EP1472277A1 - Fap-activated anti-tumor compounds - Google Patents

Fap-activated anti-tumor compounds

Info

Publication number
EP1472277A1
EP1472277A1 EP03734699A EP03734699A EP1472277A1 EP 1472277 A1 EP1472277 A1 EP 1472277A1 EP 03734699 A EP03734699 A EP 03734699A EP 03734699 A EP03734699 A EP 03734699A EP 1472277 A1 EP1472277 A1 EP 1472277A1
Authority
EP
European Patent Office
Prior art keywords
compound
formula
amino
xxx
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP03734699A
Other languages
German (de)
French (fr)
Inventor
Stefan Peters
Steffen Breitfelder
John Edward Park
Pilar Garin-Chesa
Stefan Matthias Blech
Martin Lenter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim Pharma GmbH and Co KG
Original Assignee
Boehringer Ingelheim Pharma GmbH and Co KG
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 Boehringer Ingelheim Pharma GmbH and Co KG filed Critical Boehringer Ingelheim Pharma GmbH and Co KG
Priority to EP03734699A priority Critical patent/EP1472277A1/en
Publication of EP1472277A1 publication Critical patent/EP1472277A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1024Tetrapeptides with the first amino acid being heterocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • C07K9/001Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence having less than 12 amino acids and not being part of a ring structure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to the field of tumor treatment by administration of a prodrug that is converted into a drug at the site of the tumor.
  • the invention relates to prodrugs which may be converted into a drug by the catalytic action of FAP ⁇ , their manufacture and pharmaceutical use.
  • the human fibroblast activation protein is a M r 95,000 cell surface molecule origi- nally identified with monoclonal antibody (mAb) F19 (Rettig et al. (1988) Proc. Natl. Acad. Sci. USA 85, 3110-3114; Rettig et al. (1993) Cancer Res. 53, 3327-3335).
  • the FAP ⁇ cDNA codes for a type II integral membrane protein with a large extracellular domain, trans- membrane segment, and short cytoplasmic tail (Scanlan et al. (1994) Proc. Natl. Acad. Sci. USA 91, 5657-5661; WO 97/34927).
  • FAP ⁇ shows 48 % amino acid sequence identity to the T-cell activation antigen CD26, also known as dipeptidyl peptidase IN (DPPIV; EC 3.4.14.5), a membrane-bound protein with dipeptidyl peptidase activity (Scanlan et al, loc. citf).
  • CD26 also known as dipeptidyl peptidase IN (DPPIV; EC 3.4.14.5)
  • DPPIV dipeptidyl peptidase IN
  • a membrane-bound protein with dipeptidyl peptidase activity Scanlan et al, loc. citf
  • FAP ⁇ has enzymatic activity and is a member of the serine protease family, with serine 624 being critical for enzymatic function (WO 97/34927).
  • FAP ⁇ is selectively expressed in reactive stromal fibroblasts of many histological types of human epithelial cancers, granulation tissue of healing wounds, and malignant cells of certain bone and soft tissue sarcomas. Normal adult tissues are generally devoid of detectable FAP ⁇ , but some fetal mesenchymal tissues transiently express the molecule. In contrast, most of the common types of epithelial cancers, including >90% of breast, non-small-cell lung, and colorectal carcinomas, contain FAP ⁇ -reactive stromal fibroblasts (Scanlan et al., loc. citf).
  • FAP ⁇ + fibroblasts accompany newly formed tumor blood vessels, forming a distinct cellular compartment interposed between the tumor capillary endothelium and the basal aspect of malignant epithelial cell clusters (Welt et al. (1994) J. Clin. Oncol. 12(6), 1193- 1203). While FAP ⁇ + stromal fibroblasts are found in both primary and metastatic carcinomas, the benign and premalignant epithelial lesions tested (Welt et al., loc. cit.), such as fibroadenomas of the breast and colorectal adenomas, only rarely contain FAP ⁇ + stromal cells.
  • Triggering mechanisms can be designed so that the toxic agents synthesized in their prodrug or inactive forms are rendered active when and where required, notably in the cancerous tissues (Panchal (1998) Biochem. Pharmacol 55, 247-252).
  • Triggering mechanisms may include either exogenous factors such as light or chemicals or endogenous cellular factors, such as enzymes with re- stricted expression in cancer tissues.
  • ADEPT 'antibody-directed enzyme prodrug therapy'
  • ADC 'antibody-directed catalysis'
  • an antibody directed at a tu or- associated antigen is used to target a specific enzyme to the tumor site.
  • the tumor-located enzyme converts a subsequently administered prodrug into an active cytotoxic agent.
  • the antibody-enzyme conjugate binds to a target antigen on cell membranes or to free antigen in extracellular fluid (ECF).
  • ECF extracellular fluid
  • a time interval between giving the AEC and prodrug allows for the AEC to be cleared from normal tissues so that the prodrug is not activated in the normal tissues or blood.
  • some disadvantages of ADEPT are related to the properties of the AEC (Bagshawe, loc. cit.). For example, in humans, only a small fraction of the administered dose of the targeting AEC binds to tumor tissue and the remainder is distributed through body fluids from which it is cleared with significant time delays. Even very low concentrations of unbound enzyme can catalyze enough prodrug to have toxic effects because plasma and normal ECF volumes are much greater than those of tumor ECF.
  • the AEC may also be immunogenic, thus preventing repeat administration, in many instances.
  • oligopeptides including the following penta- and hexapeptide (SEQ.ID.NOs.: 151 and 177: hArg-Tyr-Gln- Ser-Ser-Pro; hArg-Tyr-Gln-Ser-Pro;), comprising amino acid sequences, which are recognized and proteolytically cleaved by free prostate specific antigen (PSA) and therapeutic agents which comprise conjugates of such oligopeptides and known therapeutic or cytotoxic agents.
  • PSA prostate specific antigen
  • therapeutic agents which comprise conjugates of such oligopeptides and known therapeutic or cytotoxic agents.
  • the International patent application WO 00/71571 discloses prodrugs which are capable of being converted into a drug by the catalytic action of FAP ⁇ , said prodrug having a cleavage site which is recognized by FAP ⁇ , and said drug being cytotoxic or cytostatic under physiological conditions.
  • these prodrugs have comparably poor solubilities.
  • the problem underlying the present invention was to provide similar prodrugs with improved solubilities.
  • the present invention relates to prodrugs which are capable of being converted into a cytotoxic or cytostatic drug, by the catalytic action of FAP ⁇ , said prodrugs exhibit an oligomeric part comprising up to 13 amino carboxylic acid residues, the C-terminal amino carboxylic acid thereof is recognized by FAP ⁇ , and a cytotoxic or cytostatic part, wherein the N-terminal amino function of the oligomeric part is attached to a capping group (Cg) of formula II.
  • a drug shall mean a chemical compound that may be administered to humans or animals as an aid in the treatment of disease.
  • a drug is an active pharmacological agent.
  • cytotoxic compound shall mean a chemical compound which is toxic to living cells, in particular a drug that destroys or kills cells.
  • cytostatic compound shall mean a compound that suppresses cell growth and multiplication and thus inhibits the proliferation of cells.
  • examples for cytotoxic or cytostatic compounds suitable for the present invention are anthracycline derivatives such as doxorubicin, analogs of methotrexate such as methothrexate, pritrexime, trimetrexate or DDMP, melphalan, analogs of cisplatin such as cisplatin, JM216, JM335, bis(platinum) or carboplatin, analogs of purines and pyrimidines such as cytarbine, gemcitabine, azacitidine, 6-thioguanine, flurdarabine or 2- deoxycoformycin, and analogs of other chemotherapeutic agents such as 9- aminocamptothecin, D,L-aminoglutethimide, trime
  • prodrug shall mean a compound that, on administration, must undergo chemical conversion by metabolic processes before becoming an active pharmacological agent.
  • a prodrug is a precursor of a drug.
  • the prodrug is significantly less cytotoxic or cytostatic than the drug it is converted into upon the catalytic action of FAP ⁇ .
  • the expert knows methods of determining cytotoxicity of a compound, see e.g. example 6 of WO 00/71571, or Mosmann ((1983) I. Immun. Meth. 65, 55-63).
  • the prodrug is at least three times less cytotoxic as compared to the drug in an in vitro assay.
  • a "drug being cytostatic or cytotoxic under physiological conditions” shall mean a chemical compound which is cytostatic or cytotoxic in a living human or animal body, in particular a compound that kills cells or inhibits proliferation of cells within a living human or animal body.
  • a "prodrug having a cleavage site which is recognised by FAP ⁇ ” shall mean a prodrug which can act as a substrate for the enzymatic activity of FAP ⁇ .
  • the enzymatic activity of FAP ⁇ can catalyse cleavage of a covalent bond of the prodrug under physiological conditions. By cleavage of this covalent bond, the prodrug is converted into the drug, either directly or indirectly. Indirect activation would be the case if the cleavage product of the FAP ⁇ catalyzed step is not the pharmacologically active agent itself but undergoes a further reaction step, e.g. hydrolysis, to become active.
  • the cleavage site of the prodrug is specifically recognized by FAP ⁇ , but not by other proteolytic enzymes present in the human or animal body. Also preferably, the cleavage site is specifically recognised by FAP ⁇ , but not by proteolytic enzymes present in human or animal body fluids, especially plasma. In a particularly preferred embodiment, the prodrug is stable in plasma, other body fluids, or tissues, in which biologically active FAP ⁇ is not present or detectable.
  • the cleavage site should most preferably be specific for FAP ⁇ .
  • the cleavage site comprises a L-proline residue which is linked to a cytotoxic or cytostatic drug via an amide bond.
  • An example of this class is a doxorubicin-peptide conjugate.
  • FAP ⁇ may catalyse the cleavage of a peptidic bond between the C-terminal amino acid residue of the peptide, which is preferably L-proline, and the cytotoxic or cytostatic compound.
  • Preferred compounds show at least 10% conversion to free drug, under standard conditions listed below. More preferred are compounds that show at least 20% conversion to free drug, under standard conditions. Even more preferred are compounds that show at least 50% conversion to free drug, under standard conditions.
  • standard conditions are defined as follows: Each compound is dissolved in 50 mM Hepes buffer, 150 mM NaCl, pH 7.2, at a final concentration of 5 ⁇ M and incubated with 100 ng CD8FAP ⁇ (see example 4 of WO 00/71571) for 24 hours at 37 °C. Release of free drug by CD ⁇ FAP ⁇ is determined as de- scribed in example 5 of WO 00/71571 .
  • the present invention relates to a compound of formula (I)
  • R 1 represents an amino alkanoyl or oligopeptidoyl group, the N-terminal amino function of which is attached to a capping group (Cg) of formula (II)
  • R a and R b together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH 2 groups may also be replaced by NH, O or S;
  • R 3 represents H, CrC 6 -alkyl, C 3 -C 8 -cycloalkyl, aryl or heteroaryl; and Cyt' represents the residue of a cytotoxic or cytostatic compound.
  • s is an integer of 1 or 2
  • l in particular l
  • t is an integer of 1 or 2, in particular 1.
  • capping groups wherein the group HO-X 1 -(CH 2 ) s - is attached in the the meta- or ⁇ r -position with respect to the group -(CH 2 )t-X -.
  • capping groups selected from the formulae HA and DB:
  • R 1 represents a residue of formula Cg-A, Cg-B-A or Cg-(D) n -B-A, in which Cg represents a capping group of formula (II),
  • A, B and D each independently represent moieties derived from amino carboxylic acids of the formula -[NR 4 -(X) p -CO]- wherein X represents CR 5 R 6 and wherein R 4 , R 5 and R 6 each independently represent a hydrogen atom, an optionally substituted - - alkyl, C 3 -C 8 -cycloalkyl, aryl, aralkyl, heteroaryl or heteroarylalkyl group, and p is 1, 2, 3, 4, 5; or
  • A, B and D each independently represent moieties derived from cyclic amino carboxylic acids of formula
  • R 7 represents -C ⁇ -alkyl, OH, or NH 2 , n is an integer from 1 to 10; q is 0, 1 or 2; and r is 0, 1 or 2, in particular wherein R 1 represents a group selected from Cg-A, Cg-B-A- and Cg-(D) n -B-A- in which A, B and D are amino acid moieties, which are each independently selected from glycine (Gly), and the D- or L-forms of alanine (Ala), valine (Nal), leucine (Leu), isoleucine (He), phenylalanine (Phe), tyrosine (Tyr), tryptophan (Trp), cysteine (Cys), methionine (Met), serine (Ser), threonine (Thr), lysine (Lys), arginine (Arg), histidine (His), aspartatic acid (Asp), glutamic acid (Glu), asparagine
  • the unit A is preferably selected from L-proline, glycine, L-norleucine, L-cyclohexylglycine, L-5-hydroxynorleucine, L-6-hydroxynorleucine, L-5-hydroxylysine, L-arginine, and L-lysine.
  • the N-terminal unit D is preferably an amino acid, wherein the amino group thereof is a secondary amino group forming with the linkage to the capping group a tertiary amino group, in particular selected from L-proline (Pro), azetidine-2-ylcarboxylic acid and N-C ⁇ -6 alkyl amino acids such as N-methylglycin (N-MeGly), N-methylalanin (N-MeAla), N-methylvaline (N-MeVal), N-methylleucine (N-Me-Leu), N-methylisoleucine (N-Me- ⁇ e), N- methylnorleucin (N-Me e), N-methyl-2-aminobutyric acid (N-Me-Abu) and N-methyl-2- aminopentanoic acid (N-MeNva).
  • Pro L-proline
  • N-C ⁇ -6 alkyl amino acids such as N-methylglycin (N-MeGly),
  • R 8 and R 9 each independently represent a hydrogen or halogen atom or a Ci-C ⁇ -alkyl, d-C ⁇ -alkylamino, di- C ⁇ -C 6 -alkylamino, Q- -alkoxy, thiol, Ci-C 6 -alkylthio, oxo, imino, fomyl, Q-Cg-alkoxy carbonyl, amino carbonyl, C 3 -C 8 -cycloalkyl, aryl, or heteroaryl group.
  • R >3 , R , R 5 , Cyt', Cg, X and Y are as defined hereinbefore, or R 4 and R 5 together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH 2 groups may also be replaced by NH, O or S.
  • CrC ⁇ -alkyl generally represents a straight-chained or branched hydrocarbon radical having 1 to 6 carbon atoms.
  • radicals may be mentioned by way of example: Methyl, ethyl, propyl, 1-methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1- dimethylethyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 2-meth- ylpentyl, 3-methylpentyl, 4-methylpentyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimeth- ylbutyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1,1,2-trimethylpropyl
  • Ci-C 6 -alkyl is substituted with aryl or heteroaryl
  • CrC 6 -alkyl is preferably Q, more preferably a methylene group.
  • amino alkanoyl and oligopeptidoyl including “di- or tripeptidoyl” as used hereinabove or hereinbelow with respect to radical R 1 describe a radical in which an amino acid or an oligomer comprising up to 12, preferably 2 or 3 amino acid moieties is attached C- terminally to the nitrogen atom of the heterocyclic ring via an amide bond.
  • amino acids may be replaced by other homologous, isosteric and/or isolectronic amino acids wherein the biological activity of the original amino acid or oligopeptide has been conserved upon modification.
  • Certain unnatural and modified natural amino acids may also be utilized to replace the corresponding natural amino acid.
  • tyrosine may be replaced by 3-iodotyrosine, 2- or 3-methyltyrosine, 3- fluorotyrosine.
  • Heterocyclic ring as used hereinabove and hereinbelow with respect to the group formed by R a and R together with the interjacent N-C group generally represents a 3 to 7-membered, preferably 4-, 5- or 6-membered non-aromatic heterocyclic ring system, containing one nitrogen atom and optionally 1 or 2 additional heteroatoms selected from the group of nitrogen, oxygen and sulfur, which may be substituted by one or several halogen atoms or Q- C ⁇ -alkyl, Q-Ce-alkylamino, di-CrCe-alkylamino, Q-Cts-alkoxy, thiol, C!-C 6 -alkylthio, oxo, imino, fomyl, C ⁇ -C 6 -alkoxy carbonyl, amino carbonyl, C 3 -C 8 -cycloalkyl, aryl, or heteroaryl groups, which may be identical to one another or different, and which optionally may be benzo-
  • Such heterocyclic rings are preferably azetidine or are derived from a fully or partially hydrogenated pyrrole, pyridine, thiazole, isoxazole, pyrazole, imidazole, indole, benzi idazole, indazole, pyridazine, pyrimidine, pyrazin group.
  • Most preferred are azetidine, pyrrolidine, 3,4-dehydropyrrolidine, piperidine, hexahydro-lH- azepine, octahydroindole, imidazolidine, thiazolidine.
  • the substituents are preferably methyl, ethyl, propyl, 1- methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dimethyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, -COOCH 3 , -COOCH 2 CH 3 , or -CONH 2 .
  • Aryl generally represents an aromatic ring system with 6 to 10, preferably 6 carbon atoms which may optionally be substituted by one or several hydroxyl, amino, d-C 6 -alkyl-amino, di- d-Ce-alkyl-amino, d-C 6 -alkyl, C C 6 -alkyloxy, thiol, d-C 6 -alkyl-thio, -CHO, -COOH, - COOCH 3 , -COOCH 2 CH 3 , -CONH 2 , or halogen substituents, which may be identical to one another or different, and which optionally may be benzocondensed.
  • Aryl substituents may be preferably derived from benzene, preferred examples being phenyl, 2-hydroxy-phenyl, 3- hydroxy-phenyl, 4-hydroxy-phenyl, 4-amino-phenyl, 2-amino- ⁇ henyl, 3-amino-phenyl.
  • the substituents are preferably methyl, ethyl, propyl, 1 -methylethyl (iso- propyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dime- thyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, -COOCH 3 , - COOCH 2 CH 3 , or -CONH 2 .
  • Heteroaryl generally represents a 5 to 10-membered aromatic heterocyclic ring system, containing 1 to 5 heteroatoms selected from the group of nitrogen, oxygen, or sulfur, which may optionally be substituted by one or several hydroxyl, amino, d-C 6 -alkyl-amino, di- d- Ce-alkyl-amino, C C 6 -alkyl, d-C 6 -alkyloxy, thiol, C ⁇ -C 6 -alkyl-thio, -CHO, -COOH, - COOCH 3 , -COOCH 2 CH 3 , -CONH 2 , or halogen substituents, which may be identical to one another or different, and which optionally may be benzocondensed.
  • Heteroaryl substituents may preferably be derived from furane, pyrrole, thiophene, pyridine, thiazole, isoxazole, pyrazole, imidazole, benzofuran, thianaphthene, indole, benzimidazole, indazole, quinoline, pyridazine, pyrimidine, pyrazine, chinazoline, pyrane, purine, adenine, guanine, thymine, cytosine, uracil.
  • the substituents are preferably methyl, ethyl, propyl, 1 -methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dimethyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, - COOCH 3 , -COOCH 2 CH 3 , or -CONH 2 .
  • Residue of a cytotoxic or cytostatic compound means that the compound H 2 N-Cyt', which is released upon cleavage of the amide bond shown in formula (I), is either cytotoxic or cytostatic itself, or may be converted into a cytotoxic or cytostatic compound in a subsequent step.
  • -Cyt' may be a residue of formula -L-Cyt", wherein L is a linker residue derived from a bifunctional molecule, for instance a diamine H 2 N-L'-NH 2 , an amino alcohol H 2 N-L'-OH, for example p-amino-benzyl alcohol (PABOH), an amino carbonate, for example
  • -Cyt' is of formula -L-Cyt
  • the compound H 2 N-L'- Cyt' ' is generated by the enzymatic cleavage of the amide bond shown in formula (I).
  • the compound H 2 N-L'-Cyt" may be cytotoxic or cytostatic itself or the linker residue cleaved off from Cyt' ' in a subsequent step releasing the cytotoxic or cytostatic agent.
  • the compound H 2 N-L'-Cyt may be hydrolysed under physiological conditions into a compound H 2 N-L'-OH and the cytotoxic or cytostatic compound H-Cyt" , which is the active therapeutic agent (In the following, only the term Cyt' is used for both Cyt' and Cyt", and only the term L is used for both L and L', for simplicity).
  • the pharmaceutically acceptable salts of the compounds of the present invention include the conventional non-toxic salts formed from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those from inorganic acids such as hydrochloric acid, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, maleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, oxalictrifluoroacetic and the like.
  • H 2 N-Cyt' is preferably an anthracycline derivative of formula IN,
  • R c represents d-C 6 alkyl, d-C 6 hydroxyalkyl or d-C 6 alkanoyloxy C ⁇ -C 6 alkyl, in particular methyl, hydroxymethyl, diethoxyacetoxymethyl or butyryloxymethyl;
  • R d represents hydrogen, hydroxy or d-C 6 alkoxy, in particular methoxy; one of R e and R f represents a hydrogen atom; and the other represents a hydrogen atom or a hydroxy or tetrahydropyran-2-yloxy (OTHP) group.
  • Cyt' may be derived for example from methotrexate, trimetrexate, pyritrexim, 5,10- dideazatetrahydrofolatepyrimetamine, trimethoprim, 10-propargyl-5,8-dideazafolate2,4- diamino-5(3 ' ,4' -dichloropheyl)-6-methylpyrimidine, aminoglutethimide, goreserelin, melphalan, chlorambucil, analogs of other chemotherapeutic agents such as 9- aminocamptothecin (for examples see e.g. Burris HA, r. d. and S. M.
  • 9- aminocamptothecin for examples see e.g. Burris HA, r. d. and S. M.
  • Cyt' may also be a biological effector molecule which either directly or indirectly effects destruction of tumor cells, like for example TNF ⁇ .
  • Preferred anthracycline prodrugs are the compounds of formula HI wherein R a , R b , R c , R d , R e , R f and R 1 are as defined hereinabove.
  • Most preferred compounds of the invention are doxorubicin derivatives of formulae (IIIA) to
  • the compound of the invention may contain one or more disulfide bonds.
  • cytotoxic or cytostatic compounds which may be used for the present invention has a primary amino function which is available for formation of an amidic bond as shown in formula (I), like doxorubicin.
  • a linker molecule L is not necessary. If a cytostatic or cytotoxic compound does not have such an amino function, such a function may be created in such a compound by way of chemical modification, e.g. by introducing or converting a functional group or attaching a linker molecule to the compound.
  • a linker molecule may also be inserted between the oligomeric part (i.e.
  • the part comprising the amino carboxylic residues) and the cytostatic or cytotoxic part of the compound of the invention to ensure or optimize cleavage of the amide bond between the oligomeric part and the cytotoxic or cytostatic part.
  • a linker molecule is present, i.e. in compounds containing the structure L- Cyt', the bond between L and Cyt' is preferably an amidic or ester bond.
  • such a linker molecule is hydrolyzed off the cytostatic or cytotoxic compound under physiological conditions after the enzymatic cleavage and thus the free cytostatic or cytotoxic compound is generated.
  • the compound of the invention must have the property of being cleavable upon the catalytic action of FAP ⁇ and, as a direct or indirect consequence of this cleavage, releasing under physiological conditions a cytostatic or cyto- toxic compound.
  • the present invention relates to prodrug that is capable of being converted into a cytotoxic or cytostatic drug, by the catalytic action of FAP ⁇ , said prodrug exhibits an oligomeric part comprising up to 13 amino carboxylic residues, the C-terminal amino carboxylic thereof is recognized by FAP ⁇ , and a cytotoxic or cytostatic part, characterized in that the N-terminal amino function of the oligomeric part is attached to a capping group (Cg) of formula ⁇ .
  • Cg capping group
  • the oligomeric part is preferably a peptide.
  • the oligomeric part comprises two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve amino carboxylic acid residues, more preferably two, three, or four amino carboxylic residues.
  • the compounds of the invention may be synthesized by processes known in the art (E. W ⁇ nsch, Synthese von Peptiden, in “Methoden der organischen Chemie”, Houben-Weyl (Eds. E. M ⁇ ller, O. Bayer), Vol. XV, Part 1 and 2, Georg Thieme Nerlag, Stuttgart, 1974).
  • the compounds could be synthesized by condensation of the terminal amino function of the oligomeric part of a compound of formula NI with a compound of formula N,
  • R la represents an amino alkanoyl or oUgopeptidoyl group, the N-terminal amino function of which bears at least one hydrogen atom.
  • R la represents an amino alkanoyl or oUgopeptidoyl group, the N-terminal amino function of which bears at least one hydrogen atom.
  • This activation can be done by conversion of the carboxylic acid into an acid chloride or acid fluoride or by conversion of the carboxylic acid into an activated ester, for instance a N- hydroxysuccinimidyl ester or a pentafluorophenyl ester.
  • Another method of activation is the transformation into a symmetrical or unsymmetrical anhydride.
  • the formation of the amide bonds can be achieved by the use of in situ coupling reagents like benzotriazole- 1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBOP) (E.
  • the protection group PG 1 is removed at the end of the synthesis.
  • the compounds of the invention are intended for medical use.
  • these compounds are useful for the treatment of tumors which are associated with stromal fibroblasts that express FAP ⁇ and which are generally not optimally treated with available cytotoxic and/or cytostatic agents.
  • Tumors with this property are, for example, epithelial cancers, such as lung, breast, and colon carcinomas.
  • Tumors, such as bone and soft tissue sarcomas which express FAP ⁇ may also be treated with these compounds.
  • compositions comprising a compound of the present invention and optionally one or more suitable and pharmaceutically acceptable excipients, as exemplified in: Remington: the science and practice of pharmacy. 19th ed. Easton : Mack Publ, 1995.
  • the pharmaceutical compositions may be formulated as solids or solutions. Solid formulations may be for preparation of a solution before injection.
  • the pharmaceutical compositions of the invention are solutions for injection. They may be administered systemically, e.g. by intravenous injection, or topically, e.g. by direct injection into the tumor site.
  • the dosage will be adjusted according to factors like body weight and health status of the patient, nature of the underlying disease, therapeutic window of the compound to be applied, solubility, and the like. It is within the knowledge of the expert to adjust dosage appropriately.
  • the dose will preferably be in the range from 10 mg/m 2 to 2000 mg/m 2 , but also higher or lower doses may be appropriate.
  • a further aspect of the present invention is the use of a compound of the invention in the preparation of a pharmaceutical composition for the treatment of cancer.
  • an aspect of the invention is a method of treatment of cancer, comprising administering an effective amount of a pharmaceutical composition of the invention to a patient.
  • Indications include the treatment of cancer, specifically:
  • epithelial carcinomas including breast, lung, colorectal, head and neck, pancreatic, ovarian, bladder, gastric, skin, endometrial, ovarian, testicular, esophageal, prostatic and renal origin;
  • Bone and soft-tissue sarcomas Osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma (MFH), leiomyosarcoma;
  • Hematopoietic malignancies Hodgkin's and non-Hodgkin's lymphomas; 4) Neuroectodermal tumors: Peripheral nerve tumors, astrocytomas, melanomas; 5) Mesotheliomas.
  • a further aspect of the invention is a method of treatment of cancer, wherein a prodrug is administered to a patient wherein said prodrug is capable of being converted into a cytotoxic or cytostatic drug by an enzymatic activity, said enzymatic activity being the expression product of cells associated with tumor tissue.
  • said enzymatic activity is the proteolytic activity of FAP ⁇ .
  • One method of administration of the compounds is intravenous infusion.
  • Other possible routes of administration include intraperitoneal (either as a bolus or infusion), intramuscular or intratumoral injection. Where appropriate, direct application may also be possible (for example, lung fibrosis).
  • 1,4-phenylendiacetic acid (4.8 g, 25 mmol) was dissolved in acetonitrile (100 ml) and N,N- dimethylformamide (100 ml) The solution was cooled to 0 °C and N-hydroxysuccinimide (2,9 g, 25 mmol) and diisopropylethylamine (8.6 ml, 50 mmol) were added. The solution was stirred for 30 min at 0 °C. Then a solution of dicyclohexyl carbodiimide(5.2 g, 25 mmol) dissolved in acetonitrile (50 ml) was added dropwise over 1 h. The ice bath was removed an the suspension was stirred over night.
  • Buffer A lOOmM Tris HC1 pH 7.8, 100 mM NaCl
  • MNA conjugates Excitation: 355 nm, Emission: 405 nm.
  • the fluorescence measured in the samples treated with control 293 control cell extracts without FAP is subtracted from the values measured in the samples treated with 1 ng FAP enzyme.
  • H-Gly-2-chlorotrityl-resin (10 g, 11.1 mmol) were added to a reaction vessel and washed with N,N-dimethylformamide (DMF) (three times with 150 ml).
  • Fmoc-Ala-OH (20.7 g, 66.6 mmol)
  • O-(benzotriazol-l-yl)-N,N,N,N-tetramethyluronium tetrafluoroborate (TBTU) (21.4 g, 66.6 mmol)
  • 1-hydroxybenzotriazole (HOBt) (9.06 g, 66.6 mmol) and diisopropylethylamine (8 eq.) dissolved in DMF (100 ml) were added.
  • the suspension was shaken at room temperature over night.
  • the resin was washed carefully four times with DMF, dichloromethane, and DMF.
  • the cleavage of the Fmoc-group was performed with a solution of 30% piperidine in DMF for 30 minutes followed by careful washing steps with DMF (4 times), dichloromethane (3 times) and DMF (4 times). Boc-Pro-OH was incorporated in the same manner.
  • the resin was finally washed with methanol and diethyl ether and dried in a stream of nitrogen.
  • the following table shows the peptide-M ⁇ A-conjugates which have been prepared analogously and includes cleavage data by FAP.
  • solubility of A is > 10 mg/ml, whereas the solubility of B is ⁇ 2 mg/ml.

Abstract

The invention relates to a prodrug that is capable of being converted into a drug by the catalytic action of human fibroblast activation protein (FAPα), said prodrug is chemically stable under physiological conditions and can be used for the manufacture of physically stable aqueous formulations. It has a cleavage site which is recognised by FAPα, and the drug released by the enzymatic activity of FAPα is cytotoxic or cytostatic under physiological conditions.

Description

FAP-activated anti-tumor compounds
TECHNICAL FIELD OF THE INVENTION
The present invention relates to the field of tumor treatment by administration of a prodrug that is converted into a drug at the site of the tumor. In particular, the invention relates to prodrugs which may be converted into a drug by the catalytic action of FAPα, their manufacture and pharmaceutical use.
BACKGROUND OF THE INVENTION
The human fibroblast activation protein (FAPα) is a Mr 95,000 cell surface molecule origi- nally identified with monoclonal antibody (mAb) F19 (Rettig et al. (1988) Proc. Natl. Acad. Sci. USA 85, 3110-3114; Rettig et al. (1993) Cancer Res. 53, 3327-3335). The FAPα cDNA codes for a type II integral membrane protein with a large extracellular domain, trans- membrane segment, and short cytoplasmic tail (Scanlan et al. (1994) Proc. Natl. Acad. Sci. USA 91, 5657-5661; WO 97/34927). FAPα shows 48 % amino acid sequence identity to the T-cell activation antigen CD26, also known as dipeptidyl peptidase IN (DPPIV; EC 3.4.14.5), a membrane-bound protein with dipeptidyl peptidase activity (Scanlan et al, loc. citf). FAPα has enzymatic activity and is a member of the serine protease family, with serine 624 being critical for enzymatic function (WO 97/34927). Work using a membrane overlay assay revealed that FAPα dimers are able to cleave Ala-Pro-7-amino-4-trifluoromethyl coumarin, Gly-Pro-7-amino-4-trifluoromethyl coumarin, and Lys-Pro-7-amino-4-trifluoromethyl coumarin dipeptides (WO 97/34927).
FAPα is selectively expressed in reactive stromal fibroblasts of many histological types of human epithelial cancers, granulation tissue of healing wounds, and malignant cells of certain bone and soft tissue sarcomas. Normal adult tissues are generally devoid of detectable FAPα, but some fetal mesenchymal tissues transiently express the molecule. In contrast, most of the common types of epithelial cancers, including >90% of breast, non-small-cell lung, and colorectal carcinomas, contain FAPα-reactive stromal fibroblasts (Scanlan et al., loc. citf). These FAPα+ fibroblasts accompany newly formed tumor blood vessels, forming a distinct cellular compartment interposed between the tumor capillary endothelium and the basal aspect of malignant epithelial cell clusters (Welt et al. (1994) J. Clin. Oncol. 12(6), 1193- 1203). While FAPα+ stromal fibroblasts are found in both primary and metastatic carcinomas, the benign and premalignant epithelial lesions tested (Welt et al., loc. cit.), such as fibroadenomas of the breast and colorectal adenomas, only rarely contain FAPα+ stromal cells. Based on the restricted distribution pattern of FAPα in normal tissues and its uniform expression in the supporting stroma of many malignant tumors, clinical trials with 131I-labeled mAb F19 have been initiated in patients with metastatic colon carcinomas (Welt et al, loc. cit.).
For new cancer therapies based on cytotoxic or cytostatic drugs, a major consideration is to increase the therapeutic index by improving the efficacy of cancerous tissue killing and/or reducing the toxicity for normal tissue of the cytotoxic or cytostatic agents. To increase specificity of tumor tissue killing and reduce toxicity in normal tissues, trigger mechanisms can be designed so that the toxic agents synthesized in their prodrug or inactive forms are rendered active when and where required, notably in the cancerous tissues (Panchal (1998) Biochem. Pharmacol 55, 247-252). Triggering mechanisms may include either exogenous factors such as light or chemicals or endogenous cellular factors, such as enzymes with re- stricted expression in cancer tissues. Another concept, that has been further elaborated, is called 'antibody-directed enzyme prodrug therapy' (ADEPT) or 'antibody-directed catalysis' (ADC) (Huennekens (1994) Trends Biotechnol. 12, 234-239; Bagshawe (1994) Clin. Pharmacokinet. 27, 368-376; Wang et al. (1992) Cancer Res. 52, 4484-4491; Sperker et al. (1997) Clin. Pharmacokinet. 33(1), 18-31). In ADEPT, an antibody directed at a tu or- associated antigen is used to target a specific enzyme to the tumor site. The tumor-located enzyme converts a subsequently administered prodrug into an active cytotoxic agent. The antibody-enzyme conjugate (AEC) binds to a target antigen on cell membranes or to free antigen in extracellular fluid (ECF). A time interval between giving the AEC and prodrug allows for the AEC to be cleared from normal tissues so that the prodrug is not activated in the normal tissues or blood. However, some disadvantages of ADEPT are related to the properties of the AEC (Bagshawe, loc. cit.). For example, in humans, only a small fraction of the administered dose of the targeting AEC binds to tumor tissue and the remainder is distributed through body fluids from which it is cleared with significant time delays. Even very low concentrations of unbound enzyme can catalyze enough prodrug to have toxic effects because plasma and normal ECF volumes are much greater than those of tumor ECF. The AEC may also be immunogenic, thus preventing repeat administration, in many instances.
The International patent applications WO 97/12624 and WO 97/14416 disclose oligopeptides including the following penta- and hexapeptide (SEQ.ID.NOs.: 151 and 177: hArg-Tyr-Gln- Ser-Ser-Pro; hArg-Tyr-Gln-Ser-Pro;), comprising amino acid sequences, which are recognized and proteolytically cleaved by free prostate specific antigen (PSA) and therapeutic agents which comprise conjugates of such oligopeptides and known therapeutic or cytotoxic agents. These oligopeptide conjugates which comprise at least one glutamine-serine moiety are useful for treatment of prostate cancer only.
The International patent application WO 00/71571 discloses prodrugs which are capable of being converted into a drug by the catalytic action of FAPα, said prodrug having a cleavage site which is recognized by FAPα, and said drug being cytotoxic or cytostatic under physiological conditions. However, these prodrugs have comparably poor solubilities.
The problem underlying the present invention was to provide similar prodrugs with improved solubilities.
BRIEF DESCRIPTION OF THE INVENTION
Surprisingly it has been found that the prodrugs, wherein the N-terminal amino function of the oligomeric part is attached to a capping group (Cg) of formula (II)
in which
X1 represents C=O or SO2,
X2 represents C=O, SO2, NH-C=O or a single bond, s is an integer of 1 or 2, and t is 0 or an integer of 1, 2 or 3 and have high solubilities.
The present invention relates to prodrugs which are capable of being converted into a cytotoxic or cytostatic drug, by the catalytic action of FAPα, said prodrugs exhibit an oligomeric part comprising up to 13 amino carboxylic acid residues, the C-terminal amino carboxylic acid thereof is recognized by FAPα, and a cytotoxic or cytostatic part, wherein the N-terminal amino function of the oligomeric part is attached to a capping group (Cg) of formula II.
In the context of this invention, a "drug" shall mean a chemical compound that may be administered to humans or animals as an aid in the treatment of disease. In particular, a drug is an active pharmacological agent.
The term "cytotoxic compound" shall mean a chemical compound which is toxic to living cells, in particular a drug that destroys or kills cells. The term "cytostatic compound" shall mean a compound that suppresses cell growth and multiplication and thus inhibits the proliferation of cells. Examples for cytotoxic or cytostatic compounds suitable for the present invention are anthracycline derivatives such as doxorubicin, analogs of methotrexate such as methothrexate, pritrexime, trimetrexate or DDMP, melphalan, analogs of cisplatin such as cisplatin, JM216, JM335, bis(platinum) or carboplatin, analogs of purines and pyrimidines such as cytarbine, gemcitabine, azacitidine, 6-thioguanine, flurdarabine or 2- deoxycoformycin, and analogs of other chemotherapeutic agents such as 9- aminocamptothecin, D,L-aminoglutethimide, trimethoprim, pyrimethamine, mitomycin C, mitoxantrone, cyclophosphanamide, 5-fluorouracil, extramustine, podophyllotoxin, bleomycin, epothilone and derivatives of epothilone as described for example in US 6,204,388 or taxol.
A "prodrug" shall mean a compound that, on administration, must undergo chemical conversion by metabolic processes before becoming an active pharmacological agent. In par- ticular, a prodrug is a precursor of a drug. In the context of the present invention, the prodrug is significantly less cytotoxic or cytostatic than the drug it is converted into upon the catalytic action of FAPα. The expert knows methods of determining cytotoxicity of a compound, see e.g. example 6 of WO 00/71571, or Mosmann ((1983) I. Immun. Meth. 65, 55-63). Pref- erably, the prodrug is at least three times less cytotoxic as compared to the drug in an in vitro assay.
A "drug being cytostatic or cytotoxic under physiological conditions" shall mean a chemical compound which is cytostatic or cytotoxic in a living human or animal body, in particular a compound that kills cells or inhibits proliferation of cells within a living human or animal body.
A "prodrug having a cleavage site which is recognised by FAPα" shall mean a prodrug which can act as a substrate for the enzymatic activity of FAPα. In particular, the enzymatic activity of FAPα can catalyse cleavage of a covalent bond of the prodrug under physiological conditions. By cleavage of this covalent bond, the prodrug is converted into the drug, either directly or indirectly. Indirect activation would be the case if the cleavage product of the FAPα catalyzed step is not the pharmacologically active agent itself but undergoes a further reaction step, e.g. hydrolysis, to become active. More preferably, the cleavage site of the prodrug is specifically recognized by FAPα, but not by other proteolytic enzymes present in the human or animal body. Also preferably, the cleavage site is specifically recognised by FAPα, but not by proteolytic enzymes present in human or animal body fluids, especially plasma. In a particularly preferred embodiment, the prodrug is stable in plasma, other body fluids, or tissues, in which biologically active FAPα is not present or detectable. Preferably, in an in vitro assay as carried out in Example 7 of the International Patent Application WO 00/71571, the disclosure of which is fully incorporated herein by reference, more than 50%, more preferably more than 80%, more preferably more than 90% of the prodrug are still present in a solution containing 10% (v/v) of human plasma after 8 h at 37°C. The cleavage site should most preferably be specific for FAPα. In a preferred embodiment, the cleavage site comprises a L-proline residue which is linked to a cytotoxic or cytostatic drug via an amide bond. An example of this class is a doxorubicin-peptide conjugate. FAPα may catalyse the cleavage of a peptidic bond between the C-terminal amino acid residue of the peptide, which is preferably L-proline, and the cytotoxic or cytostatic compound.
Preferred compounds show at least 10% conversion to free drug, under standard conditions listed below. More preferred are compounds that show at least 20% conversion to free drug, under standard conditions. Even more preferred are compounds that show at least 50% conversion to free drug, under standard conditions. In this context, standard conditions are defined as follows: Each compound is dissolved in 50 mM Hepes buffer, 150 mM NaCl, pH 7.2, at a final concentration of 5 μM and incubated with 100 ng CD8FAPα (see example 4 of WO 00/71571) for 24 hours at 37 °C. Release of free drug by CDδFAPα is determined as de- scribed in example 5 of WO 00/71571 .
DETAILED DESCRIPTION OF THE INVENTION
Preferably, the present invention relates to a compound of formula (I)
or a pharmaceutically acceptable salt thereof, wherein
R1 represents an amino alkanoyl or oligopeptidoyl group, the N-terminal amino function of which is attached to a capping group (Cg) of formula (II)
in which
X1 represents C=O or SO2,
X2 represents C=O, SO2, NH-C=O or a single bond, s is an integer of 1 or 2, and t is 0 or an integer of 1, 2 or 3,
Ra and Rb together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH2 groups may also be replaced by NH, O or S; R3 represents H, CrC6-alkyl, C3-C8-cycloalkyl, aryl or heteroaryl; and Cyt' represents the residue of a cytotoxic or cytostatic compound.
Preferred are the compounds of formula I, wherein
X1 represents C=O or SO2, in particular C=O, X2 represents C=O or SO2, in particular C=O, s is an integer of 1 or 2, in particular l,and t is an integer of 1 or 2, in particular 1.
Particularly preferred are the capping groups, wherein the group HO-X1-(CH2)s- is attached in the the meta- or^αr -position with respect to the group -(CH2)t-X -. Most preferred are the capping groups selected from the formulae HA and DB:
Furthermore preferred are those compounds of formula I, wherein
R1 represents a residue of formula Cg-A, Cg-B-A or Cg-(D)n-B-A, in which Cg represents a capping group of formula (II),
A, B and D each independently represent moieties derived from amino carboxylic acids of the formula -[NR4-(X)p-CO]- wherein X represents CR5R6 and wherein R4, R5 and R6 each independently represent a hydrogen atom, an optionally substituted - - alkyl, C3-C8-cycloalkyl, aryl, aralkyl, heteroaryl or heteroarylalkyl group, and p is 1, 2, 3, 4, 5; or
A, B and D each independently represent moieties derived from cyclic amino carboxylic acids of formula
wherein
R7 represents -Cβ-alkyl, OH, or NH2, n is an integer from 1 to 10; q is 0, 1 or 2; and r is 0, 1 or 2, in particular wherein R1 represents a group selected from Cg-A, Cg-B-A- and Cg-(D)n-B-A- in which A, B and D are amino acid moieties, which are each independently selected from glycine (Gly), and the D- or L-forms of alanine (Ala), valine (Nal), leucine (Leu), isoleucine (He), phenylalanine (Phe), tyrosine (Tyr), tryptophan (Trp), cysteine (Cys), methionine (Met), serine (Ser), threonine (Thr), lysine (Lys), arginine (Arg), histidine (His), aspartatic acid (Asp), glutamic acid (Glu), asparagine (Asn), glutamine
(Gin), proline (Pro), 4-hydroxy-proline (Hyp), 5-hydroxy-lysine, norleucine (Νle), 5- hydroxynorleucine (Hyn), 6-hydroxynorleucine, ornithine, cyclohexylglycine (Chg), Ν-methylglycin (Ν-MeGly), Ν-methylalanin (Ν-MeAla), Ν-methylvaline (Ν-MeNal), Ν-methylleucine (Ν-MeLeu), Ν-methylisoleucine (Ν-Melle), Ν-methylnorleucin (Ν- MeΝle), Ν-methyl-2-aminobutyric acid (N-MeAbu) and N-methyl-2-aminopentanoic acid (N-MeNva).
The unit A is preferably selected from L-proline, glycine, L-norleucine, L-cyclohexylglycine, L-5-hydroxynorleucine, L-6-hydroxynorleucine, L-5-hydroxylysine, L-arginine, and L-lysine.
The N-terminal unit D is preferably an amino acid, wherein the amino group thereof is a secondary amino group forming with the linkage to the capping group a tertiary amino group, in particular selected from L-proline (Pro), azetidine-2-ylcarboxylic acid and N-Cι-6 alkyl amino acids such as N-methylglycin (N-MeGly), N-methylalanin (N-MeAla), N-methylvaline (N-MeVal), N-methylleucine (N-Me-Leu), N-methylisoleucine (N-Me-ϋe), N- methylnorleucin (N-Me e), N-methyl-2-aminobutyric acid (N-Me-Abu) and N-methyl-2- aminopentanoic acid (N-MeNva).
Furthermore preferred are those compounds of formula I, wherein the heterocyclic ring formed by Ra, Rb and the interjacent N-C is substituted by R8 and R9, wherein R8 and R9 each independently represent a hydrogen or halogen atom or a Ci-Cβ-alkyl, d-Cδ-alkylamino, di- Cι-C6-alkylamino, Q- -alkoxy, thiol, Ci-C6-alkylthio, oxo, imino, fomyl, Q-Cg-alkoxy carbonyl, amino carbonyl, C3-C8-cycloalkyl, aryl, or heteroaryl group.
Particularly preferred are the compounds of formula LA,
wherein R1, R3, R8, Cyt' are as hereinbefore, and
X-Y represents CHR9-CH2, CR9=CH, NH-CH2, CH2-NH, -CR9-, CH2-CHR9-CH2, in particular CH2-CH2.
Most preferred are the compounds of formulae IA1 and IA2,
wherein R >3 , R , R5, Cyt', Cg, X and Y are as defined hereinbefore, or R4 and R5 together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH2 groups may also be replaced by NH, O or S.
Unless indicated otherwise, the simple stereoisomers as well as mixtures or racemates of the stereoisomers are included in the invention.
"CrCό-alkyl" generally represents a straight-chained or branched hydrocarbon radical having 1 to 6 carbon atoms.
The term "optionally substituted" as used hereinabove or hereinbelow with respect to a group or a moiety refers to a group or moiety which may optionally be substituted by one or several halogen atoms, hydroxyl, amino, CrCg-alkyl-amino, di- Q-Cό-alkyl-amino, -Cό-alkyl-oxy, thiol, d-Ce-alkyl-thio, =O, =NH, -CHO, -COOH, -CONH2, -NHC(=NH)NH2, , C3-C8- cycloalkyl, aryl, or heteroaryl substituents , which may be identical to one another or different.
The following radicals may be mentioned by way of example: Methyl, ethyl, propyl, 1-methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1- dimethylethyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 2-meth- ylpentyl, 3-methylpentyl, 4-methylpentyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 1,3-dimeth- ylbutyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1 -ethyl- 1-methylpropyl and l-ethyl-2methyl- propyl, HOCH2-, CH3CH(OH)-, CH3CH(OH)CH2CH2-, HOCH2CH2CH2CH2-, H2NCH2CH2CH2-, H2NCH2CH2CH2CH2-, H2NCH2CH(OH)CH2CH2-, H2NC(=NH)NHCH2CH2CH2-, HSCH2-, CH3SCH2CH2-, HOOCCH2-, HOOCCH2CH2-, H2NC(=O)CH2-, H2NC(=O)CH2CH2-, benzyl, αra-hydroxy-benzyl, 4-(para- hydroxyphenoxy)-benzyl,
If a Q-Ce-alkyl group is substituted, the substituents are preferably hydroxyl, amino, dimethylamino, diethylamino, thiol, methyl-thiol, methoxy, ethoxy, =O, =NH, -CHO, -
COOH, -COOCH3, -COOCH2CH3, -CONH2, -NHC(=NH)NH2, cyclohexyl, phenyl, benzyl, para-hydroxy-benzyl,
If Ci-C6-alkyl is substituted with aryl or heteroaryl, CrC6-alkyl is preferably Q, more preferably a methylene group.
The terms "amino alkanoyl" and "oligopeptidoyl" including "di- or tripeptidoyl" as used hereinabove or hereinbelow with respect to radical R1 describe a radical in which an amino acid or an oligomer comprising up to 12, preferably 2 or 3 amino acid moieties is attached C- terminally to the nitrogen atom of the heterocyclic ring via an amide bond. A person of ordinary skill in the chemistry of amino acids and oligopeptides will readily appreciate that certain amino acids may be replaced by other homologous, isosteric and/or isolectronic amino acids wherein the biological activity of the original amino acid or oligopeptide has been conserved upon modification. Certain unnatural and modified natural amino acids may also be utilized to replace the corresponding natural amino acid. Thus, for example, tyrosine may be replaced by 3-iodotyrosine, 2- or 3-methyltyrosine, 3- fluorotyrosine.
"C3-C8-Cycloalkyl" generally represents cyclic hydrocarbon radical having 3 to 8 carbon atoms which may optionally be substituted by one or several hydroxyl, amino, CrCβ-alkyl- amino, di- Q-Cό-alkyl-amino, Ci-Cβ-alkyl, Q-Cό-alkyloxy, thiol, Q-Cβ-alkyl-thio, =O, =NH, -CHO, -COOH, -COOCH3, -COOCH2CH3, -CONH2, -NHC(=NH)NH2, or halogen substituents , which may be identical to one another or different.
"Heterocyclic ring" as used hereinabove and hereinbelow with respect to the group formed by Ra and R together with the interjacent N-C group generally represents a 3 to 7-membered, preferably 4-, 5- or 6-membered non-aromatic heterocyclic ring system, containing one nitrogen atom and optionally 1 or 2 additional heteroatoms selected from the group of nitrogen, oxygen and sulfur, which may be substituted by one or several halogen atoms or Q- Cβ-alkyl, Q-Ce-alkylamino, di-CrCe-alkylamino, Q-Cts-alkoxy, thiol, C!-C6-alkylthio, oxo, imino, fomyl, Cϊ-C6-alkoxy carbonyl, amino carbonyl, C3-C8-cycloalkyl, aryl, or heteroaryl groups, which may be identical to one another or different, and which optionally may be benzo- or cyclohexano-condensed. Such heterocyclic rings are preferably azetidine or are derived from a fully or partially hydrogenated pyrrole, pyridine, thiazole, isoxazole, pyrazole, imidazole, indole, benzi idazole, indazole, pyridazine, pyrimidine, pyrazin group. Most preferred are azetidine, pyrrolidine, 3,4-dehydropyrrolidine, piperidine, hexahydro-lH- azepine, octahydroindole, imidazolidine, thiazolidine.
If such heterocyclic ring is substituted, the substituents are preferably methyl, ethyl, propyl, 1- methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dimethyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, -COOCH3, -COOCH2CH3, or -CONH2. "Aryl" generally represents an aromatic ring system with 6 to 10, preferably 6 carbon atoms which may optionally be substituted by one or several hydroxyl, amino, d-C6-alkyl-amino, di- d-Ce-alkyl-amino, d-C6-alkyl, C C6-alkyloxy, thiol, d-C6-alkyl-thio, -CHO, -COOH, - COOCH3, -COOCH2CH3, -CONH2, or halogen substituents, which may be identical to one another or different, and which optionally may be benzocondensed. Aryl substituents may be preferably derived from benzene, preferred examples being phenyl, 2-hydroxy-phenyl, 3- hydroxy-phenyl, 4-hydroxy-phenyl, 4-amino-phenyl, 2-amino-ρhenyl, 3-amino-phenyl.
If aryl is substituted, the substituents are preferably methyl, ethyl, propyl, 1 -methylethyl (iso- propyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dime- thyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, -COOCH3, - COOCH2CH3, or -CONH2.
"Heteroaryl" generally represents a 5 to 10-membered aromatic heterocyclic ring system, containing 1 to 5 heteroatoms selected from the group of nitrogen, oxygen, or sulfur, which may optionally be substituted by one or several hydroxyl, amino, d-C6-alkyl-amino, di- d- Ce-alkyl-amino, C C6-alkyl, d-C6-alkyloxy, thiol, Cι-C6-alkyl-thio, -CHO, -COOH, - COOCH3, -COOCH2CH3, -CONH2, or halogen substituents, which may be identical to one another or different, and which optionally may be benzocondensed. Heteroaryl substituents may preferably be derived from furane, pyrrole, thiophene, pyridine, thiazole, isoxazole, pyrazole, imidazole, benzofuran, thianaphthene, indole, benzimidazole, indazole, quinoline, pyridazine, pyrimidine, pyrazine, chinazoline, pyrane, purine, adenine, guanine, thymine, cytosine, uracil.
If heteroaryl is substituted, the substituents are preferably methyl, ethyl, propyl, 1 -methylethyl (isopropyl), butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, hydroxyl, amino, dimethyl-amino, diethyl-amino, thiol, methyl-thiol, methoxy, ethoxy, -CHO, -COOH, - COOCH3, -COOCH2CH3, or -CONH2.
"Residue of a cytotoxic or cytostatic compound" means that the compound H2N-Cyt', which is released upon cleavage of the amide bond shown in formula (I), is either cytotoxic or cytostatic itself, or may be converted into a cytotoxic or cytostatic compound in a subsequent step. In the latter case, -Cyt' may be a residue of formula -L-Cyt", wherein L is a linker residue derived from a bifunctional molecule, for instance a diamine H2N-L'-NH2, an amino alcohol H2N-L'-OH, for example p-amino-benzyl alcohol (PABOH), an amino carbonate, for example
or an unnatural amino carboxylic acid. If -Cyt' is of formula -L-Cyt", the compound H2N-L'- Cyt' ' is generated by the enzymatic cleavage of the amide bond shown in formula (I). The compound H2N-L'-Cyt" may be cytotoxic or cytostatic itself or the linker residue cleaved off from Cyt' ' in a subsequent step releasing the cytotoxic or cytostatic agent. For example, the compound H2N-L'-Cyt" may be hydrolysed under physiological conditions into a compound H2N-L'-OH and the cytotoxic or cytostatic compound H-Cyt" , which is the active therapeutic agent (In the following, only the term Cyt' is used for both Cyt' and Cyt", and only the term L is used for both L and L', for simplicity).
The pharmaceutically acceptable salts of the compounds of the present invention include the conventional non-toxic salts formed from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those from inorganic acids such as hydrochloric acid, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, maleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, oxalictrifluoroacetic and the like.
H2N-Cyt' is preferably an anthracycline derivative of formula IN,
wherein
Rc represents d-C6 alkyl, d-C6 hydroxyalkyl or d-C6 alkanoyloxy Cι-C6 alkyl, in particular methyl, hydroxymethyl, diethoxyacetoxymethyl or butyryloxymethyl;
Rd represents hydrogen, hydroxy or d-C6 alkoxy, in particular methoxy; one of Re and Rf represents a hydrogen atom; and the other represents a hydrogen atom or a hydroxy or tetrahydropyran-2-yloxy (OTHP) group.
Paricularly preferred are the following compounds of formula II:
Rc Rα Re R1 Cyt
CH2OH OCH3 H OH doxorubicin
CH3 OCH3 H OH daunorubicin
CH2OH OCH3 OH H epirubicin
CH3 H H OH idarubicin
CH2OH OCH3 H OTHP THP
CH2OH OCH3 H H esorubicin
CH2OCOCH(OC2H5)2 OCH3 H OH detorubicin CH2OH H H OH carminorubicin
CH2OCOC4H9 OCH3 H OH
Most preferred is doxorubicin (Dox). Other cytotoxic or cytostatic residues Cyt' may be derived for example from methotrexate, trimetrexate, pyritrexim, 5,10- dideazatetrahydrofolatepyrimetamine, trimethoprim, 10-propargyl-5,8-dideazafolate2,4- diamino-5(3 ' ,4' -dichloropheyl)-6-methylpyrimidine, aminoglutethimide, goreserelin, melphalan, chlorambucil, analogs of other chemotherapeutic agents such as 9- aminocamptothecin (for examples see e.g. Burris HA, r. d. and S. M. Fields (1994). "Topoisomerase I inhibitors. An overview of the camptothecin analogs. [Review]." Hematol. Oncol Clin. North Am. 8(2): 333-355; Iyer, L. and M. J. Ratain (1998). "Clinical pharmacology of camptothecins. [Review] [137 refs]." Cancer Chemother. Pharmacol. 42 Suppl: S31-S43.) and epothilone and the derivatives thereof.
In formula (I), Cyt' may also be a biological effector molecule which either directly or indirectly effects destruction of tumor cells, like for example TNFα.
Preferred anthracycline prodrugs are the compounds of formula HI wherein Ra, Rb, Rc, Rd, Re, Rf and R1 are as defined hereinabove.
Most preferred compounds of the invention are doxorubicin derivatives of formulae (IIIA) to
(πiK):
If the part Cg-B-A or Cg-(D)m-B-A of formula (I) contains two or more sulfur atoms, the compound of the invention may contain one or more disulfide bonds.
One class of cytotoxic or cytostatic compounds which may be used for the present invention has a primary amino function which is available for formation of an amidic bond as shown in formula (I), like doxorubicin. In this case, a linker molecule L is not necessary. If a cytostatic or cytotoxic compound does not have such an amino function, such a function may be created in such a compound by way of chemical modification, e.g. by introducing or converting a functional group or attaching a linker molecule to the compound. A linker molecule may also be inserted between the oligomeric part (i.e. the part comprising the amino carboxylic residues) and the cytostatic or cytotoxic part of the compound of the invention to ensure or optimize cleavage of the amide bond between the oligomeric part and the cytotoxic or cytostatic part. If a linker molecule is present, i.e. in compounds containing the structure L- Cyt', the bond between L and Cyt' is preferably an amidic or ester bond. In a preferred embodiment, such a linker molecule is hydrolyzed off the cytostatic or cytotoxic compound under physiological conditions after the enzymatic cleavage and thus the free cytostatic or cytotoxic compound is generated. In any case, the compound of the invention must have the property of being cleavable upon the catalytic action of FAPα and, as a direct or indirect consequence of this cleavage, releasing under physiological conditions a cytostatic or cyto- toxic compound.
In a further aspect, the present invention relates to prodrug that is capable of being converted into a cytotoxic or cytostatic drug, by the catalytic action of FAPα, said prodrug exhibits an oligomeric part comprising up to 13 amino carboxylic residues, the C-terminal amino carboxylic thereof is recognized by FAPα, and a cytotoxic or cytostatic part, characterized in that the N-terminal amino function of the oligomeric part is attached to a capping group (Cg) of formula π.
The oligomeric part is preferably a peptide. Preferably, the oligomeric part comprises two, three, four, five, six, seven, eight, nine, ten, eleven, or twelve amino carboxylic acid residues, more preferably two, three, or four amino carboxylic residues.
The compounds of the invention may be synthesized by processes known in the art (E. Wϋnsch, Synthese von Peptiden, in "Methoden der organischen Chemie", Houben-Weyl (Eds. E. Mϋller, O. Bayer), Vol. XV, Part 1 and 2, Georg Thieme Nerlag, Stuttgart, 1974). For example, the compounds could be synthesized by condensation of the terminal amino function of the oligomeric part of a compound of formula NI with a compound of formula N,
wherein X1, X2, s and t have the meaning given hereinabove, and LG1 is OH or an activation leaving group, and PG1 is H or a suitable protection group according to the following reaction scheme:
wherein Cyt', Ra, Rb, R3 and R4 have the meaning given hereinabove, and Rla represents an amino alkanoyl or oUgopeptidoyl group, the N-terminal amino function of which bears at least one hydrogen atom. To achieve such an amide formation, it may be necessary to activate the carbonyl group of the carboxylic acid for a nucleophilic attack of an amine, i.e. LG1 to be an activation group or leaving group which is suitable to be substituted by an amino group. This activation can be done by conversion of the carboxylic acid into an acid chloride or acid fluoride or by conversion of the carboxylic acid into an activated ester, for instance a N- hydroxysuccinimidyl ester or a pentafluorophenyl ester. Another method of activation is the transformation into a symmetrical or unsymmetrical anhydride. Alternatively, the formation of the amide bonds can be achieved by the use of in situ coupling reagents like benzotriazole- 1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate (PyBOP) (E. Frerot et al., Tetrahedron, 1991, 47, 259-70), U'-carbonyldimidazole (GDI) (K. Akaji et al, THL, 35, 1994, 3315-18), 2-(lH-benzotriazole-l-yl)-l,l,3,3-tetramethyluronium tetrafluoroborate (TBTU) (R. Knorr et al., THL, 30, 1989, 1927-30), l-(mesitylene-2-sulfonyl)-3-nitro-lH- 1,2,4-triazole (MSNT) (B. Blankenmeyer-Menge et al., THL, 31, 1990, 1701-04).
The protection group PG1 is removed at the end of the synthesis.
The compounds of formula VI can be prepared as described in WO 00/71571, the complete disclosure of which is hereby incorporated by reference.
The compounds of the invention are intended for medical use. In particular, these compounds are useful for the treatment of tumors which are associated with stromal fibroblasts that express FAPα and which are generally not optimally treated with available cytotoxic and/or cytostatic agents. Tumors with this property are, for example, epithelial cancers, such as lung, breast, and colon carcinomas. Tumors, such as bone and soft tissue sarcomas which express FAPα, may also be treated with these compounds.
Consequently, another aspect of the present invention are pharmaceutical compositions comprising a compound of the present invention and optionally one or more suitable and pharmaceutically acceptable excipients, as exemplified in: Remington: the science and practice of pharmacy. 19th ed. Easton : Mack Publ, 1995. The pharmaceutical compositions may be formulated as solids or solutions. Solid formulations may be for preparation of a solution before injection. Preferably, the pharmaceutical compositions of the invention are solutions for injection. They may be administered systemically, e.g. by intravenous injection, or topically, e.g. by direct injection into the tumor site. The dosage will be adjusted according to factors like body weight and health status of the patient, nature of the underlying disease, therapeutic window of the compound to be applied, solubility, and the like. It is within the knowledge of the expert to adjust dosage appropriately. For doxorubicin conjugates, for example, the dose will preferably be in the range from 10 mg/m2 to 2000 mg/m2, but also higher or lower doses may be appropriate.
Accordingly, a further aspect of the present invention is the use of a compound of the invention in the preparation of a pharmaceutical composition for the treatment of cancer. Furthermore, an aspect of the invention is a method of treatment of cancer, comprising administering an effective amount of a pharmaceutical composition of the invention to a patient. Indications include the treatment of cancer, specifically:
1) The treatment of epithelial carcinomas including breast, lung, colorectal, head and neck, pancreatic, ovarian, bladder, gastric, skin, endometrial, ovarian, testicular, esophageal, prostatic and renal origin;
2) Bone and soft-tissue sarcomas: Osteosarcoma, chondrosarcoma, fibrosarcoma, malignant fibrous histiocytoma (MFH), leiomyosarcoma;
3) Hematopoietic malignancies: Hodgkin's and non-Hodgkin's lymphomas; 4) Neuroectodermal tumors: Peripheral nerve tumors, astrocytomas, melanomas; 5) Mesotheliomas.
Also included are the treatment of chronic inflammatory conditions such as rheumatoid arthritis, osteoarthritis, liver cirrhosis, lung fibrosis, arteriosclerosis, and abnormal wound healing.
A further aspect of the invention is a method of treatment of cancer, wherein a prodrug is administered to a patient wherein said prodrug is capable of being converted into a cytotoxic or cytostatic drug by an enzymatic activity, said enzymatic activity being the expression product of cells associated with tumor tissue. Preferably, said enzymatic activity is the proteolytic activity of FAPα.
One method of administration of the compounds is intravenous infusion. Other possible routes of administration include intraperitoneal (either as a bolus or infusion), intramuscular or intratumoral injection. Where appropriate, direct application may also be possible (for example, lung fibrosis).
One skilled in the art will appreciate that although specific reagents and reaction conditions are outlined in the following examples, modifications can be made which are meant to be encompassed by the scope of the invention. The following examples, therefore, are intended to further illustrate the invention and are not limiting.
Example I Synthesis of (4-Carboxymethyl-phenyl)-acetic acid -hydroxysuccinimidyl ester
1,4-phenylendiacetic acid (4.8 g, 25 mmol) was dissolved in acetonitrile (100 ml) and N,N- dimethylformamide (100 ml) The solution was cooled to 0 °C and N-hydroxysuccinimide (2,9 g, 25 mmol) and diisopropylethylamine (8.6 ml, 50 mmol) were added. The solution was stirred for 30 min at 0 °C. Then a solution of dicyclohexyl carbodiimide(5.2 g, 25 mmol) dissolved in acetonitrile (50 ml) was added dropwise over 1 h. The ice bath was removed an the suspension was stirred over night. The suspension was filtered and the solvent of the filtrate was concentrated under reduced pressure. A residual amount of dicyclohexyl urea was precipitated by addition of diethyl ether and removed by filtration. The filtrate was purified by reversed phase HPLC applying acetonitrile/water gradient. The product fraction was lyophilized to give the product as white cristalls (1.1 g, 16 %). The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [M+H]+= 292
(3-Carboxymethyl-phenyl)-acetic acid N-hydroxysuccinimidyl ester was prepared analogously.
Example 2 [2-(4-Carboxymethyl-ρhenyl)-acetyl]-Pro-Ala-Gly-Pro-Dox (III-G)
All steps were performed under nitrogen atmosphere.
2A Aloe-Pro- Ala-Gl v-Pro-Dox : Aloc-Pro-Ala-Gly-Pro-OH (190 mg 0.45 mmol) and Doxorubicin hydrochloride (260 mg 0.49 mmol) were dissolved in N,N-dimethylformamide (5 ml). Diisopropylethylamine (153 μl 0.90 mmol) was added and the solution was cooled to 0 °C. To the stirred mixture a solution of O-(7-azabenzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexfluorophosphate (HATU) (221mg 0.582 mmol) in N,N-dimethylformamide (10 ml) was added dropwise over a periode of 30 min. The solution was stirred for 1 h at 0 °C. After addition of diisopropylethylamine (25 μl, 0.14 mmol) the solvent was removed in vacuo at 30 °C. The crude product was dissolved in methanol (1 ml) and water (6 ml) and purified by reversed phase HPLC applying acetonitrile/water gradient. The product fraction was lyophilized and gave red cristalls of Aloe-Pro- Ala-Gly-Pro-Dox (345 mg, 81 %). The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [M+Νa]+= 972
2B H-Pro-Ala-Glv-Pro-Dox:
Aloc-Pro-Ala-Gly-Pro-Dox (343 mg 0.36 mmol) was dissolved dichloromethane (20 ml) and diethylamine (185 μl, 1.81 mmol) and Pd(Ph3P)4 (20.8 mg 0.02 mmol) were added. The solution was stirred at room temperature for 1 h. The solvent was removed under reduced pressure and the crude oil was dissolved water (3 ml) and acetonitrile (1 ml). The product was purified by reversed phase HPLC applying acetonitrile/water gradient and the product fraction was lyophilized to give red cristalls of H-Pro-Ala-Gly-Pro-Dox (296 mg, 95 %). The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [m+H]+= 866
2C r2-(4-Carboxymethyl-phenylVacetyl]-Pro-Ala-Glv-Pro-Dox fiπ-G):
H-Pro-Ala-Gly-Pro-Dox (100 mg 0.12 mmol) was dissolved in in N,N-dimethylformamide (5 ml). (4-Carboxymethyl-phenyl)-acetic acid N-hydroxysuccinimidyl ester (36.8 mg 0.127 mmol) and diisopropylethylamine (22 μl 0.127 mmol) were added. The solution was stirred for 48 h at room temperature. The solvent was removed under reduced pressure at 30 °C. The crude product was purified by reversed phase HPLC applying acetonitrile/water gradient and the product fraction was concentrated by lyophilization to yield the product (77 mg, 64 %). The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [M+H]+= 1043
In analogy to this method the following- compounds were prepared: [2-(4-Carboxymethyl-phenyl)-acetyl]-N-MeIle-Ala-Gly-Pro-Dox (iπ-E) [2-(4-Carboxymethyl-phenyl)-acetyl]-N-MeLeu-Ala-Gly-Pro-Dox (πi-A) [2-(4-Carboxymethyl-phenyl)-acetyl]-N-MeVal-Ala-Gly-Pro-Dox (III-F) [2-(4-Carboxymethyl-phenyl)-acetyl]-Aze-Ala-Gly-Pro-Dox (πi-H) [2-(3-Carboxymethyl-ρhenyl)-acetyl]-Pro-Ala-Gly-Pro-Dox (III-K) [2-(3-Carboxymethyl-ρhenyl)-acetyl]-N-MeIle-Ala-Gly-Pro-Dox (iπ-B) [2-(3-Carboxymethyl-phenyl)-acetyl]-N-MeLeu-Ala-Gly-Pro-Dox (iπ-C) [2-(3-Carboxymethyl-phenyl)-acetyl]-N-MeVal-Ala-Gly-Pro-Dox (m-D)
Assay for cleavage ofMNA substrates by FAP:
Buffer A: lOOmM Tris HC1 pH 7.8, 100 mM NaCl
Cell extract from 293 cells stably transfected with FAP prepared as described (see Park, et al., Fibroblast Activation Protein, a Dual Specificity Serine Protease expressed in human tumor stromal fibroblasts. (1999) J. Biol. Chem. 36505-12.). A similar extract was also prepared from parental 293 control cells without FAP. The FAP concentration in the FAP-transfected cell extract was estimated by immunoassay and 1 ng enzyme (diluted in buffer A) was used per assay. FAP-negative 293 control cell extract was used at the same dilution (also in buffer A) as a negative control. Substrate was initially dissolved in dimethylformamide at a concentration of 200 mM and diluted in buffer A to a final concentration of 2.5 mM. A few substrates were not soluble at this concentration and had to be diluted further.
Assay conditions:
10 μl 10% DMSO in buffer A
70 μl diluted FAP cell extract containing 1 ng FAP enzyme (OR control 293 cell extract without FAP)
20 μl 2.5 mM substrate
Mix, incubate at room temperature for 1 hour, and measure fluorescence in Fluorostar fluorimeter at the following wavelengths:
MNA conjugates: Excitation: 355 nm, Emission: 405 nm. The fluorescence measured in the samples treated with control 293 control cell extracts without FAP is subtracted from the values measured in the samples treated with 1 ng FAP enzyme.
e a le 3
Synthesis of [2-(4-Carboxymethyl-phenyl)-acetyl]-Pro-Ala-Gly-Pro-MNA
3A Boc-Pro-Ala-Glv-OH:
H-Gly-2-chlorotrityl-resin (10 g, 11.1 mmol) were added to a reaction vessel and washed with N,N-dimethylformamide (DMF) (three times with 150 ml). Fmoc-Ala-OH (20.7 g, 66.6 mmol), O-(benzotriazol-l-yl)-N,N,N,N-tetramethyluronium tetrafluoroborate (TBTU) (21.4 g, 66.6 mmol), 1-hydroxybenzotriazole (HOBt) (9.06 g, 66.6 mmol) and diisopropylethylamine (8 eq.) dissolved in DMF (100 ml) were added. The suspension was shaken at room temperature over night. The resin was washed carefully four times with DMF, dichloromethane, and DMF. The cleavage of the Fmoc-group was performed with a solution of 30% piperidine in DMF for 30 minutes followed by careful washing steps with DMF (4 times), dichloromethane (3 times) and DMF (4 times). Boc-Pro-OH was incorporated in the same manner. After cleavage of the Fmoc group as described above the resin was finally washed with methanol and diethyl ether and dried in a stream of nitrogen. The cleavage of the product was performed with a solution of 30 % l,l,l,3,3,3-hexafluoro-2-propanol in dichloromethane for 30 minutes at room temperature. The filtrate was concentrated in vacuo to yield the peptide which was pure enough for further reaction. The product gave satisfactory analytical data. HPLC > 95 %; ES-MS : [M+H]+ = 344.2
3B H-Pro-Ala-Glv-Pro-MΝA:
Boc-Pro-Ala-Gly-OH (3.3 g, 9.6 mmol), H-Pro-MΝA (3.2 g, 10.6 mmol), TBTU (3.1 g, 9.6 mmol), HOBt (1.3 g, 9.6 mmol) and diisopropylethylamine (3 eq.) were dissolved in DMF (20 ml) and stirred at room temperature for 4 hours. The solvent was removed under vacuum and the residual oil was dissolved with trifluoroacetic acid water 95:5 (10 ml) for 3 h. The solvent was removed under vacuum and the crude product was purified by reversed phase HPLC applying acetonitrile/water gradient. The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [M+H = 496.6
3C 4-Carboxymethylphenylacetyl-Pro-Ala-Gly-Pro-MNA:
A solution of 1,4-phenylendiacetic acid (16 mg, 80 μmol), H-Pro-Ala-Gly-Pro-MNA (40 mg, 80 μmol), hydroxybenzotriazol (11 mg, 80 μmol), O-(Benzotriazol-l-yl)-N,N,N,N- tetramethyluronium tetrafluoroborate (TBTU) (26 mg, 80 μmol ) and DIPEA (30 μl, 160 μmol) in N,N-dimethylformamide (4 ml) was shaken overnight at room temperature. The solution was concentrated to dryness under reduced pressure and the residue was purified via reversed phase HPLC applying acetonitrile/water gradient. The product containing fractions were lyophyhzed to yield 26 mg (39 μmol, 48 %) of the desired product as a white powder. The product gave satisfactory analytical data. HPLC > 95 %; ES-MS: [M+H]+= 672.7
The following table shows the peptide-MΝA-conjugates which have been prepared analogously and includes cleavage data by FAP.
MΝA-Conjugate Cleavage
[μM]
[2-(4-Carboxymethyl-phenyl)-acetyl]-Ν-MeIle-Ala-Gly-Pro-MΝA 16.36
[2-(4-Carboxymethyl-phenyl)-acetyl]-N-MeLeu-Ala-Gly-Pro-MNA 13.97
[2-(4-Carboxymethyl-phenyl)-acetyl]-N-MeVal-Ala-Gly-Pro-MNA 13.39
[2-(4-Carboxymethyl-phenyl)-acetyl]-Aze-Ala-Gly-Pro-MNA 9.54
[2-(3-Carboxymethyl-phenyl)-acetyl]-Pro-Ala-Gly-Pro-MNA 2.80
[2-(3-Carboxymethyl-phenyl)-acetyl]-N-MeIle-Ala-Gly-Pro-MNA 15.36
[2-(3-Carboxymethyl-phenyl)-acetyl]-N-MeLeu-Ala-Gly-Pro-MNA 12.00
[2-(3-Carboxymethyl-phenyl)-acetyl]-N-MeVal-Ala-Gly-Pro-MNA 9.52
[2-(4-Carboxymethyl-phenyl)-acetyl]-Pro-Ala-Gly-Pro-MNA 7.25
[2-(2-Carboxymethyl-phenyl)-acetyl]-Pro-Ala-Gly-Pro-MNA 0.94
Solubility Test
The solubilities of the compounds of formula
in a phosphate buffer have been compared.
It has been found that the solubility of A is > 10 mg/ml, whereas the solubility of B is < 2 mg/ml.

Claims

Claims
1. A compound of formula (I)
or a pharmaceutically acceptable salt thereof, wherein
R1 represents an amino alkanoyl or oUgopeptidoyl group, the N-terminal amino function of which is attached to a capping group (Cg) of formula (II)
in which
X1 represents C=O or SO2,
X2 represents C=O, SO2, NH-C=O or a single bond, s is an integer of 1 or 2, and t is 0 or an integer of 1, 2 or 3, Ra and Rb together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH2 groups may also be replaced by NH, O or S; R3 represents H, d-Cδ-alkyl, C3-C8-cycloalkyl, aryl or heteroaryl; and Cyt' represents the residue of a cytotoxic or cytostatic compound.
2. A compound of formula (I) according to claim 1, wherein X1 represents C=O or SO2, X2 represents C=O or SO2, s is an integer of 1 or 2, and t is an integer of 1 or 2.
3. A compound of formula (I) according to claim 1 or 2, wherein X1 and X2 represent C=O, s and t are 1.
4. A compound of formula (I) according to any of claims 1 to 3, wherein
R1 represents a residue of formula Cg-A, Cg-B-A or Cg-(D)n-B-A, in which
Cg represents a capping group of formula (II),
A, B and D each independently represent moieties derived from amino carboxylic acids of the formula -[NR -(X)p-CO]- wherein X represents CR5R6 and wherein R4, R5 and R6 each independently represent a hydrogen atom, an optionally substituted d-C6- alkyl, C3-C8-cycloalkyl, aryl, aralkyl, heteroaryl or heteroarylalkyl group, and p is 1, 2, 3, 4, 5; or
A, B and D each independently represent moieties derived from cyclic amino carboxylic acids of formula
wherein
R7 represents d-C6-alkyl, OH, or NH2, n is an integer from 1 to 10; q is 0, 1 or 2; and r is 0, 1 or 2.
5. A compound of formula I according to any of claims 1 to 4, wherein the heterocyclic ring formed by Ra, Rb and the interjacent N-C is substituted by R8 and R9, wherein R8 and R9 each independently represent a hydrogen or halogen atom or a Cl-C6-alkyl, C1-C6- alkylamino, di-Cl-C6-alkylamino, Cl-C6-alkoxy, thiol, Cl-C6-alkylthio, oxo, imino, fomyl, Cl-C6-alkoxy carbonyl, amino carbonyl, C3-C8-cycloalkyl, aryl, or heteroaryl group.
6. A compound of formula I A
wherein R .11, R Rs, Cyt' are as defined in any of the preceding claims, and X-Y represents CHR9-CH2, CR9=CH, NH-CH2, CH2-NH, -CR9-, CH2-CHR9-CH2.
7. A compound of formula I A 1
wherein R3, R4, R5, Cyt', Cg, X and Y are as defined in any of the preceding claims, or R4 and R5 together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring, in which one or two CH2 groups may also be replaced by NH, O or S.
8. A compound of formula IA2
wherein R , R , R , Cyt', Cg, X and Y each independently are as defined in any of the preceding claims, or
R4 and R5 together with the interjacent N-C group form an optionally substituted, optionally benzo- or cyclohexano-condensed 3- to 7-membered saturated or unsaturated heterocyclic ring.
9. A compound according to any of the preceding claim, wherein
R1 represents a group selected from Cg-A, Cg-B-A- and Cg-(D)n-B-A- in which A, B and D are amino acid moieties, which are each independently selected from glycine (Gly), and the D- or L-forms of alanine (Ala), valine (Val), leucine (Leu), isoleucine (He), phenylalanine (Phe), tyrosine (Tyr), tryptophan (Trp), cysteine (Cys), methionine (Met), serine (Ser), threonine (Thr), lysine (Lys), arginine (Arg), histidine
(His), aspartatic acid (Asp), glutamic acid (Glu), asparagine (Asn), glutamine (Gin), proline (Pro), 4-hydroxy-proline (Hyp), 5-hydroxy-lysine, norleucine (Nle), 5- hydroxynorleucine (Hyn), 6-hydroxynorleucine, ornithine, cyclohexylglycine (Chg), N- Methylglycin (N-MeGly), N-Methylalanin (N-MeAla), N-Methylvaline (N-MeVal), N- Methylleucine (N-MeLeu), N-Methylisoleucine (N-Melle), N-Methylnorleucin (N- MeNle), N-Methyl-2-aminobutyric acid (N-MeAbu) and N-Methyl-2-aminopentanoic acid (N-MeNva).
10. A compound of formula I according to any of the preceding claims, wherein the unit A is selected from L-proline, glycine, L-norleucine, L-cyclohexylglycine, L-5- hydroxynorleucine, L-6-hydroxynorleucine, L-5-hydroxylysine, L-arginine, and L-lysine.
11. A compound of formula I according to any of the preceding claims, wherein the unit D is selected from L-proline, (Pro), N-Methylglycin (N-MeGly), N-Methylalanin (N-MeAla), N-Methylvaline (N-MeNal), Ν-Methylleucine (Ν-MeLeu), Ν-Methylisoleucine (Ν- Melle), Ν-Methylnorleucin (Ν-MeΝle), Ν-Methyl-2-aminobutyric acid (N-MeAbu) and N-Methyl-2-aminopentanoic acid (N-MeNva).
12. A compound according to any of the preceding claims wherein Rl is a group selected from the formulae (1) to (14):
Cg-Gly (1)
Cg-Nle (2)
Cg-Nal (3)
Cg-Met (4)
Cg-Xxx-Gly (5)
Cg-Xxx-Hyn (6)
Cg-Xxx-Pro (7)
Cg-Xxx-His (8)
Cg-Xxx-Met (9)
Cg-Xxx-Ala (10)
Cg-Xxx-Hyn (11)
Cg-Xxx-Ala-Gly (12)
Cg-(Xxx)n-Xxx-Gly (13)
Cg-(Xxx)n-Xxx-Ala-Gly (14)
wherein Cg represents a capping group of formula II;
Xxx represents a moiety derived from an amino carboxylic acid; and n is an integer from 1 to 6.
13. A compound according to claim 12 wherein the amino alkanoic acid moieties exist in the (L)-configuration
14. A compound of any one of claims 1 to 13, wherein Cyt' is an anthracyclme group.
15. A compound of claim 14 selected from the formulae (IH-A) to (III-K):
16. A prodrug that is capable of being converted into a cytotoxic or cytostatic drug, by the catalytic action of FAPα, said prodrug exhibits an oligomeric part comprising up to 13 amino carboxylic residues, the C-terminal amino carboxylic thereof is recognised by FAPα, and a cytotoxic or cytostatic part, characterized in that the N-terminal amino function of the oligomeric part is attached to a capping group
(Cg) of formula (II) .
in which
X1 represents C=O or SO2,
X2 represents C=O, SO2, NH-C=O or a single bond, s is an integer of 1 or 2, and t is 0 or an integer of 1, 2 or 3.
17. The prodrug of claim 16, wherein the C-terminal amino carboxilic residue is selected from D-proline, L-proline, D-hydroxyproline and L-hydroxyproline and the oligomeric part comprises two, three, or four amino carboxylic acid residues.
18. A compound of any one of the preceding claims for medical use.
19. Pharmaceutical composition comprising a compound according to any one of claims 1 to 18, and optionally one or more pharmaceutically acceptable excipients.
20. Use of a compound according to any one of claims 1 to 18 in the preparation of a pharmaceutical composition for the treatment of cancer.
21. Use of a prodrug according to claim 16 or 17 for the manufacture of a stable medicament for the treatment of cancer.
EP03734699A 2002-01-30 2003-01-28 Fap-activated anti-tumor compounds Withdrawn EP1472277A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP03734699A EP1472277A1 (en) 2002-01-30 2003-01-28 Fap-activated anti-tumor compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP02002222A EP1333033A1 (en) 2002-01-30 2002-01-30 FAP-activated anti-tumor compounds
EP02002222 2002-01-30
EP03734699A EP1472277A1 (en) 2002-01-30 2003-01-28 Fap-activated anti-tumor compounds
PCT/EP2003/000815 WO2003064454A1 (en) 2002-01-30 2003-01-28 Fap-activated anti-tumor compounds

Publications (1)

Publication Number Publication Date
EP1472277A1 true EP1472277A1 (en) 2004-11-03

Family

ID=8185379

Family Applications (2)

Application Number Title Priority Date Filing Date
EP02002222A Withdrawn EP1333033A1 (en) 2002-01-30 2002-01-30 FAP-activated anti-tumor compounds
EP03734699A Withdrawn EP1472277A1 (en) 2002-01-30 2003-01-28 Fap-activated anti-tumor compounds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP02002222A Withdrawn EP1333033A1 (en) 2002-01-30 2002-01-30 FAP-activated anti-tumor compounds

Country Status (4)

Country Link
EP (2) EP1333033A1 (en)
JP (1) JP2005523264A (en)
CA (1) CA2472296A1 (en)
WO (1) WO2003064454A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7407955B2 (en) 2002-08-21 2008-08-05 Boehringer Ingelheim Pharma Gmbh & Co., Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
US7501426B2 (en) 2004-02-18 2009-03-10 Boehringer Ingelheim International Gmbh 8-[3-amino-piperidin-1-yl]-xanthines, their preparation and their use as pharmaceutical compositions
DE102004054054A1 (en) 2004-11-05 2006-05-11 Boehringer Ingelheim Pharma Gmbh & Co. Kg Process for preparing chiral 8- (3-amino-piperidin-1-yl) -xanthines
DE102005035891A1 (en) 2005-07-30 2007-02-08 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8- (3-amino-piperidin-1-yl) -xanthines, their preparation and their use as pharmaceuticals
EP2540725A1 (en) 2006-05-04 2013-01-02 Boehringer Ingelheim International GmbH Polymorphs of 1-((4-Methyl-chinazolin-2-yl)methyl)-3-methyl-7-(2-butin-1-yl)-8-(3-(R)-amino-piperidin-1-yl)xanthin
EP1852108A1 (en) 2006-05-04 2007-11-07 Boehringer Ingelheim Pharma GmbH & Co.KG DPP IV inhibitor formulations
PE20080251A1 (en) 2006-05-04 2008-04-25 Boehringer Ingelheim Int USES OF DPP IV INHIBITORS
EP3542801A1 (en) 2007-08-17 2019-09-25 Boehringer Ingelheim International GmbH Purin derivatives for use in the treatment of fap-related diseases
PE20140960A1 (en) 2008-04-03 2014-08-15 Boehringer Ingelheim Int FORMULATIONS INVOLVING A DPP4 INHIBITOR
KR20190016601A (en) 2008-08-06 2019-02-18 베링거 인겔하임 인터내셔날 게엠베하 Treatment for diabetes in patients inappropriate for metformin therapy
UY32030A (en) 2008-08-06 2010-03-26 Boehringer Ingelheim Int "TREATMENT FOR DIABETES IN INAPPROPRIATE PATIENTS FOR THERAPY WITH METFORMIN"
RU2011113823A (en) 2008-09-10 2012-10-20 БЕРИНГЕР ИНГЕЛЬХАЙМ ИНТЕРНАЦИОНАЛЬ ГмбХ (DE) COMBINED THERAPY FOR THE TREATMENT OF DIABETES AND RELATED CONDITIONS
US20200155558A1 (en) 2018-11-20 2020-05-21 Boehringer Ingelheim International Gmbh Treatment for diabetes in patients with insufficient glycemic control despite therapy with an oral antidiabetic drug
JP2012512848A (en) 2008-12-23 2012-06-07 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Salt forms of organic compounds
TW201036975A (en) 2009-01-07 2010-10-16 Boehringer Ingelheim Int Treatment for diabetes in patients with inadequate glycemic control despite metformin therapy
NZ599298A (en) 2009-11-27 2014-11-28 Boehringer Ingelheim Int Treatment of genotyped diabetic patients with dpp-iv inhibitors such as linagliptin
JP6034781B2 (en) 2010-05-05 2016-11-30 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Combination therapy
EP3725325B1 (en) 2010-06-24 2023-05-31 Boehringer Ingelheim International GmbH Diabetes therapy
US9034883B2 (en) 2010-11-15 2015-05-19 Boehringer Ingelheim International Gmbh Vasoprotective and cardioprotective antidiabetic therapy
DK2731947T3 (en) 2011-07-15 2019-04-23 Boehringer Ingelheim Int SUBSTITUTED DIMERIC QUINAZOLINE DERIVATIVE, PREPARATION AND USE thereof IN PHARMACEUTICAL COMPOSITIONS FOR TREATMENT OF TYPE I AND TYPE II DIABETES
US9555001B2 (en) 2012-03-07 2017-01-31 Boehringer Ingelheim International Gmbh Pharmaceutical composition and uses thereof
EP2849755A1 (en) 2012-05-14 2015-03-25 Boehringer Ingelheim International GmbH A xanthine derivative as dpp -4 inhibitor for use in the treatment of podocytes related disorders and/or nephrotic syndrome
WO2013174767A1 (en) 2012-05-24 2013-11-28 Boehringer Ingelheim International Gmbh A xanthine derivative as dpp -4 inhibitor for use in modifying food intake and regulating food preference
AU2013369261B2 (en) * 2012-12-28 2018-08-09 Cobiores Nv Minimally toxic prodrugs
WO2014169697A1 (en) * 2013-04-19 2014-10-23 暨南大学 Vinblastine derivatives, preparation method therefor and application thereof
JP6615109B2 (en) 2014-02-28 2019-12-04 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Medical use of DPP-4 inhibitors
US9737556B2 (en) 2014-06-13 2017-08-22 Trustees Of Tufts College FAP-activated therapeutic agents, and uses related thereto
CA3022202A1 (en) 2016-06-10 2017-12-14 Boehringer Ingelheim International Gmbh Combinations of linagliptin and metformin

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6613879B1 (en) * 1999-05-14 2003-09-02 Boehringer Ingelheim Pharma Kg FAP-activated anti-tumour compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO03064454A1 *

Also Published As

Publication number Publication date
CA2472296A1 (en) 2003-08-07
EP1333033A1 (en) 2003-08-06
WO2003064454A1 (en) 2003-08-07
JP2005523264A (en) 2005-08-04

Similar Documents

Publication Publication Date Title
WO2003064454A1 (en) Fap-activated anti-tumor compounds
US6613879B1 (en) FAP-activated anti-tumour compounds
US20030055052A1 (en) FAP-activated anti-tumor compounds
US20020155565A1 (en) FAP-activated anti-tumor compounds
WO2000069472A2 (en) Enzyme-activated anti-tumor prodrug compounds
CA2233272A1 (en) Novel peptides
JP2003526683A (en) Peptidase-cleavable targeted antineoplastic drugs and their therapeutic use
AU740597B2 (en) Conjugates useful in the treatment of prostate cancer
US6174858B1 (en) Conjugates useful in the treatment of prostate cancer
EP1317287A2 (en) Polymeric conjugates of antitumor agents
WO2002038590A1 (en) Fap-alpha-activated anti-tumor compounds
KR20010032687A (en) Conjugates useful in the treatment of prostate cancer
US20030232742A1 (en) FAP-activated anti-tumor compounds
US6127333A (en) Conjugates useful in the treatment of prostate cancer
US20030211979A1 (en) FAP-activated anti-tumor compounds
WO2003094972A2 (en) Fap-activated anti-tumor prodrugs
US20040033957A1 (en) FAP-activated anti-tumor prodrugs
US20040081659A1 (en) Conjugates useful in the treatment of prostate cancer
Su et al. Development of novel bone targeting peptide–drug conjugate of 13-aminomethyl-15-thiomatrine for osteoporosis therapy
WO2002038187A1 (en) Fap-activated anti-tumour prodrug
WO2002038591A1 (en) Fap-activated anti-tumor compounds
SK165497A3 (en) Peptides, preparation method thereof and pharmaceutical composition containing the same

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040830

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20060817