dione) (64 mg, 0.72 mmol) was added and the mixture was boiled under reflux for 2.5 hours. A further portion of succinic anhydride (32 mg, 0.36 mmol) was added and boiling under reflux continued for a further 2 hours. The mixture was cooled to ambient temperature for 16 hours. The precipitated solid was collected by
  • N-(Phenylmethoxycarbonyl)sarcosine N-(2-aminoethyl)amide N-(Phenylmethoxycarbonyl)sarcosine pentafluorophenyl ester (3.5 g, 9.2 mmol) in
  • N-(phenylmethoxycarbonyl)sarcosine, 2,4,5-trichlorophenyl ester similarly from N-(phenylmethoxycarbonyl)sarcosine, 2,4,5-trichlorophenyl ester. 2. N-(1,1-Dimethylethoxycarbonyl)glycine N-(2-(N-phenylmethoxycarbronyl)sarcosylamino)ethyl)amide.
  • N-(Phenylmethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (3.71 g, 14 mmol) was stirred with N-(1,1-dimethyl ⁇ ethoxycarbonyl)glycine 2,4,5-trichlorophenyl ester (4.96 g, 14 mmol, Example B, Intermediate C) and N,N-diisopropylethylamine (1.99 g, 15.4 mmol) in
  • N-(N-(1,1-Dimethylethoxycarbonyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)-amide N-(1,1-Dimethylethoxycarbonyl)glycine N-(2-(N- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (5.22 g, 8 mmol) was treated with excess hydrogen chloride in dichloromethane (50 mL) for 1 hour. Water (50 mL) was added and the mixture was stirred vigorously for 15 minutes. The solvent and excess reagent were
  • N-Leucylglycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)-ethyl)amide hydrochloride N-(N-(1,1-Dimethylethoxycarbonyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (3.26 g, 6.1 mmol) was treated with excess hydrogen chloride in dichloromethane (40 mL) for 1 hour.
  • N-leucylglycine N-(2-(N-(phenylmethoxycarbonyl) sarcosylamino) ethyl) amide hydrochloride (2.65 g, quantitative).
  • N-(1,1-Dimethylethoxycarbonyl)phenylahanine pentafluorophenyl ester (2.65 g, 6.1 mmol)
  • Example B Intermediate B
  • N-leucylglycine N-(2-(N- (phenylmethoxycarbonyl)- sarcosylamino) ethyl) amide hydrochloride (2.81 g, 6.16 mmol), N-N-diisopropylethylamine (1.75 g, 13.5 mmol) and 4-(dimethylamino)pyridine (10 mg) in
  • N-(N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.94 g, 2.8 mmol) was treated with excess hydrogen chloride in dichloromethane (25 mL) for 1 hour. The solvent and excess reagent were evaporated under reduced pressure. The residue was dissolved in methanol.
  • N-(1,1- Dimethylethoxycarbonyl)glycine 2,4,5-trichlorophenyl ester (1.58 g, 2.55 mmol)
  • Example B Intermediate C
  • 4-(dimethylamino)pyridine 3.1 g, 2.5 mmol
  • N-(N-phenylalanylleucyl)glycine N- (2 - (N- (phenylmethoxycarb-onyl)sarcosylamino)ethyl)amide hydrochloride (904 mg, 2.55 mmol) and N,N-diisopropylethylamine (990 mg, 7.7 mmol) in dichloromethane (20 mL).
  • the mixture was stirred for 4 days.
  • the solution was washed with cold aqueous
  • N-(N-(N-(N-(1,1 Dimethylethoxycarbonyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.29 g, 1.77 mmol) was treated with excess hydrogen chloride in dichloromethane (10 mL) for 1 hour.
  • N-(N-(N-Glycylphenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)ethyl)amide hydrochloride (1.11 g, 1.77 mmol) was added to N,N-diisopropylethylamine (683 mg, 5.3 mmol) in dichloromethane (10 mL).
  • Polymer B N-(N-(N-(N-(N-(N-sarcosyl-N-(4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoyl)lysyl)glycyl)phenyl-alanyl)leucyl)glycine
  • N-(2-sarcosylaminoethyl)amide dihydrobromide is boiled under reflux with anhydrous sodium carbonate and poly(oxyethylene)-, -bis(oxiranylmethyl) ether (prepared by the literature method [Y. Chen and M. Feng, Chinese Patent 86 104 089,1987]) in ethanol for 6 hours. The suspension is filtered and the solvent is evaporated from the filtrate under reduced pressure to give the polymer.

Abstract

A linear block copolymer comprising units of an alkylene oxide, linked to units of peptide via a linking group comprising a -CH2CHOHCH2N(R)- moiety, is useful as an imaging agent, drug, prodrug or as a delivery system for imaging agents, drugs or prodrugs.

Description

BLOCK COPOLYMERS This invention relates to linear block copolymers useful in diagnostic imaging, drug delivery, and as drugs, and in particular to such polymers having
polypeptide and polyalkylene oxide moieties in the polymer backbone.
Nathen et al, Bioconjugate Chemistry 4 : 54-62
(1993) disclose copolymers of lysine and polyethylene glycol prepared by reacting amino groups of lysine with activated ester detivatives of polyethylene glycol. The polymer is best described as a polyamide formed by ∈-amino and the α-amino of lysine.
Davis et al., U.S. Patent 4,179,337 dated December 18, 1979 disclose insulin coupled to polyethylene glycol or polypropropylene glycol having a molecular weight of 500 to 20,000.
Zilkha et al, U.S. patent 3,441,526 issued April 29, 1969 disclose an N-carboxyanhydride condensation reaction for providing polyhydroxy polymers (such as starch etc.) with pendant polypeptide side chains.
British Patent 1,469,472 discloses low molecular weight polyethylene oxide immobilized proteins, said to have low immunogenicity.
However, none of these references suggests a linear block copolymer having repeating units of an alkylene oxide linked to repeating units of a peptide through a linking group formed by the reaction of an amine
precursor and an epoxide precursor. Moreover, the prior art teaches that crosslinking (via amino acid side chains) often frustrates the desired linear
copolymerization. The invention described herein advantageously avoids such crosslinking.
The invention concerns a linear block copolymer comprising single or repeating units of poly (alkylene oxide) (PAG) linked to units of peptide. The copolymer can be tailored to produce water-soluble polymers which are stable in the blood circulation but ultimately will be degraded to allow more facile excretion of low molecular weight PAG derivatives in the urine.
Thus viewed from one aspect the present invention provides a linear block copolymer comprising units of an alkylene oxide linked to units of peptide via a linking group comprising a -CH2CHOHCH2N(R) - moiety, wherein R is a lower alkyl group, (eg. C1-6-alkyl), eg a copolymer comprising units of polyalkyleneoxide linked to
polypeptide units via a linker group comprising an amine :epoxide conjugation product.
The copolymers of the invention have a variety of end uses. In particular they may be used as diagnostic agents, eg. image contrast enhancing agents in
diagnostic imaging techniques sch as MRI and
scintigraphy, as therapeutic agents, for example in radiotherapy or drug delivery, or as targetting agents, for example in cytotoxic or imaging procedures. Thus for example the peptide units may have chelating agents coupled thereto (eg. to the peptide side chains) such that the resultant chelating moieties may be metallated with metal species useful diagnostically or
therapeutically, such as paramagnetic or radioactive metal ions. Similarly drug or pro-drug species may be coupled to the peptide side chains, so that the
copolymer acts in effect both as a targetting agent and as a reservoir for release of the drug species. The targetting delivery system effected by the copolymers of the invention is of course also especially useful for delivery of metal species useful in diagnostic imaging of body organs or tissues or as cytotoxic agents.
Accordingly, viewed from a further aspect the present invention also provides a pharmaceutical
composition comprising a copolymer according to the invention together with at least one physiologically acceptable carrier or excipient.
The compositions of the present invention may include one or more of the polymers of this invention formulated into compositions together with one or more non-toxic physiologically acceptable carriers, adjuvants or vehicles which are collectively referred to herein as carriers, for parenteral injection, for oral
administration in solid or liquid form, for rectal or topical administration, or the like.
The copolymers of the invention may be produced by a particularly elegant and simple condensation of a bisamine reagent with a bisepoxide reagent, the amine: epoxide condensation yielding the linking group moiety -CH2CHOHCH2N(R) - mentioned above. By using polymeric such reagents, one incorporating a polyalkylene oxide chain and the other a polypeptide chain, the linear block copolymer structure of the compounds of the invention is produced.
Thus, viewed from a still further aspect, the invention provides a process for the preparation of a linear block copolymer according to the invention, said process comprising reating a bisepoxide reagent with a bisamine reagent, one of said reagents incorporating said peptide units and the other incorporating said alkylene oxide units.
The linking group in the copolymers according to the invention preferably comprises a -CH2-CHOH-CH2N(CH3) - moiety, particularly preferably attached at the nitrogen end to an alkylene chain (CH2)P (where p is an integer having a value of from 1 to 6) and optionally attached at the carbon end to a phenyleneoxy moiety. Thus the linking group preferably comprises a moiety
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4CO-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4(CH2)2-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4(CH2)2NH-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4-; -NH(CH2)pN(CH3)CH2CHOHCH2OC6H4CO-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4(CH2)2-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4(CH2)2NH-;
-CONH(CH2)pNHCO(CH2)pN(CH3)CH2CHOHCH2-;
-NH(CH2)pNHCO(CH2)pN(CH3)CH2CHOHCH2-;
-NHCO(CH2)pN(CH3)CH2CHOHCH2-; or
-CO(CH2)pN(CH3)CH2CHOHCH2-.
The alkylene oxide residues, generally occuring in a polyalkyleneoxide chain, are preferably residues of lower alkylene oxides, eg. C2-6, preferably C2-4 and especially preferably ethylene oxide residues. These will generally be chain end derivatised to link up to the amine :epoxide conjugation product component of the linking group, eg. to link the terminal ether oxygens of the polyalkyleneoxide chain to epoxide-reactive amine groups or to amine-reactive epoxide groups. The nature of the chain-end derivatization is not critical and such bifunctional reagents may be represented by the formulae HNR-(PAG)-NRH or with the PAG representing the
Figure imgf000006_0001
polyalkyleneoxide group and the
enclosing brackets symbolizing any such chain-end derivatization.
The peptide units, again generally occur in a polypeptide chain containing a plurality of amino acid residues. Either synthetic or naturally occurring polypeptide units or fragments may be used and these may in and of themselves provide a therapeutic or targetting moiety or alternatively, if desired, further moieties such as drugs, prodrugs or chelating agents may be conjugated to the peptide side chains. As with the polyalkyleneoxide chains, the polypeptide chains may be chain end derivatised to link up to the amine:epoxide conjugation product component of the linking group, eg. to link carbonyl carbon or amine nitrogen termini to epoxides-reactive amines or to amine-reactive epoxides. In the formulae used herein however CO(peptide)NH is generally used to indicate a peptide group showing the terminal carbonyl and amine groups outside the brackets.
The peptide moiety in one embodiment of the
invention particularly preferably derives from the
bisepoxide
Figure imgf000007_0002
or from the bisamine NHRCH2CO(peptide)NH(CH2)pCOCH2NHR
(where p and R are as defined above, R preferably being C1-4 alkyl). The bisepoxide of formula I
Figure imgf000007_0001
are especially preferred and form a further aspect of the invention. However in general the block copolymers having the repeat unit
- (PAG)N(R)CH2CHOHCH2OC6H4CO(Peptide)NH(CH2)pC6H4OCH2CHOHCH2N(R)
Formula A
or
-(PAG)CH2CHOHCH2N(R)CH2CO(Peptide)NH(CH2)pNHCOCH2NRCH2CHOHCH2-
Formula B
(wherein
R is a 1-4 carbon alkyl; and
p is from 1 to 6) are preferred.
The copolymer compounds can be tailored for
specific uses by altering the size of the polymer or
altering the peptide composition to provide differing blood pool residence time, enzymatic breakdown rates, and tissue distributions.
As an imaging agent, the copolymer of the
invention preferably has a molecular weight of at least about 5000 and a metal ion useful as a contrast
enhancer, fluorophore or x-ray opaque ion associated therewith, thus making it suitable for use as an agent for diagnostic imaging.
An imaging metal is defined as a metal useful in x-ray imaging (eg. a metal of atomic number 50 or above) or a metal useful in magnetic resonance imaging (preferably a paramagnetic metal and more preferably a lanthanide metal or transition metal) or a metal useful in fluorescence imaging (preferably a lanthanide metal, most preferably europium).
It is a particularly advantageous feature that the polymeric chelates of this invention provide effective imaging contrast enhancement of the blood pool within the vascular system for remarkably long periods of time.
It is another advantageous feature of this
invention that polymeric compounds are provided having a specificity toward accumulation in different tissues, for example, in tumors and the liver.
As used herein, the abbreviation PAG refers to polyalkylene oxide moieties having a single type of repeating unit or differing (non-repeating) units of alkylene oxide, or a mixture thereof in each PAG. Each alkylene oxide unit in the PAG preferably contains from 2 to about 4 carbons, and is linear or branched.
Poly(alkylene oxide) units in the polymer may also differ in length and composition from each other.
Exemplary PAG moieties include poly (ethylene oxides), poly(propylene oxides) and poly (butylene oxides).
Preferred PAG moieties include poly (ethylene oxides), poly(propylene oxides) and random and block copolymers thereof. Poly(ethylene oxide) containing polymers are particularly preferred when it is desired for the final polymer to possess solubility in water. It is also contemplated that the poly(alkylene oxide) moiety can comprise glycerol poly(alkylene oxide) triethers, polyglycidols, linear, block and graft copolymers of alkylene oxides with compatible comonomers such as poly(proplene oxide-coethylene oxide), or poly(butylene oxide-co-ethylene oxide) and grafted block copolymers. These moieties can be derived from poly (alkylene oxide) moieties which are commercially available or
alternatively they can be prepared by techniques well known to those skilled in the art. A particularly preferred class of polyalkyleneoxide moieties derived from poly (ethylene oxide) can be represented by the structure:
-O(CH2CH2O)mCH2CH2O- wherein m is 1 to 750. The preferred length depends upon the desired molecular weight.
These PAG moieties and their reactive derivatives, useful in preparing the polymer of the invention, are known in the art. For example, bis (methyl amino) polyethylene glycol and its use as an intermediate in the preparation of block copolymers is known in the art. For example Mutter, Tetrahedron Letters, 31:
2839-2842 (1978) describes a procedure to convert the terminal hydroxyl groups of poly (ethyleneoxide) to reactive primary amino groups as well as the
preparation of a number of reagents bound to
poly (ethyleneoxide) amines; and Harris et al, J.
Polymer. Science, 22 : 341-352 (1984) describe various PAD derivatives including for example, amino
poly(ethyleneoxide). Other PAG derivatives may be prepared by known chemical techniques, examples of which are described hereinbelow.
As used herein, peptide refers to an amino acid chain of at least 2 amino acids, wherein each of the amino acids in the peptide may or may not be the same, and may or may not be selected from the 20 naturally occurring L-amino acids. Thus peptide units may contain D-amino acids, artificial amino acids or amino acid derivatives, such as glutamate esters, lysyKe- amino) amides and the like. This definition also includes proteins, enzymes, polypeptides and
oligopeptides, which are art recognized amino acid chains. Specifically contemplated preferred peptides include small enzymes less than 100 kD), peptide hormones, peptide recognition domains, peptide drugs, and peptides with known enzymatic breakdown rates.
Certain abbreviations appearing in the text and schemes are here defined: Boc refers to the t-butoxy carbonyl radical commonly used as a blocking agent in solid phase peptide synthesis. Conventional three letter abbreviations for amino acid residues are used throughout the specification. OPFP refers to
pentafluorophenyloxy; Bn refers to benzyl; CBZ refers to phenylmethoxycarbonyl; OTCP refers to 2,4,5-trichlorophenyloxy; and Troc refers to 2,2,2-trichloroethoxycarbonyl.
Copolymerization can occur by reaction of
bis (oxiranyl) derivatives (also known as bisepoxides) with bis (amino or alkylamino) derivatives (also known as bisamines). There are no by-products of the
polymerization reaction. The monomer units of PAG and peptide can be prepared as either bis(oxiranyl)
derivatives or bis (amino) derivatives provided that the reaction producing the copolymers is between an amine and an epoxide. Therefore there are two chemical strategies for preparing products of the invention described hereinbelow. As a consequence of reacting bisamines with bisepoxides the sense of the PAG and peptide units can be reversed.
The polymer of the invention has between its PAG and peptide subunits , a linking group. The linking group contains a -CH2CHOHCH2N(R)- diradical, typically derived from the reaction of an amine and an epoxide. It is preferred that a bisepoxide subunit be reacted with a bisamine subunit. The skilled artisan will appreciate that the recitation used throughout the specification of each type of linking group diradical can be reversed and have the same meaning. Thus the sense of the linking group can be reversed (end for end), with one terminus attached to the PAG moiety, and the other terminus attached to the peptide or vise versa, while its recitation in the specification and the claims is the same.
Peptides used to prepare the copolymers of the invention can be prepared by standard procedures known in the art. Useful peptides include those derived from native or recombinant organisms, solid phase peptide synthesis or traditional wet chemistry peptide
synthesis and the like. Each of these peptide
preparation methods are well known in the art and use conventional, known materials. Protein expression and purification from natural and recombinant sources is in the prior art (cf. Protein Expression and Purification (1990); Harris et al., Protein Purification Methods
(1989); Deutscher, M.P. Guide to Protein Purification Methods in Enzymology, Vol. 82 (1990)). Peptide synthesis is also known in the art (cf. Atherton, et al., Solid Phase Peptide Synthesis a Practical
Approach. Oxford University Press (1989)). Thus, the peptides are easily prepared by known chemistry.
Linear peptide fragments can be tailored such that they are stable in blood, but are susceptible to lysosomal degradation by commonly occurring proteases. Examples of susceptible peptide units are gly-phe-leu-gly, gly-phe-tyr-ala, ala-gly-val-phe, gly-phe-ala-gly, and others known in the art. The prior art describes such oligσpeptides as useful in preparing prodrugs, when the drug is attached to one terminus of the oligopeptide. (See generally "Polymers Containing Enzymatically Degradable Bonds" Makromol. Chem. 184
(1983) R. Duncan, H.C Cable, J.B. Lloyd, P. Rejmanov'a and J. Kopecek, in Polymers containing enzymatically degradable bonds, 7, Design of oligopeptide side-chains to promote efficient degradation by lysosomal enzymes, Makromol. Chem., 184, p. 1997-2008 (1983); and P.
Rejmanova, J. Kopecek, J. Pohl, M. Baudys and V.
Kostva, in Polymers containing enzymatically degradable bonds, 8, Degradation of oligopeptide sequences in N- ( 2- hydroxypropyl)methacrylamide copolymers by bovine spleen cathepsin B, Makromoi. Chem. 184, p. 2009-2020, (1983).) For the compounds of the present invention it is contemplated that prodrugs can be attached to functionalized side chains of the peptide, rather than the terminus of the peptide.
The concept of drug targeting has gained
importance in recent years, especially for anticancer drugs, inasmuch as toxic side effects of anticancer drugs to normal cells are a primary obstacle in cancer chemotherapy due to lack of selectivity of the
anticancer drugs to cancer cells. In the prior art, drug targeting has been accomplished by drug
conjugation with large antibodies, or encapsulation in a transporter specific to the target. Materials such as proteins, saccharides, lipids and synthetic polymers have been used for such transporters. Antibodies have been perhaps most widely used due to their target specificity and wide applicability. However, these methodologies have not been commercially exploited because the prohibitive cost of the transporter or targeting agent which can be used to target only one type of cell or tissue.
The peptide portion of the polymer can be tailored to recognize (or target) certain cells or functions of cells. Because the polymer can use more than one peptide and thus more than one type of peptide, the polymer can advantageously target more than one type of cell or tissue at once. Judicious choice of peptide allows treatment or targeting of more than one type of cancer cell, for example, or other disease state. This choice is facilitated by the prior art which contains a myriad of known oligopeptides which are antigenic to certain cells. Furthermore, the invention allows such targeting without the cost of raising antibodies to certain cells, harvesting such antibodies, conjugating antibodies to drug and further testing for maintained specificity after conjugation. The invention allows specific targeting to be achieved by short recognition sequences. Cell specific delivery can be achieved by incorporating targeting agents into the polymer.
Preferred peptides are those which have a receptor molecule specific to a ligand of interest. Thus, a specific binding reaction involving the reagent can be used for the targeting expected.
Depending upon the intended use, the peptides can be selected from a wide variety of naturally occurring or synthetically prepared materials, including, for example enzymes, proteins, peptide hormones, virus coats, or proteins derived from blood components, tissue and organ components, including haptens,
antibodies, antigenic proteinaceous materials, or fragments of any of these and others known to one skilled in the art.
Examples of these targeting peptides include: the integrin binding motif RGDS (arg-gly-asp-ser), which is present on many extracellular matrix proteins and can be used to interfere with cell adhesions involved in migration of leukocytes. Other peptide sequences which can be used to deliver the polymer include cationic sequences (ie. rich in lys or arg) which are useful in producing a DNA-binding polymer for use in supression of gene expression, antisense oligomer delivery and the like; peptide hormones such as αMSH which can be used for targeting to melanoma; and relatively low molecular weight (15-2OkDa) engineered hypervariable antibody binding domains (VH+VL constructs) raised against any target. Such sequences are obtained by synthesis, isolation from cells or bacteriophages or they can also be raised against cells, proteins, or foreign
substances in a host. Common hosts for raising
recognition sequences include rabbits, goats, mice, and the like. These and other methods of obtaining
recognition sequences are known in the art.
In certain embodiments, the above-described peptide can be an immunoreactive group, which would be found in a living organism or which finds utility in the diagnosis, treatment or genetic engineering of cellular material of living organisms. The peptide has a capacity for interaction with another component which may be found in biological fluids, cells or associated with cells to be treated or imaged, such as, for example tumor cells and the like.
Two highly preferred uses for the polymer of this invention are for the diagnostic imaging of tumors and the treatment of tumors. Preferred immunoreactive groups therefore include antibodies, or immunoreactive fragments thereof, to tumor-associated antigens.
Specific examples include B72.3 antibodies (described in U.S. Patents Nos. 4,522,918 and 4,612,282) which recognize colorectal tumors, 9.2.27 antimelanoma antibodies, D612 antibodies which recognize colorectal tumors, UJ13A antibodies which recognize small cell lung carcinomas, NRLU-10 antibodies which recognize small cell lung carcinomas and colorectal tumors (Pan- carcinoma), 7E11C5 antibodies which recognize prostate-tumors, CC49 antibodies which recognize colorectal tumors, TNT antibodies which recognize necrotic tissue, PR1A3 antibodies, which recognize colon carcinoma, ING-1 antibodies, which are known in the art and are described in International Patent Publication WO-A-90/02569, B174 antibodies which recognize squamous cell carcinomas, B43 antibodies which are reactive with certain lymphomas and leukemias and any other antibody which may be of particular interest.
Because the peptides of the polymer are linear, they can provide functional groups for coupling of diagnostic agents, drugs, or prodrugs or other
targeting moieties by the side chains of individual amino acids found in the peptide portion of the
backbone. Functional groups can also be added by reacting or derivatizing functionalizable basic groups (found for example in lysyl or argininyl residues) or acidic groups (as found in aspartate, glutamate, providing free carboxyl groups), or sulfhydryl groups, (e.g. cysteine), hydroxyl groups (such as found in serine) and the like. This coupling is done by
standard peptide chemistry known in the art.
Cytotoxic drugs can also be coupled to the polymer to produce prodrugs which are released as a drug to targeted cells or tissues. Such coupling methods are known in the art, see for example; Duncan, P.
Kopeckova-Rejmanova, J. Strohalm, I. Hume, H. C. Cable, J. Pohl, J. B. Lloyd and J. Kopecek (1987) Anti-cancer agents coupled to N-(2-hydroxypropyl)methacrylamide copolymers. I. Evaluation of daunomycin and puromycin conjugates in vitro. British J. Cancer 55: 165-174; and R. Duncan, P. Kopeckova, J. Strohalm, I. Hume, J.B. Lloyd and J. Kopecek (1988) Anti-cancer agents coupled to N-(2-hydroxypropyl)methacrylamide copolymers. II. Evaluation of daunomycin conjugates in vivo against L1210 leukaemia. British J. Cancer 57: 147-156. Drugs contemplated to be useful include any drug which can be covalently attached to the polymer and retains its activity when so attached. It is contemplated that drugs which become active only when liberated from the polymer are also useful, and as such are prodrugs.
Drugs which are contemplated to be useful in the polymer include cytotoxic agents, and immunomodulating peptides and proteins as described above.
By "cytotoxic agent", is meant any agent able to kill cells, including, chemotherapeutic agents such as cytotoxic drugs and cytotoxic antibiotics, chelated radionuclides and toxins or any agent which initiates or which leads to cell death. The term cytotoxic agents also includes agents which activate a host's immune response leading to cell death. The cytotoxic agent will be selected with reference to factors, such as the type of disease state, for example the type of cancer tumor and the efficacy of a certain chemotherapy agent for treating the cancer tumor involved, and the like. The cytotoxic agent may be selected from
alkylating agents, antimetabolites, natural products useful as cytotoxic drugs, hormones and antagonists and other types of cytotoxic compounds.
Examples of alkylating agents include the nitrogen mustards (i.e. the 2-chloroethylamines) such as, for example, chloromethine, chlorambucil, melphalan, uramustine, mannomustine, extramustine phosphate, mechlor-thaminoxide, cyclophosphamide, ifosamide and trifosfamide; alkylating agents having a substituted aziridine group such as, for example, tretamine, thiotepa, triaziquone and mitomycin; alkylating agents of the alkyl sulfonate type, such as, for example, busulfan and piposulfan; alkylating N-alkyl-N-nitrosourea derivatives such as, for example,
carmustine, lomustine, semustine or streptozotocne; alkylating agents of the mitobronitole, decarbazine and procarbazine type; and platinum complexes such as for example, cisplatin and carboplatin and others.
Examples of antimetabolites include folic acid
derivatives such as, for example, methotrexate, aminopterin and 3'-dichloromethotrexate; pyrimidine derivatives such as, for example, 5-fluorouracil, floxuridine, tegafur, cytarabine, idoxuridine, and flucytosine; purine derivatives such as, for example, mercaptopurine, thioguanine, azathioprine, tiamiprine, vidarabine, pentostatin and puromycin and others. Examples of natural products, useful as cytotoxic agents include for example vinca alkaloids, such as vinblastine and vincristine; epipodophylotoxins such as, for example, etoposide, and teniposide; antibiotics such as, for example, adrimycin, daunomycin,
dactinomycin, daunorubicin, doxorubicin, mithramycin, bleomycin and mitomycin; enzymes such as, for example, L-asparaginase; biological response modifiers such as, for example, alpha-interferon; camptothecin; taxol; and retinoids such as retinoic acid and the like.
Examples of hormones and antagonists include adrenocorticoids, such as, for example, prednisone;
progestins, such as, for example, hydroxyprogesterone acetate, medroxyprogesterone acetate and megestrol acetate; estrogens such as, for example,
diethylstilbestrol and ethinyl estradiol; antiestrogens such as for example, tamoxifen; androgens such as, for example, testosterone propionate and fluoxymestrone; antiandrogens such as, for example, flutamide; and gonadotropinreleasing hormone analogs such as, for example, leuprolide.
Examples of miscellaneous cytotoxic agents include anthracenediones such as for example, mitoxantrone;
substituted ureas such as, for example, hydroxyureas; and adrenocortical suppressants such as, for example, mitotane and aminoglutethimide. The cytotoxic agent can be ionically associated with the chelating residue. For example, in preferred embodiments, the cytotoxic agent is a radionuclide comprising a radioactive metal ion such as described below associated with a peptide-linked cheating residue. The polymer of the invention can contain one or more of a wide variety of chelating agents. As is well known, a chelating agent is a compound containing donor atoms that can combine by coordinate bonding with a cation to form a cyclic structure called a chelation complex or chelate. This class of compounds is described in the Kirk-Othmer Encyclopedia of Chemical Technology, Vol. 5, 339-368. Chelating residues may also be attached via the functionalizable side chains of the peptide via known chemistry. These chelating residues can be coupled to the polymer to produce contrast agents useful in diagnostic imaging or cytotoxic agents when complexed with the appropriate metal . The chelating residue is attached to an available amino acid side chain in the peptide portion of the polymer by a protein reacting group. By "protein reactive group" it is meant any group which can react with any functional groups typically found in proteins, especially an amino acid side chain.
Preferred protein reactive groups can be selected from but are not limited to:
(1) A group that will react directly with the amine or sulfhydryl groups on an amino acid side chain. For example, active halogen containing groups
including, for example, chloromethylphenyl groups and chloroacetyl [Cl-CH2CO-] groups, activated 2-leaving- group-substituted ethylsulfonyl and ethylcarbonyl groups such as 2-chloroethylsulfonyl and 2-chloroethylcarbonyl; vinylsulfonyl; vinylcarbonyl;
epoxy; isocyanato; isothiocyanato; aldehyde; aziridine; succinimidoxycarbonyl; activated acyl groups such as carboxylic acid halides; mixed anhydrides and the like; and other groups known to be useful in attaching molecules to proteins or crosslinking proteins and the like.
(2) A group that can react readily with modified proteins or similar biological molecules modified to contain reactive groups such as those mentioned in (1) above, for example, by oxidation of the amino acid side chain to an aldehyde or a carboxylic acid, in which case the "protein reactive group" can be selected from amino, alkylamino, arylamino, hydrazino,
alkylhydrazino, arylhydrazino, carbazido, semicarbazido, thiocarbazido, thiosemicarbazido, sulfhydryl, sulfhydrylalkyl, sulfhydrylaryl, hydroxy, carboxy, carboxyalkyl and carboxyaryl. The alkyl portions of the protein reactive group can contain from 1 to about 18 carbon atoms or be a lower alkyl group as described for R above. The aryl portions of the protein reactive group can contain from about 6 to about 20 carbon atoms.
(3) A group that can be linked to the amino acid side chain or similar biological molecule, or to the modified peptide as noted in (1) and (2) above by use of a crosslinking agent. Certain useful crosslinking agents, such as, for example, difunctional gelatin hardeners, bisisocyanates etc., which become a part of a linking group in the polymer during the crosslinking reaction. Other useful crosslinking agents, such as, for example, consumable catalysts, are not present in the final conjugate. Examples of such crosslinking agents are carbodiimide and carbamoylonium crosslinking agents as disclosed in U.S. Patent 4,421,847 and the dication ethers of U.S. Patent 4,877,724. With these crosslinking agents, one of the reactants must have a carboxyl group and the other an amine, alcohol, or sulfhydryl group. The crosslinking agent first reacts selectively with the carboxyl group, then is cleaved during reaction of the "activated" carboxyl group with, for example, an amine to form an amide linkage between the peptide portion of the polymer and metal complexing agents, thus covalently bonding the two moieties. An advantage of this approach is that crosslinking of like molecules, e.g., amino acid side chains with imino acid side chains or complexing agents with complexing agents is avoided, whereas the reaction of difunctional crosslinking agents is less selective. Especially preferred protein reactive groups include amino and isothiocyanato. Preferred chelating agent precursors have anhydride, sulfonylylchloride, alkylsulfate, vinyl sulfate, vinylsulfate, or ester functionalkyl.
The chelating residues can be derived from
chelating moieties which are selected to contain electron donating atoms which will chelate a metal, by forming coordination bonds therewith. These moieties can be selected from polyphosphates, such as sodium tripolyphosphate and hexametaphosphoric acid;
linear, branched or cyclic aminocarboxylic acids, such as ethylenediaminetetra-acetic acid, N-(2- hydroxyethyl) ethylenediaminetriacetic acid,
nitrilotriacetic acid, N,N-di (2-hydroxyethyl)glycine, ethylenebis (hydroxyphenylglycine), diethylenetriamine pentaacetic acid and the N-carboxymethylated
macrocyclic polyazacycloalkanes sch as DOTA and D03A and the phosphonomethylated analogs;
1, 3-diketones, such as acetylacetone,
trifluoroacetylacetone, and thenoyltrifluoroacetone; hydroxycarboxylic acids, such as tartaric acid, citric acid, gluconic acid, and 5-sulfosalicylic acid; polyamines, such as ethylenediamine,
diethylenetriamine, triethylenetetramine, and
triaminotriethylamine;
aminoalcohols, such as triethanolamine and N-(2-hydroxyethyl)ethylenediamine;
aromatic heterocyclic bases, such as 2,2'-dipyridyl, 2,2'-diimidazole, dipicoline amino and 1,10-phenanthroline;
phenols, such as salicylaldehyde,
disulfopyrocatechol, and chromotropic acid;
aminophenols, such as 8-hydroxyquinoline and oxinesulfonic acid;
oximes, such as dimethylglyoxime and
salicylaldoxime;
peptides containing proximal chelating
functionality such as polycysteine, polyhistidine, polyaspartic acid, polyglutamic acid, or combinations of such amino acids; Schiff bases, such as disalicylaldehyde 1,2-propylenediimine;
tetrapyrroles, such as tetraphenylporphin and phthalocyanine;
sulfur compounds, such as toluenedithiol, meso- 2,3-dimercaptosuccinic acid, dimercaptopropanol, thioglycolic acid , potassium ethyl xanthate, sodium diethyldithiocarbamate, dithizone, diethyl
dithiophosphoric acid, and thiourea;
synthetic macrocyclic compounds, such as
dibenzo[18]crown-6, (CH3)6-[14]- 4,11-diene-N4, and
(2.2.2)-cryptate; and
phosphonic acids; such as
nitrilotrimethylenephosphonic acid,
ethylenediaminetetra (methylenephosphonic acid), and hydroxyethylidenediphosphonic acid, or combinations of two or more of the above agents.
Preferred chelating residues contain polycarboxlic acid or carboxylate groups and include elements present in: ethylenediamine-N,N,N',N'-tetraacetic acid (EDTA);
N,N,N',N",N"-diethylenetriaminepentaacetic acid (DTPA);
1,4,7,10-tetraazacyclododecane-N,N',N",N"'tetraacetic acid (DOTA); 1,4,7,10-tetraazacyclododecane-N,N',N"-triacetic acid (D03A); 1-oxa-4,7,10-triazacyclo-dodecane-N,N',N"-triacetic acid (OTTA); trans(1,2)-cyclohexanodiethylenetriamine pentaacetic acid (CDTPA);
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000022_0002
; and
Figure imgf000022_0003
Such chelating compounds, including their
preparation and manipulation are well known in the art. For example, the acid and anhydride forms of EDTA and DTPA are commercially available; methods for preparing B4A, P4A and TMT are described in U.S. Patent
4,859,777; the disclosure of which is hereby
incorporated by reference; and other suitable chelating groups are known in the art, and are described in
WO-A-92/08494 and many other readily available
references.
If the chelating residue is made of multiple chelating moieties or subunits, such subunits can be linked together by a linking group. Thus, more than one chelating moiety can be used to make up the
chelating residue. If more than one chelating moiety is present in the chelating residue, these may be the same or different. Chelating moieties can be linked together using known chemistries. Thus the chelating residue can be one moiety or a "core" of chelating moieties. For example, a core of DTPA residues may be prepared by reacting DTPA dianhydride with a diamine, such as ethylene diamine, to form a "core" of DTPA chelators. Other chelating residues, made up of multiple chelating moieties are well known in the art and are prepared by known chemistries as well.
For magnetic resonance imaging applications, the chelated metal ion M(+a) preferably represents a
paramagnetic metal ion such as an ion of metals of atomic number 21 to 29, 42, 44 and 57 to 71, especially 57 to 71. Ions of the following metals are preferred: Cr, V, Mn, Fe, Co, Ni, Cu, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm and Yb. Especially preferred are
Cr3+, Cr2+, V2+, Mn3+, Mn2+, Fe3+, Fe2+, Co2+, Gd3+ and Dy+3. It is a particularly advantageous feature that polymers can be provided exhibiting a high substitution ratio, i.e., containing relatively large numbers of
paramagnetic metal ions per molecule.
The cytotoxic agent can be a radioactive isotope, preferably a radioactive metal ion isotope. This radioactive metal isotope can be an ion of an isotope of a metal selected, for example, from Sc, Fe, Pb, Ga, Y, Bi, Mn, Cu, Cr, Zn, Ge, Mo, Tc, Ru, In, Sn, Re, Sr, Sm, Lu, Du, Sb, W, Re, Po, Ta and Tl ions. In a prefered embodiment, radioisotopes which are also useful in diagnostic imaging applications are
specifically contemplated. Thus this embodiment finds utility in imaging and therapy where either procedure can be performed in conjunction with or ancillary to the other. Preferred isotopes of radioactive metal ions for this embodiment include 44Sc, 64,67Cu, 111In, 212Pb, 68Ga, 90y, 153Sm, 212Bi, 99mTc and 188Re for
therapeutic and diagnostic imaging applications.
If a metal is chelated by the polymer, as for example, in imaging or therapy as described above, the metal content in the polymer can vary from about 0.1 up to about 20% based on the total weight of the polymer. For example in a magnetic resonance imaging embodiment, the polymer preferably contains a paramagnetic metal ion in an amount of from 1 to 25%, more preferably 2-20% by weight. In a therapeutic embodiment the
radionuclide metal ion is present in roughly the same amounts as for imaging.
The PAG moiety in this composition can be capped at the terminus with a capping moiety selected from a hydrogen, hydroxy, alkyl, amino, or alkoxy. Preferred capping groups are hydrogen or hydroxyl groups. Thus capping is done by known chemistry, and precapped prepolymers are available. It is further contemplated that cyclic copolymers can be prepared.
The copolymers of this invention can be prepared in water-soluble, water dispersible or water-insoluble forms depending upon the intended application. The copolymer can have a molecular weight ranging from 10,000 to 1 million preferably 11,000 to 80,000. The preferred molecular weight varies according to the application as described below.
In addition to targeted delivery of the polymers of the invention, the polymer can be selectively delivered to specific cells, tissue types, or organs with or without the aid of a targeting agent. When no targeting agent is used such targeted delivery is based on size (hydrodynamic radius) and charge alone. The charge of the polymer can be altered by judicious choice of the aminoacids used in the peptide component of the copolymer to suit the application. Of course, the size of the polymer can be chosen by altering the size of PAG or peptide used to prepare the polymer or by altering the degree of polymerization. The
mechanism of the targeted delivery of polymer is thought to be based upon the passive biodistribution in tissues of the polymer. It is thought that this passive biodistribution can occur because the PAG component of the polymers allows free distribution of the polymers within the circulatory system, with low antigenicity or without interference by the mononuclear phagocytic system. Unlike hydrophobic polymers known in the art, which are taken up by the
reticuloendothelial system, the polymers of the
invention can be designed to be distributed to tissues without being metabolized. Thus the size and charge of the polymer in the tissue is a function of the size and charge of the polymer administered. Distribution of the unmetabolized polymer to tissues will be influenced by the nature of the local vascular endothelium in each tissue and the prescence or absence of a lymphatic system. Three general categories of vascular
endothelium are sinusoidal epithelium, characterized by discontinuity and little or no basement membrane;
fenestrated vascular endothelium; and continuous vascular endothelium, characterized by tight junctions and basement membrane. The lymphatic system is known to recirculate proteins and other molecules which can float freely in the plasma, but escape the circulatory system, exist for a time in tissue and then are
returned to the circulatory system via the lymphatic system. The skilled artisan can determine which tissues will be passively targeted by the polymer by approximating the molecular weight or more preferably the hydrodynamic radius of known proteins diffusing through the tissue in a known given period.
Tissues such as bone marrow, liver and spleen tissue are characterized by sinusoidal endothelium, (which allows escape of large molecules from the circulating system into the surrounding tissue) thus larger polymer molecules are useful in passively targeting such tissues. Tissues such as those found in the GI tract, kidney glomeruli, and endocrine gland tissue are characterized by fenestrated endothelium
(which allows escape of smaller macromolecules from the circulatory system), thus slightly smaller polymer molecules are useful in passively targeting such tissues. Tissues such as muscle and lung tissue are characterized by continuous vascular endothelium (which allows small molecules to escape from the circulatory system into the surrounding tissue), thus smaller polymer molecules are useful in passively targeting these tissues.
For example, the hydrodynamic radius of albumin is approximately 37 A, its molecular weight is 67 Kd, and its charge is known. It is known that the average half life for albumin circulation through tissue is
approximately 24 hours, but this half life is longer in some tissues and shorter in others. Moreover, the concentration of albumin in certain tissues is
appreciable and in other tissues albumin is nearly absent altogether. The skilled artisan can prepare a polymer of approximately the same size, or preferably the same hydrodynamic radius and charge, and expect a similar half life and concentration in tissues.
The skilled artisan will recognize that
inflammation of tissues will perturb the normal
physiology of that tissue and thus the half life and concentration of macromolecules, such as proteins or the polymer of the invention, in an inflamed tissue or inflamed tissue site. Thus the polymer finds utility in imaging and/or treating such inflamed tissues or inflamed tissue sites.
The skilled artisan will also appreciate that the absence of a lymphatic system in a tissue will perturb the concentration and increase the half life of macromolecules in a tissue because no convenient mechanism is provided for the scavenging of such macromolecules. Such is the case in growing tumors. One can deliver a cytotoxic agent, a pro-drug, or an imaging moiety to the growing tumor surface based on size of the polymer and on vasculature of the
surrounding targeted tissue as described above. Thus dosing a cytotoxic agent will result in accumulation of such agent in the growing surface of the tumor.
Thus molecular weight and charge of the polymer may be tailored to the specific application based on tissue type, presence or absence of inflammation, tumor and/or vasculature type and presence or absence of a lymphatic system to provide a polymer with the correct characteristics for targeting the desired tissue.
The general synthetic methods for production of linear alternating polymers follow two related schemes (A and B) involving the reaction of a bis- (methylamino)-monomer with a bis(oxiranyl)monomer, described below. Compounds of the invention are prepared by chemical transformations which are
conventional and known to those skilled in the art of chemistry. Furthermore, known transformations can be used for effecting changes in functional groups in the polymer or compounds used in preparing the polymer of the invention. For example, acylation of hydroxy- or amino-substituted species to prepare the corresponding esters or amides, respectively; simple aromatic and heterocyclic substitutions or displacements; cleavage of alkyl or benzyl ethers to produce the corresponding alcohols or phenols; and hydrolysis of esters or amides to produce the corresponding acids, alcohols or amines, preparation of anhydrides, acid halides, aldehydes, simple aromatic alkylation and the like as desired can be carried aromatic out.
Such transformations will provide suitable
chelating agents and precursors thereof containing reactive functionality, including, for example,
polycarboxylic acids in dianhydride form, di(sulfonyl chlorides), di(alkyl sulfates), di(vinyl sulfones), diesters, and the like. Such known transformations are also useful in attaching the chelator to the polymer or polymer precursor, and in preparing the polymer itself. However, as will be recognized by one skilled in the art, obtaining the desired product by some reactions will be better facilitated by blocking or rendering certain functional groups inert. This practice is well recognized in the art, see for example, Theodora
Greene, Protective Groups in Organic Synthesis (1991). Thus when reaction conditions are such that they may cause undesired reactions with other parts of the molecule, for example in portions of the chelator intended to become ligands, the skilled artisan will appreciate the need to protect these reactive regions of the molecule and will act accordingly. For example, the chelating residue containing reactive functionality can be prevented from reacting to form undesired products by suitably blocking the chelating residue precursor which can be contacted with the reactive poly (alkylene oxide) moiety to form the polymer, and then the blocking group can be subsequently removed by techniques known in the art. For example, if hydroxy substituents are to be selectively present in the final polymer, they preferably should be temporarily blocked during polymerization, such as by formation of an alkyl ether from the hydroxyl by conventional blocking techniques to minimize formation of undesirable by products. However, by products which contain one or more linkages formed by unblocked reactive precursor groups in the backbone of the polymer are contemplated to be useful.
Small proteins or peptides may be incorporated into the polymer by methods as described hereinbelow. An advantage of this chemistry is that the N and C terminus of the peptide can be reversed or randomized in the polymer of the invention, reducing
immunogenicity or masking peptide activity until the peptide is liberated.
Scheme A
Bis-(oxiranyl)-peptide monomers (Apep) are reacted with bis-(alkylamino)-PAG derivative monomers (Apag).
A linking group precursor is added to the PAG monomers at the terminal hydroxy. The reaction of the known linking group precursor with the known PAG moiety forms a (PAG)-linking group precursor radical. The precursor radical is conveniently chosen from
aminoalkylamino, N-sarcosyl-aminoalkyl-amino, or N-sarcosylaminoalkylamino-N'-carboxy.
In this scheme, the peptide monomers conveniently have 4-(oxiranylmethoxy) aryl radicals connected as linking group precursors using carboxy funtionality to attach to the N terminus of the peptide or amino functionality to attach to the C terminus of the peptide, thus forming amide bonds with the N terminus with the C terminus of the peptide monomer with the one end of each linking group precursor, and having an oxirane at the other end of each linking group
precursor as shown by the example below:
Figure imgf000029_0001
This oxirane functionalized peptide is referred to as Apep. As an example Apep can be;
Figure imgf000030_0002
and is combined with Bis (amino) PAG monomers (Apag) , such as :
Figure imgf000030_0001
wherein R is lower alkyl.
Such PAG derivatives are prepared by known
chemistry, for example; the preparation of an acid chloride from PAG monomers by SOCl2, COCl2 and the like, with subsequent reaction with a suitable diamine, or another suitable linking group, such as
-N(R)CH2CONHCH2CH2NH2, or the like.
Scheme B
Alternatively, oxiranyl functionality can be used on PAG derivative monomers while using amino
functionality on peptide derivative monomers. In this scheme, bis(alkylamino)-peptide monomers (Bpep) are reacted with bis-(oxiranyl)-PAG monomers (Bpag). The peptide has a linking group precursor radical attached to the C and N termini so as to provide terminal amine functionality. Glycine or sarcosine can be used as the linking group precursor for the N terminus. The C terminus is attached to a -NH(CH2)pNHCOCH2NH(R) or
-NH(CH2)DNHCOCH2NH2 linking group precursor radical which is derived from a diamine (wherein p is one to six, R is an alkyl radical, linear or branched, of 1 to about 4 carbons) and glycine or sarcosine. Thus the peptide is attached to the linking group precursor via amide linkages at both the N and C termini.
An example of Bpep is:
Figure imgf000031_0001
wherein p is 1 to about 6.
The bis (oxiranyl) PAG monomers (Bpag) of formula;
Figure imgf000031_0002
are known in the art. (See Y. Chen and M. Feng,
Chinese Patent 86/104,089 (1987)).
Thus it will be appreciated that the bis
(alkylamine) and bis (oxiranyl) functionality may be on either the PAG moiety or the peptide moiety; so long as the polymerization takes place between a peptide and PAG, using the reaction of an amine and an epoxide.
Before, during or after polymerization, suitable chelating agents and precursors thereof may be attached to the polymer or polymer precursor. As described previously, a suitably blocked progenitor to the chelating agent or precursor thereof containing
reactive functionality can be contacted with the reactive amino acid side chain incorporated into the polymer or polymer precursor to form the chelate-polymer or chelate polymer precursor, and then any blocking groups can be subsequently removed by
techniques known in the art, thus avoiding formation of undesired by products. The metallized polymer can be formed by contacting the unmetallized polymer sequentially or simultaneously with one or more sources of metal ions. This can be conveniently accomplished by adding one or more metal ion solutions or one or more metal ion solid salts or metal ion oxides, preferably sequentially, to a
solution, preferably an aqueous solution, of the polymer. Thereafter, or between sequential addition of metal ions, the chelated polymer preferably is
diafiltered in water to remove excess unbound metal.
The copolymer preferably is prepared in a water soluble, for example, an injectable form when used as magnetic resonance contrast agent for blood pool imaging, as a composition intended to be administered intravenously, and the like. The preparation of water-soluble compounds of molecular weight 10,000 to 50,000 can be accomplished by known methods by one skilled in the art.
Where the copolymer carries an overall charge, it will conveniently be used in the form of a salt with a physiologically acceptable counterion, for example an ammonium, substituted ammonium, alkali metal or
alkaline earth metal (eg. calcium) cation or an anion deriving from an inorganic or organic acid. In this regard, meglumine salts are particularly preferred.
In the compositions of the invention the copolymer may be formulated with conventional pharmaceutical or veterinary formulation aids, for example stabilizers, antioxidants, osmolality adjusting agents, buffers, pH adjusting agents, etc. and may be in a form suitable for parenteral or enteral administration, for example injection or infusion or administration directly or after dispersion or dilution with a physiologically tolerable medium, eg. water for injections into a body cavity having an external escape duct, for example the gastrointestinal tract, the bladder or the uterus.
Thus the compositions of the present invention may be in conventional pharmaceutical administration forms such as powders, solutions, suspensions, dispersions, etc; however, solutions, suspensions and dispersions in physiologically acceptable carrier media, for example water for injections, will generally be preferred.
The copolymers according to the invention may therefore be formulated for administration using physiologically acceptable carriers or excipients in a manner fully within the skill of the art. For example, the copolymers, optionally with the addition of
pharmaceutically acceptable excipients, may be
suspended or dissolved in an aqueous medium, with the resulting solution or suspensions then being
sterilized. Suitable additives include, for example, physiologically biocompatible buffers (as for example, tromethamine hydrochloride), additions (eg. 0.01 to 10 mole percent) of chelants (such as, for example, DTPA) or calcium chelate complexes (as for example calcium DTPA, CaNaDTPA-bisamide, or calcium salts) or,
optionally, additions (eg., 1 to 50 mole percent) of calcium of sodium salts (for example, calcium chloride, calcium ascorbate, calcium gluconate or calcium lactate and the like).
If the copolymers are to be formulated in
suspension form, eg., in water or physiological saline for oral administration, a small amount of soluble chelate may be mixed with one or more of the inactive ingredients traditionally present in oral solutions and/or surfactants and/or aromatics for flavouring.
For MRI and for X-ray imaging of some portions of the body the most preferred mode for administering metal chelates as contrast agents is parentral, eg., intravenous administration. Parenterally administrable forms, eg., intravenous solutions, should be sterile and free from physiologically unacceptable agents, and should have low osmolality to minimize irritation of other adverse effects upon administration, and thus the contrast medium should preferably be isotonic or slightly hypertonic. Suitable vehicles include aqueous vehicles customarily used for administering parenteral solutions such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection and other solutions such as are described in Remington's Pharmaceutical Sciences, 15th ed., Easton: Mack
Publishing Co., pp. 1405-1412 and 1461-1487 (1975) and The National Formulary XIV, 14th ed. Washington:
American Pharmaceutical Association (1975). The solutions can contain preservatives, antimicrobial agents, buffers and antioxidants conventionally used for parenteral solutions, excipients and other
additives which are compatible with the copolymers and which will not interfere with the manufacture, storage or use of products.
Where the copolymer comprises a chelated toxic metal species, eg. a heavy metal ion, it may be
desirable to include within the formulation a slight excess of a chelating agent, eg. as discussed by
Schering in DE-A-3640708, or more preferably a slight excess of the calcium salt of a chelating agent.
Actual levels of active ingredient in administered compositions of the present invention may be varied so as to obtain an amount of active ingredient that is effective to obtain the desired effect for a particular composition and method of administration. The selected dosage level therefore depends upon the desired effect, on the route of administration, on the desired duration of treatment and other commonly considered factors.
The dosages of the contrast agent used according to the method of the present invention will vary according to the precise nature of the contrast agent used. Preferably however, the dosage should be kept as low as is consistent with achieving contrast enhanced imaging and volumes minimized for IV drip or bolus injection. In this way, the toxicity potential is minimized.
For MR-diagnostic examination, the diagnostic agent of the present invention, if in solution, suspension or dispersion form, will generally contain the chelated metal at concentration in the range 1 micromole to 1.5 mole per litre, preferably 0.1 to 700mM. The composition may however be supplied in a more concentrated form for dilution prior to
administration.
For most MR contrast agents the appropriate dosage will generally range from 0.02 to 3 mmol paramagnetic metal/kg body weight, especially 0.05 to 1.5 mmol/kg, particularly 0.08 to 0.5, more especially 0.1 to 0.4 mmol/kg. It is well within the skill of the average practitioner in this field to determine the optimum dosage for any particular contrast agent for both in vivo or in vitro applications.
For X-ray examination, the dose of the contrast agent should generally be higher and for scintigraphic examination the dose should generally be lower than for MR examination. For radiotherapy and drug release therapy, conventional or sub conventional dosages may be used.
For cytotoxic therapy the total daily dose of the compounds of this invention administered to a host in single or divided dose may be in amounts, for example, of from about 1 picomol to about 10 millimoles of cytotoxic agent per kilogram of body weight. Dosage unit compositions may contain such amounts or such submultiples thereof as may be used to make up the daily dose. It will be understood, however, that the specific dose level for any particular patient will depend upon a variety of factors including the body weight, general health, sex, diet, time and route of administration, rates of absorption and excretion, combination with other drugs and the severity of the particular disease being treated.
Viewed from a further aspect, the present
invention provides a method of generating enhanced images of the human or non-human animal body, which method comprises administering to said body a
diagnostic composition according to the present
invention and generating an X-ray, MR, ultrasound or scintigraphic image of at least a part of said body into which said copolymer distributes.
Viewed from a further aspect, the present
invention provides a method of therapy practised on the human or non-human animal body, which method comprises administering to said body a therapeutically effective copolymer according to the invention, eg. one
incorporating a drug or prodrug or a therapeutically effective, eg. cytotoxic, chelated metal.
The present invention may include one or more of the polymers of this invention formulated into
compositions together with one or more non-toxic physiologically acceptable carriers, adjuvants or vehicles which are collectively referred to herein as carriers, for parenteral injection, for oral
administration in solid or liquid form, for rectal or topical administration, or the like.
The compositions can be administered to humans and animals either orally, rectally, parenterally
(intravenous, intramuscularly or subcutaneously), intracisternally, intravaginally, intraperitoneally, locally (powders, ointments or drops), or as a buccal or nasal spray.
Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions and sterile powders and lyophilizates for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and
nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of
dispersions and by the use of surfactants.
These compositions may also contain adjuvants such as preserving, wetting, emulsifying, cryoprotecting, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens,
chlorobutanol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or
carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, gelatin, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glylcerol, (d) disintegrating agents, as for example, agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate, (h)
adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate or mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethyleneglycols, and the like.
Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain
opacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions which can be used are polymeric substances and waxes.
The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions,
solutions, suspensions, syrups and elixirs. In
addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol,
tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan or mixtures of these substances, and the like. Besides such inert diluents, the composition can also include adjuvants, such as wetting agents,
emulsifying and suspending agents, sweetening,
flavoring and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents, as for example,
ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline
cellulose, aluminum metahydroxide, bentonite, agar and tragacanth, or mixtures of these substances, and the like.
Compositions for rectal or vaginal administrations are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt in the rectum or vaginal cavity and release the active component.
Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays and inhalants. The active component is admixed under sterile conditions with a physiologically
acceptable carrier and any preservatives, buffers or propellants as may be required. Ophthalmic
formulations, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
Viewed from a yet further aspect, the present invention also provides the use of the copolymers according to the invention for the manufacture of diagnostic or therapeutic agents for use in methods of image generation or therapy practised on the human or non-human animal body.
Viewed from a still further aspect, the present invention provides a process for the preparation of a chelated metal bearing copolymer, said process comprising metallating a chelating moiety containing copolymer according to the invention, eg. by admixing the copolymer in a solvent together with an at least sparingly soluble compound of the metal, for example a chloride, oxide, acetate or carbonate.
Viewed from a yet still further aspect, the present invention provides a process for the
preparation of a therapeutic copolymer according to the present invention, which comprises conjugating a drug or prodrug to a copolymer according to the invention.
The following non-limiting example illustrates the preparation of an example of a compound of formula A.
Example A
The synthesis of the compound prepared by Method
A, giving a compound of formula A is achieved according to the following scheme;
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
EXAMPLE A
Intermediate A 1. N-(N-(1,1-Dimethyl-ethoxycarbonyl)-phenylalanyl)leucine pentafluorophenylester. N-(N-(1,1-Dimethylethoxycarbonyl)-phenylalanyl)leucine (23.0 g, 61 mmol) (prepared by a literature method [Anderson, G.W.; McGregor, A.C., t-Butoxycarbonyl amino acids and their use in peptide synthesis, J. Am. Chem. Soc, 1957, 79, 6180-6183]) was stirred with
pentafluorophenol (11.2 g, 61 mmol) and
dicyclohexylcarbodiimide (12.5 g, 61 mmol) in
tetrahydrofuran (170 mL) for 1 hour at 0°C. The suspension was filtered. The solvent was evaporated from the filtrate under reduced pressure. The residue, in dichloromethane, was washed twice with saturated aqueous sodium hydrogen carbonate and with water. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure to give N-(N-(1,1-dimethylethoxycarbonyl)phenylalanyl)-leucinepentafluorophenyl ester (28.0 g, 85%).
Intermediate B
1. 4(Phenylmethoxy)benzoic acid. In a modification of the literature method [E.L. Elied, R.P. Anderson, Reactions of esters with targeting amines. I. Benzyl esters from methyl esters and benzyldimethylamine, J. Am. Chem. Soc, 1952, 74, 547-549] a mixture of 4-hydroxybenzoic acid (27.6 g, 200 mmol), chloromethyl- benzene (57.0 g, 450 mmol), potassium carbonate (50 g) and sodium iodide (25 g) was boiled under reflux in acetonitrile (500 mL) for 16 hours. The suspension was filtered and the solvent was evaporated from the filtrate under reduced pressure. The residue was recrystallised from ethanol to give phenylmethyl 4- (phenylmethoxy)benzoate (48.8 g, 76%). Phenylmethyl 4- (phenylmethoxy)benzoate (48.8 g, 150 mmol) was boiled under reflux with aqueous sodium hydroxide (2 M; 250 mL) and ethanol (250 mL) for 4 hours. The ethanol was evaporated under reduced pressure. Water (1000 mL) was added. The white solid was collected by filtration, warmed to 65°C with aqueous sulphuric acid (2 M; 300 mL) for 1 hour and extracted with warm ethyl acetate. The ethyl acetate solution was dried with anhydrous
magnesium sulphate and the solvent was evaporated under reduced pressure to give 4-(phenylmethoxy) benzoic acid
(27.15 g, 80%). The filtrate was washed twice with diethyl ether, acidified by addition of sulphuric acid
(2 M) and extracted with diethyl ether. Evaporation of the diethyl ether gave a further portion of 4-(phenylmethoxy)benzoic acid (6.0 g, 18%). The total yield was 98%.
2. 4-(Phenylmethoxy)benzoyl chloride. 4-(Phenylmethoxy)benzoic acid (500 mg, 2.2 mmol) was stirred with oxalyl chloride (280 mg, 2.2 mmol) and
dimethylformamide (25 mg) in 1,4-dioxan (25 mL) for 20 minutes. The solvent and catalyst were evaporated under reduced pressure. The residue was recrystallised from hexanes to give 4-(phenylmethoxy)benzoyl chloride (460 mg, 85%).
3. N-(4-(Phenylmethoxy)benzoyl)glycine methyl ester. 4-(Phenylmethoxy)benzoyl chloride (13.64 g, 55.5 mmol) in dichloromethane (90 mL) was added dropwise to glycine methyl ester hydrochloride (7.66 g, 61 mmol) and triethylamine (11.78 g, 116.5 mmol) in
dichloromethane (250 mL). The mixture was stirred for 16 hours. The suspension was filtered. The solvent was evaporated from the filtrate under reduced
pressure. The residue was recrystallised from
dichloromethane/hexane to give N-(4- (phenylmethoxy)benzoyl)glycine methyl ester (14.75 g, 89%).
4. N-(4-(Phenylmethoxy)benzoyl)glycine
pentafluorophenyl ester. N-(4-(Phenylmethoxy)benzoyl)-glycine methyl ester (14.75 g, 49.2 mmol) was boiled under reflux with methanolic sodium hydroxide (1M) (80 mL) for 2 hours. The solvent was evaporated under reduced pressure. The residue was dissolved in water and was acidified by addition of aqueous hydrochloric acid. The suspension was extracted with ethyl acetate. The extract was washed with saturated brine and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure to give N-(4-(phenylmethoxy)benzoyl)glycine (6.59 g, 47%).
Dicyclohexylcarbodiimide (720 mg, 3.5 mmol) was added to N-(4-(phenylmethoxy)benzoyl)glycine (100 g, 3.5 mmol) in dry tetrahydrofuran (100 mL) and the mixture was taken to 0°C. Pentafluorophenol (640 g, 3.5 mmol) was added dropwise and the mixture was stirred for 17 hours at 0°C. The suspension was filtered and the solvent was evaporated from the filtrate under reduced pressure. The residue was dissolved in ethyl acetate (200 mL) and was washed with saturated aqueous sodium hydrogen carbonate (2 × 75 mL), with aqueous sulphuric acid (10%) and with water. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure to give N-(4- (phenylmethoxy)benzoyl)glycine pentafluorophenyl ester (Intermediate B) (1.5 g, 95%).
Intermediate C
1. 1-(2-Nitroethenyl)-4-(phenylmethoxy)benzene. In a modification of the literature method [M. Hoequanx, B. Macot, G. Recleuith, C. Viel, M. Brunaub, J. Nauamo, C. Lacoun and C. Cozeubon, Diazoestrones and analogs. I. Pharmacological study and syntheses of heterosteroid analogs to establish structure analgesic activity relationships, Eur. J. Med. Chem., 1983, 18, 319-329], to 4-(phenylmethoxy)benzaldehyde (28 g, 132 mmol) in ethanol (900 mL) at 5°C was added nitromethane (16.1 g, 264 mmol). Sodium hydroxide (13.2 g, 330 mmol) in ethanol (200 mL) was added dropwise and the mixture was stirred for 30 minutes at 5°C. The mixture was poured into a mixture of hydrochloric acid (9 M; 136 mL) and water (208 mL)). The precipitate was collected by filtration and was recrystallised from ethanol to give 1-(2-nitroethenyl)-4-(phenylmethoxy)benzene (14.0 g, 42%). 2. 2-(4-(Phenylmethoxy)phenyl)ethylamine. Lithium aluminum hydride (8.48 g, 223 mmol) was suspended in dry diethyl ether (600 mL). 1-(2-Nitroethenyl)-4-(phenylmethoxy)benzene (13.9 g, 55 mmol) was extracted into this mixture using a Soxhlet apparatus. The mixture was boiled under reflux for 16 hours. Water (7.38 mL) was added, followed by aqueous sodium
hydroxide (20%, 5.53 mL) and water (27.8 mL). The suspension was filtered. The solvent was evaporated from the filtrate under reduced pressure to give 2-(4-(phenylmethoxy)phenyl)-ethylamine (11.25 g, 91%).
Intermediate P
1. N-(1,1-Dimethylethoxvcarbonyl)glycine N-(2-(4-phenylmethoxy)phenyl)ethyl)amide. N-(1,1- Dimethylethoxy-carbonyl)glycine (850 mg, 4.85 mmol) was stirred with dicyclohexylcarbodiimide (1.00 g, 4.85 mmol) and 2-(4-(phenylmethoxy)phenyl)ethylamine
(Intermediate C) (1.00 g, 4.4 mmol) in dry
tetrahydrofuran (30 mL) for 16 hours. The suspension was filtered and the solvent was evaporated from the filtrate under reduced pressure. The residue was dissolved in ethyl acetate and was washed with aqueous sulphuric acid (10%) and with saturated brine. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure to give N-(1,1-dimethylethoxycarbonyl)glycine N-(2-(-(phenylmethoxy)phenyl)ethyl)amide (1.65 g, 98%).
2. Glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)amide. N-(1,1-Dimethylethoxycarbonyl) glycine N-(2-(4-(phenyl)-ethyl)amide (2.01 g, 5.23 mmol) was treated with excess hydrogen chloride in 1,4-dioxan (45 mL) for 2 hours. The solid was collected by filtration and was dissolved in water and ethyl acetate. Aqueous sodium hydroxide was added to basify the solution to pH 9. The ethyl acetate solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure to give glycine N-(2-(4-(phenylmethoxy)-phenyl)ethyl)amide (1.15 g, 77%). 3. N-(N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanyl)-leucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)-amide. N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanyl)-leucine pentafluorophenyl ester (1.19 g, 2.18 mmol) (Example A, Intermediate A) in tetrahydrofuran (30 mL) was added dropwise to glycine N-(2-(4-(phenylmethoxy)-phenyl)ethyl)amide (620 mg, 2.18 mmol), N,N-diisopropylethylamine (310 mg, 2.4 mmol) and 1-hydroxybenzotriazole (20 mg) in tetrahydrofuran (30 mL) and the mixture was stirred for 16 hours. The solvent was evaporated under reduced pressure. The residue, in ethyl acetate, was washed with aqueous sulphuric acid (10%) and with saturated aqueous sodium hydrogen carbonate. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure. The residue was triturated with diethyl ether and the solid was collected by filtration to give N- (N- (N- (1 , 1 -dimethylethoxy-carbonyl)- phenylalanyl)leucyl)glycine N-(2-(4- (phenylmethoxy)phenyl)-ethyl)amide (360 mg, 26%)
(Intermediate D). Intermediate E
1. N-(1,1-Dimethylethoxycarbonyl)sarcosine 2,4,5-trichlorophenyl ester. N-(1,1-Dimethylethoxycarbonyl)-sarcosine (10.0 g, 53 mmol) was stirred with 2,4,5-trichlorophenol (10.6 g, 53 mmol) and
dicyclohexylcarbodiimi.de (10.9 g, 53 mmol) in ethyl acetate (100 mL) at -10°C for 2.5 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was
filtered and the solvent was evaporated under reduced pressure from the filtrate to give N-(1,1-dimethyl-ethoxycarbonyl)sarcosine 2,4,5-trichlorophenyl ester (19.3 g, 98%).
2. N-(1,1-Dimethylethoxycarbonyl)sarcosine N-(2-aminoethyl)amide. N-(1,1-Dimethylethoxycarbonyl)-sarcosine 2,4,5-trichlorophenyl ester (12.7 g, 34.5 mmol) in dichloromethane (50 mL) was added during 30 minutes to ethane-1,2-diamine (20.7 g, 345 mmol) in dichloromethane (150 mL) and the solution was stirred for a further 2 hours. The solution was washed with water and with 10% aqueous sodium carbonate and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure to give N-(1,1-dimethylethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (6.9 g, 86%).
3. Bis(2-( 2 -(N-(2-N-(1,1-Dimethylethoxycarbonyl)- sarcosyl)aminoethyl)-aminocarhoxy)ethoxy)ethoxy)ethane. Bis(2-hydroxyethoxy)ethoxy)ethane (5.0 g, 21 mmol) was boiled in toluene (120 mL) for 20 hours with azeotropic removal of water. The resulting solution was cooled to 20°C. Dichloromethane (35 mL) was added, followed by phosgene (1.93 M in dichloromethane, 109 mL, 210 mmol). The solution was stirred for 4 hours. The solvent and excess reagent were evaporated under reduced pressure from a portion (30 mL) of this solution to give crude bis(2-(2-(chlorocarboxy)ethoxy)ethoxy)ethane (900 mg, 2.5 mmol). This material was dissolved in
dichloromethane (50 mL). To this solution was added triethylamine (1.26 g, 12.5 mmol) and 4- (dimethylamino)pyridine (20 mg). N-(1,1-dimethylethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (1.73 g, 7.5 mmol) (Intermediate E2) in dichloromethane (100 mL) was then added dropwise during 40 minutes.
The solution was stirred for 20 hours before being washed with water, 10% aqueous sulphuric acid and water. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure to give bis(2-(2-(N-(2-(N-(1,1 dimethylethoxycarbonyl)sarcosyl)aminoethyl)aminocarboxy)ethoxy)-ethoxy)-ethane (1.1 g, 59%).
4. Bis(2-(2-(N-(2-sarcosylaminoethyl)aminocarboxy)-ethoxy)ethoxy)ethane dihydrochloride. Bis(2-(2-(N-(2- (N-(1,1-dimethylethoxycarbonyl)sarccsyl)aminoethyl)-aminocarboxy)ethoxy)ethoxy)ethane (752 mg, 1 mmol) was treated with excess hydrogen chloride in
dichloromethane for 2 hours. Evaporation of the solvent gave bis (2- (2- (N- (2-sarcosylaminoethyl) amino-carboxy) ethoxy) ethoxy) ethane dihydro-chloride (550 mg, quantitative).
Preparation of the peptide portion of Example A 1. N-(N-Phenylalanylleucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)amide hydrochloride. N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanylleucyl)glycine N-(2- (4-(phenylmethoxy) phenyl)ethyl)amide (3.89 g, 6.05 mmol) was treated with excess hydrogen chloride in dichloromethane (200 mL) for 3 hours. The solvent and excess reagent were evaporated under reduced pressure. The residual oil was triturated with diethyl ether to give N-(N-phenylalanylleucyl)glycine N-(2-(4-(phenyl methoxy) phenyl) ethyl) amide hydrochloride (3.26 g, 93%).
2. N-(N-(N-(N-(4-(Phenylmethoxy)benzoyl)glycyl)-Phenylalanyl)leucyl)glycine N-(2-(4-(phenylmethoxy)-phenyl)ethyl)amide. N-(N-Phenylalanylleucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)amide hydrochloride (165 mg, 284 mol) was stirred with N,N-diisopropylethyl amine (100 mg, 774 mol), 4-(dimethylamino)pyridine (10 mg) and 1-hydroxybenzotriazole (10 mg) in dry
dichloromethane (5 mL) until all solid dissolved. N-(4-(Phenylmethoxy)benzoyl) glycine pentafluorophenyl ester (117 mg, 258 mol) (Example A, Intermediate B) in chloroform (10 mL) was added dropwise during 30 minutes and the reaction mixture was stirred for 5 hours. The solvent was evaporated under reduced pressure. Column chromatography (silica gel, chloroform/methanol 50:1) of the residue gave N-(N-(N-(N-(4-(Phenylmethoxy)-benzoyl)glycyl)phenylalanyl)leucyl)-glycine N-(2-(4- (phenylmethoxy)phenyl)ethyl)amide (170 mg, 81%).
3. N-(N-(N-(N- (4-Hydroxyhenzoy )glycyl)phenylalanyl)-leucyl)glycine N-(2-(4-hydroxyphenyl)ethyl)amide. N-(N- (N-(N-(4-(Phenylmethoxy)benzoyl)glycyl)phenyalanyl)-leucyl)glycine N-(2-(4-(phenylmethoxy)phenyl)ethyl)-amide (444 mg, 547 mol) in ethanol (45 mL) was stirred vigorously with palladium on charcoal (10%; 50 mg) and hydrogen for 12 hours. The suspension was filtered through diatomaceous earth. The solvent was evaporated from the filtrate under reduced pressure to give N-(N- (N-(N-(4-hydroxybenzoyl)glycyl)phenylalanyl)leucyl)-glycine N-(2-(4-hydroxyphenyl)ethyl)-amide (304 mg, 88 % ) .
4. N-(N-(N-(N-(4-(Oxiranylmethoxy)benzoyl)glycyl)-phenylalanyl)leucyl)glycine N-(2-(4-(oxiranylmethoxy)-phenyl)ethyl)amide. N-(N-(N-(N-(4-Hydroxybenzoyl)-glycyl)phenylalanyl)leucyl)glycine N-(2-(4-hydroxyphenyl)ethyl)amide (106 mg, 0.131 mol) was suspended in water (12 mL) containing sodium hydroxide (52.3 mg, 1.31 mmol). Chloromethyloxirane (604 mg, 6.5 mmol) in methanol (10 mL) was added, followed by phenylmethyltrimethylammonium hydroxide (40% aqueous solution, 90 mg). The solution was stirred for 48 hours at 40°C. The solvent and excess reagent were evaporated under reduced pressure. The residue was dissolved in ethyl acetate and was washed with water. The solution was dried with anhydrous magnesium
sulphate. The solvent was evaporated under reduced pressure. Column chromatography (silica gel; ethyl acetate, then echyl acetate/methanol 39:1, then ethyl acetate/methanol 19:1, then ethyl acetate/methanol 9:1) gave N-(N-(N-(N-(4-(oxiranylmethoxy)benzoyl)glycyl)-phenylalanyl)leucyl)glycine Ν-(2-(4(oxiranyl-methoxy)-phenyl)ethyl)amide (26.5 mg, 27%). 5. Polymer A. It is contemplated that Ν-(Ν-(Ν-(Ν-(4- (Oxiranylmethoxy)benzoyl)-glycyl)phenylalanyl)leucyl)-glycine Ν-(2-(4-(oxiranylmethoxy)phenyl)ethyl)amide is boiled under reflux with anhydrous sodium carbonate and bis(2-(2-(Ν-(2-sarcosylaminoethyl)aminocarboxy)ethoxy)-ethoxy)ethane dihydrochloride (Intermediate E) in ethanol for 6 hours, giving the polymer of formula A.
Figure imgf000053_0001
Figure imgf000054_0001
EXAMPLE B
PREPARATION OF INTERMEDIATES
Intermediate A N-(1,1-Dimethylethoxycarbonyl)leucine 2,4,5-trichlorophenyl ester. N-(1,1-Dimethylethoxycarbonyl)leucine (6.32 g, 15 mmol) was stirred with 2,4, 5-trichlorophenol (3.01 g, 15.2 mmol) and
dicyclohexylcarbodiimide (3.14 g, 15.2 mmol) in ethyl acetate (50 mL) at -10°C for 4 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was
filtered and the solvent was evaporated under reduced pressure from the filtrate to give N-(1,1-dimethylethoxycarbonyl) leucine 2,4,5-trichlorophenyl ester (6.2 g, 99%).
Intermediate B N-(1,1 Pimethylethoxycarbonyl)-phenylalanine pentafluorophenyl ester. N-(1,1-Dimethylethoxycarbonyl)phenylalanine (6.36 g, 24 mmol) in ethyl acetate (50 mL) at 0°C was added to
dicyclohexylcarbodiimide (4.95 g, 24 mmol) and
pentafluorophenol (4.42 g, 24 mmol) in ethyl acetate (50 mL) at 0°C. The mixture was stirred for 2.75 hours at 0°C. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate to give N-(1 , 1-dimethylethoxycarbonyl)phenylalanine pentafluorophenyl ester (10.32 g, quantitative).
Intermediate C N-(1,1-Dimethylethoxycarbonyl)glycine
2,4,5-trichlorophenyl ester. N-(1,1-Dimethylethoxycarbonyl)glycine (6.12 g, 35 mmol) was stirred with 2, 4, 5-trichlorophenol (6.91 g, 35 mmol) and
dicyclohexylcarbodiimide (7.22 g, 35 mmol) in ethyl acetate (100 mL) at 0°C for 4 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate to give N-(1,1-dimethylethoxycarbonyl)glycine 2,4,5-trichlorophenyl ester (12.4 g, quantitative).
Intermediate D
1. Nα-(1.1-Dimethylethoxycarbonyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)lysine. In a modification of a literature method [Yajima, H.; Watanabe, H.; Okamoto, M., Studies on peptides. XXXIII. Nϐβ,β,β-Trichloroethyloxycarbonyllysine, Chem. Pharm. Bull,
1971, 19, 2185-2189], lysine monohydrochloride (9.14 g, 50 mmol) was stirred under reflux with copper (II) carbonate (21.6 g, 75 mmol) in water (180 mL) for 3 hours. The solution was filtered while hot and the filtrate was cooled to 20ºC. 2,2,2-Trichloroethyl chloroformate (15.9 g, 75 mmol) and aqueous sodium carbonate (13.3 g, 125 mmol in 40 mL) were added alternately in portions to the filtrate during 30 minutes and the mixture was stirred vigorously at 0ºC for 20 hours. The blue precipitate was collected and was boiled under reflux with ethylenediaminetetraacetic acid disodium salt (18.6 g, 100 mmol) in water (200 mL) for 2 hours. The solution was cooled to 0°C for 20 hours and crude Nϐ-(2,2,2- trichloroethoxycarbonyl)-lysine was collected as a gummy solid. This material was dissolved in water (75 mL) and triethylamine (20.2 g, 200 mmol) was added, followed by di-t-butyl
dicarbonate (13.64 g, 62 mmol) and 1,4-dioxan (30 mL). The mixture was stirred vigorously for 3 days. The mixture was washed with diethyl ether. Ethyl acetate was added to the aqueous phase and the mixture was acidified by careful addition of cold 10% aqueous sulphuric acid. The ethyl acetate phase was washed with water and dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure to give Nα-(1,1-dimethylethoxycarbonyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)lysine (12.32 g, 55%).
2. Nα-(1,1-Dimethylethoxycarbonyl)-Nϐ(2,2,2-trichloroethoxycarbonyl)lysine 2,4,5-trichlorophenyl ester. Nα-(1,1- Dimethylethoxycarbonyl)-Nϐ-(2,2 , 2 -trichloroethoxy-carbonyl)lysine (6.32 g, 15 mmol) was stirred with 2, 4, 5-trichlorophenol (2.96 g, 15 mmol) and dicyclohexylcarbodiimide (3.10 g, 15 mmol) in ethyl acetate (100 mL) at 0°C for 20 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was
filtered and the solvent was evaporated under reduced pressure from the filtrate to give Nα-(1,1-dimethylethoxycarbonyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)lysine 2,4,5-trichlorophenylester (7.50 g, 97%).
Intermediate E 1. N-(Phenylmethoxycarbonyl)sarcosine 2,4,5-trichlorophenylester. N-(Phenylmethoxycarbonyl)-sarcosine (4.0 g, 18 mmol) was stirred with 2,4,5-trichlorophenol (3.53 g, 18 mmol) and
dicyclohexylcarbodiimide (3.69 g, 18 mmol) in ethyl acetate (40 mL) at -10°C for 1 hour, then at 20°C for 20 hours. The suspension was cooled to 0°C. The
suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate to give N-(phenylmethoxycarbonyl) sarcosine 2,4,5-trichlorophenyl ester (7.2 g, quantitative).
2. N-(Phenylmethoxycarbonyl)sarcosine
pentafluorophenyl ester. Ν-(Phenylmethoxycarbonyl)-sarcosine (3.0 g, 13.4 mmol) was stirred with
pentafluorophenol (2.46 g, 13.4 mmol) and
dicyclohexylcarbodiimide (2.32 g, 13.4 mmol) in ethyl acetate (30 mL) at 0°C for 2 hours. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate. The residue was dissolved in ethyl acetate. The suspension was filtered and the solvent was evaporated under reduced pressure from the filtrate to give Ν-(phenylmethoxycarbonyl)sarcosine pentafluorophenyl ester (4.66 g, 89%).
Intermediate F
1. 5-(4-Νitrophenyl)-10,15,20-triphenyl-21H,23H-porphine. Fuming nitric acid (density 1.5 mL-1) (2.26 mL) was added during 2 hours to 5,10,15,20-tetraphenyl-21H,23H-porphine (2.00 g, 3.26 mmol) in chloroform (ethanol-free) (300 mL). The mixture was washed with water (5 × 300 mL) and was dried with anhydrous sodium carbonate and anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure.
Chromatography (silica gel; dichloromethane/hexane 2:1) of the residue gave 5-(4-nitrophenyl)-10,15,20-triphenyl-21H,23H-porphine (1.17 g, 55%). 2. 4-(10,15,20-Triphenyl-21H,23H-porphin-5-yl)-benzeneamine. Tin(II) chloride dihydrate (595 mg, 2.6 mmol) was added to 5-(4-nitrophenyl)-10,15,20-triphenyl-21H,23H-porphine (580 mg, 0.88 mmol) in aqueous hydrochloric acid (9 M. 20 mL) and the mixture was stirred at 65ºC for 2 hours. The solution was allowed to cool and was added to water (70 mL).
Concentrated aqueous ammonia was added until the solution was basified to pH 8. The suspension was extracted with chloroform (9 × 75 mL). The chloroform fractions were combined and were dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure. Chromatography (silica gel;
dichloromethane/hexane 5:1) of the residue gave 4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)benzeneamine (462 mg, 84%). 3. 4-Oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5- yl)phenylamino)butanoic acid. 4-(10,15,20-Triphenyl-21H,23H-porphin-5-yl)benzeneamine (450 mg, 0.72 mmol) was dissolved in chloroform (ethanol-free) (10 mL) with warming. Succinic anhydride (tetrahydrofuran-2,5-dione) (64 mg, 0.72 mmol) was added and the mixture was boiled under reflux for 2.5 hours. A further portion of succinic anhydride (32 mg, 0.36 mmol) was added and boiling under reflux continued for a further 2 hours. The mixture was cooled to ambient temperature for 16 hours. The precipitated solid was collected by
filtration to give 4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoic acid (460 mg, 89%). Preparation of Peptide Portion of Example B
1. N-(Phenylmethoxycarbonyl)sarcosine N-(2-aminoethyl)amide. N-(Phenylmethoxycarbonyl)sarcosine pentafluorophenyl ester (3.5 g, 9.2 mmol) in
dichloromethane (40 mL) was added during 30 minutes to ethane-1,2-diamine (10.8 g, 180 mmol) in
dichloromethane (300 mL) and the solution was stirred for a further 2 hours. The solution was washed with water and with 10% aqueous sodium carbonate and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure to give N- (phenylmethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (2.1 g, 88%). This material was also prepared
similarly from N-(phenylmethoxycarbonyl)sarcosine, 2,4,5-trichlorophenyl ester. 2. N-(1,1-Dimethylethoxycarbonyl)glycine N-(2-(N-phenylmethoxycarbronyl)sarcosylamino)ethyl)amide. N-(Phenylmethoxycarbonyl)sarcosine N-(2-aminoethyl)amide (3.71 g, 14 mmol) was stirred with N-(1,1-dimethyl¬ethoxycarbonyl)glycine 2,4,5-trichlorophenyl ester (4.96 g, 14 mmol, Example B, Intermediate C) and N,N-diisopropylethylamine (1.99 g, 15.4 mmol) in
dichloromethane (100 mL) for 20 hours. The solution was washed with cold 10% aqueous sulphuric acid (2 x) and with saturated aqueous sodium hydrogen carbonate and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure.
Chromatography (silica gel; ethyl acetate/methanol 10:1, then ethyl acetate/methanol 5:1, then ethyl acetate/methanol 3:1) of the residue gave N-(1,1-dimethylethoxycarbonyl)glycine N-(2-(N-phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (2.12 g, 37%).
3. Glycine N-(2-(N-phenylmethoxycarbonvl)sarcosylamino)ethyl)amide hydrochioride. N-(1,1-Dimethylethoxycarbonyl)glycine N-(2-(N-phenylmethoxycarbonyl)-sarcosylamino)ethyl)amide (2.04 g, 4.95 mmol) was treated with excess hydrogen chloride in
dichloromethane (50 mL) for 1 hours. The solvent and excess reagent were evaporated under reduced pressure to give glycine N-(2-(N-phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride (1.5 g, quantitative).
4. N-(N-(1,1-Dimethylethoxycarbonyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)-amide. N-(1,1-Dimethylethoxycarbonyl)glycine N-(2-(N- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (5.22 g, 8 mmol) was treated with excess hydrogen chloride in dichloromethane (50 mL) for 1 hour. Water (50 mL) was added and the mixture was stirred vigorously for 15 minutes. The solvent and excess reagent were
evaporated from the aqueous layer under reduced
pressure to give crude glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride as a white solid. This material was stirred with N,N-diisopropylethylamine (3.231 g, 25 mmol) and N-(1,1-dimethylethoxycarbonyl)leucine 2,4,5-trichlorophenyl ester (3.19 g, 7.8 mmol) (Example II, Intermediate A) in dimethylformamide (30 mL) for 3 days. The solvent was evaporated under reduced pressure. The residue was dissolved in ethyl acetate and was washed with aqueous sodium hydroxide (5%), aqueous sulphuric acid (10%) and water and was dried with anhydrous magnesium sulphate. Evaporation of the solvent under reduced pressure gave N-(N-(1,1-dimethylethoxycarbonyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (3.26 g, 78%).
5. N-Leucylglycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)-ethyl)amide hydrochloride. N-(N-(1,1-Dimethylethoxycarbonyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (3.26 g, 6.1 mmol) was treated with excess hydrogen chloride in dichloromethane (40 mL) for 1 hour. The solvent and excess reagent were evaporated under reduced pressure to give N-leucylglycine N-(2-(N-(phenylmethoxycarbonyl) sarcosylamino) ethyl) amide hydrochloride (2.65 g, quantitative).
6. N-(N-(N-(1,1 Dimethylethoxycarbonyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)esthyl)amide. N-(1,1-Dimethylethoxycarbonyl)phenylahanine pentafluorophenyl ester (2.65 g, 6.1 mmol) (Example B, Intermediate B) was added to N-leucylglycine N-(2-(N- (phenylmethoxycarbonyl)- sarcosylamino) ethyl) amide hydrochloride (2.81 g, 6.16 mmol), N-N-diisopropylethylamine (1.75 g, 13.5 mmol) and 4-(dimethylamino)pyridine (10 mg) in
dichloromethane (30 mL) and the mixture was stirred for 2 days. The solution was then washed with cold aqueous sulphuric acid (10%), aqueous sodium carbonate (10%) and saturated brine. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure. Chromatography (silica gel; chloroform/methanol 1:1) gave N-(N-(N- (1,1-dimethylethoxycarbonyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)-amide (1.94 g, 46%). 7. N-(N-Phenylalanylleucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amidehydrochloririe. N-(N-(N-(1,1-Dimethylethoxycarbonyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.94 g, 2.8 mmol) was treated with excess hydrogen chloride in dichloromethane (25 mL) for 1 hour. The solvent and excess reagent were evaporated under reduced pressure. The residue was dissolved in methanol. Evaporation of the solvent under reduced pressure gave N-(N-phenylalanylleucyl)glycine N-(2-(N- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride (1.67 g, 95%).
8. N-(N-(N-(N-(1,1-Dimethylethoxycarbonyl)glycyl)-phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxy-aarbonyl)sarcosylamino)ethyl)amide. N-(1,1- Dimethylethoxycarbonyl)glycine 2,4,5-trichlorophenyl ester (1.58 g, 2.55 mmol) (Example B, Intermediate C) and 4-(dimethylamino)pyridine (3.1 g, 2.5 mmol) were added to N-(N-phenylalanylleucyl)glycine N- (2 - (N- (phenylmethoxycarb-onyl)sarcosylamino)ethyl)amide hydrochloride (904 mg, 2.55 mmol) and N,N-diisopropylethylamine (990 mg, 7.7 mmol) in dichloromethane (20 mL). The mixture was stirred for 4 days. The solution was washed with cold aqueous
sulphuric acid (10%), aqueous sodium carbonate (10%) and saturated brine. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure. Chromatography (silica gel; chloroform, then chloroform/methanol 10:1) of the residue gave N-(N-(N-(N-(1,1-dimethylethoxycarbonyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.14 g, 61%).
9. N-(N-(N-glycylphenylalanyl)leucyl)glycine N-(2- (N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride. N-(N-(N-(N-(1,1 Dimethylethoxycarbonyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.29 g, 1.77 mmol) was treated with excess hydrogen chloride in dichloromethane (10 mL) for 1 hour. Methanol (1 mL) was added and the solvents and excess reagents were evaporated under reduced pressure to give N-(N-(N-glycylphenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino) ethyl) amide hydrochloride (1.1 g, quantitative).
10. N-(N-(N-(N-(Nα-(1.1-Dimethylethoxycarbonyl)-Nϐ- (2.2.2-trichloroethoxy-carbonyl)lysyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)ethyl)amide. N-(N-(N-Glycylphenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)ethyl)amide hydrochloride (1.11 g, 1.77 mmol) was added to N,N-diisopropylethylamine (683 mg, 5.3 mmol) in dichloromethane (10 mL). To this mixture was added Nα-(1,1-dimethylethoxycarbonyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)lysine 2,4,5-trichlorophenyl ester (950mg, 1.77 mmol) (Example B, Intermediate D) in dichloromethane (20 mL) and 4-(dimethylamino)pyridine (10 mg). The mixture was stirred for 3 days. The solution was washed with cold aqueous sulphuric acid (10%) , aqueous sodium carbonate (10%) and saturated brine. The solution was dried with anhydrous magnesium sulphate and the solvent was evaporated under reduced pressure. Chromatography (silica gel; chloroform, then chloroform/methanol 10:1) of the residue gave N-(N-(N- (N-(Nα-(1,1- dimethylethoxycarbonyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)- lysyl)glycyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)-sarcosylamino)ethyl)amide. (1.44 g, 78%).
11. N-(N-(N-(N-(Nϐ-(2,2,2-Trichloroethoxycarbonyl)-lysyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(Ν- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride. Ν-(Ν-(Ν-(Ν-(Nα(1,1-Dimethylethoxycarbonyl)-Νϐ-(2,2,2- trichloroethoxycarbonyl)lysyl)-glycyl)phenylalanyl)leucyl)glycine Ν-(2-(Ν-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (1.32 g, 1.27 mmol) was treated with excess hydrogen chloride in dichloromethane (20 mL) for 1 hour. Methanol (1.0 mL) was added and the mixture was filtered. The solvent was evaporated from the filtrate under reduced pressure to give Ν-(Ν-(Ν-(Ν-(Nϐ-(2,2,2-trichloroethoxycarbonyl)-lysyl)glycyl)phenylalanyl)leucyl)glycine Ν-(2-(Ν- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide
hydrochloride (1.14 g, 92%).
12. Ν-(Ν-(Ν-(N- (Nα-(Ν-(Phenylmethoxycarbony])sarcosyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)Iysyl)glycyl)phenylalanyl)-leucyl)glycine Ν-(2-(Ν-(phenylmethoxycarbonyl) -sarcosylamino)ethyl)amide. Ν-(Ν-(Ν-(Ν-(Nϐ-(2 , 2 , 2 -Trichloroethoxycarbonyl)lysyl)glycyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide hydrochloride (980 mg, 1.0 mmol) was stirred with N,N-diisopropylethylamine (402 mg, 3.1 mmol), N-(phenylmethoxycarbonyl)sarcosine 2,4,5- trichlorophenyl ester (418 mg, 1.0 mmol) (Example B, Intermediate E) and 4- (dimethylamino) pyridine (10 mg) in dichloromethane (30 mL) for 24 hours. The solution was washed with saturated aqueous sodium hydrogen carbonate and with aqueous sulphuric acid (2M) and was dried with anhydrous magnesium sulphate. The solvent was evaporated under reduced pressure. Chromatography (silica gel, chloroform/methanol 20:1, then
chloroform/methanol 10:1) of the residue gave N-(N-(N- (N-(ISP-(N-(phenylmethoxycarbonyl)sarcosyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)lysyl)glycyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide (418 mg, 36%). 13. N-(N-(N-(N-(Nα-(N-(Phenylmethoxycarbonyl)sarcosyl)-lysyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N- (phenvl-methoxycarbonyl)sarcosylamino)ethyl)-amide. It is contemplated that N-(N-(N-(N-(Nα-(N-(Phenylmethoxycarbonyl)sarcosyl)-Nϐ-(2,2,2-trichloroethoxycarbonyl)-lysyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N- (phenylmethoxycarbonyl)sarcosylamino)ethyl)amide is boiled under reflux with zinc powder in methanol for 2 hours. The solvent is evaporated under reduced
pressure. Ethyl acetate is added to the residue. The suspension is filtered and the filtrate is washed twice with water. The solution is dried with anhydrous magnesium sulphate and the solvent is evaporated under reduced pressure to give N-(N-(N-(N-(KP-(N-(phenylmethoxycarbonyl)sarcosyl)lysyl)glycyl)phenylalanyl)-leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)amide.
14. N-(N-(N-(N-(N-(N-(Phenylmethoxycarbonyl)sarcosyl)-N-(4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)-phenylamino)butanovl)lysyl)glycyl)phenylalanyl)- leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)-amide. It is contemplated that 4-oxo-4- (4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoic acid (Example B Intermediate E 3) is stirred with pentafluorophenol and
dicyclohexylcarbodiimide in dimethylformamide for 16 hours at 4°C. The suspension is filtered and the filtrate was added to N-(N-(N-(N-(N-(N-(phenylmethoxycarbonyl)sarcosyl)lysyl)glycyl)phenylalanyl)leucyl)-glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)-ethyl)amide and 4-(dimethylamino) pyridine in
tetrahydrofuran. The mixture is stirred for 2 days. Ethyl acetate is added and the solution is washed thrice with water, twice with 10% aqueous sodium carbonate solution and once with saturated brine. The solution is dried with anhydrous magnesium sulphate and the solvent is evaporated under reduced pressure.
Chromatography (silica gel) of the residue gives N-(N- (N-(N-(N-(N-(phenylmethoxycarbonyl)sarcosyl)-N-(4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoyl)lysyl)glycyl)phenylalanyl)leucyl)glycine N-(2-(N-(phenylmethoxycarbonyl)sarcosylamino)ethyl)-amide.
15. N-(N-(N-(Ν-(Ν-Sarcosyl-Ν-(4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoyl)-lysyl)glycyl)phenylalanyl)leucyl)glycine Ν-(2-sarcosylaminoethyl)amide dihydrobromide. It is contemplated that Ν-(Ν-(Ν-(Ν-(Ν-(Ν-(phenylmethoxycarbonyl)sarcosyl)-N- (4-oxo-4- (4- (10 , 15 , 20-triphenyl-21H, 23H-porphin-5 -yl)-phenylamino)butanoyl)lysyl)glycyl)phenylalanyl)-leucyl)-glycine Ν-(2-(Ν-(phenylmethoxycarbonyl)-sarcosylamino)-ethyl)amide stirred with 30% hydrogen bromide in acetic acid for 1 hour. The solvent and excess reagent is evaporated under reduced pressure. Trituration of the residue with five portions of dry diethylether give Ν-(Ν-(Ν-(Ν-(Ν-sarcosyl-Ν-(4-oxo-4-(4- (10,15,20-triphenyl-21H,23H-porphin-5-yl)-phenylamino)butanoyl)-lysyl)glycyl)phenylalanyl)- leucyl) glycine N-(2-sarcosyl-aminoethyl)amide
dihydrobromide.
16. Polymer B. It is contemplated that N-(N-(N-(N-(N-sarcosyl-N-(4-oxo-4-(4-(10,15,20-triphenyl-21H,23H-porphin-5-yl)phenylamino)butanoyl)lysyl)glycyl)phenyl-alanyl)leucyl)glycine N-(2-sarcosylaminoethyl)amide dihydrobromide is boiled under reflux with anhydrous sodium carbonate and poly(oxyethylene)-, -bis(oxiranylmethyl) ether (prepared by the literature method [Y. Chen and M. Feng, Chinese Patent 86 104 089,1987]) in ethanol for 6 hours. The suspension is filtered and the solvent is evaporated from the filtrate under reduced pressure to give the polymer.

Claims

1. A linear block copolymer comprising units of an alkylene oxide, linked to units of peptide via a linking group comprising a -CH2CHOHCH2N(R)- moiety, wherein R is a lower alkyl group.
2. A copolymer as claimed in claim 1 comprising units of polyalkyleneoxide linked to polypeptide units via a linker group comprising an amine:epoxide conjugation product.
3. A copolymer as claimed in either of claims 1 and 2, wherein the linking group comprises a moiety
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4CO-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4(CH2)2-;
-CONH(CH2)pNHCOCH2N(CH3)CH2CHOHCH2OC6H4(CH2)2NH-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4CO-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4(CH2)2-;
-NH(CH2)pN(CH3)CH2CHOHCH2OC6H4(CH2)2NH-;
-CONH(CH2)pNHCO(CH2)pN(CH3)CH2CHOHCH2- ;
-NH(CH2)pNHCO(CH2)pN(CH3)CH2CHOHCH2-;
-NHCO(CH2)pN(CH3)CH2CHOHCH2-; or
- CO(CH2)pN(CH3)CH2CHOHCH2- wherein p is an integer having a value of from 1 to 6.
4. A copolymer as claimed in any one of claims 1 to 3 wherein the peptide is of about 3 to about 50 amino acid residues in length.
5. A copolymer as claimed in any one of claims 1 to 4 wherein the units of alkylene oxide comprise ethylene oxide residues.
6. A copolymer as claimed in any one of claims 1 to 5 having a molecular weight of from about 10,000 to about 1 million.
7. A copolymer as claimed in any one of claims 1 to 6 wherein a peptide unit is conjugated to a chelating agent moiety.
8. A copolymer as claimed in claim 7 wherein said chelating agent moiety is metallated.
9. A copolymer as claimed in claim 7 wherein said chelating agent moiety is metallated with a
paramagnetic metal species.
10. A copolymer as claimed in claim 7 wherein said chelating agent moiety is metallated with a metal radionuclide species.
11. A copolymer as claimed in any one of claims 1 to 10 comprising a comprising a repeat unit comprising a moiety of formula
-(PAG)N(R')CH2CHOHCH2OC6H4CO(Peptide)NH(CH2) C6H4OCH2CHOHCH2N(R')- or
-(PAG)CH2CHOHCH2N(R')CH2CO(Peptide)NH(CH2)pNHCOCH2N(R')-CH2CHOHCH2-
(wherein
R' is a C1-4-alkyl group;
p is an integer having a value of from 1 to 6;
PAG comprises a polyethyleneoxide chain; and
peptide is a Gly-Phe-Leu-Gly or Lys-Gly-Phe-Leu-Gly residue).
12. A pharmaceutical composition comprising a
copolymer as claimed in any one of claims 1 to 11 together with at least one physiologically acceptable carrier or excipient.
13. A process for the preparation of a copolymer as claimed in claim 1 said process comprising reacting a bis epoxide reagent with a bis amine reagent, one of said reagents incorporating said peptide units and the other incorporating said alkylene oxide units.
14. A process for the preparation of a chelated metal bearing copolymer, said process comprising metallating a chelating moiety containing copolymer as defined in claim 7.
15. A process for the preparation of a therapeutic copolymer, said process comprising conjugating a copolymer according to claim 1 to a drug or prodrug.
16. A method of generating an enhanced image of the human or non-human animal body, said method comprising administering to said body a contrast-enhancing
copolymer as defined in claim 1 and generating an image of at least a part of said body into which said
copolymer distributes.
17. Use of a copolymer as defined in any one of claims 1 to 11 for the manufacture of a diagnostic or
therapeutic agent.
18. A compound of formula
Figure imgf000070_0001
(wherein peptide is a peptide residue).
PCT/GB1995/000418 1994-02-28 1995-02-28 Block copolymers WO1995022991A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
MX9603680A MX9603680A (en) 1994-02-28 1995-02-28 Block copolymers.
AU18170/95A AU1817095A (en) 1994-02-28 1995-02-28 Block copolymers
EP95909864A EP0748226A1 (en) 1994-02-28 1995-02-28 Block copolymers
JP7522220A JPH09509443A (en) 1994-02-28 1995-02-28 Block copolymer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/203,106 US5618528A (en) 1994-02-28 1994-02-28 Biologically compatible linear block copolymers of polyalkylene oxide and peptide units
US08/203,106 1994-02-28

Publications (2)

Publication Number Publication Date
WO1995022991A2 true WO1995022991A2 (en) 1995-08-31
WO1995022991A3 WO1995022991A3 (en) 1995-10-12

Family

ID=22752532

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1995/000418 WO1995022991A2 (en) 1994-02-28 1995-02-28 Block copolymers

Country Status (8)

Country Link
US (2) US5618528A (en)
EP (1) EP0748226A1 (en)
JP (1) JPH09509443A (en)
CN (1) CN1146726A (en)
AU (1) AU1817095A (en)
CA (1) CA2183766A1 (en)
MX (1) MX9603680A (en)
WO (1) WO1995022991A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2863619A1 (en) * 2003-12-11 2005-06-17 Oreal New copolymers containing ROD and COIL blocks, useful as surfactants and rheological agents, particularly in cosmetic compositions, in which the ROD component is a poly(amino acid)
US7354980B1 (en) 2004-03-12 2008-04-08 Key Medical Technologies, Inc. High refractive index polymers for ophthalmic applications
US7446157B2 (en) 2004-12-07 2008-11-04 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications

Families Citing this family (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100361933B1 (en) * 1993-09-08 2003-02-14 라 졸라 파마슈티칼 컴파니 Chemically defined nonpolymeric bonds form the platform molecule and its conjugate
ATE194920T1 (en) * 1993-09-22 2000-08-15 Hoechst Ag PRO-PRODRUGS, THEIR PRODUCTION AND USE
US6770261B2 (en) * 1995-06-02 2004-08-03 Research Corporation Technologies Magnetic resonance imaging agents for the detection of physiological agents
US20020064546A1 (en) * 1996-09-13 2002-05-30 J. Milton Harris Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
US6258351B1 (en) * 1996-11-06 2001-07-10 Shearwater Corporation Delivery of poly(ethylene glycol)-modified molecules from degradable hydrogels
US20030215421A1 (en) * 1999-07-21 2003-11-20 Mcdonald John R. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US7157418B1 (en) 1998-07-22 2007-01-02 Osprey Pharmaceuticals, Ltd. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US6299860B1 (en) * 1998-10-15 2001-10-09 Fluoro Probe, Inc. Method for viewing diseased tissue located within a body cavity
US6652836B2 (en) 1998-10-15 2003-11-25 Fluoroprobe, Inc. Method for viewing tumor tissue located within a body cavity
US6458953B1 (en) * 1998-12-09 2002-10-01 La Jolla Pharmaceutical Company Valency platform molecules comprising carbamate linkages
CA2376057A1 (en) * 1999-06-08 2000-12-14 La Jolla Pharmaceutical Company Valency platform molecules comprising aminooxy groups
US6713454B1 (en) * 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
AU2001227679A1 (en) * 2000-02-25 2001-09-03 General Atomics Mutant nucleic binding enzymes and use thereof in diagnostic, detection and purification methods
CN1418112A (en) * 2000-03-15 2003-05-14 布里斯托尔-迈尔斯斯奎布药品公司 Peptidase-cleavable, targeted antineoplastic drugs and their therapeutic use
WO2001071352A2 (en) * 2000-03-17 2001-09-27 The Salk Institute For Biological Studies Compositions associated with complex formation
US6610504B1 (en) 2000-04-10 2003-08-26 General Atomics Methods of determining SAM-dependent methyltransferase activity using a mutant SAH hydrolase
US6951939B2 (en) 2000-06-08 2005-10-04 La Jolla Pharmaceutical Company Multivalent platform molecules comprising high molecular weight polyethylene oxide
MXPA03000310A (en) * 2000-07-12 2004-12-13 Gryphon Therapeutics Inc Polymer-modified bioactive synthetic chemokines, and methods for their manufacture and use.
US7118737B2 (en) * 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
EP1318827A4 (en) * 2000-09-08 2005-09-14 Gryphon Therapeutics Inc Polymer-modified synthetic proteins
EA200300473A1 (en) * 2000-10-16 2003-08-28 Неофарм, Инк. THERAPEUTIC COMPOSITION ON THE BASIS OF MITOXANTRON (OPTIONS) AND LIPID PREPARATION, THE METHOD OF ITS OBTAINING AND THE METHOD OF TREATING THE MAMMALISM DISEASE WITH ITS USE
US7265186B2 (en) * 2001-01-19 2007-09-04 Nektar Therapeutics Al, Corporation Multi-arm block copolymers as drug delivery vehicles
TWI246524B (en) 2001-01-19 2006-01-01 Shearwater Corp Multi-arm block copolymers as drug delivery vehicles
GB0105929D0 (en) * 2001-03-09 2001-04-25 Btg Int Ltd Physiologically activated prodrugs
US20040077835A1 (en) * 2001-07-12 2004-04-22 Robin Offord Chemokine receptor modulators, production and use
EP1616003A4 (en) * 2002-12-30 2007-06-20 Gryphon Therapeutics Inc Water-soluble thioester and selenoester compounds and methods for making and using the same
US7316811B2 (en) * 2002-12-30 2008-01-08 Nektar Therapeutics Al, Corporation Multi-arm polypeptide-poly (ethylene glycol) block copolymers as drug delivery vehicles
WO2004062588A2 (en) * 2003-01-06 2004-07-29 University Of Utah Water-soluble polymeric bone-targeting drug delivery system
US20050090804A1 (en) * 2003-10-22 2005-04-28 Trivascular, Inc. Endoluminal prosthesis endoleak management
US7384760B2 (en) * 2004-04-30 2008-06-10 General Atomics Methods for assaying inhibitors of S-adenosylhomocysteine (SAH) hydrolase and S-adenosylmethionine (SAM)-dependent methyltransferase
WO2005115457A2 (en) * 2004-05-10 2005-12-08 University Of Utah Research Foundation Combined active and passive targeting of biologically active agents
EP1868652A2 (en) 2005-04-05 2007-12-26 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Method for shielding functional sites or epitopes on proteins
EP1926768B1 (en) 2005-07-18 2011-09-14 Nektar Therapeutics Branched functionalized polymers using branched polyol cores
CN100365043C (en) * 2006-04-06 2008-01-30 北京理工大学 Synthesis of ABA polypeptide -b- polytetrahydrofuran-b-polypeptide triblock copolymer
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
US8956825B2 (en) 2007-05-24 2015-02-17 The United States Of America As Represented By The Department Of Veterans Affairs Intranuclear protein transduction through a nucleoside salvage pathway
CN101801180A (en) * 2007-07-16 2010-08-11 艾尔维奥血管公司 antimicrobial constructs
EP2197340A4 (en) * 2007-09-19 2015-10-21 Oncofluor Inc Method for imaging and treating organs and tissues
KR101666631B1 (en) 2008-05-27 2016-10-17 젠자임 코포레이션 Peptide analogs of alpha-melanocyte stimulating hormone
JP2012511032A (en) 2008-12-05 2012-05-17 アブラクシス バイオサイエンス リミテッド ライアビリティー カンパニー SPARC binding ScFv
CA2799608C (en) 2009-05-28 2023-02-28 Richard H. Weisbart Amino acid sequences which enhance peptide conjugate solubility
GB201012410D0 (en) 2010-07-23 2010-09-08 Medical Res Council Intracellular immunity
PT2717917T (en) 2011-07-05 2016-07-27 Bioasis Technologies Inc P97-antibody conjugates
GB201202268D0 (en) 2012-02-09 2012-03-28 Medical Res Council Intracellular immunity
US9532866B2 (en) * 2012-03-15 2017-01-03 L&C Bio Co., Ltd. Acellular dermal graft
AU2013296557B2 (en) 2012-07-31 2019-04-18 Bioasis Technologies Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
EP2916835A4 (en) 2012-11-12 2016-07-27 Redwood Bioscience Inc Compounds and methods for producing a conjugate
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
WO2014078733A1 (en) 2012-11-16 2014-05-22 The Regents Of The University Of California Pictet-spengler ligation for protein chemical modification
NZ711373A (en) 2013-03-13 2020-07-31 Bioasis Technologies Inc Fragments of p97 and uses thereof
AU2014312190A1 (en) 2013-08-28 2016-02-18 Bioasis Technologies Inc. CNS-targeted conjugates of antibodies
US9493413B2 (en) 2013-11-27 2016-11-15 Redwood Bioscience, Inc. Hydrazinyl-pyrrolo compounds and methods for producing a conjugate
EP3215537B1 (en) 2014-11-05 2022-01-05 Nanyang Technological University Stabilized and autonomous antibody vh domain
WO2017218824A1 (en) 2016-06-15 2017-12-21 Yale University Anti-guanosine antibody as a molecular delivery vehicle
JP2019518040A (en) 2016-06-15 2019-06-27 イェール ユニバーシティーYale University Antibody-Mediated Autocatalytic Delivery of Nanocarriers Targeted to Tumors
EP3732254A4 (en) 2017-12-26 2021-12-22 Becton, Dickinson and Company Deep ultraviolet-excitable water-solvated polymeric dyes
US20210054102A1 (en) 2018-02-01 2021-02-25 Yale University Compositions and methods for enhancing nuclear translocation
US10844228B2 (en) 2018-03-30 2020-11-24 Becton, Dickinson And Company Water-soluble polymeric dyes having pendant chromophores
CA3107332A1 (en) 2018-07-22 2020-01-30 Bioasis Technologies Inc. Treatment of lymphatic metastases
US20210355468A1 (en) 2020-05-18 2021-11-18 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
US20210393787A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
EP4182475A2 (en) 2020-07-17 2023-05-24 Onena Medicines S.L. Antibodies against lefty proteins
WO2023010060A2 (en) 2021-07-27 2023-02-02 Novab, Inc. Engineered vlrb antibodies with immune effector functions
EP4155349A1 (en) 2021-09-24 2023-03-29 Becton, Dickinson and Company Water-soluble yellow green absorbing dyes
WO2023056450A1 (en) 2021-09-30 2023-04-06 Yale University Compositions and methods for the treatment of autosomal dominant polycystic kidney disease and other diseases having upregulated mtor activity
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties
WO2024050524A1 (en) 2022-09-01 2024-03-07 University Of Georgia Research Foundation, Inc. Compositions and methods for directing apolipoprotein l1 to induce mammalian cell death
WO2024064752A2 (en) 2022-09-20 2024-03-28 Yale University Compositions of wet adhesives derived from vibrio cholerae biofilm adhesins and methods thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DD287949A5 (en) * 1989-09-15 1991-03-14 Adw Zi F. Molekularbiologie,De PROCESS FOR THE PRODUCTION OF COMPOUNDS WITH BIOLOGICAL ACTIVITY
WO1992000748A1 (en) * 1990-07-06 1992-01-23 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
WO1993002712A1 (en) * 1991-08-01 1993-02-18 Danbiosyst Uk Limited Preparation of microparticles

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1302066B (en) * 1963-06-10 1969-11-13 Avny Yair Method for grafting polypeptide chains onto polyhydroxy polymers
DE2433883C2 (en) * 1973-07-20 1986-03-27 Research Corp., New York, N.Y. Use of physiologically active polypeptides
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DD287949A5 (en) * 1989-09-15 1991-03-14 Adw Zi F. Molekularbiologie,De PROCESS FOR THE PRODUCTION OF COMPOUNDS WITH BIOLOGICAL ACTIVITY
WO1992000748A1 (en) * 1990-07-06 1992-01-23 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
WO1993002712A1 (en) * 1991-08-01 1993-02-18 Danbiosyst Uk Limited Preparation of microparticles

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BIOCHEM BIOPHYS RES COMMUN, NOV 15 1989, , VOL. 164, NO. 3, PAGE(S) 1234-9, US, YOKOYAMA M ET AL 'Stabilization of disulfide linkage in drug-polymer-immunoglobulin conjugate by microenvironmental control.' *
BIOCONJUG CHEM, JUL-AUG 1992, , VOL. 3, NO. 4, PAGE(S) 295-301, US, YOKOYAMA M ET AL 'Preparation of micelle-forming polymer-drug conjugates' *
BIOKHIMIIA, JUL 1993, VOL. 58, NO. 7, PAGE(S) 1071-6, EFREMOVA NV ET AL 'Supramolecular structure based on protein conjugates with polyalkyleneoxides. Complexes with beta-cyclodextrinÜ' *
BIOMED. SCI., 1991, VOL. 2, NO. 6, PAGES 562-8, TOPCHIEVA, I. N. ET AL 'The interaction of block copolymers of ethylene oxide and propylene oxide and their polymer-protein conjugates with lipids' *
BIOTECHNOL. APPL. BIOCHEM., 1993,, vol. 18, no. 3, pages 329-339, KIRILLOVA, G. P. ET AL 'The influence of pluronics and their conjugates with proteins on the rate of oxygen consumption by liver mitochondria and thymus lymphocytes' *
DATABASE WPI Section Ch, Week 9132 Derwent Publications Ltd., London, GB; Class A96, AN 91-230961 & DD,A,287 949 ( ZENT MOLEKULAR AG) , 14 March 1991 *
J. MED. CHEM., 1990, VOL. 33, NO. 6, PAGE(S) 1620-34, TOUS G ET AL 'O'-(epoxyalkyl)tyrosines and (epoxyalkyl)phenylalanine as irreversible inactivators of serine proteases: synthesis and inhibition mechanism' *
PEPT. 1990, PROC. EUR. PEPT. SYMP., 21ST, 1991, PAGES 849-50, RAPP, W. ET AL 'Comparative study of antibody titers induced by a peptide epitope conjugated with protein, lipopeptide, polyoxyethylene and polyoxyethylene-polystyrene graft copolymer' *
VESTN. MOSK. UNIV., SER. 2: KHIM., 1992, VOL. 33, NO. 6, PAGES 600-4, EFREMOVA, N. V. ET AL 'Conjugates of.alpha.-chymotrypsin with polyalkylene oxides' *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2863619A1 (en) * 2003-12-11 2005-06-17 Oreal New copolymers containing ROD and COIL blocks, useful as surfactants and rheological agents, particularly in cosmetic compositions, in which the ROD component is a poly(amino acid)
WO2005059032A1 (en) * 2003-12-11 2005-06-30 L'oreal Cosmetic application of rod-coil polymers
US7354980B1 (en) 2004-03-12 2008-04-08 Key Medical Technologies, Inc. High refractive index polymers for ophthalmic applications
US7446157B2 (en) 2004-12-07 2008-11-04 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications
US7745555B2 (en) 2004-12-07 2010-06-29 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications
US8048972B2 (en) 2004-12-07 2011-11-01 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications
US9056934B2 (en) 2004-12-07 2015-06-16 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications
US10421830B2 (en) 2004-12-07 2019-09-24 Key Medical Technologies, Inc. Nanohybrid polymers for ophthalmic applications

Also Published As

Publication number Publication date
CN1146726A (en) 1997-04-02
MX9603680A (en) 1997-06-28
WO1995022991A3 (en) 1995-10-12
EP0748226A1 (en) 1996-12-18
US5853713A (en) 1998-12-29
US5618528A (en) 1997-04-08
CA2183766A1 (en) 1995-08-31
AU1817095A (en) 1995-09-11
JPH09509443A (en) 1997-09-22

Similar Documents

Publication Publication Date Title
US5618528A (en) Biologically compatible linear block copolymers of polyalkylene oxide and peptide units
EP0664713B1 (en) Therapeutic and diagnostic imaging compositions and methods
CA2437983C (en) Transporters comprising spaced arginine moieties
EP0752890B1 (en) Segmented chelating polymers as imaging and therapeutic agents
JP2002543111A (en) Amplification of folate-mediated targeting to tumor cells using polymers
US20100292148A1 (en) Targeted polylysine dendrimer therapeutic agent
US20130149355A1 (en) Hydroxyapatite-targeting poly(ethylene glycol) and related polymers
WO1998013059A1 (en) Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
WO1995026754A9 (en) Segmented chelating polymers as imaging and therapeutic agents
US9415114B2 (en) Conformations of divergent peptides with mineral binding affinity
JP2011523415A (en) Novel dual-targeting anti-tumor complex
JP4216715B2 (en) Peptide conjugates, their derivatives including metal complexes and their use for magnetic resonance imaging (MRI)
US20030195152A1 (en) Polymeric conjugates of antitumor agents
US20020076378A1 (en) Polymers
US20050107543A1 (en) Fluoro linkers and their use as linkers for enzyme-activated drug conjugates
EP1011736A2 (en) Polymers
WO1995020603A1 (en) Inhibitors of serine proteases, bearing a chelating group
US20070232639A1 (en) Camptothecin Derivatives Conjugated in Position 20 with Integrin Antagonists
MXPA96003680A (en) Block copolymers
US7745590B1 (en) Degradable macromolecular magnetic resonance imaging contrast agents and methods thereof
EP0489220A1 (en) Cytotoxic N,N'-bis (succinyl-peptide-) derivatives of 1,4-bis (aminoalkyl)-5,8-dihydroxyanthraquinones and antibody conjugates thereof
MXPA96003679A (en) Poliquelan

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 95192754.X

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NL NO NZ PL PT RO RU SD SE SG SI SK TJ TT UA UG US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

AK Designated states

Kind code of ref document: A3

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NL NO NZ PL PT RO RU SD SE SG SI SK TJ TT UA UG US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2183766

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995909864

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995909864

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1995909864

Country of ref document: EP