WO2010140148A1 - Prodrugs containing albumin binding probe - Google Patents

Prodrugs containing albumin binding probe Download PDF

Info

Publication number
WO2010140148A1
WO2010140148A1 PCT/IL2010/000433 IL2010000433W WO2010140148A1 WO 2010140148 A1 WO2010140148 A1 WO 2010140148A1 IL 2010000433 W IL2010000433 W IL 2010000433W WO 2010140148 A1 WO2010140148 A1 WO 2010140148A1
Authority
WO
WIPO (PCT)
Prior art keywords
cooh
group
formula
conjugate
alkyl
Prior art date
Application number
PCT/IL2010/000433
Other languages
French (fr)
Inventor
Yoram Shechter
Matityahu Fridkin
Original Assignee
Yeda Research And Development Co . Ltd
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 Yeda Research And Development Co . Ltd filed Critical Yeda Research And Development Co . Ltd
Priority to EP10726604.1A priority Critical patent/EP2437786B1/en
Priority to US13/375,637 priority patent/US20120134977A1/en
Publication of WO2010140148A1 publication Critical patent/WO2010140148A1/en
Priority to IL216726A priority patent/IL216726A0/en
Priority to US14/564,469 priority patent/US20150093370A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/7036Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin having at least one amino group directly attached to the carbocyclic ring, e.g. streptomycin, gentamycin, amikacin, validamycin, fortimicins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • A61K38/37Factors VIII
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • the present invention relates to albumin-binding probes capable of converting short-lived amino-containing drugs into inactive reactivable prodrugs having prolonged lifetime profiles in vivo.
  • polypeptide drugs in particular nonglycosylated proteins of molecular mass less than 50 kDa, are short-lived species in vivo having circulatory half lives of 5-20 min.
  • the short lifetime of proteins in vivo is attributed to several mechanisms including glomerular filtration in the kidneys and proteolysis at several levels (Goodman and Gilman, 1995).
  • Pegylated proteins are long- lived species in vivo having higher stability and aqueous solubility, as well as lower immunogenicity and antigenicity, compared with the corresponding non-pegylated proteins, and the potential for specific cell targeting (Clark et al, 1996; Delgado et al, 1992; Reddy, 2000; Bailon et al, 2001). In spite of those profound clinical properties obtained by pegylation, only a limited number of pegylated proteins is in clinical use.
  • PEG polyethylene glycol
  • the inactive conjugate thus obtained is transformed into a long-acting prodrug that gradually releases its pharmacologically active constituent upon incubation at physiological conditions (Marcus et al, 2008; Nesher et al, 2008; Peleg-Shulman et al, 2004; Shechter et al, 2005a; Tsubery et al, 2004).
  • R 1 is a radical containing a protein or a polymer carrier moiety
  • R 2 is selected from hydrogen, (CrC 8 )alkyl, (CrQ)alkoxy, (CrCg)alkoxyalkyl, (C 6 - C 10 )aryl, (C r C 8 )alkaryl, (C 6 -C 10 )ar(C r C 8 )alkyl, halogen, nitro, -SO 3 H, -SO 2 NHR, amino, ammonium, carboxyl, PO 3 H 2 , or OPO 3 H 2
  • R is selected from hydrogen, (CrQ)alkyl or (C 6 -C 10 )aryl
  • R 3 and R 4 are each selected from hydrogen, (CrC 8 )alkyl or (C 6 -C 10 )aryl
  • A is a covalent bond when the radical is linked to a carboxyl, phosphate or mercapto group of the drug Y,
  • said protein carrier may be, e.g., albumin such as human serum albumin (HSA), said polymer carrier may be, e.g., a linear or branched PEG, and said drug containing at least one free amino group may be a non-peptidic drug or a peptide or protein drug, most preferably of low or medium molecular weight.
  • albumin such as human serum albumin (HSA)
  • said polymer carrier may be, e.g., a linear or branched PEG
  • said drug containing at least one free amino group may be a non-peptidic drug or a peptide or protein drug, most preferably of low or medium molecular weight.
  • albumin Since albumin is long-lived in vivo, drugs and endogenous substances that tightly associate with albumin have lower clearance rates than that of the unbound substances, and exhibit prolonged lifetime profiles in vivo (Taylor and Granger, 1984).
  • Long-chain fatty acids (LCFAs) bind tightly to albumin (Carter and Ho, 1994), and this provided the impetus for designing an insulin derivative, in which LCFA-like probe has been integrated into the insulin molecule (Kurtzhals et al, 1995, 1996, 1997).
  • the optimal derivative thus obtained (insulin-detemir) possessing protracted action in vivo, in part due to its associating affinity to endogenous albumin (Kurtzhals et al, 1995, 1996, 1997).
  • Insulin detemir (Levemir®, NovoNordisk) is a long-acting human insulin analog with up to 24 hours duration of action.
  • it is an insulin analog in which the amino acid threonine in position B30 is omitted and myristic acid has been attached to the amino acid lysine in position B29 via the acyl group, i.e., N 6B29 - tetradecanoyl des(B30) insulin.
  • insulin-detemir binds to albumin through the alkyl residue of the myristic acid and it is then slowly dissociated from this complex.
  • Insulin detemir as well as other similar derivatives of insulin are disclosed in US Patent Nos. 5,750,497, 6,011,007 and 6,869,930, and in US Patent Publication Nos. 20040110664 and 20060030518.
  • These publications disclose an insulin derivative in which (i) the amino acids at positions A21 and B3 are, independently, any amino acid residue which can be coded for by the genetic code except Lys, Arg and Cys; (ii) the amino acid at position Bl is Phe or is deleted; (iii) the amino acid at position B30 is any amino acid residue which can be coded for by the genetic code except Lys, Arg and Cys, or is deleted; and (iv) the amino e-amino group of Lys B29 is substituted with an acyl group having at least 10 carbon atoms or a lipophilic substituent having at least 6 carbon atoms, wherein the insulin derivative is a Zn 2+ complex and the Zn 2+ complex of the insulin derivative is more water soluble than
  • the technology disclosed in these publications is directed to insulin derivatives only, wherein the lipophilic substituent is linked to the insulin derivative via an amino group on the insulin molecule, preferably the €-amino of the amino acid lysine at position B29, and the insulin derivative is bound to albumin, upon administration, mainly via binding groups present in the albumin molecule capable of binding aliphatic chains.
  • US 7,186,797 discloses polypeptide conjugates having extended half life in vivo, comprising a polypeptide conjugated to a binding moiety having affinity for albumin.
  • the binding moiety disclosed has two arms, wherein each one of these arms binds to albumin via a certain linking group that is either an aryl moiety or a non-aromatic moiety having 1-10 carbon atoms.
  • WO 2008053360 discloses portable albumin binders, capable of binding to albumin through a functional group that is negatively charged or may be deprotonated to yield a negative charge, e.g., a carboxyl group, which are said to be useful for improving the pharmacokinetic properties of diagnostic or therapeutic agents, e.g., by increasing their circulation lifetime.
  • the present invention is based on a concept according to which a long chain fatty acid (LCFA) like albumin-binding compound is covalently linked to a short-lived amino-containing drug to form a drug conjugate capable of non-covalent association with albumin in vivo, i.e., a long-lived prodrug that gradually releases the pharmacologically active constituent.
  • LCFA long chain fatty acid
  • the present invention thus relates to a compound of the formula I:
  • Ri is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO 2 NH-, -O-, - OCO-, -CO-NH-, -CS-NH-, -CO(CH 2 ) M -, or -R 8 -CO-, wherein R 8 is (C r C 8 )alkyl optionally interrupted by a heteroatom selected from O, S or N;
  • R 2 is selected from >
  • R 9 or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO 3 H group;
  • R 3 is absent or an acidic group having at least one hydroxyl group such as - COOH, -SO 3 H or -0-PO 3 H 2 ;
  • R 4 is an electron withdrawing group such as -SO 3 H, -CN, -CO-(C rC 8 )alkyl, -CO-(C 6 -Cio)aryl, -NO 2 , -OPO 3 H 2 , -N(R) 3 + , -SO 2 NH 2 , or halogen, wherein R is selected from (C r C 8 )alkyl or (C 6 -C 10 )ar(C r C 8 )alkyl; R 5 and R 6 , each independently is selected from hydrogen, -(CrC 8 )alkyl or (C 6 -C 10 )aryl;
  • R 7 is a leaving group such as -O-(CH 2 ) 2 -CN, -Cl,
  • R 9 is selected from (Ci 3 -C 20 )alkylene, (C 13 -C 2 o)alkenylene or (C 13 - C 20 )alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(d-C 8 alkyl)-, -N(C 6 -C 10 aryl)-, or -(C 6 -C 10 )arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R 2 is a peptide moiety, R 3 is absent.
  • the present invention relates to a conjugate of the formula
  • Y is a moiety of a drug containing at least one amino group, linked through said at least one amino group;
  • R 1 is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO 2 NH-, -O-, - OCO-, -CO-NH-, -CS-NH-, -CO(CH 2 ) 1-4 -, or -R 8 -CO-, wherein R 8 is (C r C 8 )alkyl optionally interrupted by a heteroatom selected from O, S or N;
  • R 2 is selected from >
  • Rg or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, lie or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO 3 H group;
  • R 3 is absent or an acidic group having at least one hydroxyl group such as - COOH, -SO 3 H or -0-PO 3 H 2 ;
  • R 4 is an electron withdrawing group such as -SO 3 H, -CN, -CO-(C 1 -C 8 )alkyl, -CO-(C 6 -C 10 )aryl, -NO 2 , -OPO 3 H 2 , -N(R) 3 + , -SO 2 NH 2 , or halogen, wherein R is selected from (C r C 8 )alkyl or (C 6 -C 10 )ar(CrC 8 )alkyl;
  • R 5 and R 6 each independently is selected from hydrogen, -(Q-C 8 )alkyl or - (C 6 -C 10 )aryl;
  • R 9 is selected from (Ci 3 -C 20 )alkylene, (C 13 -C 2 o)alkenylene or (C 13 - C 20 )alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(C r C 8 alkyl)-, -N(C 6 -C 10 ary I)- or -(C 6 -C 10 )arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R 2 is a peptide moiety, R 3 is absent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a conjugate of the formula II as defined above, i.e., a conjugate obtained by nucleophilic substitution of a compound of formula I with an amino group of the drug Y, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions of the present invention can be used for treatment of various diseases, disorders and conditions, in which administration of the drug Y might be useful.
  • the present invention provides a method for treatment of diabetes mellitus or hyperglycemia comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is insulin, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of insulin.
  • the present invention provides a method for treatment of insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, or gestational diabetes mellitus, or for prevention of hyperglycemia, said method comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is exendin-4, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of exendin-4.
  • the present invention provides a method for treatment of a bacterial infection comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is gentamicin, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of gentamicin.
  • the present invention provides a method for treating a patient in need of Factor Vila or Factor VIII therapy, comprising administering to said patient an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is Factor Vila or Factor VIII, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of Factor Vila or Factor VIE, respectively.
  • Figs. 1A-1B show the binding affinity of insulin-detemir to human serum albumin (HSA) as determined by ITC-200.
  • HSA human serum albumin
  • the processed data has been derived from 20 automatic injections (2.4 ⁇ l each, IA) of insulin-detemir (400 ⁇ M in phosphate buffer saline, PBS, buffer, pH 7.4) into the sample cell containing HSA at a concentration of 10 ⁇ M in PBS buffer, and was translated to a binding isotherm
  • Figs. 2A-2B show simulated binding isotherm for the association of PEG 5 - MAL-S-(CH 2 ) 15 -COOH with HSA.
  • the data was obtained for 15 automatic injections each of 2.7 ⁇ l.
  • the total duration of the experiments was 45 min (2A).
  • the concentration of PEG 5 -MAL-S-(CH 2 ) 15 -COOH in the injection syringe was 400 ⁇ M.
  • Fig. 3 shows HPLC analysis of purified insulin-FMS-MAL-S-(CH 2 ) 15 - COOH.
  • HPLC-purified insulin-FMS-MAL-S-(CH 2 ) 15 -COOH 50 ⁇ g was loaded on a chromolith Rp- 18e (100 mm X 4 mm) column and run with a linear gradient from 0 to 100% solution A (0.1% trifluoroacetic acid, TFA) to solution B (acetonitrile-H 2 O, 75:25 in 0.1% TFA) over 10 min, and then over 4 min in solution B at a rate of 3 ml/min.
  • the effluent was monitored at 220 nm.
  • Fig. 4 shows prolonged residence time of 125 I-insulin-FMS-MAL-S-(CH 2 ) 15 - COOH following intravenous administration in rats.
  • blood aliquots 50-70 mg
  • FIG. 5 shows circulating glucose levels in CDl -mice following a single subcutaneous administration of insulin-FMS-MAL-S-(CH 2 )i 5 -COOH.
  • Mice were subcutaneously injected with PBS-buffer (-•-; 0.2 ml/mouse), Zn 2+ free insulin (- ⁇ -; 0.17 nmol/mouse in 0.2 ml PBS buffer) or insulin-FMS-MAL-S-(CH 2 ) 15 -COOH (- O-; 0.17 nmol/mouse in 0.2 ml PBS buffer).
  • FIG. 6 shows circulating glucose levels in CDl -mice following a single subcutaneous administration of either insulin-FMS-MAL-S-(CH 2 ) 15 -COOH or insulin-detemir.
  • Fig. 7 shows glucose lowering pattern of exendin-4-FMS-MAL-S-(CH 2 ) 15 - COOH, following a single subcutaneous administration to CDl -mice.
  • Fig. 8 shows time course of in vitro reactivation of gentamicin-FMS-MAL- S-(CH 2 ) 15 -COOH.
  • Gentamicin-FMS-MAL-S-(CH 2 )i 5 -COOH (0.16 ⁇ moles/ml) was incubated in PBS, pH 7.4, containing 2% (w/v) HSA at 37 0 C. Aliquots were withdrawn at the indicated time points and analyzed at several concentrations in the antibacterial assay.
  • Fig. 9 shows schematically the principle of converting short-lived drugs into long-lived species in-vivo using a certain albumin-binding probe of the present invention.
  • Inactive albumin-associated conjugates bind to serum albumin and therefore exhibit prolonged residence time in situ, during which the parent amino containing molecules are released from the inactive conjugates in their native-active form, at a slow rate over many hours following administration.
  • Albumin is the most abundant protein in the blood, at a concentration of approximately 600 ⁇ M.
  • One of the physiological roles of albumin is to act as a carrier of fatty acids, due to the fact that long chain fatty acids (LCFAs) bind tightly to albumin, wherein the terminal carboxylate (-CH 2 -COOH) serves as an albumin- binding ligand.
  • LCFAs long chain fatty acids
  • -CH 2 -COOH the terminal carboxylate
  • the present invention relates to a compound of the formula I, i.e., to an albumin-binding ligand, as defined above.
  • (Ci-C 8 )alkyl typically means a straight or branched hydrocarbon radical having 1-8 carbon atoms and includes, e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2,2- dimethylpropyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • (Ci 3 - C 20 )alkylene refers to a straight or branched divalent hydrocarbon radical having 13-20 carbon atoms and includes, e.g., n-tridecanylene, n-tetradecanylene, n- pentadecanylene, n-hexadecanylene, n-heptadecanylene, n-octadecanylene, n- nonadecanylene, icosanylene, and the like.
  • (C 13 -C 20 )alkenylene and "(C 13 -C 2 o)alkynylene” typically mean straight or branched divalent hydrocarbon radicals having 13-20 carbon atoms and one or more double or triple bonds, respectively, wherein each one of said double or triple bonds is not a terminal double or triple bond.
  • Non-limiting examples of such radicals include 2-, 3-, 4-, 5- and 6-tridecenylene, tetradecenylenes such as myristoleylene, 2-, 3-, 4-, 5-, 6- and 7-pentadecenylene, hexadecenylenes such as palmitoleylene, 2-, 3-, 4-, 5-, 6-, 7- and 8-heptadecenylene, octadecenylenes such as oleylene, linoleylene, ⁇ - linoleylene, nonadecenylene, icosenylenes such as arachidonylene and eicosapentylene, and the like.
  • (C 6 -C 10 )aryl denotes an aromatic carbocyclic group having 6-10 carbon atoms consisting of a single ring or condensed multiple rings such as, but not limited to, phenyl and naphthyl;
  • aromatic(Ci-Cg)alkyl denotes an arylalkyl radical such as benzyl and phenetyl;
  • (C 6 -C i O )arylene-diyl denotes a divalent aromatic carbocyclic group having 6-10 carbon atoms consisting of either a single ring or condensed multiple rings such as, but not limited to, phenylene and naphthylene.
  • amino acid residue refers to any natural or synthetic, i.e., non-natural, amino acid residue in its both L- and D-stereoisomers. While a natural amino acid is any one of the twenty amino acid residues typically occurring in proteins, the term synthetic/non-natural amino acid refers to any amino acid, modified amino acid and/or an analog thereof, that is not one of the twenty natural amino acids.
  • aliphatic hydrophobic amino acid residue refers to an amino acid residue having an aliphatic hydrocarbyl side chain.
  • Non-limiting examples of aliphatic hydrophobic amino acids include the natural amino acids leucine, isoleucine and valine, as well as the non-natural amino acids norvaline (Nva), norleucine (NIe), homovaline and homoleucine.
  • aromatic amino acid residue refers to an amino acid residue in which the side chain contains an aromatic ring. Examples of aromatic amino acids, without being limited to, include the natural amino acid phenylalanine as well as the non-natural amino acids bipyridyl alanine, p-carboxymethyl-L-phenylalanine and p-nitro-L-phenylalanine.
  • amino acid analog comprising -COOH or -SO 3 H group refers to any amino acid analog having amino group as well as -COOH, -SO 3 H or both groups, such as, without being limited to, taurine.
  • leaving group refers to any functional group or atom, which can be displaced by another functional group or atom in a substitution reaction, e.g., a nucleophilic substitution reaction.
  • Non-limiting examples of leaving groups include -0-(CH) 2 -CN, 2,5-dioxopyrrolidin-l-olate also known as N- hydroxysuccinimide (herein designated -OSu), 4-nitrophenoxy, 2-nitrophenoxy, 2,3,4,5,6-pentachlorophenoxy, isoindoline-l,3-dione-2-oxy, and benzenesulfanyl, wherein -OSu is preferred.
  • the compound of the present invention is a compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, and R 7 is -OSu.
  • the compound of the present invention is a compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R, is -NH-CO- or -NH-, preferably -NH-CO-, at position 7 of the fluorene ring, R 2 is Rg or >
  • R 9 is selected from (Ci 3 -C 20 )alkylene, (Ci 3 -C 20 )alkenylene or (C 13 -C 20 )alkynylene, preferably (C 13 -C 20 )alkylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO- NH-, -N(C r C 8 alkyl)-, -N(C 6 -C 10 ary I)- or -(C 6 -C 10 )arylene-diyl, and R 3 is -COOH or SO 3 H.
  • these heteroatoms may be either identical or different heteroatoms, and can be linked sequentially forming, e.g., -S-S- (disulfide), -N-N- or -O-S- bond, or at any two positions of the alkylene, alkenylene or alkynylene.
  • the compound of the present invention is a compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO- at position 7 of the fluorene ring, R 2 is either R 9 or >
  • R 9 is (C 13 -C 2 o)alkylene optionally interrupted by two sulfur atoms forming disulfide bond or by -CO-NH-, and R 3 is -COOH or SO 3 H.
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R) is -NH-CO-, R 2 is >
  • R 3 is -COOH
  • R 9 is -(CH 2 ) 15 -, i.e., 16-(l-(3-(9-(((2,5-dioxopyrrolidin-l- yloxy)carbonyloxy)-methyl)-7-sulfo-9H-fluoren-2-ylamino)-3-oxopropyl)-2,5- dioxopyrrolidin-3-ylthio) hexadecanoic acid (herein identified SuO-FMS-MAL-S- (CH 2 ) 15 -COOH or compound 1).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 • R 3 is -COOH, and R 9 is -(CH 2 ) 15 -CO-NH-(CH 2 ) 5 -, i.e., 6-(16-(l-(3-(9-(((2,5- dioxopyrrolidin-l-yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-3- oxopropy l)-2 , 5 -dioxopyrrolidin-3 -y lthio)hexadecanamido)hexanoic acid (herein identified SuO-FMS-MAL-S-(CH 2 ) 15 -CO-NH-(CH 2 ) 5 ,
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring,
  • R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is
  • R 3 is -COOH
  • R 9 is -(CH 2 ) 10 -S-S-(CH 2 ) 10 -, i.e., l l-((10-(l-(3-(9-(((2,5- dioxopyrrolidin- 1 -yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-3- oxopropyl)-2,5-dioxopyrrolidin-3-ylthio)decyl)disulfanyl)undecanoic acid (herein identified SuO-FMS-MAL-S-(CH 2 ) 10 -S-S-(CH 2 ) 10 -COOH or compound 3).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is >
  • R 3 is -SO 3 H
  • R 9 is -(CH 2 ) 15 -CO-NH-(CH 2 ) 2 -, i.e., 7-(3-(2,5-dioxo-3-(16-oxo- 16-(2-sulfoethylamino)hexadecylthio)pyrrolidin- 1 -yl)propanamido)-9-(((2,5- dioxopyrrolidin- l-yloxy)carbonyloxy)methyl)-9H-fluorene-2-sulfonic acid (herein identified SuO-FMS-MAL-S-(CH 2 ) 15 -CO-NH-(CH 2 ) 2 -SO 3 H or compound 4).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is R 9 , R 3 is - COOH, and R 9 is -(CH 2 ) 15 -, i.e., 17-(9-(((2,5-dioxo ⁇ yrrolidin-l- yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-17-oxoheptadecanoic acid (herein identified SuO-FMS-(CH 2 ) 15 -COOH or compound 5).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is R 9 , R 3 is -COOH, and R 9 is -(CH 2 ) 15 -CO-NH-(CH 2 ) 5 -, i.e., 6-(17-(9-(((2,5-dioxopyrrolidin-l- yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)- 17-oxoheptadecan- amido)hexanoic acid (herein identified SuO-FMS-(CH 2 ) 15 -CO-NH-(CH 2 ) 5 -COOH or compound 6).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is R 9 , R 3 is - COOH, and R 9 is -(CH 2 ) 10 -S-S-(CH 2 ) 10 -, i.e., l l-((l l-(9-(((2,5-dioxopyrrolidin-l- yloxy)-carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)- 11 -oxoundecyl) disulfanyl)-undecanoic acid (herein identified SuO-FMS-(CH 2 ) 10 -S-S-(CH 2 ) 10 - COOH or compound 7).
  • the compound of the present invention is the compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is -OSu, R 1 is -NH-CO-, R 2 is R 9 , R 3 is -SO 3 H, and R 9 is -(CH 2 ) 15 -CO-NH-(CH 2 ) 2 -, i.e., 9-(((2,5-dioxopyrrolidin-l-yloxy) carbonyl oxy)methyl)-7-( 17-oxo- 17-(2-sulfoethylamino)heptadecanamido)-9H-fluorene-2- sulfonic acid (herein identified SuO-FMS-(CH 2 ) 15 - CO-NH-(CH 2 ) 2 -SO 3 H or compound 8).
  • R 4 is -SO 3 H at position 2 of the fluorene ring
  • the compound of the present invention is a compound of formula I, wherein R 4 is -SO 3 H at position 2 of the fluorene ring, R 5 and R 6 each is hydrogen, R 7 is OSu, R 1 is selected from -NH-CO-, -OCO-, or -R 8 - CO-, wherein R 8 is (CrC 8 )alkyl optionally interrupted by a heteroatom selected from O, S or N, and R 2 is a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or -SO 3 H group such as taurine.
  • R 4 is -SO 3 H at position 2 of the fluorene ring
  • R 5 and R 6 each is hydrogen
  • R 7 is OSu
  • R 1 is selected from -NH-CO-, -OCO-, or -R 8 - CO
  • the compounds of the present invention may be prepared according to any technology or procedure known in the art, e.g., as described in detail in Tsubery et al. (2004) and in various additional publications of the scientific groups of the inventors (Peleg-Shulman et al, 2004; Shechter et al, 2005a; Shechter et al, 2001a; Nesher et al, 2008; Shechter et al, 2007; Shechter, 2005b).
  • the hydrolyzable heterobifunctional intermediate compound 7-(3-(2,5-dioxo-2,5- dihydro- 1 H-pyrrol- 1 -yl)propanamido)-9-(((2,5-dioxopyrrolidin- 1 -yloxy)carbonyl oxy) methyl)-9H-fiuorene-3-sulfonic acid, herein designated MAL-FMS-OSu, can be prepared as described in Tsubery et al.
  • the binding properties of the compounds of formula I to human serum albumin (HSA) can be evaluated by any suitable technique, e.g., by isothermal scanning calorimetry (ITC) as exemplified in the Example section hereinafter.
  • ITC isothermal scanning calorimetry
  • both 10-(2,5-dioxopyrrolidin-3-ylthio)decanoic acid and 16-(2,5-dioxopyrrolidin-3-ylthio)hexadecanoic acid herein designated MAL-S-(CH 2 ) 10 -COOH and MAL-S-(CH 2 ) 15 -COOH, respectively, were prepared by reacting maleimide moiety with 11-mercaptoundecanoic acid or 16- mercaptohexadecanoic acid, respectively, and associated with HSA yielding Ka values of 1.3 to l. ⁇ xlO 5 M "1 .
  • PEG 5 polyethylene glycol molecule
  • PEG 5 -MAL-S-(CH 2 )i 5 -COOH effectively associated with HSA, yielding a Ka value of 1.95xlO 5 M "1 indicating that the length of the LCFA like molecule may significantly influence its ability, when conjugated with a macromolecule such as a drug, to associate with HSA.
  • preferred compounds according to the present invention are those in which the shortest chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin, i.e., the hydroxyl group of R 3 or the terminal hydroxyl group of the peptide moiety in cases R 2 is a peptide moiety and R 3 is absent, is of 15 to 30 atoms.
  • chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin refers to any chain of atoms formed by the sequence R 1 -R 2 -R 3 in the compound of formula I, which links the fluorene ring and either the hydroxyl group of R 3 or the terminal hydroxyl group of the peptide moiety, in cases R 2 is a peptide moiety and R 3 is absent, through which said compound binds to albumin.
  • said chain of atoms may be interrupted by one or more heteroatoms independently selected from oxygen, nitrogen or sulfur; functional groups such as -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO 2 NH-, -O-, -OCO-, -CO- NH-, -CS-NH-, -CO-; or a cyclic aliphatic or aromatic ring such as -OSu or phenyl, respectively.
  • Said cyclic aliphatic ring can be linked through any position of the ring, e.g., through positions 1 and 3 in a 5-membered aliphatic ring such as 3- mercaptopyrrolidine-2,5-dione, also known as 3-mercaptosuccineimide (herein designated MAL-S), or through any two positions being located ortho, meta or para one to another, in a 6-membered aromatic ring such as phenyl.
  • MAL-S 3-mercaptopyrrolidine-2,5-dione
  • MAL-S 3-mercaptosuccineimide
  • R 2 does not contain a cyclic aliphatic or aromatic ring, only one chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin exists.
  • a cyclic aliphatic or aromatic ring interrupts the backbone of R 2 , e.g., in the case wherein R 1 is -NH-CO-, R 3 is -COOH and R 2 is ° ⁇ S _ (C H 2)T3 _ , two chains of atoms linking the fluorene ring and the hydroxyl group of R 3 exist, wherein one of said chains consists of the sequence -NH-CO-CH 2 -N-CO-CH-S- (CH 2 )o-CO ⁇ , i.e., is of 21 atoms, and the other chain consists of the sequence -NH- CO-CH 2 -N-CO-CH 2 -CH-S-(CH 2 )J 3 -CO-,
  • the shortest chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin thus refers to the chain of atoms linking the fluorene ring and said terminal acidic group in cases a single such chain exists or, alternatively, to the chain of atoms linking the fluorene ring and said acidic group, having the lowest number of atoms in its backbone, in cases more than one such chains exist.
  • ligand-protein affinities are affected by non-covalent intermolecular interactions between the two molecules such as hydrogen bonding, electrostatic interactions, hydrophobic and Van der Waals forces, and may also be affected by high concentrations of other macromolecules that cause macromolecular crowding.
  • low molecular-weight drugs suffer a massive loss of pharmacological potency upon conjugation.
  • gentamicin representing a low molecular-weight drug, when introduced with SuO-FMS-MAL-S-(CH 2 ) 15 -COOH to form a conjugate of formula II, regains its full potency upon incubation at physiological conditions.
  • the present invention thus relates to a conjugate of the formula II as defined above.
  • This conjugate may be obtained by nucleophilic substitution of a compound of the formula I, as defined above, with any amino- containing drug, i.e., by nucleophilic substitution of R 7 in the compound of formula I with the amino group of said drug.
  • the drug according to the present invention may be any drug containing at least one amino group.
  • the drug is an aminoglycoside antibiotic such as gentamicin or amphotericin, an antineoplastic drug such as aminolevulinic acid, or an anthracycline chemo therapeutic agent such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone and valrubicin.
  • an aminoglycoside antibiotic such as gentamicin or amphotericin
  • an antineoplastic drug such as aminolevulinic acid
  • an anthracycline chemo therapeutic agent such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone and valrubicin.
  • the drug is a peptide or a protein drug of low or medium molecular weight such as insulin, an interferon, preferably IFN- ⁇ 2, a peptide YY (PYY) agonist, preferably the peptide PYY 3-36 , an exendin, preferably exendin-3 or exendin-4, an exendin analog or exendin agonist, atrial natriuretic peptide (ANP), human growth hormone (hGH), erythropoietin, TNF- ⁇ , calcitonin, gonadotropin releasing hormone (GnRH), a GnRH analogue, hirudin, glucagon, a coagulation factor such as Factor Vila and Factor VIII, and a monoclonal antibody fragment, preferably anti-TNF- ⁇ monoclonal antibody fragment.
  • a peptide YY (PYY) agonist preferably the peptide PYY 3-36
  • an exendin preferably exendin-3 or
  • Insulin is the predominant drug for diabetes mellitus, a group of syndromes characterized by hyperglycemia, altered metabolism of lipids, carbohydrates and proteins, and an increased risk of complications from vascular diseases. Most patients can clinically be classified as having either insulin-dependent (Type I) or insulin-independent diabetes mellitus (Type II). About 90% of diabetic patients in the Western world have Type II diabetes, and about 70% of the Type II diabetics in the United States are also obese, a factor that significantly contributes to insulin resistance.
  • Type II diabetes there is an extensive and selective loss of pancreatic /J-cells and a state of hypoinsulinemia
  • pancreatic /J-cells there is no significant loss of ⁇ - cells from the islets in Type II diabetic patients, in which patients the mean plasma concentration of insulin over a 24-hour period is essentially normal or even elevated because of peripheral resistance to the action of the hormone.
  • individuals with Type II diabetes are relatively insulin deficient, as a normal pancreatic /3-cell should be capable of secreting amounts of insulin that are considerably greater than normal when confronted with hyperglycemia, thus allowing an individual to maintain euglycemia in the face of moderate resistance to insulin.
  • the half-life of insulin in plasma is about 5-6 min, wherein the degradation of insulin occurs primarily in liver and to a lesser extent in kidney and muscle. Proteolytic degradation of insulin in the liver is primarily receptor mediated.
  • Various modifications have been described in order to create insulin analogs having ' longer half-lives in the blood circulation, in particular, prodrugs capable of releasing active insulin into the circulation over a relatively long time period, i.e., 8-24 hours, intended to provide the required basal level of insulin for a whole day.
  • insulin detemir is an insulin analog in which the amino acid threonine in position B30 is omitted and myristic acid has been attached to the amino acid lysine in position B29, i.e., N £B29 -tetradecanoyl des(B30) insulin.
  • insulin-detemir binds to albumin through the acyl group at position B29 and it is then slowly dissociated from this complex.
  • a conjugate according to the present invention formed by introducing insulin to a compound of formula I, in particular, the conjugate herein designated insulin-FMS-MAL-S-(CH 2 ) 15 -COOH, formed by introducing insulin to SuO-FMS-MAL-S-(CH 2 ) 15 -COOH, had about 10% the efficacy of insulin to activate lipogenesis in rat adipocytes yielding an half-maximal effect (ED 50 ) at a concentration of 1.03 ⁇ 0.1 nM; however, it has regained its full lipogenic potency (ED 50 O. l ⁇ O.02 nM) following 4 hours of incubation under conditions that completely release insulin from the conjugate.
  • the circulating level of insulin declined yielding a tVi value of 3.3 ⁇ 0.4 hours
  • the circulating level of said conjugate increased over a period of 2 hours reaching a value of 31,000 ⁇ l,000 cpm/ml blood, which was stably maintained over a period of 6 hours and than declined with a X 1 A value of 17 ⁇ 1 hours, and a significant amount, in particular, -10,000 cpm/ml blood, was still evident 30 hours after intravenous administration.
  • said conjugate had a flat glucose-lowering pattern that was by about two folds prolonged than that of insulin.
  • the aforesaid conjugate was subcutaneously administered at a dose of 0.68 nmol/mouse and as shown, it was highly potent in reducing blood glucose level over prolong time period with a tY ⁇ value of 6 ⁇ 1 hours, wherein low blood glucose level was still evident 24 hours following administration.
  • the area under the curve could not accurately integrated, it exceeded five or more times that obtained by similar dose of subcutaneously administered insulin-detemir.
  • the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of insulin.
  • Exendins are peptides found in the venom of the Gila-monster, a lizard found in Arizona, and the Mexican Beaded Lizard. Exendin-3 is present in the venom of Heloderma horridum, and exendin-4 is present in the venom of Helodermasuspectum.
  • the exendins have some sequence similarity to several members of the glucagon-like peptide family, with the highest homology, 53%, being to GLP-1[7-36]NH 2 , which is also known as proglucagon, and has an insulinotropic effect, stimulating insulin secretion from pancreatic /3-cells.
  • Exendin- 4 is composed of 39 amino acid residues with the carboxy terminus amidated.
  • Exendin-4 potently binds at GLP-I receptors on insulin-secreting /3TCl cells, at dispersed acinar cells from guinea pig pancreas, and at parietal cells from stomach.
  • exendin-3 and exendin-4 as insulinotrophic agents for the treatment of diabetes mellitus and the prevention of hyperglycemia has been previously proposed, e.g., in US 5,424,286.
  • the glucose-lowering profile of native exendin-4 was compared with that of an exendin-4-based conjugate according to the present invention, in particular, the conjugate herein designated exendin-4-FMS-MAL-S-(CH 2 ) 15 -COOH, formed by introducing exendin-4 to SuO-FMS-MAL-S-(CH 2 ) 15 -COOH, when subcutaneously administered at a dose of 0.24 nmol/CDl mouse, and as shown in Example 9, circulating glucose reached its lowest concentration 3 hours following administration of said conjugate and this level was preserved over a period of 20 hours. Returning to initial glucose level took place with a iVi value of 28 ⁇ 2h, which is 4.7 times longer than that obtained by the same dose of the native hormone.
  • the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of exendin-4.
  • HSA-binding probe(s) each having a size of about 760 daltons, with the preservation of significant amount of their biological/pharmacological potencies (Shechter et al, 2001b; Shechter et al., 2007), low molecular-weight amino containing compounds suffer a massive loss of pharmacological potency upon conjugation, and are thus impractical under these circumstances, unless may be reactivated upon administration.
  • gentamicin-based conjugate in particular, the conjugate herein designated gentamicin-FMS-MAL-S-(CH 2 ) 15 -COOH, formed by introducing gentamicin to SuO-FMS-MAL-S-(CH 2 ) 15 -COOH, was used.
  • Gentamicin is an aminoglycoside antibiotic, used in treatment of many types of bacterial infections, particularly those caused by Gram-negative bacteria. Gentamicin works by binding the 30S subunit of the bacterial ribosome, thus interrupting protein synthesis.
  • the conjugate gentamicin- FMS-MAL-S-(CH 2 )i 5 -COOH that was incubated in PBS (pH 7.4) containing 2% (w/v) HSA at 37 0 C had ⁇ 3 ⁇ 0.7% the antibacterial potency of native gentamicin; however, upon incubation in PBS buffer (pH 7.4) containing 20 mg/ml HSA, the antibacterial potency was generated with a t /2 value of 7.1 ⁇ 0.2 h, regaining full (100%) antibacterial potency following 30 hours of incubation.
  • the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of gentamicin.
  • FVII Factor VII
  • proconvertin is a vitamin K dependent enzyme of the serine protease class, produced in the liver, and is one of the central proteins in the coagulation cascade.
  • the main role of FVII is to initiate the process of coagulation in conjunction with tissue factor, which is found on the outside of blood vessels, normally not exposed to the bloodstream. Upon vessel injury, tissue factor is exposed to the blood and circulating FVII. Once bound to tissue factor, FVII is activated to activated FVII (FVIIa) by different proteases, among which are thrombin (Factor Ha), activated Factor X and the FVIIa-tissue factor complex itself.
  • the most important substrates for FVIIa-tissue factor are Factors X (FX) and IX (FIX).
  • Recombinant human FVIIa has been introduced for use in uncontrollable bleeding in hemophilia patients with Factor VIII (FVIII) or FIX deficiency, who have developed inhibitors against replacement coagulation factor. This factor is increasingly used in uncontrollable hemorrhage, as it induces coagulation only in those sites where tissue factor is present as well.
  • FVIII Factor VIII
  • FIX deficiency FIX deficiency
  • FVIII is another essential blood clotting factor. In fact, it is a cofactor for activated FIX which, in the presence of Ca +2 and phospholipids, forms a complex that converts FX to the activated form thereof. In human, FVIII is encoded by the F8 gene, and therefore defects in this gene result in hemophilia A, a common recessive X-linked coagulation disorder.
  • transcript variant 1 encodes a large glycoprotein, isoform a, which circulates in plasma, associates with von Willebrand factor in a noncovalent complex and undergoes multiple cleavage events
  • transcript variant 2 encodes a putative small protein, isoform b, which consists primarily of the phospholipid binding domain of FVIIIc that is essential for coagulant activity.
  • US 7,199,223 discloses conjugates of a FVIII moiety and one or more water- soluble polymers, each having a molecular weight in the range of 6 to 150 kDa, preferably conjugates wherein each one of the polymers is a poly(alkylene oxide), more preferably a PEG, and the FVIII moiety is either recombinantly produced or blood-derived FVIII, FVIHa, FVIILC, FVIILvWF, and B-domain deleted FVIII. As described in this patent, these conjugates may be used for treating patients in need of FVIII therapy such as patients suffering from hemophilia A. US Publication Nos.
  • the technology of the present invention may further be applied FVIIa and to FVIII, and the pharmacokinetic pattern of the specific conjugates formed by introducing these coagulation factors to SuO-FMS- MAL-S-(CH 2 ) 15 -COOH, i.e., FVIIa-FMS-MAL-S-(CH 2 ) 15 -COOH and FVIII-FMS- MAL-S-(CH 2 ) I5 -COOH can be studied both in vitro as well as in in vivo experimental systems.
  • the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of FVIIa or FVIII. It should be noted that both FVIIa and FVIII, according to the present invention, might be either natural or recombinant.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a conjugate of the formula II as defined above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • compositions of the present invention comprise conjugates obtained by nucleophilic substitution of a compound of the formula I, preferably any one of compounds 1 to 8, with insulin, exendin-4, gentamicin or coagulation factors such as FVIIa and FVIII, or a pharmaceutically acceptable salts thereof.
  • compositions of the present invention may be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy, 19 th Ed., 1995.
  • the composition may be in solid, semisolid or liquid form and may further include pharmaceutically acceptable fillers, carriers or diluents, and other inert ingredients and excipients.
  • the pharmaceutical composition can be designed for a slow release of the conjugate.
  • the composition can be administered by any suitable route, e.g. intravenously, orally, parenterally, rectally, or transdermally. The dosage will depend on the state of the patient, and will be determined as deemed appropriate by the practitioner.
  • the route of administration may be any route that effectively transports the active compound to the appropriate or desired site of action, the oral and the intravenous routes being preferred.
  • a solid carrier is used for oral administration, the preparation may be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a lozenge.
  • a liquid carrier is used, the preparation may be in the form of a syrup, emulsion or soft gelatin capsule. Tablets, dragees or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application.
  • Preferable carriers for tablets, dragees or capsules include lactose, cornstarch and/or potato starch.
  • the pharmaceutical compositions of the present invention can be used for treatment of various diseases, disorders or conditions, in which administration of the drug designated Y in the formula II might be useful.
  • the present invention provides a method for treatment of diabetes mellitus or hyperglycemia comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably, any one of compounds 1 to 8, with any of the amino groups of insulin.
  • the present invention provides a method for treatment of insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, or gestational diabetes mellitus, or for prevention of hyperglycemia, said method comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of exendin-4.
  • the present invention provides a method for treatment of a bacterial infection, preferably a bacterial infection caused by Gram-negative bacteria, comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of gentamicin.
  • the present invention provides a method for treating a patient in need of Factor Vila or Factor VIII therapy, comprising administering to said patient an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of FVIIa or FVIII, respectively.
  • this method is used for treatment of patients who suffer from hemophilia A.
  • the present invention provides a novel technology according to which any amino-containing short-lived drug can be converted, upon administration, into a long-lived prodrug, which gradually releases the pharmacologically active constituent under physiological conditions. This property is particularly achieved by introducing said short-lived drug with a LCFA like molecule capable of associating with HSA in vivo, which contains a spontaneously hydrolysable bond.
  • D-[U- 14 C] glucose (4-7 mci/mol) was obtained from Du Pont-NEN (Boston, Ma)
  • type I collagenase 134 U/mg
  • gentamicin sulfate was purchased from Sigma Chemical Co. (Ness- Ziona, Israel)
  • polyethylene glycol 5 kDa-maleimide PEG 5 -MAL was obtained from Shearwater Group Inc. (Ra'anana, Israel).
  • Exendin-4 (HGEGTFTSDLSKQM EEEAVRLFIEWLKNGGPSSGAPPPS-NH 2 ) was synthesized by the solid phase method using the multiple peptide synthesizer AMS 422 (Abimed Analysentechnik, GmbH). 11-mercapto undecanoic acid, 16-mercaptohexadecanoic acid, decan-l,10-dithiol, taurine and trityl chloride were all purchased from Sigma- Aldrich Ltd. 6-amino-n-hexanoic acid purchased from BDH Ltd. All other materials used were of analytical grade.
  • PEG 5 -CO-NH-(CH 2 ) 3 -NH 2 was prepared by dissolving PEG 5 -N- hydroxysuccinimide ester (PEG 5 -OSu, Shearwater product) at a concentration of 20 mg/ml in 0.1 M NaHCO 3 containing 1 M of 1,3-diaminopropane dihydrochloride (Aldrich). The reaction was carried out for 1 hour at 25 0 C, and the product was extensively dialyzed against H 2 O, lyophilized and kept at 7°C until used.
  • Example 1 Applying isothermal scanning calorimetry for evaluating various ligands-human serum albumin associating affinities
  • insulin detemir (Levemir®, NovoNordisk) is a long-acting human insulin analog, in which the amino acid threonine in position B30 has been omitted and myristic acid has been attached to the amino acid lysine in position B29, i.e., N' B29 -tetradecanoyl des(B30) insulin.
  • insulin-detemir binds to HSA through the acyl group at position B29 and it is then slowly dissociated from the complex.
  • Figs. 1A-1B show the binding affinity of insulin-detemir to HSA, as determined by ITC-200. The primary ITC data was translated to a binding isotherm (Fig.
  • LCFA long-chain fatty acid
  • MAL maleimide
  • a required part of any such molecule is a terminal carboxylate (-CH 2 -COOH) that is essential for LCFAs' association with albumin (Peters, 1996).
  • PEG 5 -maleimide PEG 5 - MAL
  • PEG 5 - MAL PEG 5 -maleimide
  • PEG 5 -NH 2 was used as well for linking the appropriate HSA binding probe through our hydrolyzable heterobifunctional agent.
  • SuO-FMS-MAL (58.3 mg, 100 ⁇ mol) and 16-sulfanylhexadecanoic acid (HS-(CH 2 ), 5 -COOH, 38.5 mg, 120 ⁇ mol) were dissolved in 1.0 ml DMF, followed by addition of pyridine (20 ⁇ l, 248 ⁇ mol). The reaction mixture was stirred for 40 min at 25°C, and product formation was monitored by the decrease in the maleimide moiety in aliquots withdrawn during synthesis.
  • HS-(CH 2 ), 5 -COOH 16-sulfanylhexadecanoic acid
  • the derivative formed SuO-FMS-MAL-S-(CH 2 ) 15 -COOH was added to an aqueous solution of insulin 6 mg/ml (1 ⁇ mol/ml) dissolved in 0.1 M NaHCO 3 (pH 8.5) at three molar excess over the protein (30 ⁇ l).
  • the reaction was carried out for 2 h at 0 0 C, and the mixture was then dialyzed against H 2 O at 7°C.
  • Monomodified derivative of insulin-linked to FMS-MAL-S-CH 2 ) i5 -COOH was purified from un-reacted insulin and from residual bismodified derivative, using semi-preparative HPLC (RP-4 column, Hesperia CA, 20-100% solution B (acetonitrile-H 2 O, 75:25 in 0.1% TFA) over 60 min with a flow rate of 10 ml/rnin).
  • the fraction corresponding to monomodified insulin-FMS-MAL-S-(CH 2 ) 15 -COOH was collected, redialyzed against H 2 O and lyophilized.
  • both 10-(2,5-dioxopyrrolidin-3- ylthio)decanoic acid and 16-(2,5-dioxopyrrolidin-3-ylthio)hexadecanoic acid prepared by reacting MAL with 11-sulfanylundecanoic acid and 16- sulfanylhexadecanoic acid, and herein designated MAL-S-(CH 2 ) IO -COOH and MAL-S-(CH 2 ) 15 -COOH, respectively, associated with HSA yielding Ka values of 1.3 to 1.6xlO 5 M "1 .
  • the MAL-S -(CH 2 ) 10 -COOH lost the capability to associate with HSA when linked to PEG 5 (PEG 5 -MAL-S-(CH 2 ) I0 -COOH)
  • the MAL-S-(CH 2 ), 5 -COOH linked to PEG 5 i.e., PEG 5 -MAL-S-(CH 2 ) 15 -COOH
  • effectively associated with HSA yielding a Ka value of 1.95xlO 5 M "1 , as further shown in Fig. 2B, and thus was selected for further designing of the HSA- associating probe.
  • SuO- FMS-MAL-S-(CH 2 ) 15 -COOH was reacted with an amino side chain of either a small peptide, i.e., Gly-His-Lys, or of a larger polypeptide such as exendin-4 (4.2 kDa) or insulin (5.8 kDa), forming Gly-His-Lys-FMS-MAL-S-(CH 2 ) 15 -COOH, exendin-4-FMS-MAL-S-(CH 2 ) 15 -COOH or
  • one of the sulfanyl groups of 1,11-dithio undecanoic acid is first protected with trytyl, by reacting with trytyl chloride to obtain Tr-S-(CH 2 ) 10 -SH, which is then reacted with dithiodipyridyl to obtain Tr-S-(CH 2 ) 10 -S-S-pyridyl.
  • Tr-S-(CH 2 )i 0 -S-S-pyridyl is reacted with HS-(CH 2 ) 10 -COOH to obtain Tr-S-(CH 2 ) 10 -S-S-(CH 2 ) 10 -COOH, which is then deprotected with DCM, 2% TFA and 5% trimethyl silane, for 30 min, to obtain H- S-(CH 2 ) 10 -S-S-(CH 2 ) 10 -COOH.
  • Compound 3 is obtained by reacting obtain H-S- (CH 2 ) 10 -S-S-(CH 2 ) 10 -COOH with the maleimido of the SuO-FMS-MAL spacer.
  • Example 5 Synthesis of SuO-FMS-MAL-S-(CH 2 ) l5 -CO-NH-(CH 2 ) 2 -SO 3 H, 4
  • Tr-S-(CH 2 ) 15 CO-NH-(CH 2 ) 2 -SO 3 H was achieved as described above in Example 3 (section 3.3) for HS-(CH 2 ) 15 -CO-NH-(CH 2 ) 5 -COOH, resulting with the crude HS-(CH 2 ) 15 CO-NH-(CH 2 ) 2 -SO 3 H.
  • Insulin-FMS-MAL-S-(CH 2 ) 15 -COOH is a monomodified derivative having molecular-weight of 6570 daltons (calculated value is 6565.5 daltons) as verified by mass spectroscopy.
  • PBS phosphate buffer saline
  • Table 3 hereinafter summarizes the characteristic features of HPLC-purif ⁇ ed insulin-FMS-(CH 2 ) 15 -COOH.
  • insulin-FMS-MAL-S-(CH 2 ) 15 -COOH has about 10% the efficacy of insulin to activate lipogenesis in rat adipocytes yielding an half-maximal effect (ED 50 ) at a concentration of 1.03 ⁇ 0.1 nM.
  • ED 50 half-maximal effect
  • the biological potency of such albumin associated insulin derivative may be significantly reduced, due to the presence of bovine serum albumin (BSA) (10 mg/ml) in this particular assay, as previously noted with insulin- detemir (data not shown).
  • BSA bovine serum albumin
  • insulin-FMS-MAL-S-(CH 2 ) 15 -COOH was compared with that of Zn 2+ - free insulin, both administered at a low and similar dose (0.17 nmol/mouse in 0.2 ml PBS buffer).
  • insulin-FMS-MAL-S- (CH 2 ) 15 -COOH had a flat glucose-lowering pattern that was about two folds prolonged than that of insulin.
  • insulin- FMS-MAL-S-(CH 2 ) 15 -COOH resembled that of the native hormone, although the former has, in vitro, only 10% the biological potency of insulin, as shown in Table 3 hereinabove.
  • insulin-FMS-MAL-S-(CH 2 ) 15 -COOH equals native insulin.
  • FIG. 6 shows the glucose lowering pattern of insulin-FMS-MAL- S-(CH 2 ) 15 -COOH as compared to that of insulin-detemir, when subcutaneously administered at a dose of 0.68 nmol/mouse.
  • insulin-FMS-MAL-S- (CH 2 ) I5 -COOH was highly potent in reducing blood glucose level and it did so over prolong period with a X 1 A value of 6 ⁇ 1 hours.
  • Low blood glucose level was still evident 24 hours following administration.
  • the area under the curve could not, therefore, be accurately integrated; however, it exceeded five or more times that obtained by similar dose of subcutaneously administered insulin-detemir.
  • subcutaneous administration of Zn 2+ -free insulin to mice at this dose is severely hypoglycemic.
  • the CDl strain of mice reflects well the action of exendin-4, a glucagon- like peptide- 1 agonist, in healthy and in type II diabetic patients, in the sense that at any dosage applied, circulating blood glucose level never falls below a threshold level which in CDl -mice amounts to a decrease of 27 ⁇ 3%.
  • Fig. 7 shows the glucose-lowering profile of native exendin-4 vs. exendin-4- FMS-MAL-S-(CH 2 ) 15 -COOH, both subcutaneously administered at a dose of 0.24 nmol/CD 1 mouse.
  • exendin-4- FMS-MAL-S-(CH 2 ) 15 -COOH circulating glucose reached its lowest concentration 3 hours after administration and this level was preserved over a period of 20 hours.
  • Returning to initial glucose level took place with a tVz value of 28 ⁇ 2 hours, which is 4.7 times longer than that obtained by the same dose of the native hormone.
  • Gentamicin-FMS-MAL-S-(CH 2 ) 15 -COOH is an inactive- reactivable prodrug
  • Fig. 8 shows time course of in vitro reactivation of gentamicin-FMS-MAL-
  • gentamicin-FMS-MAL-S-(CH 2 ) 15 -COOH had ⁇ 3 ⁇ 0.7% the antibacterial potency of native gentamicin; however, upon incubation in PBS buffer (pH 7.4) containing 20 mg/ml HSA, the antibacterial potency of this conjugate was generated with a X 1 A value of 7.1 ⁇ 0.2 hours, regaining full (100%) antibacterial potency following 30 hours of incubation.
  • Derivatization of the proteins is . carried out in 0.1 M Hepes (pH 7.4) containing 1 mg/ml of rFVIII or rFVIIa. SuO-FMS-MAL-S-(CH 2 ) 15 -COOH is added at 2 to 10 molar excess. Proteins are then examined for their biological potency prior to (time 0) and following their incubation at 37 0 C in 0.1 M Hepes pH 7.4, containing 140 mM NaCl and 20 mg/ml BSA.
  • rFV ⁇ I and its derivative are diluted to a final concentration of 1 ng/ml, prior of being assayed, and their biological potencies are estimated with a Coatest-SP4 FVIII (Chromogenix) kit.
  • Biological potencies of rFVIIa and its derivative are obtained by the clotting assay using "Activated Factor Vila, STACLOT® VIIa-Rtf ' (Agis, Bnei-Brak, Israel).
  • both rFVIII and rFVIIa derivatized with excess of SuO- FMS-MAL-S-(CH 2 )] 5 -COOH will have a significantly reduced biological activity at time 0 but will undergo reactivation with a tl/2 value of several hours under physiological conditions.
  • the pharmacokinetic patterns of these derivatives can be studied in in vivo experimental systems such as increased half-life after derivatives administration in FVIH-deficient mice or estimating FVIIa activity in ret serum following FVII derivative administration in rats, and it is expected that such derivatives will show considerable half-life prolongation, due to their capability to associate with serum albumin, to thereby substantially decrease their clearing rates in situ.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention provides albumin-binding probes capable of reversibly linking to short-lived amiho-containing drugs and non-covalently associating with albumin in-vivo, thereby converting said drugs into inactive reactivable prodrugs having prolonged lifetime in-vivo. The invention further provides conjugates of said probes with amino-containing drugs, as well as pharmaceutical compositions and uses thereof.

Description

PRODRUGS CONTAINING ALBUMIN BINDING PROBE
TECHNICAL FIELD
The present invention relates to albumin-binding probes capable of converting short-lived amino-containing drugs into inactive reactivable prodrugs having prolonged lifetime profiles in vivo.
BACKGROUND ART
Most polypeptide drugs, in particular nonglycosylated proteins of molecular mass less than 50 kDa, are short-lived species in vivo having circulatory half lives of 5-20 min. The short lifetime of proteins in vivo is attributed to several mechanisms including glomerular filtration in the kidneys and proteolysis at several levels (Goodman and Gilman, 1995).
An attractive strategy for improving clinical properties of small protein drugs has come to be known as PEGylation (or pegylation). Pegylated proteins are long- lived species in vivo having higher stability and aqueous solubility, as well as lower immunogenicity and antigenicity, compared with the corresponding non-pegylated proteins, and the potential for specific cell targeting (Clark et al, 1996; Delgado et al, 1992; Reddy, 2000; Bailon et al, 2001). In spite of those profound clinical properties obtained by pegylation, only a limited number of pegylated proteins is in clinical use. The covalent attachment of polyethylene glycol (PEG) chains to proteins often results in a marked reduction in biological/pharmacological potency, and this is obviously valid with regard to low molecular-weight drugs (Marcus et al, 2008). This deficiency can principally be overcome by introducing the PEG chain via a chemical bond that is sensitive to hydrolysis, or can be cleaved enzymatically by serum proteases or esterases. Clearly, a consistent rate of hydrolysis is crucial, and a prerequisite condition is therefore that the hydrolysis of the PEG chains from the conjugate is to take place at a slow rate and in a homogenous fashion in vivo.
In recent years we have developed a strategy of reversible pegylation according to which PEG chains containing sulfhydryl-moieties are covalently linked to amino groups of drug compounds through the heterobifunctional agent (2- (3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl)propanamido)-9H-fluoren-9-yl)methyl 2,5-dioxopyrrolidin-l-yl carbonate, herein designated MAL-Fmoc-OSu, or 7-(3- (2,5-dioxo-2,5-dihydro- lH-pyrrol- 1 -yl)propanamido)-9-(((2,5-dioxopyrrolidin- 1 - yloxy)carbonyloxy)methyl)-9H-fluorene-2-sulfonic acid, herein designated MAL- FMS-OSu, which contains a spontaneously hydrolyzable bond. The inactive conjugate thus obtained is transformed into a long-acting prodrug that gradually releases its pharmacologically active constituent upon incubation at physiological conditions (Marcus et al, 2008; Nesher et al, 2008; Peleg-Shulman et al, 2004; Shechter et al, 2005a; Tsubery et al, 2004).
In particular, US 7,585,837, herewith incorporated by reference in its entirety as if fully disclosed herein, discloses a compound of the formula (X)n-Y, where Y is a moiety of a drug bearing at least one functional, e.g., amino, group, and X is a radical such as:
Figure imgf000003_0001
wherein R1 is a radical containing a protein or a polymer carrier moiety; R2 is selected from hydrogen, (CrC8)alkyl, (CrQ)alkoxy, (CrCg)alkoxyalkyl, (C6- C10)aryl, (CrC8)alkaryl, (C6-C10)ar(CrC8)alkyl, halogen, nitro, -SO3H, -SO2NHR, amino, ammonium, carboxyl, PO3H2, or OPO3H2, and R is selected from hydrogen, (CrQ)alkyl or (C6-C10)aryl; R3 and R4, the same or different, are each selected from hydrogen, (CrC8)alkyl or (C6-C10)aryl; A is a covalent bond when the radical is linked to a carboxyl, phosphate or mercapto group of the drug Y, or A is OCO- when the radical is linked to an amino or hydroxyl group of the drug Y; n is an integer of at least one, and pharmaceutically acceptable salts thereof. As disclosed in this publication, said protein carrier may be, e.g., albumin such as human serum albumin (HSA), said polymer carrier may be, e.g., a linear or branched PEG, and said drug containing at least one free amino group may be a non-peptidic drug or a peptide or protein drug, most preferably of low or medium molecular weight.
Since albumin is long-lived in vivo, drugs and endogenous substances that tightly associate with albumin have lower clearance rates than that of the unbound substances, and exhibit prolonged lifetime profiles in vivo (Taylor and Granger, 1984). Long-chain fatty acids (LCFAs) bind tightly to albumin (Carter and Ho, 1994), and this provided the impetus for designing an insulin derivative, in which LCFA-like probe has been integrated into the insulin molecule (Kurtzhals et al, 1995, 1996, 1997). The optimal derivative thus obtained (insulin-detemir) possessing protracted action in vivo, in part due to its associating affinity to endogenous albumin (Kurtzhals et al, 1995, 1996, 1997).
Insulin detemir (Levemir®, NovoNordisk) is a long-acting human insulin analog with up to 24 hours duration of action. In particular, it is an insulin analog in which the amino acid threonine in position B30 is omitted and myristic acid has been attached to the amino acid lysine in position B29 via the acyl group, i.e., N6B29- tetradecanoyl des(B30) insulin. In the blood, insulin-detemir binds to albumin through the alkyl residue of the myristic acid and it is then slowly dissociated from this complex.
Insulin detemir as well as other similar derivatives of insulin are disclosed in US Patent Nos. 5,750,497, 6,011,007 and 6,869,930, and in US Patent Publication Nos. 20040110664 and 20060030518. These publications disclose an insulin derivative in which (i) the amino acids at positions A21 and B3 are, independently, any amino acid residue which can be coded for by the genetic code except Lys, Arg and Cys; (ii) the amino acid at position Bl is Phe or is deleted; (iii) the amino acid at position B30 is any amino acid residue which can be coded for by the genetic code except Lys, Arg and Cys, or is deleted; and (iv) the amino e-amino group of LysB29 is substituted with an acyl group having at least 10 carbon atoms or a lipophilic substituent having at least 6 carbon atoms, wherein the insulin derivative is a Zn2+ complex and the Zn2+ complex of the insulin derivative is more water soluble than the insulin derivative without Zn2+. The technology disclosed in these publications is directed to insulin derivatives only, wherein the lipophilic substituent is linked to the insulin derivative via an amino group on the insulin molecule, preferably the €-amino of the amino acid lysine at position B29, and the insulin derivative is bound to albumin, upon administration, mainly via binding groups present in the albumin molecule capable of binding aliphatic chains.
US 7,186,797 discloses polypeptide conjugates having extended half life in vivo, comprising a polypeptide conjugated to a binding moiety having affinity for albumin. The binding moiety disclosed has two arms, wherein each one of these arms binds to albumin via a certain linking group that is either an aryl moiety or a non-aromatic moiety having 1-10 carbon atoms.
WO 2008053360 discloses portable albumin binders, capable of binding to albumin through a functional group that is negatively charged or may be deprotonated to yield a negative charge, e.g., a carboxyl group, which are said to be useful for improving the pharmacokinetic properties of diagnostic or therapeutic agents, e.g., by increasing their circulation lifetime.
SUMMARY OF INVENTION
Contrary to the concept disclosed in the aforesaid US 7,585,837, according to which a polymer carrier, e.g., PEG, or a protein carrier, e.g., albumin, is covalently linked to a drug via a heterobifunctional agent such as FMS or Fmoc, the present invention is based on a concept according to which a long chain fatty acid (LCFA) like albumin-binding compound is covalently linked to a short-lived amino-containing drug to form a drug conjugate capable of non-covalent association with albumin in vivo, i.e., a long-lived prodrug that gradually releases the pharmacologically active constituent. In particular, it has been found, in accordance with the present invention, that by introducing a certain LCFA like albumin-binding compound, in particular, the 16-(l-(3-(9-(((2,5-dioxopyrrolidin-l-yloxy) carbonyloxy)-methyi)-7-sulfo-9H- fluoren-2-ylamino)-3-oxopropyl)-2,5-dioxopyrrolidin-3-ylthio) hexadecanoic acid, herein designated SuO-FMS-MAL-S-(CH2)15-COOH, to various short-lived amino- containing drugs such as insulin, exendin-4, gentamicin, Factor Vila and Factor VIII, a conjugate having associating affinity to albumin is formed. The albumin- associating affinity of the conjugate formed is in the range of Ka=2.0-2.6xl05 M" , i.e., sufficient to turn short-lived molecules to long-lived species in vivo. Furthermore, although low molecular-weight drugs suffer from a massive loss of pharmacological potency upon conjugation, such drugs, when conjugated to said albumin-binding compound, regain their full potency upon incubation at physiological conditions.
In one aspect, the present invention thus relates to a compound of the formula I:
Figure imgf000006_0001
wherein
Ri is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO2NH-, -O-, - OCO-, -CO-NH-, -CS-NH-, -CO(CH2)M-, or -R8-CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N;
R2 is selected from >
Figure imgf000006_0002
R9, or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO3H group;
R3 is absent or an acidic group having at least one hydroxyl group such as - COOH, -SO3H or -0-PO3H2;
R4 is an electron withdrawing group such as -SO3H, -CN, -CO-(C rC8)alkyl, -CO-(C6-Cio)aryl, -NO2, -OPO3H2, -N(R)3 +, -SO2NH2, or halogen, wherein R is selected from (CrC8)alkyl or (C6-C10)ar(CrC8)alkyl; R5 and R6, each independently is selected from hydrogen, -(CrC8)alkyl or (C6-C10)aryl;
R7 is a leaving group such as -O-(CH2)2-CN, -Cl,
Figure imgf000007_0001
R9 is selected from (Ci3-C20)alkylene, (C13-C2o)alkenylene or (C13- C20)alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(d-C8alkyl)-, -N(C6-C 10aryl)-, or -(C6-C 10)arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R2 is a peptide moiety, R3 is absent.
In another aspect, the present invention relates to a conjugate of the formula
Figure imgf000007_0002
wherein
Y is a moiety of a drug containing at least one amino group, linked through said at least one amino group; R1 is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO2NH-, -O-, - OCO-, -CO-NH-, -CS-NH-, -CO(CH2)1-4-, or -R8-CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N;
R2 is selected from >
Figure imgf000008_0001
Rg, or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, lie or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO3H group;
R3 is absent or an acidic group having at least one hydroxyl group such as - COOH, -SO3H or -0-PO3H2;
R4 is an electron withdrawing group such as -SO3H, -CN, -CO-(C 1-C8)alkyl, -CO-(C6-C10)aryl, -NO2, -OPO3H2, -N(R)3 +, -SO2NH2, or halogen, wherein R is selected from (CrC8)alkyl or (C6-C10)ar(CrC8)alkyl;
R5 and R6, each independently is selected from hydrogen, -(Q-C8)alkyl or - (C6-C10)aryl; and
R9 is selected from (Ci3-C20)alkylene, (C13-C2o)alkenylene or (C13- C20)alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10ary I)- or -(C6-C10)arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R2 is a peptide moiety, R3 is absent.
In a further aspect, the present invention provides a pharmaceutical composition comprising a conjugate of the formula II as defined above, i.e., a conjugate obtained by nucleophilic substitution of a compound of formula I with an amino group of the drug Y, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The pharmaceutical compositions of the present invention can be used for treatment of various diseases, disorders and conditions, in which administration of the drug Y might be useful.
Thus, in still a further aspect, the present invention provides a method for treatment of diabetes mellitus or hyperglycemia comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is insulin, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of insulin.
In yet a further aspect, the present invention provides a method for treatment of insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, or gestational diabetes mellitus, or for prevention of hyperglycemia, said method comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is exendin-4, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of exendin-4. In still another aspect, the present invention provides a method for treatment of a bacterial infection comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is gentamicin, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of gentamicin. In yet another aspect, the present invention provides a method for treating a patient in need of Factor Vila or Factor VIII therapy, comprising administering to said patient an effective amount of a conjugate of formula II, as defined above, wherein the drug Y is Factor Vila or Factor VIII, obtained by nucleophilic substitution of a compound of formula I, as defined above, with an amino group of Factor Vila or Factor VIE, respectively.
BRIEF DESCRIPTION OF DRAWINGS
Figs. 1A-1B show the binding affinity of insulin-detemir to human serum albumin (HSA) as determined by ITC-200. The processed data has been derived from 20 automatic injections (2.4 μl each, IA) of insulin-detemir (400 μM in phosphate buffer saline, PBS, buffer, pH 7.4) into the sample cell containing HSA at a concentration of 10 μM in PBS buffer, and was translated to a binding isotherm
(IB). Data: HSA NDH; model: OneSites; chiA2/DoF=2.235xl04; N=0.693±0.0430 sites; K=6.87xl04±1.60xl04 M"1; ΔH=-4575±406.7 cal/mol; ΔS=6.79 (cal/mol)/deg.
Figs. 2A-2B show simulated binding isotherm for the association of PEG5- MAL-S-(CH2)15-COOH with HSA. The data was obtained for 15 automatic injections each of 2.7 μl. The total duration of the experiments was 45 min (2A).
The concentration of PEG5-MAL-S-(CH2)15-COOH in the injection syringe was 400 μM. Sample cell contained HSA at a concentration of 10 μM. Both components were dissolved in PBS-buffer pH=7.4. The experiment was conducted at 23°C. Data: A400HSA10_NDH; model: OneSites; chiΛ2/DoF=3.454xl04; N=1.76±0.0915 sites; K=1.95xl05±3.17xl04 M"1; ΔH=-1.0374xl04±712.6 cal/mol; ΔS=-10.6 (cal/mol)/deg.
Fig. 3 shows HPLC analysis of purified insulin-FMS-MAL-S-(CH2)15- COOH. HPLC-purified insulin-FMS-MAL-S-(CH2)15-COOH (50 μg) was loaded on a chromolith Rp- 18e (100 mm X 4 mm) column and run with a linear gradient from 0 to 100% solution A (0.1% trifluoroacetic acid, TFA) to solution B (acetonitrile-H2O, 75:25 in 0.1% TFA) over 10 min, and then over 4 min in solution B at a rate of 3 ml/min. The effluent was monitored at 220 nm.
Fig. 4 shows prolonged residence time of 125I-insulin-FMS-MAL-S-(CH2)15- COOH following intravenous administration in rats. Two groups of rats (n=3 per group) received either 125I-insulin (-D-) or HPLC-purified 125I-insulin-FMS-MAL-S- (CH2)15-COOH (-■-; 8.4±0.4 x 106 CPM per 220±10 grams rat). At the indicated time points, blood aliquots (50-70 mg) were drawn and counted for their radioactive content. Fig. 5 shows circulating glucose levels in CDl -mice following a single subcutaneous administration of insulin-FMS-MAL-S-(CH2)i5-COOH. Mice were subcutaneously injected with PBS-buffer (-•-; 0.2 ml/mouse), Zn2+ free insulin (-■-; 0.17 nmol/mouse in 0.2 ml PBS buffer) or insulin-FMS-MAL-S-(CH2)15-COOH (- O-; 0.17 nmol/mouse in 0.2 ml PBS buffer). Blood glucose levels were determined at the indicated time points. Each point is the arithmetic mean of n=5 mice±SE. Fig. 6 shows circulating glucose levels in CDl -mice following a single subcutaneous administration of either insulin-FMS-MAL-S-(CH2)15-COOH or insulin-detemir. Mice were subcutaneously injected with PBS-buffer (-•-), insulin- FMS-MAL-S-(CH2)15-COOH (-O-) or insulin-detemir (-■-; 0.68 nmol/mouse in 0.2 ml PBS buffer), and blood glucose levels were determined at the indicated time points. Each point is the arithmetic mean of n=5 mice±SE.
Fig. 7 shows glucose lowering pattern of exendin-4-FMS-MAL-S-(CH2)15- COOH, following a single subcutaneous administration to CDl -mice. Three groups of mice (n=6 per group) underwent one subcutaneous administration of PBS buffer (-D-) pH 7.4, native exendin-4 (-O-; 0.24 nmol/mouse) or exendin-4-FMS-MAL-S- (CH2)15-COOH (-O-; 0.24 nmol/mouse), and circulating glucose levels were then monitored at the time points indicated in this figure. Each point is the arithmetic mean of n=6 mice±SE.
Fig. 8 shows time course of in vitro reactivation of gentamicin-FMS-MAL- S-(CH2)15-COOH. Gentamicin-FMS-MAL-S-(CH2)i 5-COOH (0.16 μmoles/ml) was incubated in PBS, pH 7.4, containing 2% (w/v) HSA at 370C. Aliquots were withdrawn at the indicated time points and analyzed at several concentrations in the antibacterial assay. Gentamicin inhibits E. CoIi replication with IC5o=2.1±O.2 μM. An aliquot with ICso=21±2 μM is considered as having 10% of the native gentamicin antibacterial potency.
Fig. 9 shows schematically the principle of converting short-lived drugs into long-lived species in-vivo using a certain albumin-binding probe of the present invention. Inactive albumin-associated conjugates bind to serum albumin and therefore exhibit prolonged residence time in situ, during which the parent amino containing molecules are released from the inactive conjugates in their native-active form, at a slow rate over many hours following administration.
MODES FOR CARRYING OUT THE INVENTION
Albumin is the most abundant protein in the blood, at a concentration of approximately 600 μM. One of the physiological roles of albumin is to act as a carrier of fatty acids, due to the fact that long chain fatty acids (LCFAs) bind tightly to albumin, wherein the terminal carboxylate (-CH2-COOH) serves as an albumin- binding ligand. According to the present invention, this specific property of LCFA like compounds has been exploited in order to associate any amino-containing drug with albumin, thus prolonging the life of said drug in the blood circulation.
In one aspect, the present invention relates to a compound of the formula I, i.e., to an albumin-binding ligand, as defined above.
The term "(Ci-C8)alkyl", as used herein, typically means a straight or branched hydrocarbon radical having 1-8 carbon atoms and includes, e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, 2,2- dimethylpropyl, n-hexyl, n-heptyl, n-octyl, and the like. The terms "(Ci3- C20)alkylene" refers to a straight or branched divalent hydrocarbon radical having 13-20 carbon atoms and includes, e.g., n-tridecanylene, n-tetradecanylene, n- pentadecanylene, n-hexadecanylene, n-heptadecanylene, n-octadecanylene, n- nonadecanylene, icosanylene, and the like. The terms "(C13-C20)alkenylene" and "(C13-C2o)alkynylene" typically mean straight or branched divalent hydrocarbon radicals having 13-20 carbon atoms and one or more double or triple bonds, respectively, wherein each one of said double or triple bonds is not a terminal double or triple bond. Non-limiting examples of such radicals include 2-, 3-, 4-, 5- and 6-tridecenylene, tetradecenylenes such as myristoleylene, 2-, 3-, 4-, 5-, 6- and 7-pentadecenylene, hexadecenylenes such as palmitoleylene, 2-, 3-, 4-, 5-, 6-, 7- and 8-heptadecenylene, octadecenylenes such as oleylene, linoleylene, α- linoleylene, nonadecenylene, icosenylenes such as arachidonylene and eicosapentylene, and the like. The term "(C6-C 10)aryl" denotes an aromatic carbocyclic group having 6-10 carbon atoms consisting of a single ring or condensed multiple rings such as, but not limited to, phenyl and naphthyl; the term "ar(Ci-Cg)alkyl" denotes an arylalkyl radical such as benzyl and phenetyl; and the term "(C6-C iO)arylene-diyl" denotes a divalent aromatic carbocyclic group having 6-10 carbon atoms consisting of either a single ring or condensed multiple rings such as, but not limited to, phenylene and naphthylene.
The term "amino acid residue", as used herein, refers to any natural or synthetic, i.e., non-natural, amino acid residue in its both L- and D-stereoisomers. While a natural amino acid is any one of the twenty amino acid residues typically occurring in proteins, the term synthetic/non-natural amino acid refers to any amino acid, modified amino acid and/or an analog thereof, that is not one of the twenty natural amino acids. The term "aliphatic hydrophobic amino acid residue" refers to an amino acid residue having an aliphatic hydrocarbyl side chain. Non-limiting examples of aliphatic hydrophobic amino acids include the natural amino acids leucine, isoleucine and valine, as well as the non-natural amino acids norvaline (Nva), norleucine (NIe), homovaline and homoleucine. The term "aromatic amino acid residue" refers to an amino acid residue in which the side chain contains an aromatic ring. Examples of aromatic amino acids, without being limited to, include the natural amino acid phenylalanine as well as the non-natural amino acids bipyridyl alanine, p-carboxymethyl-L-phenylalanine and p-nitro-L-phenylalanine. The term "amino acid analog comprising -COOH or -SO3H group" refers to any amino acid analog having amino group as well as -COOH, -SO3H or both groups, such as, without being limited to, taurine. The term "leaving group", as used herein, refers to any functional group or atom, which can be displaced by another functional group or atom in a substitution reaction, e.g., a nucleophilic substitution reaction. Non-limiting examples of leaving groups include -0-(CH)2-CN, 2,5-dioxopyrrolidin-l-olate also known as N- hydroxysuccinimide (herein designated -OSu), 4-nitrophenoxy, 2-nitrophenoxy, 2,3,4,5,6-pentachlorophenoxy, isoindoline-l,3-dione-2-oxy, and benzenesulfanyl, wherein -OSu is preferred.
In certain embodiments, the compound of the present invention is a compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, and R7 is -OSu. In particular embodiments, the compound of the present invention is a compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R, is -NH-CO- or -NH-, preferably -NH-CO-, at position 7 of the fluorene ring, R2 is Rg or >
Figure imgf000014_0001
R9 is selected from (Ci3-C20)alkylene, (Ci3-C20)alkenylene or (C13-C20)alkynylene, preferably (C13-C20)alkylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO- NH-, -N(CrC8alkyl)-, -N(C6-C 10ary I)- or -(C6-C 10)arylene-diyl, and R3 is -COOH or SO3H. As defined above, it should be noted that in cases wherein the alkylene, alkenylene or alkynylene is interrupted by more than one, e.g., two, heteroatoms, these heteroatoms may be either identical or different heteroatoms, and can be linked sequentially forming, e.g., -S-S- (disulfide), -N-N- or -O-S- bond, or at any two positions of the alkylene, alkenylene or alkynylene.
In certain particular embodiments, the compound of the present invention is a compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO- at position 7 of the fluorene ring, R2 is either R9 or >
Figure imgf000014_0002
R9 is (C13-C2o)alkylene optionally interrupted by two sulfur atoms forming disulfide bond or by -CO-NH-, and R3 is -COOH or SO3H.
The specific compounds of the formula I described in the specification are herein identified by the Arabic numbers 1-8 in bold, wherein their full chemical structures are depicted in Table 1 hereinafter. In one specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R) is -NH-CO-, R2 is >
Figure imgf000015_0001
R3 is -COOH, and R9 is -(CH2)15-, i.e., 16-(l-(3-(9-(((2,5-dioxopyrrolidin-l- yloxy)carbonyloxy)-methyl)-7-sulfo-9H-fluoren-2-ylamino)-3-oxopropyl)-2,5- dioxopyrrolidin-3-ylthio) hexadecanoic acid (herein identified SuO-FMS-MAL-S- (CH2)15-COOH or compound 1). In another specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2
Figure imgf000015_0002
R3 is -COOH, and R9 is -(CH2) 15-CO-NH-(CH2)5-, i.e., 6-(16-(l-(3-(9-(((2,5- dioxopyrrolidin-l-yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-3- oxopropy l)-2 , 5 -dioxopyrrolidin-3 -y lthio)hexadecanamido)hexanoic acid (herein identified SuO-FMS-MAL-S-(CH2)15-CO-NH-(CH2)5-COOH or compound 2).
In a further specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring,
R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is
Figure imgf000015_0003
R3 is -COOH, and R9 is -(CH2)10-S-S-(CH2)10-, i.e., l l-((10-(l-(3-(9-(((2,5- dioxopyrrolidin- 1 -yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-3- oxopropyl)-2,5-dioxopyrrolidin-3-ylthio)decyl)disulfanyl)undecanoic acid (herein identified SuO-FMS-MAL-S-(CH2)10-S-S-(CH2)10-COOH or compound 3). In another specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is >
Figure imgf000016_0001
R3 is -SO3H, and R9 is -(CH2) 15-CO-NH-(CH2)2-, i.e., 7-(3-(2,5-dioxo-3-(16-oxo- 16-(2-sulfoethylamino)hexadecylthio)pyrrolidin- 1 -yl)propanamido)-9-(((2,5- dioxopyrrolidin- l-yloxy)carbonyloxy)methyl)-9H-fluorene-2-sulfonic acid (herein identified SuO-FMS-MAL-S-(CH2)15-CO-NH-(CH2)2-SO3H or compound 4). In still another specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is R9, R3 is - COOH, and R9 is -(CH2)15-, i.e., 17-(9-(((2,5-dioxoρyrrolidin-l- yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-17-oxoheptadecanoic acid (herein identified SuO-FMS-(CH2)15-COOH or compound 5).
In yet another specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is R9, R3 is -COOH, and R9 is -(CH2)15-CO-NH-(CH2)5-, i.e., 6-(17-(9-(((2,5-dioxopyrrolidin-l- yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)- 17-oxoheptadecan- amido)hexanoic acid (herein identified SuO-FMS-(CH2)15-CO-NH-(CH2)5-COOH or compound 6).
In still a further specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is R9, R3 is - COOH, and R9 is -(CH2)10-S-S-(CH2)10-, i.e., l l-((l l-(9-(((2,5-dioxopyrrolidin-l- yloxy)-carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)- 11 -oxoundecyl) disulfanyl)-undecanoic acid (herein identified SuO-FMS-(CH2)10-S-S-(CH2)10- COOH or compound 7). In yet a further specific embodiment, the compound of the present invention is the compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is -OSu, R1 is -NH-CO-, R2 is R9, R3 is -SO3H, and R9 is -(CH2) 15-CO-NH-(CH2)2-, i.e., 9-(((2,5-dioxopyrrolidin-l-yloxy) carbonyl oxy)methyl)-7-( 17-oxo- 17-(2-sulfoethylamino)heptadecanamido)-9H-fluorene-2- sulfonic acid (herein identified SuO-FMS-(CH2)15- CO-NH-(CH2)2-SO3H or compound 8).
In other particular embodiments, the compound of the present invention is a compound of formula I, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, R7 is OSu, R1 is selected from -NH-CO-, -OCO-, or -R8- CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N, and R2 is a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or -SO3H group such as taurine.
Table 1: Specific compounds of the invention described in the specification
Figure imgf000017_0001
Structure
Figure imgf000018_0001
The compounds of the present invention may be prepared according to any technology or procedure known in the art, e.g., as described in detail in Tsubery et al. (2004) and in various additional publications of the scientific groups of the inventors (Peleg-Shulman et al, 2004; Shechter et al, 2005a; Shechter et al, 2001a; Nesher et al, 2008; Shechter et al, 2007; Shechter, 2005b). For example, the hydrolyzable heterobifunctional intermediate compound 7-(3-(2,5-dioxo-2,5- dihydro- 1 H-pyrrol- 1 -yl)propanamido)-9-(((2,5-dioxopyrrolidin- 1 -yloxy)carbonyl oxy) methyl)-9H-fiuorene-3-sulfonic acid, herein designated MAL-FMS-OSu, can be prepared as described in Tsubery et al. (2004) starting from 9-hydroxymethyl-2- aminofluorene, and the compounds of the formula I may then be obtained by reacting said MAL-FMS-OSu with, e.g., a HS-(C13-C20)alkylene-COOH, HS-(C13- C20)alkenylene-COOH or HS-(C13-C20)alkynylene-COOH, thus obtaining the corresponding derivatives of the compound of formula I having the general formula SuO-FMS-MAL-S-(C13-C20)alkylene-COOH, SuO-FMS-MAL-S-(C 13-C20) alkenylene-COOH or SuO-FMS-MAL-S-(Ci3-C20)alkynylene-COOH, respectively, which are, in fact, LCFA like molecules containing sulfhydryl moieties, where the terminal carboxylate (-CH2-COOH) serves as an albumin-binding ligand.
Particular procedures for the preparation of various compounds according to the present invention, in particular, compounds 1, 2, 3 and 4, are exemplified in Examples 2-5 hereinafter.
The binding properties of the compounds of formula I to human serum albumin (HSA) can be evaluated by any suitable technique, e.g., by isothermal scanning calorimetry (ITC) as exemplified in the Example section hereinafter. As particularly shown in Example 2, both 10-(2,5-dioxopyrrolidin-3-ylthio)decanoic acid and 16-(2,5-dioxopyrrolidin-3-ylthio)hexadecanoic acid, herein designated MAL-S-(CH2)10-COOH and MAL-S-(CH2)15-COOH, respectively, were prepared by reacting maleimide moiety with 11-mercaptoundecanoic acid or 16- mercaptohexadecanoic acid, respectively, and associated with HSA yielding Ka values of 1.3 to l.όxlO5 M"1. However, while the former completely lost this capability when linked to a 5 kDa polyethylene glycol molecule (PEG5), i.e., a macromolecule representing a drug, PEG5-MAL-S-(CH2)i5-COOH effectively associated with HSA, yielding a Ka value of 1.95xlO5 M"1 indicating that the length of the LCFA like molecule may significantly influence its ability, when conjugated with a macromolecule such as a drug, to associate with HSA. Thus, preferred compounds according to the present invention are those in which the shortest chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin, i.e., the hydroxyl group of R3 or the terminal hydroxyl group of the peptide moiety in cases R2 is a peptide moiety and R3 is absent, is of 15 to 30 atoms. The term "chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin", as used herein, refers to any chain of atoms formed by the sequence R1-R2-R3 in the compound of formula I, which links the fluorene ring and either the hydroxyl group of R3 or the terminal hydroxyl group of the peptide moiety, in cases R2 is a peptide moiety and R3 is absent, through which said compound binds to albumin. In view of the definitions of R1, R2 and R3, said chain of atoms may be interrupted by one or more heteroatoms independently selected from oxygen, nitrogen or sulfur; functional groups such as -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO2NH-, -O-, -OCO-, -CO- NH-, -CS-NH-, -CO-; or a cyclic aliphatic or aromatic ring such as -OSu or phenyl, respectively. Said cyclic aliphatic ring can be linked through any position of the ring, e.g., through positions 1 and 3 in a 5-membered aliphatic ring such as 3- mercaptopyrrolidine-2,5-dione, also known as 3-mercaptosuccineimide (herein designated MAL-S), or through any two positions being located ortho, meta or para one to another, in a 6-membered aromatic ring such as phenyl.
It should be noted that in cases the backbone of R2 does not contain a cyclic aliphatic or aromatic ring, only one chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin exists. However, in cases a cyclic aliphatic or aromatic ring interrupts the backbone of R2, e.g., in the case wherein R1 is -NH-CO-, R3 is -COOH and R2 is °Λ S_(CH2)T3_ ,
Figure imgf000020_0001
two chains of atoms linking the fluorene ring and the hydroxyl group of R3 exist, wherein one of said chains consists of the sequence -NH-CO-CH2-N-CO-CH-S- (CH2)o-CO~, i.e., is of 21 atoms, and the other chain consists of the sequence -NH- CO-CH2-N-CO-CH2-CH-S-(CH2)J3-CO-, i.e., is of 22 atoms. The term "the shortest chain of atoms linking the fluorene ring and the terminal acidic group associating with albumin" thus refers to the chain of atoms linking the fluorene ring and said terminal acidic group in cases a single such chain exists or, alternatively, to the chain of atoms linking the fluorene ring and said acidic group, having the lowest number of atoms in its backbone, in cases more than one such chains exist. As stated above and shown in the Example section hereinafter, conjugates of formula II formed by introducing a compound of formula I, in particular, SuO- FMS-MAL-S-(CH2)15-COOH, with various short-lived amino-containing drugs, in particular, insulin, exendin-4, gentamicin, Factor Vila and Factor VIII, have associating affinity to HSA that is in the range of Ka=2.0-2.6xl05 M"1, i.e., sufficient to turn said short-lived drugs to long-lived species in vivo. It is expected that corresponding conjugates formed by introducing a compound of formula I having a terminal sulfonyl (-SO3H) instead of carboxyl, e.g., compounds 4 and 8, with such short-lived amino-containing drugs, would have even higher associating affinity to HSA, due to the higher affinity of the sulfonyl group to HSA compared with that of the carboxyl group.
As commonly known, ligand-protein affinities are affected by non-covalent intermolecular interactions between the two molecules such as hydrogen bonding, electrostatic interactions, hydrophobic and Van der Waals forces, and may also be affected by high concentrations of other macromolecules that cause macromolecular crowding. With this respect, it is particularly known that low molecular-weight drugs suffer a massive loss of pharmacological potency upon conjugation. As further shown herein, gentamicin representing a low molecular-weight drug, when introduced with SuO-FMS-MAL-S-(CH2)15-COOH to form a conjugate of formula II, regains its full potency upon incubation at physiological conditions. In another aspect, the present invention thus relates to a conjugate of the formula II as defined above. This conjugate may be obtained by nucleophilic substitution of a compound of the formula I, as defined above, with any amino- containing drug, i.e., by nucleophilic substitution of R7 in the compound of formula I with the amino group of said drug.
The drug according to the present invention may be any drug containing at least one amino group.
In one embodiment, the drug is an aminoglycoside antibiotic such as gentamicin or amphotericin, an antineoplastic drug such as aminolevulinic acid, or an anthracycline chemo therapeutic agent such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone and valrubicin.
In another embodiment, the drug is a peptide or a protein drug of low or medium molecular weight such as insulin, an interferon, preferably IFN-α2, a peptide YY (PYY) agonist, preferably the peptide PYY3-36, an exendin, preferably exendin-3 or exendin-4, an exendin analog or exendin agonist, atrial natriuretic peptide (ANP), human growth hormone (hGH), erythropoietin, TNF-α, calcitonin, gonadotropin releasing hormone (GnRH), a GnRH analogue, hirudin, glucagon, a coagulation factor such as Factor Vila and Factor VIII, and a monoclonal antibody fragment, preferably anti-TNF-α monoclonal antibody fragment. Insulin is the predominant drug for diabetes mellitus, a group of syndromes characterized by hyperglycemia, altered metabolism of lipids, carbohydrates and proteins, and an increased risk of complications from vascular diseases. Most patients can clinically be classified as having either insulin-dependent (Type I) or insulin-independent diabetes mellitus (Type II). About 90% of diabetic patients in the Western world have Type II diabetes, and about 70% of the Type II diabetics in the United States are also obese, a factor that significantly contributes to insulin resistance. Whereas in Type I diabetes, there is an extensive and selective loss of pancreatic /J-cells and a state of hypoinsulinemia, there is no significant loss of β- cells from the islets in Type II diabetic patients, in which patients the mean plasma concentration of insulin over a 24-hour period is essentially normal or even elevated because of peripheral resistance to the action of the hormone. Nevertheless, individuals with Type II diabetes are relatively insulin deficient, as a normal pancreatic /3-cell should be capable of secreting amounts of insulin that are considerably greater than normal when confronted with hyperglycemia, thus allowing an individual to maintain euglycemia in the face of moderate resistance to insulin.
Virtually all forms of diabetes mellitus are due to either a decrease in the circulating concentration of insulin (insulin deficiency) or a decrease in response of peripheral tissues to insulin (insulin resistance), in association with an excess of hormones with actions opposite to those of insulin, i.e., glucagon, growth hormone, Cortisol and catecholamines.
The half-life of insulin in plasma is about 5-6 min, wherein the degradation of insulin occurs primarily in liver and to a lesser extent in kidney and muscle. Proteolytic degradation of insulin in the liver is primarily receptor mediated. Various modifications have been described in order to create insulin analogs having ' longer half-lives in the blood circulation, in particular, prodrugs capable of releasing active insulin into the circulation over a relatively long time period, i.e., 8-24 hours, intended to provide the required basal level of insulin for a whole day. One of such long-acting human insulin analogs is the aforesaid insulin detemir (Levemir®, NovoNordisk), produced by a process including expression of recombinant DNA in Saccharomyces cerevisiae followed by chemical modification, which said to have up to 24 hours duration of action. In particular, insulin detemir is an insulin analog in which the amino acid threonine in position B30 is omitted and myristic acid has been attached to the amino acid lysine in position B29, i.e., N£B29-tetradecanoyl des(B30) insulin. In the blood, insulin-detemir binds to albumin through the acyl group at position B29 and it is then slowly dissociated from this complex.
As shown in Examples 6-8, a conjugate according to the present invention formed by introducing insulin to a compound of formula I, in particular, the conjugate herein designated insulin-FMS-MAL-S-(CH2)15-COOH, formed by introducing insulin to SuO-FMS-MAL-S-(CH2)15-COOH, had about 10% the efficacy of insulin to activate lipogenesis in rat adipocytes yielding an half-maximal effect (ED50) at a concentration of 1.03±0.1 nM; however, it has regained its full lipogenic potency (ED50=O. l±O.02 nM) following 4 hours of incubation under conditions that completely release insulin from the conjugate. Furthermore, while the circulating level of insulin declined yielding a tVi value of 3.3±0.4 hours, the circulating level of said conjugate increased over a period of 2 hours reaching a value of 31,000±l,000 cpm/ml blood, which was stably maintained over a period of 6 hours and than declined with a X1A value of 17±1 hours, and a significant amount, in particular, -10,000 cpm/ml blood, was still evident 30 hours after intravenous administration. In addition, said conjugate had a flat glucose-lowering pattern that was by about two folds prolonged than that of insulin.
The aforesaid conjugate was subcutaneously administered at a dose of 0.68 nmol/mouse and as shown, it was highly potent in reducing blood glucose level over prolong time period with a tY≥ value of 6±1 hours, wherein low blood glucose level was still evident 24 hours following administration. In particular, although the area under the curve could not accurately integrated, it exceeded five or more times that obtained by similar dose of subcutaneously administered insulin-detemir.
Thus, in certain embodiments, the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of insulin.
Exendins are peptides found in the venom of the Gila-monster, a lizard found in Arizona, and the Mexican Beaded Lizard. Exendin-3 is present in the venom of Heloderma horridum, and exendin-4 is present in the venom of Helodermasuspectum. The exendins have some sequence similarity to several members of the glucagon-like peptide family, with the highest homology, 53%, being to GLP-1[7-36]NH2, which is also known as proglucagon, and has an insulinotropic effect, stimulating insulin secretion from pancreatic /3-cells. Exendin- 4 is composed of 39 amino acid residues with the carboxy terminus amidated. Exendin-4 potently binds at GLP-I receptors on insulin-secreting /3TCl cells, at dispersed acinar cells from guinea pig pancreas, and at parietal cells from stomach. The use of exendin-3 and exendin-4 as insulinotrophic agents for the treatment of diabetes mellitus and the prevention of hyperglycemia has been previously proposed, e.g., in US 5,424,286.
The glucose-lowering profile of native exendin-4 was compared with that of an exendin-4-based conjugate according to the present invention, in particular, the conjugate herein designated exendin-4-FMS-MAL-S-(CH2)15-COOH, formed by introducing exendin-4 to SuO-FMS-MAL-S-(CH2)15-COOH, when subcutaneously administered at a dose of 0.24 nmol/CDl mouse, and as shown in Example 9, circulating glucose reached its lowest concentration 3 hours following administration of said conjugate and this level was preserved over a period of 20 hours. Returning to initial glucose level took place with a iVi value of 28±2h, which is 4.7 times longer than that obtained by the same dose of the native hormone.
Thus, in other certain embodiments, the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of exendin-4.
While large protein drugs can accommodate one or two HSA-binding probe(s), each having a size of about 760 daltons, with the preservation of significant amount of their biological/pharmacological potencies (Shechter et al, 2001b; Shechter et al., 2007), low molecular-weight amino containing compounds suffer a massive loss of pharmacological potency upon conjugation, and are thus impractical under these circumstances, unless may be reactivated upon administration. In order to test whether the approach of the present invention may be used for low molecular-weight amino containing compounds as well, a gentamicin-based conjugate according to the present invention, in particular, the conjugate herein designated gentamicin-FMS-MAL-S-(CH2)15-COOH, formed by introducing gentamicin to SuO-FMS-MAL-S-(CH2)15-COOH, was used.
Gentamicin is an aminoglycoside antibiotic, used in treatment of many types of bacterial infections, particularly those caused by Gram-negative bacteria. Gentamicin works by binding the 30S subunit of the bacterial ribosome, thus interrupting protein synthesis. As shown in Example 10, the conjugate gentamicin- FMS-MAL-S-(CH2)i5-COOH that was incubated in PBS (pH 7.4) containing 2% (w/v) HSA at 370C had ~3±0.7% the antibacterial potency of native gentamicin; however, upon incubation in PBS buffer (pH 7.4) containing 20 mg/ml HSA, the antibacterial potency was generated with a t/2 value of 7.1±0.2 h, regaining full (100%) antibacterial potency following 30 hours of incubation.
Thus, in further certain embodiments, the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of gentamicin.
Factor VII (FVII), formerly known as proconvertin, is a vitamin K dependent enzyme of the serine protease class, produced in the liver, and is one of the central proteins in the coagulation cascade. The main role of FVII is to initiate the process of coagulation in conjunction with tissue factor, which is found on the outside of blood vessels, normally not exposed to the bloodstream. Upon vessel injury, tissue factor is exposed to the blood and circulating FVII. Once bound to tissue factor, FVII is activated to activated FVII (FVIIa) by different proteases, among which are thrombin (Factor Ha), activated Factor X and the FVIIa-tissue factor complex itself. The most important substrates for FVIIa-tissue factor are Factors X (FX) and IX (FIX).
Recombinant human FVIIa has been introduced for use in uncontrollable bleeding in hemophilia patients with Factor VIII (FVIII) or FIX deficiency, who have developed inhibitors against replacement coagulation factor. This factor is increasingly used in uncontrollable hemorrhage, as it induces coagulation only in those sites where tissue factor is present as well. In addition, according to Mayer et al. (2005), recombinant human FVII improves outcomes in acute intracerebral hemorrhage.
FVIII is another essential blood clotting factor. In fact, it is a cofactor for activated FIX which, in the presence of Ca+2 and phospholipids, forms a complex that converts FX to the activated form thereof. In human, FVIII is encoded by the F8 gene, and therefore defects in this gene result in hemophilia A, a common recessive X-linked coagulation disorder. The FVIII gene produces two alternatively spliced transcripts, wherein transcript variant 1 encodes a large glycoprotein, isoform a, which circulates in plasma, associates with von Willebrand factor in a noncovalent complex and undergoes multiple cleavage events, and transcript variant 2 encodes a putative small protein, isoform b, which consists primarily of the phospholipid binding domain of FVIIIc that is essential for coagulant activity.
US 7,199,223 discloses conjugates of a FVIII moiety and one or more water- soluble polymers, each having a molecular weight in the range of 6 to 150 kDa, preferably conjugates wherein each one of the polymers is a poly(alkylene oxide), more preferably a PEG, and the FVIII moiety is either recombinantly produced or blood-derived FVIII, FVIHa, FVIILC, FVIILvWF, and B-domain deleted FVIII. As described in this patent, these conjugates may be used for treating patients in need of FVIII therapy such as patients suffering from hemophilia A. US Publication Nos. 20080058504 and 20090041714, both continuation applications of US 7,199,223, discloses similar conjugates, wherein a water-soluble polymer is covalently attached to the FVIII moiety via either a degradable linkage such as a physiologically hydrolyzable or enzymatically degradable linkage, or a thiol group of a cysteine residue contained within said FVIII moiety.
As described in Example 11, the technology of the present invention may further be applied FVIIa and to FVIII, and the pharmacokinetic pattern of the specific conjugates formed by introducing these coagulation factors to SuO-FMS- MAL-S-(CH2)15-COOH, i.e., FVIIa-FMS-MAL-S-(CH2)15-COOH and FVIII-FMS- MAL-S-(CH2) I5-COOH can be studied both in vitro as well as in in vivo experimental systems. It is expected that both FVIII and FVIIa derivatized with excess of SuO-FMS-MAL-S-(CH2),5-COOH will have significantly reduced biological activities at time 0 but will undergo reactivation with a ti/2 value of several hours under physiological conditions.
Thus, in still further certain embodiments, the conjugate of the present invention is obtained by nucleophilic substitution of a compound of the formula I, preferably, any one of compounds 1 to 8, with any of the amino groups of FVIIa or FVIII. It should be noted that both FVIIa and FVIII, according to the present invention, might be either natural or recombinant.
In a further aspect, the present invention provides a pharmaceutical composition comprising a conjugate of the formula II as defined above, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In certain embodiments, the pharmaceutical compositions of the present invention comprise conjugates obtained by nucleophilic substitution of a compound of the formula I, preferably any one of compounds 1 to 8, with insulin, exendin-4, gentamicin or coagulation factors such as FVIIa and FVIII, or a pharmaceutically acceptable salts thereof.
The pharmaceutical compositions of the present invention may be prepared by conventional techniques, e.g. as described in Remington: The Science and Practice of Pharmacy, 19th Ed., 1995. The composition may be in solid, semisolid or liquid form and may further include pharmaceutically acceptable fillers, carriers or diluents, and other inert ingredients and excipients. Furthermore, the pharmaceutical composition can be designed for a slow release of the conjugate. The composition can be administered by any suitable route, e.g. intravenously, orally, parenterally, rectally, or transdermally. The dosage will depend on the state of the patient, and will be determined as deemed appropriate by the practitioner. The route of administration may be any route that effectively transports the active compound to the appropriate or desired site of action, the oral and the intravenous routes being preferred. If a solid carrier is used for oral administration, the preparation may be tabletted, placed in a hard gelatin capsule in powder or pellet form or it can be in the form of a lozenge. If a liquid carrier is used, the preparation may be in the form of a syrup, emulsion or soft gelatin capsule. Tablets, dragees or capsules having talc and/or a carbohydrate carrier or binder or the like are particularly suitable for oral application. Preferable carriers for tablets, dragees or capsules include lactose, cornstarch and/or potato starch. The pharmaceutical compositions of the present invention can be used for treatment of various diseases, disorders or conditions, in which administration of the drug designated Y in the formula II might be useful.
In particular, in still another aspect, the present invention provides a method for treatment of diabetes mellitus or hyperglycemia comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably, any one of compounds 1 to 8, with any of the amino groups of insulin. In yet another aspect, the present invention provides a method for treatment of insulin-dependent diabetes mellitus, non-insulin-dependent diabetes mellitus, or gestational diabetes mellitus, or for prevention of hyperglycemia, said method comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of exendin-4.
In still a further aspect, the present invention provides a method for treatment of a bacterial infection, preferably a bacterial infection caused by Gram-negative bacteria, comprising administering to an individual in need an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of gentamicin.
In yet a further aspect, the present invention provides a method for treating a patient in need of Factor Vila or Factor VIII therapy, comprising administering to said patient an effective amount of a conjugate of formula II, as defined above, obtained by nucleophilic substitution of a compound of formula I, as defined above, preferably any one of compounds 1 to 8, with any of the amino groups of FVIIa or FVIII, respectively. In certain embodiments, this method is used for treatment of patients who suffer from hemophilia A. In summary, the present invention provides a novel technology according to which any amino-containing short-lived drug can be converted, upon administration, into a long-lived prodrug, which gradually releases the pharmacologically active constituent under physiological conditions. This property is particularly achieved by introducing said short-lived drug with a LCFA like molecule capable of associating with HSA in vivo, which contains a spontaneously hydrolysable bond.
The strategy disclosed herein has not been materialized before because of two main barriers, i.e., (i) the difficulty to obtain an albumin-binding probe having sufficient associating affinity to albumin, as most small molecules that bind tightly to this carrier protein lose this capability when linked to a macromolecule such as drugs; and (ii) the lack of "reversibility principle" of the type developed in the laboratories of the inventors, which is a prerequisite condition in case the albumin associated conjugate formed in vivo has lost its biological/pharmacological potency upon conjugation. It should further be understood that this conjugate-reactivation becomes irrelevant, if does not take place in body fluids at a slow rate with a desirable pharmacokinetic pattern.
The invention will now be illustrated by the following non-limiting Examples.
EXAMPLES
Materials and Methods
(i) Materials. Human (Zn2+-free) insulin was donated by NovoNordisk (Bagsvalrd, Denemark) or by Biotechnology General (Rehovot, Israel). Insulin- detemir (Levemir®, NovoNordisk) was extensively dialyzed against 0.01 M NaHCO3 and stored at 7°C until used. The concentration of the insulin-detemir was determined by its absorbance at 280 run (ε280=5900) and/or by acid hydrolyzing an aliquot (in 6 M HCl, for 22 h at HO0C) followed by quantitative amino acid analysis. D-[U-14C] glucose (4-7 mci/mol) was obtained from Du Pont-NEN (Boston, Ma), type I collagenase (134 U/mg) was purchased from Worthington (Freehold, NY), gentamicin sulfate was purchased from Sigma Chemical Co. (Ness- Ziona, Israel) and polyethylene glycol 5 kDa-maleimide (PEG5-MAL) was obtained from Shearwater Group Inc. (Ra'anana, Israel). Exendin-4 (HGEGTFTSDLSKQM EEEAVRLFIEWLKNGGPSSGAPPPS-NH2) was synthesized by the solid phase method using the multiple peptide synthesizer AMS 422 (Abimed Analysen Technik, GmbH). 11-mercapto undecanoic acid, 16-mercaptohexadecanoic acid, decan-l,10-dithiol, taurine and trityl chloride were all purchased from Sigma- Aldrich Ltd. 6-amino-n-hexanoic acid purchased from BDH Ltd. All other materials used were of analytical grade. (H) MAL-FMS-OSu, i.e., 7-(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l-yl) propanamido)-9-(((2,5-dioxopyrrolidin- 1 -yloxy)carbonyloxy) methyl)-9H-fluorene- 2-sulfonic acid, was synthesized as described in Tsubery et al. (2004), starting from 9-hydroxymethyl-2-aminofluorene, and the final product was obtained in 65% yield following four steps of synthesis. (Hi) PEG5-NH2 is a 5 kDa polyethylene glycol (PEG5) moiety, in particular,
PEG5-CO-NH-(CH2)3-NH2, and it was prepared by dissolving PEG5-N- hydroxysuccinimide ester (PEG5-OSu, Shearwater product) at a concentration of 20 mg/ml in 0.1 M NaHCO3 containing 1 M of 1,3-diaminopropane dihydrochloride (Aldrich). The reaction was carried out for 1 hour at 250C, and the product was extensively dialyzed against H2O, lyophilized and kept at 7°C until used.
(iv) Isothermal scanning calorimetry measurements were performed with iTC200 microcalorimeter (MicroCal LLC, Nothampton MA, USA), according to the manufacturer's instructions.
(v) Iodination of peptides/proteins using [125I] iodine was performed using the chloramine T method (Hunter and Greenwood, 1962). Rat adipocytes were prepared from the fat pads of male Wistar rats (100-200 g) by collagenase digestion
(Rodbell, 1964). Lipogenesis, during which [U-14C] glucose was incorporated into the lipids, was carried out as described by Moody et al. (1974). (vi) Antibacterial potency of gentamicin and derivatives was determined by E. CoIi replication inhibition. As previously disclosed (Shechter et al., 2002; Marcus et al., 2008), native gentamicin inhibits replication of E. CoIi (IC50) at a concentration of 2.1±0.2 μM. (vii) Blood glucose levels were monitored at varying time points following administration of insulin, exendin-4 and derivatives thereof in blood aliquots taken from the tail vein, with a glucose analyzer (Beckman Instruments Fullerton, CA) by the glucose oxidase method. Groups consisted of five or six mice each. Data are presented as means±SE. (viii) Radioactive content in rat blood following intravenous administration of 125I-labeled peptides/proteins was monitored in blood samples taken at varying time points from the tail vein. Blood aliquots (50-70 mg) were absorbed to preweight Whatman 3MM filters that were reweight immediately after immersing blood samples.
Example 1. Applying isothermal scanning calorimetry for evaluating various ligands-human serum albumin associating affinities
Several versatile procedures were used in the last three decades in order to evaluate the associating affinities of various ligands to albumin (Peters et al, 1996). In this experiment we have applied isothermal scanning calorimetry (ITC), which is a modern approach for determining binding enthalpies, stochiometries and constants for ligand-protein interactions (Chaires, 2008); however, we first wished to confirm that the Ka values for ligand-human serum albumin (HSA) association, provided by the ITC approach, are close to those obtained using other binding strategies.
As stated above, insulin detemir (Levemir®, NovoNordisk) is a long-acting human insulin analog, in which the amino acid threonine in position B30 has been omitted and myristic acid has been attached to the amino acid lysine in position B29, i.e., N'B29-tetradecanoyl des(B30) insulin. In the blood, insulin-detemir binds to HSA through the acyl group at position B29 and it is then slowly dissociated from the complex. Figs. 1A-1B show the binding affinity of insulin-detemir to HSA, as determined by ITC-200. The primary ITC data was translated to a binding isotherm (Fig. IB), providing a binding constant of Ka=0.687±0.16xl05 M"1; however, a significantly higher value, i.e.,
Figure imgf000033_0001
was previously obtained for the binding affinity of insulin-detemir to HSA using the immobilized HSA binding strategy (Markusen et al, 1996). Whereas the immobilized HSA binding strategy assumes same binding constants to either immobilized or free albumin (Reed et al, 1975), it appears that ITC yields binding constants that are more accurate than those obtained by partitioning procedures between macromolecules and free ligands (Wiseman et al, 1989). With ITC measurements, ligands or macromolecules "signal" directly the accuracy of the binding process. Spectroscopic signaling can directly reflects associating events as well; however, this depends on the presence of chromophores or fluorophores that alter their properties in the binding state (Wiseman et al, 1989).
Example 2. Developing a reversible HSA binding probe
In order to design the HSA binding probe, we first searched for long-chain fatty acid (LCFA) like molecules containing sulfhydryl-moieties that can ultimately be linked to the maleimide (MAL) moiety of our hydrolyzable heterobifunctional spacer MAL-FMS-OSu. A required part of any such molecule is a terminal carboxylate (-CH2-COOH) that is essential for LCFAs' association with albumin (Peters, 1996). Once such compound is found, it should further be evaluated whether it preserves its HSA-associating affinity following its attachment to a macromolecule. As a model for a macromolecule we used PEG5-maleimide (PEG5- MAL), i.e., a 5 kDa polyethylene glycol chain that reacts with sulfhydryl containing molecule in a 1:1 stoichiometry. PEG5-NH2 was used as well for linking the appropriate HSA binding probe through our hydrolyzable heterobifunctional agent.
2.1 Preparation OfPEGs-MAL-S-(CH2)Ie-COOH
To a stirred solution of PEG5-MAL (50 mg in 2 ml H2O), 2.3 mg of 11- sulfanylundecanoic acid were added (0.1 ml from a fresh solution of HS-(CH2)10- COOH in dimethylformamide, DMF, 23 mg/ml). After 7 min, 10 mg of solid NaHCO3 were added. The reaction was carried out for, 1 h, and after centrifugation the supernatant was dialyzed overnight against H2O and lyophilized. The product thus obtained is 5,5'-bisdithio(2-nitrobenzoic acid) (DTNB; Ellman's reagent)- negative.
2.2 Preparation of monomodified FMS-MAL-S-(CH2) is-COOH containing derivatives of insulin and exendin-4.
SuO-FMS-MAL (58.3 mg, 100 μmol) and 16-sulfanylhexadecanoic acid (HS-(CH2), 5-COOH, 38.5 mg, 120 μmol) were dissolved in 1.0 ml DMF, followed by addition of pyridine (20 μl, 248 μmol). The reaction mixture was stirred for 40 min at 25°C, and product formation was monitored by the decrease in the maleimide moiety in aliquots withdrawn during synthesis. Following completion of the reaction (at 4 h) the derivative formed SuO-FMS-MAL-S-(CH2)15-COOH was added to an aqueous solution of insulin 6 mg/ml (1 μmol/ml) dissolved in 0.1 M NaHCO3 (pH 8.5) at three molar excess over the protein (30 μl). The reaction was carried out for 2 h at 00C, and the mixture was then dialyzed against H2O at 7°C. Monomodified derivative of insulin-linked to FMS-MAL-S-CH2)i5-COOH was purified from un-reacted insulin and from residual bismodified derivative, using semi-preparative HPLC (RP-4 column, Hesperia CA, 20-100% solution B (acetonitrile-H2O, 75:25 in 0.1% TFA) over 60 min with a flow rate of 10 ml/rnin). The fraction corresponding to monomodified insulin-FMS-MAL-S-(CH2)15-COOH was collected, redialyzed against H2O and lyophilized.
Monomodified derivative of exendin-4 (exendin-4-FMS-MAL-S-(CH2)15- COOH) was prepared under identical conditions, by adding 3 fold molar excess of the reagent to an aqueous solution of exendin-4 (4.2 mg/ml in 0.1 M NaHCO3, pH 8.5). The reaction was carried out for 3 h at 00C, and the reaction mixture was then dialyzed overnight, purified by preparative HPLC, redialyzed and lyophilized. 2.3 Discussion
As shown in Table 2 hereinafter, both 10-(2,5-dioxopyrrolidin-3- ylthio)decanoic acid and 16-(2,5-dioxopyrrolidin-3-ylthio)hexadecanoic acid, prepared by reacting MAL with 11-sulfanylundecanoic acid and 16- sulfanylhexadecanoic acid, and herein designated MAL-S-(CH2) IO-COOH and MAL-S-(CH2) 15-COOH, respectively, associated with HSA yielding Ka values of 1.3 to 1.6xlO5 M"1. However, while the MAL-S -(CH2) 10-COOH lost the capability to associate with HSA when linked to PEG5 (PEG5-MAL-S-(CH2)I0-COOH), the MAL-S-(CH2),5-COOH linked to PEG5, i.e., PEG5-MAL-S-(CH2)15-COOH, effectively associated with HSA yielding a Ka value of 1.95xlO5 M"1, as further shown in Fig. 2B, and thus was selected for further designing of the HSA- associating probe.
As further shown in Table 2, 2-(((2-(3-(2,5-dioxo-2,5-dihydro-lH-pyrrol-l- yl)propanamido)-7-sulfo-9H-fluoren-9-yl)methoxy)carbonylamino)acetic acid, herein designated glycine-FMS-MAL, associated with HSA with a Ka value of 1.4x105 M"1, but lost its associating affinity towards HSA when linked to a 5 kDa PEG5-NH2 chain. This association capacity was regained upon linking HS-(CH2)15- COOH to the MAL-moiety of the spacer, and as further shown, PEG5-NH-FMS- MAL-S-(CH2)15-COOH associated with HSA with a Ka value of 2.57xlO5 M"1. Similar HSA-associating affinities were obtained when 16-(l-(3-(9-(((2,5- dioxopyrrolidin-l-yloxy)carbonyloxy)methyl)-7-sulfo-9H-fluoren-2-ylamino)-3- oxopropyl)-2,5-dioxopyrrolidin-3-ylthio)hexadecanoic acid, herein designated SuO- FMS-MAL-S-(CH2)15-COOH was reacted with an amino side chain of either a small peptide, i.e., Gly-His-Lys, or of a larger polypeptide such as exendin-4 (4.2 kDa) or insulin (5.8 kDa), forming Gly-His-Lys-FMS-MAL-S-(CH2)15-COOH, exendin-4-FMS-MAL-S-(CH2)15-COOH or InSuHn-FMS-MAL-S-(CH2)! 5-COOH, respectively. It is thus concluded that the covalent introduction of FMS-MAL-S- (CH2)15-COOH, a specific HSA-binding probe, to peptides/proteins of a variable size, provided them with HSA associating affinity in the range of Ka=2.0-2.6xl05 M"1, exceeding about 3.5 times the associating affinity of insulin-detemir to HSA, as shown in Fig. IB. The structures of some of the compounds and conjugates listed in Table 2 are shown in Appendix 1.
Table 2. Binding affinities of various compounds/conjugates to HSA
Figure imgf000036_0001
Simulated binding isotherms for the association of all compounds and conjugates were carried out under the experimental conditions specified in Fig. 2. Each value is the arithmetic meaniSEM of 4-6 determinations.
Example 3. Synthesis of SuO-FMS-MAL-S-(CH2)15-CO-NH-(CH2)5-COOH, 2
3.1 Synthesis oftrityl-mercaptohexadecanoic acid
A solution of trityl (Tr) chloride (0.53 gr, 1.9 mmol) in dry dichloromethane (DCM, 5 ml) was added dropwise to a solution of 16-mercaptohexadecanoic acid
(0.5 gr, 1.7 mmol) and 2,4,6 trimethyl pyridine (452 μl) in dry DCM (5 ml). The mixture was stirred at room temperature under argon for 4 hr. The organic solution was washed with 5% HCl solution and with 2x brine solution, and was then dried with MgSO4 and evaporated. The reaction was followed by analytical HPLC and thin layer chromatography (TLC). The obtained product was purified by flash chromatography (hexane/ethyl acetate 9:1) receiving 480 mg of trityl- mercaptohexadecanoic acid (53% yield). 1H NMR (250MHz, CDCl3) δ: 1.26-1.43
(m, 24H) 1.63-1.68 (m, 2H), 2.15 (t, 2H, J=6 Hz), 2.37 (t, 2H, J=6.25Hz), 7.19-
7.32, 7.42-7.45 (m, 15H). Analyzed by mass spectroscopy. [M-H]+=530g/mol. 3.2 Synthesis of Tr-S-(CH2) ^CO-NH-(CH2) rCOOH
To a stirred solution of trityl-mercaptohexadecanoic acid (200 mg, 0.38 mmol) in dimethylformamide (DMF, 1 ml), N-hydroxysuccinimide (44 mg, 0.38 mmol) and dicyclohexylcarbodiimide (DCC) (376 μl) were added and mixed for 4 hr at room temperature. The reaction vial was centrifuged and the solution was added to 6-amino-n-hexanoic acid (54 mg, 0.4 mmol) in water (0.5 ml) and sodium bicarbonate (pH adjusted to ~7). The mixture was stirred over night at room temperature. Water were added and the precipitate was centrifuged. This operation was repeated 3 times. The product, Tr-S-(CH2)15-CO-NH-(CH2)5-COOH, was analyzed by analytical HPLC, mass spectroscopy [M-H]+=643 g/mol, and 1H NMR (250 MHz, CDCl3) δ 1.19-1.69 (m, 30H), 1.95 (m, 2H), 2.15 (m, 2H), 2.35 (m, 2H), 3.26 (m, 2H), 3.45 (m, 2H), 7.21-7.31, 7.41-7.435 (m, 15H).
3.3 Synthesis of HS-(CH2) 15-CO-NH-(CH2)5-COOH
A solution of Tr-S-(CH2)15-CO-NH-(CH2)5-COOH in dichloromethane/ trifluoroacetic acid/triethyl silane (93:2:5) (5 ml) was stirred for 30 min, and the reaction completion was tested according to the disappearance of the starting material. The solvent was evaporated, producing 140 mg crude HS-(CH2)15-CO- NH-(CH2)5-COOH (yield 91%), which was analyzed by mass spectroscopy [M- H]+=400 g/mol. 3.4 Synthesis of SUO-FMS-MAL-S-(CH2) I5-CO-NH-(CH2)S-COOH, 2
SuO-FMS-MAL (40 mg, 0.07 mmol) and HS-(CH2) 15-CO-NH-(CH2)5- COOH (85 mg, 0.2 mmol) were mixed in DMF (1 ml) and 2,4,6 trimethyl pyridine (40 μl) for 30 min. The desired product, compound 2, was purified by semi- preparative HPLC (C 18 column) and analyzed by mass spectrometry [M-H]+=983 g/mol. Example 4. Synthesis of SuO-FMS-MAL-S-(CH2)io-S-S-(CH2)io-COOH, 3
4.1 Synthesis
Figure imgf000038_0001
SuO-FMS-MAL (25 mg, 4.3xlO"5 mol) and decane-l,10-dithiol (19 μl, 8.8χlO~5 mol) were mixed in DMF (0.3 ml) and 2,4,6 trimethyl pyridine (pH adjusted to ~7) for 2 hr at room temperature. Purification was done by semi- preparative HPLC (C 18 column) receiving 6.3 mg of SuO-FMS-MAL-S-(CH2)i0- SH (18% yield), which was analyzed by mass spectrometry [M-H]+=788 g/mol.
4.2 Synthesis ofundecanoic acid dithio-pyridyl
To a stirred solution of 11-mercapto undecanoic acid (10 mg, 4.6xlO"5 mol) in DMF (0.5 ml), 4,4-dithiodipyridyl (50 mg, 0.23 mmol) and 2,4,6-trimethyl pyridine (pH adjusted to ~7) were added and mixed for 2 hr at room temperature.
The reaction was followed by analytical HPLC. Upon water addition, the product precipitated. After washing 3 times with water and centrifugation, 7.2 mg of undecanoic acid dithio-pyridyl were received (yield 48%), and was then analyzed by mass spectrometry [M-H]+=326 g/mol.
4.3 Synthesis of SUO-FMS-MAL-S-(CH2)] 0-S-S-(CH2)I0-COOH, 3
SuO-FMS-MAL-S-(CH2)10-SH (3 mg, 3.8xlO"6 mol) and undecanoic acid dithio-pyridyl (2 mg, 5.7xlO"6 mol) are mixed in DMF (0.2 ml) and 2,4,6 trimethyl pyridine (pH adjusted to ~7) for 2 hr at room temperature, and the desired product, compound 3, is purified using semi-preparative HPLC (C 18 column).
Alternatively, in order to obtain compound 3, one of the sulfanyl groups of 1,11-dithio undecanoic acid is first protected with trytyl, by reacting with trytyl chloride to obtain Tr-S-(CH2)10-SH, which is then reacted with dithiodipyridyl to obtain Tr-S-(CH2)10-S-S-pyridyl. Next, Tr-S-(CH2)i0-S-S-pyridyl is reacted with HS-(CH2)10-COOH to obtain Tr-S-(CH2)10-S-S-(CH2)10-COOH, which is then deprotected with DCM, 2% TFA and 5% trimethyl silane, for 30 min, to obtain H- S-(CH2)10-S-S-(CH2)10-COOH. Compound 3 is obtained by reacting obtain H-S- (CH2)10-S-S-(CH2)10-COOH with the maleimido of the SuO-FMS-MAL spacer. Example 5. Synthesis of SuO-FMS-MAL-S-(CH2)l5-CO-NH-(CH2)2-SO3H, 4
The synthesis of SuO-FMS-MAL-S-(CH2)15-CO-NH-(CH2)2-SO3H was similar to that of compound 2, described in Example 3, by first preparing Tr-S- (CH2) 15CO-NH-(CH2)2-SO3H, which was analyzed by mass spectrometry [M- H]+=637 g/mol and HPLC, and then reacting it with SuO-FMS-MAL.
5.1 Synthesis of Tr-S-(CH2) J5CO-NH-(CH2) ^SO3H
To a stirred solution of trityl-mercaptohexadecanoic acid (27 mg, 5X10"5 mol) in DMF (0.5 ml), N-hydroxysuccinimide (7 mg, 6x10 5 mol) and DCC (40 μl) were added and mixed for 4 hr at room temperature. The reaction vial was centrifuged and the solution was added to taurine (8 mg, 6x10~5 mol) in water (0.3 ml), resulting in pH of ~7. The mixture was stirred over night at room temperature. Water was added and the mixture lyophilized. The product, Tr-S-(CH2)15CO-NH- (CH2)2-SO3H, was analyzed by both analytical HPLC and mass spectroscopy [M- H]+=637 g/mol. 5.2 Synthesis of HS-(CH2) J5CO-NH-(CH2) rSOjH
The deprotection step of Tr-S-(CH2)15CO-NH-(CH2)2-SO3H was achieved as described above in Example 3 (section 3.3) for HS-(CH2)15-CO-NH-(CH2)5-COOH, resulting with the crude HS-(CH2)15CO-NH-(CH2)2-SO3H.
5.5 Synthesis of SUO-FMS-MAL-S-(CH2) J5-CO-NH-(CH2) ΓSOJH, 4 Compound 4 was obtained by reacting the crude HS-(CH2)15CO-NH-(CH2)2-
SO3H with SuO-FMS-MAL, as described above in Example 3 (section 3.4) for compound 2.
Example 6. Chemical and biological features of insulin-FMS-MAL-S-(CH2)is- COOH Insulin-FMS-MAL-S-(CH2)15-COOH is a monomodified derivative having molecular-weight of 6570 daltons (calculated value is 6565.5 daltons) as verified by mass spectroscopy. The derivative is soluble in phosphate buffer saline (PBS)- buffer at a concentration of ~4±1 mg/ml, and it has high absorbance at 280 nm, ε280=23,300±1000, amounting to 86% of the calculated value. Insulin-FMS-MAL- S-(CH2)i5-COOH emerged as a single symmetric peak on analytical high- performance liquid chromatography (HPLC)-column with retention time (ta) of 9.052 min, as shown in Fig. 3. Under the same experimental conditions, native insulin elutes with tR=7.28 min.
Table 3 hereinafter summarizes the characteristic features of HPLC-purifϊed insulin-FMS-(CH2)15-COOH. As shown, insulin-FMS-MAL-S-(CH2)15-COOH has about 10% the efficacy of insulin to activate lipogenesis in rat adipocytes yielding an half-maximal effect (ED50) at a concentration of 1.03±0.1 nM. It should be noted, however, that the biological potency of such albumin associated insulin derivative may be significantly reduced, due to the presence of bovine serum albumin (BSA) (10 mg/ml) in this particular assay, as previously noted with insulin- detemir (data not shown). As further shown in Table 3, insulin-FMS-MAL-S- (CH2)15-COOH has regained its full lipogenic potency (ED50=O. l±0.02 nM) following 4 hours of incubation at pH 10.3 and 25°C, i.e., under conditions that completely release insulin from the conjugate (Shechter et al., 2005b).
Table 3. Chemical and biological features of InSuInI-FMS-MAL-S-(CH2)! 5-COOH
Figure imgf000040_0001
(a) Mass spectra were determined using electrospray single quatropole mass spectroscopy (ESMS).
(b) Was calculated by combining the ε280 values for insulin (ε280=5800) and MAL-FMS-OSu (ε28o=21,200).
(c) Was determined by hydrolyzing an aliquot in 6 M HCl (110 0C, 22h) followed by quantitative amino-acid analysis.
(d) Analytical HPLC-analysis was carried out under the experimental conditions described in Materials and Methods. Under these conditions, insulin elutes with tR=7.28 min and has a surface area of 240,600±8,000 mav/μg of insulin. Example 7. Insulin-FMS-MAL-S-(CH2)i5-COOH has prolonged life time following intravenous administration to rats
In this experiment, two groups of rats (n=3 per group) were intravenously administered with either 125I-insulin or HPLC-purified 125I-insulin-FMS-MAL-S- (CH2)i5-COOH (8.4±0.4xl06 counts per minute (CPM) per 220±10 grams rat), and at indicated time points, blood aliquots (50-70 mg) were drawn and counted for their radioactive content in cpm/ml blood. As shown in Fig. 4, the circulating level of radioactive-labeled-insulin declined yielding a XXA value of 3.3±0.4 h; while the circulating level of radioactive-labeled-insulin-FMS- MAL-S-(CH2)i5-COOH increased over a period of two hours reaching a value of 31,000±l,000 cpm/ml blood, which was stably maintained over a period of 6 hours and than declined with a VA value of 17±1 hours. A significant amount, in particular, ~ 10,000 cpm/ml blood, was still evident 30 hours after intravenous administration.
Example 8. Glucose lowering pattern in mice following subcutaneous administration of insulin-FMS-MAL-S-(CH2)i5-COOH
In this experiment, the glucose-lowering pattern obtained after a single subcutaneous administration of insulin-FMS-MAL-S-(CH2)15-COOH was compared with that of Zn2+- free insulin, both administered at a low and similar dose (0.17 nmol/mouse in 0.2 ml PBS buffer). As shown in Fig. 5, insulin-FMS-MAL-S- (CH2) 15-COOH had a flat glucose-lowering pattern that was about two folds prolonged than that of insulin. Interestingly, the area under the curve of insulin- FMS-MAL-S-(CH2)15-COOH resembled that of the native hormone, although the former has, in vitro, only 10% the biological potency of insulin, as shown in Table 3 hereinabove. Thus, in terms of units/mg, insulin-FMS-MAL-S-(CH2)15-COOH equals native insulin.
It should be noted that this glucose lowering pattern was previously determined for new insulin preparations in rabbits (Kahn and Shechter, 1990); however, we do not recommend doing so in this species with an insulin derivative that associates with albumin, as rabbit albumin exhibits extraordinarily high affinity to fatty-acid acylated insulins (Kurtzhals et al, 1996). As shown in Figs. 1A-1B and Table 2, utilizing ITC measurements it was revealed that insulin-FMS-MAL-S-(CH2)15-COOH has a binding affinity to HSA that is 3.5 times higher than that of insulin-detemir, i.e., NeB29-tetradecanoyl des(B30) insulin. Fig. 6 shows the glucose lowering pattern of insulin-FMS-MAL- S-(CH2)15-COOH as compared to that of insulin-detemir, when subcutaneously administered at a dose of 0.68 nmol/mouse. At this dose, insulin-FMS-MAL-S- (CH2) I5-COOH was highly potent in reducing blood glucose level and it did so over prolong period with a X1A value of 6±1 hours. Low blood glucose level was still evident 24 hours following administration. The area under the curve could not, therefore, be accurately integrated; however, it exceeded five or more times that obtained by similar dose of subcutaneously administered insulin-detemir. As described in Shechter et al. (2003), subcutaneous administration of Zn2+-free insulin to mice at this dose is severely hypoglycemic.
Example 9. Exendin-4-FMS-MAL-S-(CH2)15-COOH facilitates prolonged glucose-lowering effect in CDl mice
As previously described (Tsubery et al., 2004; Shechter et al., 2003), the CDl strain of mice reflects well the action of exendin-4, a glucagon- like peptide- 1 agonist, in healthy and in type II diabetic patients, in the sense that at any dosage applied, circulating blood glucose level never falls below a threshold level which in CDl -mice amounts to a decrease of 27±3%.
Fig. 7 shows the glucose-lowering profile of native exendin-4 vs. exendin-4- FMS-MAL-S-(CH2)15-COOH, both subcutaneously administered at a dose of 0.24 nmol/CD 1 mouse. As particularly shown, following administration of exendin-4- FMS-MAL-S-(CH2)15-COOH, circulating glucose reached its lowest concentration 3 hours after administration and this level was preserved over a period of 20 hours. Returning to initial glucose level took place with a tVz value of 28±2 hours, which is 4.7 times longer than that obtained by the same dose of the native hormone. Example 10. Gentamicin-FMS-MAL-S-(CH2)15-COOH is an inactive- reactivable prodrug
While large protein drugs can accommodate one or two HSA-binding probe(s), each having a size of about 760 daltons, with the preservation of significant amount of their biological/pharmacological potencies (Shechter et al, 2001b; Shechter et al, 2007), low molecular- weight amino containing compounds suffer a massive loss of pharmacological potency upon conjugation. Under these circumstances, low molecular-weight compounds are thus impractical, unless may be reactivated upon administration. Fig. 8 shows time course of in vitro reactivation of gentamicin-FMS-MAL-
S-(CH2)15-COOH. In particular, gentamicin-FMS-MAL-S-(CH2)15-COOH (0.16 μmoles/ml) was incubated in PBS, pH 7.4, containing 2% (w/v) HSA at 37°C, and aliquots were withdrawn at certain time points and analyzed at several concentrations in the antibacterial assay. As particularly shown, at time 0, gentamicin-FMS-MAL-S-(CH2)15-COOH had ~3±0.7% the antibacterial potency of native gentamicin; however, upon incubation in PBS buffer (pH 7.4) containing 20 mg/ml HSA, the antibacterial potency of this conjugate was generated with a X1A value of 7.1±0.2 hours, regaining full (100%) antibacterial potency following 30 hours of incubation.
Example 11. Factor Vila and Factor VIII derivatized with OSu-FMS-MAL-S- (CH2)15-COOH, 1
In this experiment, the technology described in the Examples above is applied to recombinant Factor VIII (rFVIII), an essential protein for normal blood coagulation, and to recombinant Factor Vila (rFVIIa), a protease that participates in the clotting cascade as well. In particular, each of these two proteins is treated with 2 to 10 molar excess of SuO-FMS-MAL-S-(CH2)15-COOH, obtaining derivatives having low biological potencies of the native proteins, which are expected to regain 70-90% biological activity upon incubation at physiological conditions.
Derivatization of the proteins is . carried out in 0.1 M Hepes (pH 7.4) containing 1 mg/ml of rFVIII or rFVIIa. SuO-FMS-MAL-S-(CH2)15-COOH is added at 2 to 10 molar excess. Proteins are then examined for their biological potency prior to (time 0) and following their incubation at 370C in 0.1 M Hepes pH 7.4, containing 140 mM NaCl and 20 mg/ml BSA. Native rFVΗI and its derivative are diluted to a final concentration of 1 ng/ml, prior of being assayed, and their biological potencies are estimated with a Coatest-SP4 FVIII (Chromogenix) kit. Biological potencies of rFVIIa and its derivative are obtained by the clotting assay using "Activated Factor Vila, STACLOT® VIIa-Rtf ' (Agis, Bnei-Brak, Israel).
It is expected that both rFVIII and rFVIIa derivatized with excess of SuO- FMS-MAL-S-(CH2)]5-COOH will have a significantly reduced biological activity at time 0 but will undergo reactivation with a tl/2 value of several hours under physiological conditions. The pharmacokinetic patterns of these derivatives can be studied in in vivo experimental systems such as increased half-life after derivatives administration in FVIH-deficient mice or estimating FVIIa activity in ret serum following FVII derivative administration in rats, and it is expected that such derivatives will show considerable half-life prolongation, due to their capability to associate with serum albumin, to thereby substantially decrease their clearing rates in situ.
APPENDIX
Figure imgf000045_0001
Figure imgf000046_0001
REFERENCES
Bailon P., Palleroni A., Schaffer C.A., Spence C.L., Fung W.J., Porter J.E.,
Ehrlich G.K., Pan W., Xu Z.X., Modi M. W., Farid A., Graves m., Rational design of a potent, long-lasting form of interferon: a 40 kDa branched polyethylene glycol- conjugated interferon alpha-2a for the treatment of hepatitis C, Bioconjug Chem.,
2001, 12, 195-202
Carter D. C, Ho J.X., Structure of serum albumin. In advances in Protein Chemistry (Schumaker, V. N., Ed.), 1994, 153-203, Academic Press, Inc., San Diego, CA Chaires J.B., Calorimetry and Thermodynamics in Drug Design. Ann. Rev.
Biophys., 2008, 37, 135-151
Clark R., Olson K., Fuh G., Marian M., Mortensen D., Teshima G., Chang S., Chu H., Mukku V., Canova-Davis E., Somers T., Cronin M., Winkler M., Wells J. A., Long-acting growth hormones produced by conjugation with polyethylene glycol, J Biol Chem., 1996, 271, 21969-21977
Delgado C, Sancho P., Mendieta J., Luque J., Ligand-receptor interactions in affinity cell partitioning. Studies with transferrin covalently linked to monomethoxypoly(ethylene glycol) and rat reticulocytes, J Chromatogr., 1992, 594, 97-103 Goodman L., Gilman A.G., The Pharmacological Basis of Therapeutics,
McGraw-Hill, New York, 1995
Hunter W.M., Greenwood F. C, Preparation of iodine-131 labeled human growth hormone of high specific activity, Nature, 1962, 194, 495-496
Kahn C.R., Shechter Y., Insulin, oral hypoglycemic agents and pharmacology of the endocrine pancreas, in A. G. Gilman, T. W. Rail, A.S. Nies and P. Taylor (eds.), Goodman and Gilman Handbook of Pharmacology, New York/Oxford, Pergamon Press, 1990, 1463-1495
Kurtzhals P., Haveland S., Jonassen I., Kiehr B., Larsen U.D., Ribel V., Markussen J., Albumin binding of insulins acylated with fatty acids: Characterization of the ligand-protein interaction and correlation between binding affinity and timing of the insulin effect in vivo, BiochemicalJ., 1995, 312, 725-731
Kurtzhals P., Haveland S., Jonassen L, Kiehr B., Ribel U., Markussen J., Albumin binding and time action of acylated insulins in various species. J. Pharmaceut. ScL, 1996, 85, 304-308
Kurtzhals P., Haveland S., Jonassen LB. , Markussen J., Effect of fatty acids and selected drugs on the albumin binding of a long-acting, acylated insulin analogue, J. Pharmaceut. ScL, 1997, 86, 1365-1368
Marcus Y., Sasson K., Fridkin M., Shechter Y., Turning low-molecular- weight drugs into prolonged acting prodrugs by reversible pegylation: a study with gentamicin, J. Med. Chem., 2008, 51, 4300-4305
Markusen J., Havelund S., Kurtzhals P., Andersen A.S., Halstrøm E., Hasselager E., Larsen V.D., Ribbel U., Schafter L., Jonassen V.I., Soluble, fatty acid acylated insulins bind to albumin and show protracted action in pigs, Diabetologia, 1996, 39, 281-288
Mayer S., Brun N., Begtrup K., Broderick J., Davis S., Diringer M., Skolnick B., Steiner T., Recombinant activated factor VII for acute intracerebral hemorrhage, N. Engl. J. Med., 2005, 352, 777-785
Moody A.J., Stan M., Gliemann J., A simple free fat cell bioassay for insulin, Horm. Metab. Res., 1974, 6, 12-16
Νesher M., Vachutinsky Y., Fridkin G., Schwarz Y., Sasson K., Fridkin M., Shechter Y., Lichtstein D., Reversible pegylation prolongs the hypotensive effect of atrial natriuretic peptide. Biochonjugate Chem., 2008, 19, 342-348
Peleg-Shulman T., Tsubery H., Mironchik M., Fridkin M., Schreiber G., Shechter Y., reversible PEGylation: A novel technology to release native interferon alpha2 over a prolonged time period, J. Med. Chem., 2004, 47, 4897-4904
Peters T.J., The albumin molecule: Its structure and chemical properties. All about albumin, Biochemistry, Genetics and medical applications, 1996, 24-54, Academic Press, Inc., San Diego, CA Reddy K.R., Controlled-release, pegylation, liposomal formulations: new mechanisms in the delivery of injectable drugs, Ann Pharmacother., 2000, 34, 915- 923
Reed R.G., Gates T., Peters TJn., Albumin immobilizing on agarose as a tool for measuring ligand binding of proteins or peptides, Anal. Biochem., 1975, 69, 361-371
Rodbell M., Metabolism of isolated fat cells: effects of hormones on glucose metabolism and lipolysis, J. Biol. Chem., 1964, 239, 375-380
Shechter Y., Goldwaser L, Lavon I., Gershonov E., Mester B., Mironchik M., Patt L.P., Fridkin M., A new approach for prolonging the half-life of peptides, proteins and low-molecular-weigh drugs in vivo, Drugs of the Future, 2001a, 26, 669-676
Shechter Y., Patt L., Schreiber G., Fridkin M., Prolonging the half-life of human interferone-α2 In circulation: Design, preparation, and analysis of FMS7- interferone-α2. Proc. Natl. Acad. ScL USA, 2001b, 98, 1212-1217
Shechter Y., Tsubery H., Fridkin M., N-[(2-sulfo)-9-fluorenylmethoxy carbonyl)3-gentamicin Cl is a long-acting prodrug derivative, J. Med. Chem, 2002, 45, 4264-4270
Shechter Y., Tsubery H., Fridkin M., Suspensions of pro-drug insulin greatly prolong normoglycemic patterns in diabetic rats, Biochem Biophys Res Commun., 2003, 307, 315-321
Shechter Y., Tsubery H., Mironchik M., Rubinstein M., Fridkin M., Reversible PEGylation of peptide YY3-36 prolongs its inhibition of food intake in mice, FEBS Letts., 2005a, 579, 2439-2444 Shechter Y., Morinchik M., Rubinraut S., Saul A., Tsubery H., Fridkin M.,
Albumin- insulin conjugate releasing insulin slowly under physiological conditions: A new concept for long-acting insulin, Bioconjugate Chem., 2005b, 16, 913-920
Shechter Y., Mironchik M., Saul A., Gershonov E., Patt L.P., Sasson K., Tsubery H., Mester B., Kapitokovsky A., Rubinraut S., Vachutinski Y., Fridkin G., Fridkin M., New technologies to prolong life-time of peptide and protein drugs in vivo, Intl. J. Peptide Res. and Therapeutics, 2007, 13, 105-117
Shechter Y., Morinchik M., Rubinraut S., Tsubery H., Sasson K., Marcus Y., Fridkin M., Reversible pegylation of insulin facilitates its prolonged action in vivo, Eur. J. Pharm. Biopharm., 2008, In press
Taylor A., Granger D.N., Exchange of macromolecules across the microcirculation, In Handbook of Physiology. E.M. Renkin and CC. Michel, editors. American Physiological Society, Bethesda, 1984, 467-520
Tsubery H., Mironchik M., Fridkin M., Shechter Y., Prolonging the action of protein and peptide drugs by a novel approach of reversible polyethylene glycol modification, J. Biol. Chem., 2004, 279, 38118-38124
Wiseman T., Williston S., Brandts J.F., Nan Lin Lung, Rapid measurements of binding constants and heats of binding using a new titration calorimeter, Analytical Biochemistry, 1989, 179, 131-137

Claims

Claim 1. A compound of the formula I:
Figure imgf000051_0001
wherein
R1 is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO2NH-, -O-, -OCO-, -CO-NH-, -CS-NH-, -CO(CH2)M-, or -R8-CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N;
R2 is selected from >
Figure imgf000051_0002
Rg, or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO3H group;
R3 is absent or an acidic group having at least one hydroxyl group such as -COOH, -SO3H or -0-PO3H2;
R4 is an electron withdrawing group such as -SO3H, -CN, -CO-(C1- C8)alkyl, -CO-(C6-C,o)aryl, -NO2, -OPO3H2, -N(R)3 +, -SO2NH2, or halogen, wherein R is selected from (CrC8)alkyl or (Q-CuOa^CrC^alkyl;
R5 and R6, each independently is selected from hydrogen, -(CrC8)alkyl or -(C6-C10)aryl;
R7 is a leaving group such as -O-(CH2)2-CN, -Cl,
Figure imgf000051_0003
Figure imgf000052_0001
R9 is selected from (C13-C20)alkylene, (C13-C20)alkenylene or (C13-
C20)alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10aryl)-, or -(C6-Ci0)arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R2 is a peptide moiety, R3 is absent.
Claim 2. The compound of claim 1, wherein R4 is -SO3H at position 2 of the fluorene ring, R5 and R6 each is hydrogen, and R7 is N-hydroxysuccinimide (-OSu).
Claim 3. The compound of claim 2, wherein R1 is -NH-CO- or -NH- at position 7 of the fluorene ring, R2 is R9 or >
Figure imgf000052_0002
R9 is selected from (C13-C20)alkylene, (C13-C20)alkenylene or (C13-C20)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6- C10aryl)- or -(C6-C10)arylene-diyl-, and R3 is -COOH or -SO3H.
Claim 4. The compound of claim 3, wherein R1 is -NH-CO-, R2 is
Figure imgf000052_0003
and R9 is (C13-C2o)alkylene, (C13-C2O)alkenylene or (C13-C2O)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10aryl)- or -(C6- Cio)arylene-diyl-.
Claim 5. The compound of claim 4, wherein R9 is (C13-C2o)alkylene optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10ary I)- or -(C6-
C \ o)arylene-diy 1- .
, and:
Figure imgf000053_0001
(i) R3 is -COOH, and Rg is -(CH2)15- (herein identified compound 1), of the formula:
Figure imgf000053_0002
(ii) R3 is -COOH, and R9 is -(CH2)! 5-CO-NH-(CH2)5- group (herein identified compound 2), of the formula:
Figure imgf000053_0003
(iii) R3 is -COOH, and R9 is -(CH2)10-S-S-(CH2)10- group (herein identified compound 3), of the formula:
Figure imgf000053_0004
(iv) R3 is -SO3H, and R9 is -(CH2)15-CO-NH-(CH2)2- group (herein identified compound 4), of the formula:
Figure imgf000054_0001
Claim 7. The compound of claim 3, wherein Ri is -NH-CO-, R2 is R9, and R9 is (C13-C2o)alkylene, (C13-C20)alkenylene or (Ci3-C20)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrQalkyl)-, -N(C6-C iOaryl)- or -(C6- CiO)arylene-diyl-.
Claim 8. The compound of claim 7, wherein R9 is (C13-C2o)alkylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10ary I)- or -(C6- Cio)arylene-diyl-.
Claim 9. The compound of claim 8, wherein:
(i) R3 is -COOH, and R9 is -(CH2)] 5- (herein identified compound 5), of the formula:
Figure imgf000054_0002
(ii) R3 is -COOH, and R9 is -(CH2) 15-CO-NH-(CH2)5- group (herein identified compound 6), of the formula:
Figure imgf000054_0003
(iii) R3 is -COOH, and R9 is -(CH2)10-S-S-(CH2)10- group (herein identified compound 7), of the formula:
Figure imgf000055_0001
(iv) R3 is -SO3H, and R9 is -(CH2)15-CO-NH-(CH2)2- (herein identified compound 8), of the formula:
Figure imgf000055_0002
Claim 10. The compound of claim 2, wherein R1 is selected from -NH-CO-, - OCO-, or -R8-CO-, wherein R8 is (d-Cg)alkyl optionally interrupted by a heteroatom selected from O, S or N, and R2 is a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or -SO3H group such as taurine.
Claim 11. A conjugate of the formula II:
Figure imgf000055_0003
wherein
Y is a moiety of a drug containing at least one amino group, linked through said at least one amino group; R1 is selected from -NH-, -NH-CO-, -NH-CO-NH-, -S-, -SO2NH-, -O-, -OCO-, -CO-NH-, -CS-NH-, -CO(CH2)M-, or -R8-CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N;
R2 is selected from ,
Figure imgf000056_0001
R9, or a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or - SO3H group;
R3 is absent or an acidic group having at least one hydroxyl group such as -COOH, -SO3H or -0-PO3H2;
R4 is an electron withdrawing group such as -SO3H, -CN, -CO-(C1- QOalkyl, -CO-(C6-C 10)aryl, -NO2, -OPO3H2, -N(R)3 +, -SO2NH2, or halogen, wherein R is selected from (C]-C8)alkyl or (Cό-C^a^Q-C^alkyl;
R5 and R6, each independently is selected from hydrogen, -(CrC8)alkyl or -(C6-C10)aryl; and
R9 is selected from (C13-C20)alkylene, (C13-C2o)alkenylene or (C]3- C2o)alkynylene, optionally interrupted by one or more identical or different heteroatoms selected from S, O or N, and/or at least one group selected from -NH- CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C10aryl)- or -(C6-Ci0)arylene-diyl-, wherein said alkenylene or alkynylene comprises one or more double or triple bond, respectively, and said one or more double or triple bond is not a terminal double or triple bond, provided that when R2 is a peptide moiety, R3 is absent.
Claim 12. The conjugate of claim 11, wherein R4 is -SO3H at position 2 of the fluorene ring, and R5 and R6 each is hydrogen.
Claim 13. The conjugate of claim 12, wherein R1 is -NH-CO- or -NH- at position
7 of the fluorene ring, R2 is R9 or »
Figure imgf000057_0001
R9 is selected from (C13-C20)alkylene, (C13-C2o)alkenylene or (Ci3-C2o)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6- C10aryl)- or -(C6-C10)arylene-diyl-, and R3 is -COOH or -SO3H.
Claim 14. The conjugate of claim 13, wherein R1 is -NH-CO-, R2 is
Figure imgf000057_0002
and R9 is (C13-C2o)alkylene, (C13-C20)alkenylene or (C13-C20)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10aryl)- or -(C6- Ct0)arylene-diyl-.
Claim 15. The conjugate of claim 14, wherein R9 is (C13-C2o)alkylene optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C 10ary I)- or -(C6- C10)arylene-diyl-.
Claim 16. The conjugate of claim 15, wherein R2 is
Figure imgf000057_0003
(i) R3 is -COOH, and R9 is -(CH2)15-, of the formula:
;
Figure imgf000057_0004
(ii) R3 is -COOH, and R9 is -(CH2) 15-CO-NH-(CH2)5- group, of the formula:
Figure imgf000058_0001
(iii) R3 is -COOH, and R9 is -(CH2)10-S-S-(CH2)i0- group, of the formula:
Figure imgf000058_0002
(iv) R3 is -SO3H, and R9 is -(CH2)15-CO-NH-(CH2)2-, of the formula:
Figure imgf000058_0003
Claim 17. The conjugate of claim 13, wherein R1 is -NH-CO-, R2 is R9, and R9 is (Ci3-C20)alkylene, (C13-C20)alkenylene or (Ci3-C2o)alkynylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(C,-C8alkyl)-, -N(C6-C iOary I)- or -(C6- Cio)arylene-diyl-. /
Claim 18. The conjugate of claim 17, wherein R9 is (Ci3-C20)alkylene, optionally interrupted by one or more heteroatoms selected from S, O or N, and/or at least one group selected from -NH-CO-, -CO-NH-, -N(CrC8alkyl)-, -N(C6-C ,oary I)- or -(C6- Cio)arylene-diyl-.
Claim 19. The conjugate of claim 18, wherein:
(i) R3 is -COOH, and R9 is -(CH2)15-, of the formula:
Figure imgf000059_0001
(ii) R3 is -COOH, and R9 is -(CH2)15-CO-NH-(CH2)5- group, of the formula:
Figure imgf000059_0002
(iii) R3 is -COOH, and R9 is -(CH2)10-S-S-(CH2)10- group, of the formula:
Figure imgf000059_0003
(iv) R3 is -SO3H, and R9 is -(CH2) 15-CO-NH-(CH2)2-, of the formula:
Figure imgf000059_0004
Claim 20. The conjugate of claim 12, wherein R1 is selected from -NH-CO-, - OCO-, or -R8-CO-, wherein R8 is (CrC8)alkyl optionally interrupted by a heteroatom selected from O, S or N, and R2 is a peptide moiety consisting of 3 to 5 amino acid residues each independently is an aliphatic hydrophobic amino acid residue such as Leu, He or VaI, an aromatic amino acid residue such as Phe, or an amino acid analog comprising -COOH or -SO3H group such as taurine.
Claim 21. The conjugate of any one of claims 11 to 20, wherein Y is an antibiotic aminoglycoside such as gentamicin or amphotericin, an antineoplastic drug such as aminolevulinic acid, or an anthracycline chemotherapeutic agent such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone and valrubicin.
Claim 22. The conjugate of any one of claims 11 to 20, wherein Y is a peptide or a protein drug of low or medium molecular weight such as insulin, an interferon, preferably IFN-α2, a PYY agonist, preferably the peptide PYY3-36, an exendin, preferably exendin-3 or exendin-4, an exendin analogue or exendin agonist, atrial natriuretic peptide (ANP), human growth hormone (hGH), erythropoietin, TNF-α, calcitonin, gonadotropin releasing hormone (GnRH), a GnRH analogue, hirudin, glucagon, a coagulation factor such as Factor Vila and Factor VIII, and a monoclonal antibody fragment, preferably anti-TNF-α monoclonal antibody fragment.
Claim 23. The conjugate of claim 16 or 19, wherein Y is insulin, exendin-4, gentamicin, Factor Vila or Factor VIII.
Claim 24. A pharmaceutical composition comprising a conjugate according to any one of claims 11 to 23, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
Claim 25. The pharmaceutical composition of claim 24, comprising a conjugate of claim 23.
Claim 26. A method for the treatment of diabetes mellitus or hyperglycemia comprising administering to an individual in need an effective amount of a conjugate of formula II according to any one of claims 11 to 23, preferably according to claim 16 or 19, wherein Y is insulin.
Claim 27. A method for treatment of insulin-dependent diabetes mellitus, non- insulin-dependent diabetes mellitus, or gestational diabetes mellitus, or for prevention of hyperglycemia, said method comprising administering to an individual in need an effective amount of a conjugate of formula II according to any one of claims 11 to 23, preferably according to claim 16. or 19, wherein Y is exendin-4.
Claim 28. A method for treatment of a bacterial infection comprising administering to an individual in need an effective amount of a conjugate of formula II according to any one of claims 11 to 23, preferably according to claim 16 or 19, wherein Y is gentamicin.
Claim 29. The method of claim 28, wherein said bacterial infection is caused by gram-negative bacteria.
Claim 30. A method for treating a patient in need of Factor Vila or Factor VIII therapy, comprising administering to said patient an effective amount of a conjugate of formula II according to any one of claims 11 to 23, preferably according to claim 16 or 19, wherein Y is Factor Vila or Factor VIII, respectively.
Claim 31. The method of claim 30, wherein said patient is suffering from hemophilia A.
PCT/IL2010/000433 2009-06-01 2010-06-01 Prodrugs containing albumin binding probe WO2010140148A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP10726604.1A EP2437786B1 (en) 2009-06-01 2010-06-01 Prodrugs containing albumin binding probe
US13/375,637 US20120134977A1 (en) 2009-06-01 2010-06-01 Prodrugs containing albumin binding probe
IL216726A IL216726A0 (en) 2009-06-01 2011-12-01 Prodrugs containing albumin binding probe
US14/564,469 US20150093370A1 (en) 2009-06-01 2014-12-09 Prodrugs containing albumin binding probe

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18291009P 2009-06-01 2009-06-01
US61/182,910 2009-06-01

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/375,637 A-371-Of-International US20120134977A1 (en) 2009-06-01 2010-06-01 Prodrugs containing albumin binding probe
US14/564,469 Division US20150093370A1 (en) 2009-06-01 2014-12-09 Prodrugs containing albumin binding probe

Publications (1)

Publication Number Publication Date
WO2010140148A1 true WO2010140148A1 (en) 2010-12-09

Family

ID=42644140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2010/000433 WO2010140148A1 (en) 2009-06-01 2010-06-01 Prodrugs containing albumin binding probe

Country Status (3)

Country Link
US (2) US20120134977A1 (en)
EP (1) EP2437786B1 (en)
WO (1) WO2010140148A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011013128A3 (en) * 2009-07-31 2011-03-24 Yeda Research And Development Co. Ltd. Vectors for delivery of neurotherapeutics to the central nervous system
WO2012140647A2 (en) 2011-04-11 2012-10-18 Yeda Research And Development Co. Ltd Albumin binding probes and drug conjugates thereof
WO2013185114A2 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
WO2013185113A1 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Procoagulant compounds
WO2014127215A1 (en) 2013-02-15 2014-08-21 Biogen Idec Ma Inc. Optimized factor viii gene
WO2014210029A1 (en) * 2013-06-24 2014-12-31 Riogin Corporation Double binding constructs
US8945897B2 (en) 2010-07-26 2015-02-03 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
WO2016061286A2 (en) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
WO2016065301A1 (en) 2014-10-23 2016-04-28 Biogen Ma Inc. Anti-gpiib/iiia antibodies and uses thereof
WO2016070050A1 (en) 2014-10-31 2016-05-06 Biogen Ma Inc. ANTI-GLYCOPROTEIN IIb/IIIa ANTIBODIES
WO2017136358A1 (en) 2016-02-01 2017-08-10 Bioverativ Therapeutics Inc. Optimized factor viii genes
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
WO2019152692A1 (en) 2018-02-01 2019-08-08 Bioverativ Therapeutics, Inc. Use of lentiviral vectors expressing factor viii
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
WO2021067389A1 (en) 2019-09-30 2021-04-08 Bioverativ Therapeutics Inc. Lentiviral vector formulations
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
WO2023012305A1 (en) * 2021-08-04 2023-02-09 Cytacoat Ab Disulfide compounds as medicaments

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US5750497A (en) 1993-09-17 1998-05-12 Novo Nordisk A/S Acylated insulin
US6011007A (en) 1993-09-17 2000-01-04 Novo Nordisk A/S Acylated insulin
US20040110664A1 (en) 1993-09-17 2004-06-10 Svend Havelund Acylated insulin
WO2004089280A2 (en) * 2003-04-08 2004-10-21 Yeda Research And Development Co. Ltd. Reversible pegylated drugs
WO2005117984A2 (en) * 2004-06-01 2005-12-15 Celltech R & D Limited Albumin-binding conjugates comprising a fatty acid and peg
WO2006105201A2 (en) * 2005-03-30 2006-10-05 Trimeris, Inc. Conjugates comprised of fatty acid and hiv gp41-derived peptide
US7186797B2 (en) 2001-08-10 2007-03-06 Epix Pharmaceuticals, Inc. Polypeptide conjugates with extended circulating half-lives
US7199223B2 (en) 2003-02-26 2007-04-03 Nektar Therapeutics Al, Corporation Polymer-factor VIII moiety conjugates
WO2008053360A2 (en) 2006-11-03 2008-05-08 Philochem Ag Albumin binding molecules and uses thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4356165A (en) * 1980-04-14 1982-10-26 Burroughs Wellcome Co. Bupropion radioimmunoassay, and kit
US4481298A (en) * 1981-04-13 1984-11-06 Amf Incorporated Pre-precipitated double antibody immunoassay method
US4557931A (en) * 1982-12-02 1985-12-10 Regents Of The University Of California Antigenic compositions and methods for using same
US4585740A (en) * 1983-05-03 1986-04-29 The Whittier Institute For Diabetes And Endocrinology Prolactin immunoassay using synthetic peptide
US5302681A (en) * 1992-12-10 1994-04-12 Nalco Chemical Company Polymerization inhibition by fullerenes
WO2011013128A2 (en) * 2009-07-31 2011-02-03 Yeda Research And Development Co. Ltd. Vectors for delivery of neurotherapeutics to the central nervous system

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5424286A (en) 1993-05-24 1995-06-13 Eng; John Exendin-3 and exendin-4 polypeptides, and pharmaceutical compositions comprising same
US20060030518A1 (en) 1993-09-17 2006-02-09 Svend Havelund Acylated insulin
US5750497A (en) 1993-09-17 1998-05-12 Novo Nordisk A/S Acylated insulin
US6011007A (en) 1993-09-17 2000-01-04 Novo Nordisk A/S Acylated insulin
US20040110664A1 (en) 1993-09-17 2004-06-10 Svend Havelund Acylated insulin
US6869930B1 (en) 1993-09-17 2005-03-22 Novo Nordisk A/S Acylated insulin
US7186797B2 (en) 2001-08-10 2007-03-06 Epix Pharmaceuticals, Inc. Polypeptide conjugates with extended circulating half-lives
US7199223B2 (en) 2003-02-26 2007-04-03 Nektar Therapeutics Al, Corporation Polymer-factor VIII moiety conjugates
US20080058504A1 (en) 2003-02-26 2008-03-06 Bossard Mary J Polymer-factor VIII moiety conjugates
US20090041714A1 (en) 2003-02-26 2009-02-12 Nektar Therapeutics Al, Corporation Polymer-factor VIII moiety conjugates
WO2004089280A2 (en) * 2003-04-08 2004-10-21 Yeda Research And Development Co. Ltd. Reversible pegylated drugs
US7585837B2 (en) 2003-04-08 2009-09-08 Yeda Research And Development Co. Ltd. Reversible pegylated drugs
WO2005117984A2 (en) * 2004-06-01 2005-12-15 Celltech R & D Limited Albumin-binding conjugates comprising a fatty acid and peg
WO2006105201A2 (en) * 2005-03-30 2006-10-05 Trimeris, Inc. Conjugates comprised of fatty acid and hiv gp41-derived peptide
WO2008053360A2 (en) 2006-11-03 2008-05-08 Philochem Ag Albumin binding molecules and uses thereof

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
BAILON P.; PALLERONI A.; SCHAFFER C.A.; SPENCE C.L.; FUNG W.J.; PORTER J.E.; EHRLICH G.K.; PAN W.; XU Z.X.; MODI M.W.: "Rational design of a potent, long-lasting form of interferon: a 40 kDa branched polyethylene glycol- conjugated interferon alpha-2a for the treatment of hepatitis C", BIOCONJUG CHEM., vol. 12, 2001, pages 195 - 202
CHAIRES J.B.: "Calorimetry and Thermodynamics in Drug Design", ANN. REV. BIOPHYS., vol. 37, 2008, pages 135 - 151
CLARK R.; OLSON K.; FUH G.; MARIAN M.; MORTENSEN D.; TESHIMA G.; CHANG S.; CHU H.; MUKKU V.; CANOVA-DAVIS E.: "Long-acting growth hormones produced by conjugation with polyethylene glycol", J BIOL CHEM., vol. 271, 1996, pages 21969 - 21977
DELGADO C.; SANCHO P.; MENDIETA J.; LUQUE J.: "Ligand-receptor interactions in affinity cell partitioning. Studies with transferrin covalently linked to monomethoxypoly(ethylene glycol) and rat reticulocytes", J CHROMATOGR., vol. 594, 1992, pages 97 - 103
HUNTER W.M.; GREENWOOD F.C.: "Preparation of iodine-131 labeled human growth hormone of high specific activity", NATURE, vol. 194, 1962, pages 495 - 496
KURTZHALS P.; HAVELAND S.; JONASSEN I.; KIEHR B.; LARSEN U.D.; RIBEL V.; MARKUSSEN J.: "Albumin binding of insulins acylated with fatty acids: Characterization of the ligand-protein interaction and correlation between binding affinity and timing of the insulin effect in vivo", BIOCHEMICAL J., vol. 312, 1995, pages 725 - 731
KURTZHALS P.; HAVELAND S.; JONASSEN I.; KIEHR B.; RIBEL U.; MARKUSSEN J.: "Albumin binding and time action of acylated insulins in various species", J PHARMACEUT. SCI., vol. 85, 1996, pages 304 - 308
KURTZHALS P.; HAVELAND S.; JONASSEN I.B.; MARKUSSEN J.: "Effect of fatty acids and selected drugs on the albumin binding of a long-acting, acylated insulin analogue", J PHARMACEUT. SCI., vol. 86, 1997, pages 1365 - 1368
MARCUS Y.; SASSON K.; FRIDKIN M.; SHECHTER Y.: "Turning low-molecular-weight drugs into prolonged acting prodrugs by reversible pegylation: a study with gentamicin", J MED. CHEM., vol. 51, 2008, pages 4300 - 4305
MARKUSEN J.; HAVELUND S.; KURTZHALS P.; ANDERSEN A.S.; HALSTROM E.; HASSELAGER E.; LARSEN V.D.; RIBBEL U.; SCHAFTER L.; JONASSEN V: "Soluble, fatty acid acylated insulins bind to albumin and show protracted action in pigs", DIABETOLOGIA, vol. 39, 1996, pages 281 - 288
MAYER S.; BRUN N.; BEGTRUP K.; BRODERICK J.; DAVIS S.; DIRINGER M.; SKOLNICK B.; STEINER T.: "Recombinant activated factor VII for acute intracerebral hemorrhage", N. ENGL. J MED., vol. 352, 2005, pages 777 - 785
MOODY A.J.; STAN M.; GLIEMANN J.: "A simple free fat cell bioassay for insulin", HORM. METAB. RES., vol. 6, 1974, pages 12 - 16
NESHER M.; VACHUTINSKY Y.; FRIDKIN G.; SCHWARZ Y.; SASSON K.; FRIDKIN M.; SHECHTER Y.; LICHTSTEIN D.: "Reversible pegylation prolongs the hypotensive effect of atrial natriuretic peptide", BIOCHONJUGATE CHEM., vol. 19, 2008, pages 342 - 348
PELEG-SHULMAN T.; TSUBERY H.; MIRONCHIK M.; FRIDKIN M.; SCHREIBER G.; SHECHTER Y.: "reversible PEGylation: A novel technology to release native interferon alpha2 over a prolonged time period", J MED. CHEM., vol. 47, 2004, pages 4897 - 4904
REDDY K.R.: "Controlled-release, pegylation, liposomal formulations: new mechanisms in the delivery of injectable drugs", ANN PHARMACOTHER., vol. 34, 2000, pages 915 - 923
REED R.G.; GATES T.; PETERS T.JN.: "Albumin immobilizing on agarose as a tool for measuring ligand binding of proteins or peptides", ANAL. BIOCHEM., vol. 69, 1975, pages 361 - 371
RODBELL M.: "Metabolism of isolated fat cells: effects of hormones on glucose metabolism and lipolysis", J BIOL. CHEM., vol. 239, 1964, pages 375 - 380
SASSON K ET AL: "Engineering prolonged-acting prodrugs employing an albumin-binding probe that undergoes slow hydrolysis at physiological conditions", JOURNAL OF CONTROLLED RELEASE, ELSEVIER, AMSTERDAM, NL LNKD- DOI:10.1016/J.JCONREL.2009.10.028, vol. 142, no. 2, 3 March 2010 (2010-03-03), pages 214 - 220, XP026905283, ISSN: 0168-3659, [retrieved on 20091030] *
SHECHTER Y.; GOLDWASER I.; LAVON I.; GERSHONOV E.; MESTER B.; MIRONCHIK M.; PATT L.P.; FRIDKIN M.: "A new approach for prolonging the half-life of peptides, proteins and low-molecular-weigh drugs in vivo", DRUGS OF THE FUTURE, vol. 26, 2001, pages 669 - 676
SHECHTER Y.; MIRONCHIK M.; SAUL A.; GERSHONOV E.; PATT L.P.; SASSON K.; TSUBERY H.; MESTER B.; KAPITOKOVSKY A.; RUBINRAUT S.: "New technologies to prolong life-time of peptide and protein drugs in vivo", INTL. J PEPTIDE RES. AND THERAPEUTICS, vol. 13, 2007, pages 105 - 117
SHECHTER Y.; MORINCHIK M.; RUBINRAUT S.; SAUL A.; TSUBERY H.; FRIDKIN M.: "Albumin-insulin conjugate releasing insulin slowly under physiological conditions: A new concept for long-acting insulin", BIOCONJUGATE CHEM., vol. 16, 2005, pages 913 - 920
SHECHTER Y.; MORINCHIK M.; RUBINRAUT S.; TSUBERY H.; SASSON K.; MARCUS Y.; FRIDKIN M.: "Reversible pegylation of insulin facilitates its prolonged action in vivo", EUR. J PHARM. BIOPHARM., 2008
SHECHTER Y.; PATT L.; SCHREIBER G.; FRIDKIN M.: "Prolonging the half-life of human interferone-a2 In circulation: Design, preparation, and analysis of FMS7- interferone-a2", PROC. NATL. ACAD. SCI. USA, vol. 98, 2001, pages 1212 - 1217
SHECHTER Y.; TSUBERY H.; FRIDKIN M.: "N-[(2-sulfo)-9-fluorenylmethoxy carbonyl)3-gentamicin C1 is a long-acting prodrug derivative", J MED. CHEM, vol. 45, 2002, pages 4264 - 4270
SHECHTER Y.; TSUBERY H.; FRIDKIN M.: "Suspensions of pro-drug insulin greatly prolong normoglycemic patterns in diabetic rats", BIOCHEM BIOPHYS RES COMMUN., vol. 307, 2003, pages 315 - 321
SHECHTER Y.; TSUBERY H.; MIRONCHIK M.; RUBINSTEIN M.; FRIDKIN M.: "Reversible PEGylation of peptide YY3-36 prolongs its inhibition of food intake in mice", FEBS LETTS., vol. 579, 2005, pages 2439 - 2444
TSUBERY H.; MIRONCHIK M.; FRIDKIN M.; SHECHTER Y.: "Prolonging the action of protein and peptide drugs by a novel approach of reversible polyethylene glycol modification", J BIOL. CHEM., vol. 279, 2004, pages 38118 - 38124
WISEMAN T.; WILLISTON S.; BRANDTS J.F.; NAN LIN LUNG: "Rapid measurements of binding constants and heats of binding using a new titration calorimeter", ANALYTICAL BIOCHEMISTRY, vol. 179, 1989, pages 131 - 137

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011013128A3 (en) * 2009-07-31 2011-03-24 Yeda Research And Development Co. Ltd. Vectors for delivery of neurotherapeutics to the central nervous system
US8945897B2 (en) 2010-07-26 2015-02-03 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
WO2012140647A2 (en) 2011-04-11 2012-10-18 Yeda Research And Development Co. Ltd Albumin binding probes and drug conjugates thereof
WO2012140647A3 (en) * 2011-04-11 2013-03-28 Yeda Research And Development Co. Ltd Albumin binding probes and drug conjugates thereof
US9480751B2 (en) 2011-04-11 2016-11-01 Yeda Research And Development Co. Ltd. Albumin binding probes and drug conjugates thereof
US11168316B2 (en) 2012-06-08 2021-11-09 Bioverativ Therapeutics, Inc. Chimeric clotting factors
US10202595B2 (en) 2012-06-08 2019-02-12 Bioverativ Therapeutics Inc. Chimeric clotting factors
WO2013185114A2 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Chimeric clotting factors
EP4079316A1 (en) 2012-06-08 2022-10-26 Bioverativ Therapeutics Inc. Procoagulant compounds
EP3693000A1 (en) 2012-06-08 2020-08-12 Bioverativ Therapeutics Inc. Procoagulant compounds
US11261437B2 (en) 2012-06-08 2022-03-01 Bioverativ Therapeutics Inc. Procoagulant compounds
WO2013185113A1 (en) 2012-06-08 2013-12-12 Biogen Idec Ma Inc. Procoagulant compounds
US10287564B2 (en) 2012-06-08 2019-05-14 Bioverativ Therapeutics Inc. Procoagulant compounds
EP4223772A2 (en) 2013-02-15 2023-08-09 Bioverativ Therapeutics Inc. Optimized factor viii gene
US11787851B2 (en) 2013-02-15 2023-10-17 Bioverativ Therapeutics Inc. Optimized factor VIII gene
US10370431B2 (en) 2013-02-15 2019-08-06 Bioverativ Therapeutics Inc. Optimized factor VIII gene
EP3889173A1 (en) 2013-02-15 2021-10-06 Bioverativ Therapeutics Inc. Optimized factor viii gene
WO2014127215A1 (en) 2013-02-15 2014-08-21 Biogen Idec Ma Inc. Optimized factor viii gene
WO2014210029A1 (en) * 2013-06-24 2014-12-31 Riogin Corporation Double binding constructs
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
US9969998B2 (en) 2014-10-14 2018-05-15 Halozyme, Inc. Compositions of adenosine deaminase-2 (ADA2), variants thereof and methods of using same
US11584923B2 (en) 2014-10-14 2023-02-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ADA2), variants thereof and methods of using same
WO2016061286A2 (en) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
WO2016065301A1 (en) 2014-10-23 2016-04-28 Biogen Ma Inc. Anti-gpiib/iiia antibodies and uses thereof
WO2016070050A1 (en) 2014-10-31 2016-05-06 Biogen Ma Inc. ANTI-GLYCOPROTEIN IIb/IIIa ANTIBODIES
WO2017136358A1 (en) 2016-02-01 2017-08-10 Bioverativ Therapeutics Inc. Optimized factor viii genes
EP4137570A1 (en) 2016-02-01 2023-02-22 Bioverativ Therapeutics Inc. Optimized factor viii genes
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
WO2019152692A1 (en) 2018-02-01 2019-08-08 Bioverativ Therapeutics, Inc. Use of lentiviral vectors expressing factor viii
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
WO2021067389A1 (en) 2019-09-30 2021-04-08 Bioverativ Therapeutics Inc. Lentiviral vector formulations
WO2023012305A1 (en) * 2021-08-04 2023-02-09 Cytacoat Ab Disulfide compounds as medicaments

Also Published As

Publication number Publication date
US20150093370A1 (en) 2015-04-02
US20120134977A1 (en) 2012-05-31
EP2437786A1 (en) 2012-04-11
EP2437786B1 (en) 2016-05-18

Similar Documents

Publication Publication Date Title
EP2437786B1 (en) Prodrugs containing albumin binding probe
US9480751B2 (en) Albumin binding probes and drug conjugates thereof
US9956295B2 (en) Reversible pegylated drugs
ES2328579T3 (en) LONG-TERM INSULIN DERIVATIVES AND ASSOCIATED PROCEDURES.
AU2004220727B2 (en) Modified exendins and exendin agonists
US8415296B2 (en) Oxyntomodulin peptide analogue
US9861706B2 (en) GLP-1 receptor agonist peptide gastrin conjugates
KR20140070612A (en) Novel glucagon analogues
IL172980A (en) Insulin derivatives, pharmaceutical compositions comprising them and uses thereof
US20080318845A1 (en) Selective Vpac2 Receptor Peptide Agonists
KR20210126088A (en) long-acting GLP-2 analogues
Shechter et al. Reversible pegylation of insulin facilitates its prolonged action in vivo
US20040092443A1 (en) Long-acting exendins and exendin agonists
Shechter et al. Newly designed modifier prolongs the action of short-lived peptides and proteins by allowing their binding to serum albumin

Legal Events

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

Ref document number: 10726604

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010726604

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13375637

Country of ref document: US