WO2023144053A1 - Dérivés hétérocycliques - Google Patents

Dérivés hétérocycliques Download PDF

Info

Publication number
WO2023144053A1
WO2023144053A1 PCT/EP2023/051464 EP2023051464W WO2023144053A1 WO 2023144053 A1 WO2023144053 A1 WO 2023144053A1 EP 2023051464 W EP2023051464 W EP 2023051464W WO 2023144053 A1 WO2023144053 A1 WO 2023144053A1
Authority
WO
WIPO (PCT)
Prior art keywords
denotes
het
atoms
pharmaceutically acceptable
salts
Prior art date
Application number
PCT/EP2023/051464
Other languages
English (en)
Inventor
Timo Heinrich
Sarah SCHLESIGER
Original Assignee
Merck Patent Gmbh
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 Merck Patent Gmbh filed Critical Merck Patent Gmbh
Publication of WO2023144053A1 publication Critical patent/WO2023144053A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the invention had the object of finding novel compounds having valuable properties, in particular those which can be used for the preparation of medicaments.
  • the present invention relates to E3 ligase binding compounds which degrade target proteins, preferably MetAP-2.
  • the compounds of this invention are heterocyclic derivatives and are useful in treating diseases such as of tumours, tumour metastases, proliferative dis- eases of the mesangial cells, haemangioma, proliferative retinopathy, rheumatoid arthritis, atherosclerotic neovascularisation, psoriasis, ocular neovascularisation, osteoporosis, diabetes and obesity, lymphoid leukaemia, lymphoma, malaria and prostate hypertrophy.
  • the present invention also provides methods for preparing these compounds and pharmaceutical compositions comprising these compounds.
  • Small molecule degraders are increasingly utilized as tools to examine the functional roles of proteins and emerged as a novel therapeutic modality. Operating at the post-translational level, these molecules provide the potential for differentiated biological responses in comparison to classical inhibitors and expand the repertoire of methods for protein knock down beyond genetic approaches (e.g.: knock-out, siRNA).
  • Degrader molecules provide an example of a chemical genetic technique capable of more generally targeting the proteome. These chimeric molecules are designed to induce the degradation of their target proteins via the ubiquitin proteasome system (UPS), thereby eliminating pre-existing proteins.
  • UPS ubiquitin proteasome system
  • the UPS is the major intracellular pathway for protein degradation in which a series of enzymes known as E1s (ubiquitin activating enzymes), E2s (ubiquitin conjugating enzymes) and E3s (ubiquitin ligases) carry out covalent linkage of the 9kDa, 76 amino acid protein ubiquitin to a target protein. Subsequent enzymatic reactions result in the formation of a polyubiquitin chain, which targets the protein for degradation by the 26S proteasome.
  • E1s ubiquitin activating enzymes
  • E2s ubiquitin conjugating enzymes
  • E3s ubiquitin ligases
  • Bifunctional degraders comprise an E3 ligase-binding motif that is linked to a target protein binding moiety. Consequently, these molecules hijack the cell’s own degradation machinery by recruiting an E3 ligase in vicinity of the target protein.
  • the spatial proximity enables ubiquitination of the protein and subsequent recognition and depletion by the UPS through the formation of a stable ternary complex.
  • E3 ligase Li W, et al. PLoS One. 2008; 3:e14857
  • the first generation degraders were successfully developed using peptides as an E3 ubiquitin ligase-recognizing motif, they were either not cell-permeable or made cell-permeable by adding a cell-permeating motif such as the TAT peptide (Sakamoto KM, et al Proc Natl Acad Sci U S A. 2001 ; 98:8554-8559; Zhang D, et al. Bioorg Med Chem Lett. 2004; 14:645-648; Schneekloth JS Jr.
  • Androgen receptor (AR) and estrogen receptor (ER) targeting degraders have been developed into clinical candidates, demonstrating the potential applications of these molecules in the treatment of prostate and breast cancers (Rodriguez-Gonzalez A, et al. Oncogene. 2008; 27:7201-7211 ; Cyrus K, et al. Chembiochem. 2010; 11 :1531-1534).
  • reversible MetAP-2 ligands conjugated to E3 ligase recruiting moieties are disclosed that degrade MetAP-2 in a human cancer cell line in a dose- and time-dependent manner.
  • thalidomide (2-(2,6-dioxopiperidine-3-yl)-2,3-dihydro- 1 H-isoindole-1 ,3-dione) and one hydroxyproline-derivative ((2S,4R)-1 -[(2S)-2- amino-3,3-dimethylbutanoyl]-4-hydroxy-N- ⁇ [4-(4-methyl-1 ,3-thiazol-5- yl)phenyl]methyl ⁇ pyrrolidine-2-carboxamide) are used as E3 ligase recognizing moieties.
  • nutlin (4-[(4S,5R)-4,5-bis(4-chlorophenyl)-2-[4-hydroxy-3- (propan-2-yloxy)phenyl]imidazolidine-1 -carbonyl]piperazin-2-one for MDM2) or (2S)-2-[(2S,3R)-3-amino-2-hydroxy-4-phenylbutanamido]-4-methylpentanoic acid (for clAP1 ) are described.
  • the present invention specifically relates to compounds of the formula I which degrade the target protein MetAP-2, to compositions which comprise these compounds, and to processes for the use thereof for the treatment of diseases and complaints.
  • the host or patient can belong to any mammalian species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cows, dogs, cats, etc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease.
  • the susceptibility of a particular cell to treatment with the compounds according to the invention can be determined by in vitro tests.
  • a culture of the cell is combined with a compound according to the invention at various concentrations for a period of time which is sufficient to allow active agents such as anti IgM to induce a cellular response such as expression of a surface marker, usually between about one hour and one week.
  • In vitro testing can be carried out using cultivated cells from blood or from a biopsy sample. The amount of surface marker expressed is assessed by flow cytometry using specific antibodies recognising the marker.
  • the dose varies depending on the specific compound used, the specific disease, the patient status, etc.
  • a therapeutic dose is typically sufficient considerably to reduce the undesired cell population in the target tissue while the viability of the patient is maintained.
  • the treatment is generally continued until a considerable reduction has occurred, for example an at least about 50% reduction in the cell burden, and may be continued until essentially no more undesired cells are detected in the body.
  • Cyclic amides are described as MetAP-2 inhibitors in WO 2012/048775 A1 , WO 2013/149704 A1 and WO 2016/020031 A1.
  • MetAP-2 degraders are described in WO 2020/152067 A1 .
  • the invention relates to compounds of the formula I
  • Q denotes O, NH, or CH2,
  • Q 2 denotes unbranched alkylene having 4-25 C atoms, in which one or more single bonds between C atoms may be replaced by a triple bond, and in which 1-8 non-adjacent CH2 groups may be replaced by 0, CONH and/or NHCO, and in which one CH2 group may be replaced by
  • Y denotes CO or CH2
  • R 1 denotes (CH 2 )n, [C(R 4 ) 2 ]nAr 1 -, (CH 2 ) n Het- (CH 2 )nCyc-,
  • R 2 denotes H, [C(R 4 ) 2 ]nAr 2 , (CH 2 )nCOHet 1 , (CH 2 )nCOAr 2 , (CH 2 )mNA 2 , (CH 2 )nCyc or (CH 2 )nHet 1 ,
  • R 3 denotes OH or OCOA
  • R 4 denotes H or alkyl having 1 , 2, 3 or 4 C-atoms
  • R 2 and R 4 together also denote alkylene having 2, 3, 4 or 5 C-atoms, where a CH2 group may also be replaced by N(CH2)mOH or SO2,
  • R 5 , R 6 each, independently of one another H, F or A,
  • R 5 and R 6 together also denote alkylene having 2, 3, 4 or 5 C-atoms, where a CH2 group may also be replaced by NCOA or 0,
  • R 7 denotes H, Hal or A
  • Ar 1 denotes phenyl which is unsubstituted or mono-, di- tri-, tetra- or pentasubstituted by Hal, OH, OA, CONH 2 , CONHA, CONA 2 , NHSO2A, CONHCyc, NHSO 2 Cyc, CONHAr 2 , Het 1 , COHet 1 and/or NASO2A,
  • Ar 2 denotes phenyl which is unsubstituted or mono-, di-, tri-, tetra- or pentasubstituted by Hal, A, CONH2, and/or OAr 3 ,
  • Het 1 denotes pyridazinyl, pyrazolyl, pyridyl, piperazinyl, morpholinyl, pyrimidinyl, furyl, thienyl, imidazolyl, pyrrolyl, oxazolyl, oxadiazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, thiadiazole, piperidin-1 -yl, pyrrolidin-1 -yl, tetrahydropyranyl, 1 ,2-oxazinan-2-yl, 1 ,2,5-oxadiazinan- 2-yl, 1 ,3-oxazinan-3-yl or hexahydropyrimidinyl, each of which is unsubstituted or mono-, di- or trisubstituted by A and/or OA,
  • Het 2 denotes isoindolyl
  • A denotes unbranched or branched alkyl having 1 -10 C atoms, in which 1 -7 H atoms may be replaced by F, Cl, Br, OH, CHO, COA, COOA, CN, CONA 2 , CONHA and/or CONH 2 , and/or in which one or two non-adjacent CH and/or CH2 groups may be replaced by 0, or Cyc,
  • Aik denotes alkenyl having 2, 3, 4, 5 or 6 C atoms
  • Cyc denotes cyclic alkyl having 3-7 C atoms which is unsubstituted or mono-, di- or trisubstituted by NHCOA, NHSO2, OH, OA, A, NH2, NHA, NA 2 , COOA, COOH and/or CONHA,
  • Hal denotes F, Cl, Br or I
  • m denotes 1 , 2, 3 or 4
  • n denotes 0, 1 , 2, 3 or 4
  • p denotes 1 , 2 or 3
  • enzymes that are known to induce target molecule degradation can be recruited by conjugation of the MetAP-2 binding moiety to respective enzyme-binding chemical structures such as 8-(benzoylsulfanyl)-3- ⁇ [2-(1 ,3- thiazol-2-yl)ethyl]carbamoyl ⁇ quinoline-6-carboxylate or (2E)-2-cyano-N-[1 - (4-hydroxyphenyl)butyl]-3-(1 ,3-thiazol-2-yl)prop-2-enamide instead of the Q 1 moiety defined above.
  • enzyme-binding chemical structures such as 8-(benzoylsulfanyl)-3- ⁇ [2-(1 ,3- thiazol-2-yl)ethyl]carbamoyl ⁇ quinoline-6-carboxylate or (2E)-2-cyano-N-[1 - (4-hydroxyphenyl)butyl]-3-(1 ,3-thiazol-2-yl)prop-2-en
  • the invention also relates to the optically active forms (stereoisomers), the enantiomers, the racemates, the diastereomers and the hydrates and sol- vates of these compounds.
  • the invention relates to pharmaceutically acceptable derivatives of compounds of formula I.
  • solvates of the compounds is taken to mean adductions of inert solvent molecules onto the compounds which form owing to their mutual attractive force.
  • Solvates are, for example, mono- or dihydrates or alkoxides.
  • the invention also relates to the solvates of the salts.
  • pharmaceutically acceptable derivatives is taken to mean, for example, the salts of the compounds according to the invention and also so-called prodrug compounds.
  • prodrug means a derivative of a compound of formula I that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a compound of formula I.
  • prodrugs include, but are not limited to, derivatives and metabolites of a compound of formula I that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid.
  • the carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule.
  • Prodrugs can typically be prepared using well- known methods.
  • an effective amount denotes the amount of a medicament or of a pharmaceutical active ingredient which causes in a tissue, system, animal or human a biological or medical response which is sought or de- sired, for example, by a researcher or physician.
  • the expression "therapeutically effective amount” denotes an amount which, compared with a corresponding subject who has not re- ceived this amount, has the following consequence: improved treatment, healing, prevention or elimination of a disease, syn- drome, condition, complaint, disorder or side-effects or also the reduction in the advance of a disease, complaint or disorder.
  • terapéuticaally effective amount also encompasses the amounts which are effective for increasing normal physiological function.
  • the invention also relates to the use of mixtures of the compounds of the formula I, for example mixtures of two diastereomers, for example in the ratio 1 :1 , 1 :2, 1 :3, 1 :4, 1 :5, 1 :10, 1 :100 or 1 :1000.
  • Tautomers refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution.
  • the invention relates to the compounds of the formula I and salts thereof and to a process for the preparation of compounds of the formula I, wherein L denotes CONR 4 , and pharmaceutically acceptable salts, sol- vates, tautomers and stereoisomers thereof, characterised in that a compound of formula II in which X, R, R 1 , R 3 , R 5 , R 6 , R 7 and p have the meanings indicated in
  • A denotes alkyl, this is unbranched (linear) or branched, and has 1 , 2, 3, 4, 5, 6, 7 or 8 C atoms.
  • A preferably denotes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl or tert-butyl, furthermore also pentyl, 1-, 2- or 3-methylbutyl, 1 ,1- , 1 ,2- or 2,2-dimethylpropyl, 1 -ethylpropyl, hexyl, 1 - , 2- , 3- or 4-methylpentyl, 1 ,1 - , 1 ,2- , 1 ,3- , 2,2- , 2,3- or 3,3-dimethylbutyl, 1 - or 2-ethylbutyl, 1 -ethyl-1 -methylpropyl, 1 -ethyl-2-methylpropyl, 1 ,1 ,2- or 1
  • A denotes preferably CH2OCH3, CH2CH2OH or CH2CH2OCH3.
  • Cyclic alkyl preferably denotes cyclopropyl, cyclobutyl, cyclopentyl, cyclo- hexyl or cycloheptyl.
  • X preferably denotes CO, furthermore CH2.
  • Y preferably denotes CO, furthermore CH2.
  • R 1 preferably denotes (CH2)n, [C(R 4 )2]nAr 1 -, (CH2)nHet- or (CH2)nCyc-, furthermore [C(R 4 ) 2 ]nCONHAr 1 or [C(R 4 ) 2 ]nNA-.
  • Substituent L is directly connected to Ar 1 , Het or Cyc and not to the (CH2)n or [C(R 4 )2]n moiety.
  • R 1 particularly preferably denotes o-, m- or p-phenylen, indole-diyl or benzimidazole-diyl.
  • R 2 preferably denotes [C(R 4 )2]nAr 2 , (CH2)nCyc or (CH2)nHet 1 .
  • R 3 preferably denotes OH.
  • R 4 preferably denotes H, methyl, ethyl or propyl, very particularly preferably H or methyl.
  • R 5 , R 6 preferably denote H.
  • R 7 preferably denotes H, F or CH3.
  • Ar 1 preferably denotes phenyl, o-, m- or p-fluorophenyl, o-, m- or p-bromo- phenyl, o-, m- or p-chlorophenyl, o-, m- or p-hydroxyphenyl, o-, m- or p- methoxyphenyl, o-, m- or p-aminocarbonylphenyl, further preferably 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-difluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5- dichlorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-dibromophenyl, 2,3,4-, 2,3,5-, 2,3,6-, 2,4,6- or 3,4,5-trichlorophenyl, p-iod
  • Ar 1 preferably denotes phenyl.
  • Ar 2 preferably denotes phenyl, o-, m- or p-tolyl, o-, m- or p-ethylphenyl, o-, m- or p-propylphenyl, o-, m- or p-isopropylphenyl, o-, m- or p-tert-butyl- phenyl, o- m- or p-trifluoromethylphenyl, o-, m- or p-fluorophenyl, o- m- or p-bromophenyl, o-, m- or p-chlorophenyl, o-, m- or p-aminocarbonylphenyl, further preferably 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or 3,5-difluorophenyl, 2,3-, 2,4-, 2,5-, 2,6-, 3,4- or
  • Ar 2 preferably denotes phenyl which is unsubstituted or mono-, di-, tri- or tetra-substituted by Hal.
  • Ar 2 furthermore particularly preferably denotes phenyl which is mono- or disubstituted by Hal.
  • Het preferably denotes 2- or 3-furyl, 2- or
  • 3-thienyl 1-, 2- or 3-pyrrolyl, 1-, 2, 4- or 5-imidazolyl, 1 -, 3-, 4- or 5-pyrazolyl, 2-, 4- or 5-oxazolyl, 3-, 4- or 5-isoxazolyl, 2-, 4- or 5-thiazoly 1 , 3-, 4- or 5-isothiazolyl, 2-, 3- or 4-pyridyl, 2-, 4-, 5- or 6-pyrimidinyl, furthermore preferably 1 ,2,3-triazol-1 -, -4- or -5-yl, 1 ,2,4-triazol-1 -, -3- or 5-yl, 1 - or
  • 6- or 7-benzopyrazolyl 2-, 4-, 5-, 6- or 7-benzoxazolyl, 3-, 4-, 5-, 6- or
  • heterocyclic radicals may also be partially or fully hydrogenated.
  • Unsubstituted Het can thus also denote, for example, 2,3-dihydro-2-, -3-, -4- or -5-furyl, 2,5-dihydro-2-, -3-, -4- or 5-furyl, tetrahydro-2- or -3-furyl, 1 ,3-di- oxolan-4-yl, tetrahydro-2- or -3-thienyl, 2,3-dihydro-1 -, -2-, -3-, -4- or -5-pyr- rolyl, 2,5-dihydro-1 -, -2- -3-, -4- or -5-pyrrolyl, 1-, 2- or 3-pyrrolidinyl, tetra- hydro-1-, -2- or -4-imidazolyl, 2,3-dihydro-1 -, -2-, -3-, -4- or -5
  • Het furthermore preferably denotes pyrazinyl, pyrazolyl, benzimidazolyl, pyridyl, indolyl, dihydroindolyl, benzofuranyl, tetrahydropyranyl, dihydro- quinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetrahydro- isoquinolinyl, indazolyl, imidazolyl, pyrrolyl, oxazolyl, oxadiazolyl, isoxazolyl, benzothiazolyl, piperidin-1 -yl, pyrrolidin-1 -yl, 3,4-dihydro-2H- pyrido[3,2-b]-1 ,4-oxazinyl, 3,4-dihydro-2H-benzo-1 ,4-oxazinyl, benzofuranyl, azetidinyl,
  • Het furthermore preferably denotes benzimidazolyl or indolyl, each of which is unsubstituted or monosubstituted by Hal.
  • Het 1 preferably denotes pyridazinyl, pyrazolyl, pyridyl, piperazinyl, morpholinyl, pyrimidinyl, furyl, thienyl, imidazolyl, pyrrolyl, oxazolyl, oxadiazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, thiadiazole, piperidin-1 -yl, pyrrolidin-1 -yl, tetrahydropyranyl, 1 ,2-oxazinan-2-yl, 1 ,2,5-oxadiazinan-2-yl, 1 ,3-oxazinan-3-yl or hexahydropyrimidinyl, each of which is un
  • Het 1 furthermore preferably denotes pyridyl, pyrimidinyl, furyl, thienyl, imidazolyl, pyrrolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl or tetrazolyl. Het 1 furthermore particularly preferably denotes pyridyl, furyl, thienyl, imidazolyl or pyrrolyl.
  • Q 2 preferably denotes CH2OCH2CH2OCH2CH2O
  • radicals which occur more than once may be identical or different, i.e. are independent of one another.
  • the compounds of the formula I may have one or more chiral centres and can therefore occur in various stereoisomeric forms.
  • the formula I encom- passes all these forms.
  • the invention relates, in particular, to the compounds of the formula I in which at least one of the said radicals has one of the preferred meanings indicated above.
  • Some preferred groups of compounds may be expressed by the following sub-formulae la to le, which conform to the for- mula I and in which the radicals not designated in greater detail have the meaning indicated for the formula I, but in which in la Het denotes pyrazinyl, pyrazolyl, benzimidazolyl, pyridyl, thienyl, furanyl, indolyl, dihydroindolyl, benzofuranyl, tetrahydropyranyl, dihydroquinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetra- hydroisoquinolinyl, indazolyl, imidazolyl, pyrrolyl, oxazolyl, oxadiazolyl, isoxazolyl, be
  • Q denotes 0, NH, or CH2,
  • Q 2 denotes unbranched alkylene having 4-25 C atoms, in which one or more single bonds between C atoms may be replaced by a triple bond, and in which 1-8 non-adjacent CH2 groups may be replaced by O, CONH and/or NHCO, and in which one CH2 group may be replaced by
  • R denotes NR 2 R 4 ,
  • X denotes CO or CH2
  • Y denotes CO or CH2
  • R 1 denotes (CH 2 )n, [C(R 4 ) 2 ]nAr 1 - or (CH 2 ) n Het- wherein substituent L directly is connected to Ar 1 , Het or Cyc,
  • R 2 denotes [C(R 4 ) 2 ]nAr 2 , (CH 2 )nCyc or (CH 2 )nHet 1 ,
  • R 3 denotes OH
  • R 4 denotes H or alkyl having 1 , 2, 3 or 4 C-atoms
  • R 5 , R 6 denote H
  • R 7 denotes H, Hal or A
  • Ar 1 denotes phenyl
  • Ar 2 denotes phenyl which is unsubstituted or mono-, di-, tri- or tetra-substituted by Hal, Het denotes pyrazinyl, pyrazolyl, benzimidazolyl, pyridyl, thienyl, furanyl, indolyl, dihydroindolyl, benzofuranyl, tetrahydropyranyl, dihydroquinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indazolyl, imidazolyl, pyrrolyl, oxazolyl, oxadiazolyl, isoxazolyl, benzothiazolyl, piperidin-1 -yl, pyrrolidin-1 -yl, 3,4-dihydro- 2H-pyrido[3,2-b]-1 ,4-ox
  • Het 1 denotes pyridyl, furyl, thienyl, imidazolyl or pyrrolyl,
  • A denotes unbranched or branched alkyl having 1 -6 C atoms, in which 1 -5 H atoms may be replaced by F, Cl and/or OH,
  • Cyc denotes cyclic alkyl having 3-7 C atoms, Hal denotes F, Cl, Br or I, n denotes 0, 1 , 2, 3 or 4, p denotes 1 , 2 or 3; and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.
  • Q 3 is
  • Alternative MetAP-2 binding chemical structures that can be used instead of this Q 3 moiety are described in
  • E3 ligases e.g. proline- or further hydroxy-proline derivatives, halogenated Thalidominde or Lenalidomide analogs or piperidine-2, 6-dione isomers, as well as 3-(2-oxo- 2, 3-dihydro-1 H-1 ,3-benzodiazol-1 -yl)piperidine-2, 6-dione derivatives, , N- benzyl-2-chloro-N-[4-(phenoxy-4-yl)phenyl]acetamide derivatives, 3-[2,4- dihydroxy-5-(propan-2-yl)phenyl]-4-(phenyl-4-yl)-4,5-dihydro-1 H-1 ,2,4- triazol-5-one derivatives, Idasanutlin-, bardoxolone-, Indisulam-, chloro
  • the compounds of the formula I and also the starting materials for their preparation are, in addition, prepared by methods known per se, as des- cribed in the literature (for example in the standard works, such as Houben-Weyl, Methoden der organischen Chemie [Methods of Organic Chemistry], Georg-Thieme-Verlag, Stuttgart), to be precise under reaction conditions which are known and suitable for the said reactions. Use can also be made here of variants known per se which are not mentioned here in greater detail.
  • Compounds of the formula I can preferably be obtained by reacting com- pounds of the formula II with a compound of the formula III.
  • the compounds of the formula II and of the formula III are generally known or can be prepared by methods known per se.
  • L preferably denotes Cl, Br, I or a free or a reactively modified OH group, such as, for example, an activated ester, an imidazolide or alkylsulfonyloxy having 1-6 C atoms (preferably methylsulfonyloxy or trifluoromethylsulfonyloxy) or arylsulfonyloxy having 6-10 C atoms (preferably phenyl- or p-tolylsulfonyloxy).
  • an activated ester an imidazolide or alkylsulfonyloxy having 1-6 C atoms (preferably methylsulfonyloxy or trifluoromethylsulfonyloxy) or arylsulfonyloxy having 6-10 C atoms (preferably phenyl- or p-tolylsulfonyloxy).
  • the reaction preferably succeeds in the presence of a dehydrating agent, such as, for example, a carbodiimide, such as N,N'-dicyclohexyl- carbodiimide (“DCCI”), 1 ,1'-carbonyldiimidazole or N-3-dimethylamino- propyl-N'-ethylcarbodiimide (“DAPECI”), furthermore propanephosphonic anhydride T3P (cf. Angew. Chem. 92, 129 (1980)), diphenylphosphoryl azide or 2-ethoxy-N-ethoxycarbonyl-1 ,2-dihydroquinoline, optionally in the presence of N-hydroxybenzotriaole;
  • a dehydrating agent such as, for example, a carbodiimide, such as N,N'-dicyclohexyl- carbodiimide (“DCCI”), 1 ,1'-carbonyldiimidazole or N-3-dimethyla
  • HATU O-(7-Azabenzotriazol-1-yl)- A/,A/,A/',A/'-tetramethyluronium-hexafluorphosphat
  • the reaction is carried out in an inert solvent and is generally carried out in the presence of an acid-binding agent, preferably an organic base, such as DIPEA, 4-methylmorpholine, triethylamine, dimethylaniline, pyridine or quinoline.
  • an acid-binding agent preferably an organic base, such as DIPEA, 4-methylmorpholine, triethylamine, dimethylaniline, pyridine or quinoline.
  • alkali or alkaline-earth metal hydroxide, carbonate or bi- carbonate or another salt of a weak acid of the alkali or alkaline-earth metals preferably of potassium, sodium, calcium or caesium, may also be favourable.
  • the reaction time is between a few minutes and 14 days
  • the reaction temperature is between about -15° and 150°, normally between 0° and 120°, particularly preferably between 20° and 40°C.
  • Suitable inert solvents are, for example, hydrocarbons, such as hexane, petroleum ether, benzene, toluene or xylene; chlorinated hydrocarbons, such as trichloroethylene, 1 ,2-dichloroethane, carbon tetrachloride, chloro- form or dichloromethane; alcohols, such as methanol, ethanol, isopropanol, n-propanol, n-butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran (THF) or dioxane; glycol ethers, such as ethylene glycol monomethyl or monoethyl ether, ethylene glycol dimethyl ether (diglyme); ketones, such as acetone or butanone; amides, such as acetamide, dimethylacetamide or dimethylformamide (DMF); nit
  • the said compounds according to the invention can be used in their final non-salt form.
  • the present invention also encompasses the use of these compounds in the form of their pharmaceutically accept- able salts, which can be derived from various organic and inorganic acids and bases by procedures known in the art.
  • Pharmaceutically acceptable salt forms of the compounds of the formula I are for the most part prepared by conventional methods. If the compound of the formula I contains a car- boxyl group, one of its suitable salts can be formed by reacting the com- pound with a suitable base to give the corresponding base-addition salt.
  • Such bases are, for example, alkali metal hydroxides, including potassium hydroxide, sodium hydroxide and lithium hydroxide; alkaline earth metal hydroxides, such as barium hydroxide and calcium hydroxide; alkali metal alkoxides, for example potassium ethoxide and sodium propoxide; and various organic bases, such as piperidine, diethanolamine and N-methyl- glutamine.
  • alkali metal hydroxides including potassium hydroxide, sodium hydroxide and lithium hydroxide
  • alkaline earth metal hydroxides such as barium hydroxide and calcium hydroxide
  • alkali metal alkoxides for example potassium ethoxide and sodium propoxide
  • organic bases such as piperidine, diethanolamine and N-methyl- glutamine.
  • the aluminium salts of the compounds of the formula I are like- wise included.
  • acid-addi- tion salts can be formed by treating these compounds with pharmaceuti- cally acceptable organic and inorganic acids, for example hydrogen hal- ides, such as hydrogen chloride, hydrogen bromide or hydrogen iodide, other mineral acids and corresponding salts thereof, such as sulfate, nitrate or phosphate and the like, and alkyl- and monoarylsulfonates, such as ethanesulfonate, toluenesulfonate and benzenesulfonate, and other organic acids and corresponding salts thereof, such as acetate, trifluoro- acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate, ascor- bate and the like.
  • organic and inorganic acids for example hydrogen hal- ides, such as hydrogen chloride, hydrogen bromide or hydrogen iodide, other mineral acids and corresponding salts thereof, such as sulfate, nitrate or phosphat
  • pharmaceutically acceptable acid-addition salts of the compounds of the formula I include the following: acetate, adi- pate, alginate, arginate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, citrate, cyclopentanepropionate, diglu- conate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, ethane- sulfonate, fumarate, formate, galacterate (from mucic acid), galacturonate, glucoheptanoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydro- bromide, hydro
  • the base salts of the compounds according to the invention include aluminium, ammonium, calcium, copper, iron(lll), iron(ll), lithium, magnesium, manganese(lll), manganese(ll), potassium, sodium and zinc salts, but this is not intended to represent a restriction.
  • Salts of the compounds of the formula I which are derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines, also including naturally occurring substituted amines, cyclic amines, and basic ion ex- changer resins, for example arginine, betaine, caffeine, chloroprocaine, choline, N,N'-dibenzylethylenediamine (benzathine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylaminoethanol, 2-dimethylamino- ethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethyl- piperidine, glucamine, glucosamine, histidine, hydrabamine, isopropyl- amine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine
  • Compounds of the present invention which contain basic nitrogen-contain- ing groups can be quaternised using agents such as (C1-C4)alkyl halides, for example methyl, ethyl, isopropyl and tert-butyl chloride, bromide and iodide; di(C 1 -C 4 )alkyl sulfates, for example dimethyl, diethyl and diamyl sulfate; (C10-C18)alkyl halides, for example decyl, dodecyl, lauryl, myristyl and stearyl chloride, bromide and iodide; and aryl(C1-C4)alkyl halides, for example benzyl chloride and phenethyl bromide.
  • agents such as (C1-C4)alkyl halides, for example methyl, ethyl, isopropyl and tert-butyl chloride,
  • Both water- and oil-solu- ble compounds according to the invention can be prepared using such salts.
  • the above-mentioned pharmaceutical salts which are preferred include acetate, trifluoroacetate, besylate, citrate, fumarate, gluconate, hemisucci- nate, hippurate, hydrochloride, hydrobromide, isethionate, mandelate, me- glumine, nitrate, oleate, phosphonate, pivalate, sodium phosphate, stea- rate, sulfate, sulfosalicylate, tartrate, thiomalate, tosylate and trometh- amine, but this is not intended to represent a restriction.
  • the acid-addition salts of basic compounds of the formula I are prepared by bringing the free base form into contact with a sufficient amount of the desired acid, causing the formation of the salt in a conventional manner.
  • the free base can be regenerated by bringing the salt form into contact with a base and isolating the free base in a conventional manner.
  • the free base forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts other- wise correspond to the respective free base forms thereof.
  • the pharmaceutically acceptable base-addition salts of the compounds of the formula I are formed with metals or amines, such as alkali metals and alkaline earth metals or organic amines.
  • metals are sodium, potassium, magnesium and calcium.
  • Preferred organic amines are N,N’-dibenzylethylenediamine, chloroprocaine, choline, diethanol- amine, ethylenediamine, N-methyl-D-glucamine and procaine.
  • the base-addition salts of acidic compounds according to the invention are prepared by bringing the free acid form into contact with a sufficient amount of the desired base, causing the formation of the salt in a conven- tional manner.
  • the free acid can be regenerated by bringing the salt form into contact with an acid and isolating the free acid in a conventional man- ner.
  • the free acid forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solu- bility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free acid forms thereof.
  • a compound according to the invention contains more than one group which is capable of forming pharmaceutically acceptable salts of this type, the invention also encompasses multiple salts.
  • Typical multiple salt forms include, for example, bitartrate, diacetate, difumarate, dimeglumine, di- phosphate, disodium and trihydrochloride, but this is not intended to repre- sent a restriction.
  • the expression "phar- maceutically acceptable salt” in the present connection is taken to mean an active ingredient which comprises a compound of the formula I in the form of one of its salts, in particular if this salt form imparts improved pharma- cokinetic properties on the active ingredient compared with the free form of the active ingredient or any other salt form of the active ingredient used earlier.
  • the pharmaceutically acceptable salt form of the active ingredient can also provide this active ingredient for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active ingredient with respect to its therapeutic efficacy in the body.
  • a compound of the formula I includes isotope-labelled forms thereof.
  • An isotope-labelled form of a compound of the formula I is identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally.
  • isotopes which are readily commercially available and which can be incorporated into a compound of the formula I by well-known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 CI, respectively.
  • a compound of the formula I, a prodrug, thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other iso-topes of other atoms is intended to be part of the present invention.
  • An isotope-labelled compound of the formula I can be used in a number of beneficial ways.
  • an isotope-labelled compound of the formula I into which, for example, a radioisotope, such as 3 H or 14 C, has been incorporated is suitable for medicament and/or substrate tissue distribution assays.
  • radioisotopes i.e. tritium ( 3 H) and carbon-14 ( 14 C)
  • 3 H tritium
  • 14 C carbon-14
  • Incorporation of heavier isotopes, for example deuterium ( 2 H) into a compound of the formula I has therapeutic advantages owing to the higher metabolic stability of this isotope-labelled compound. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention.
  • An isotope-labelled compound of the formula I can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related description, in the example part and in the preparation part in the present text, replacing a non-isotope-labelled reactant by a readily available isotope- labelled reactant.
  • Deuterium ( 2 H) can also be incorporated into a compound of the formula I for the purpose in order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect.
  • the primary kinetic isotope effect is a change of the rate for a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange.
  • Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus cause a reduction in the rate in rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi-product reaction, the product distribution ratios can be altered substantially.
  • a compound of the formula I which has multiple potential sites of attack for oxidative metabolism for example benzylic hydrogen atoms and hydrogen atoms bonded to a nitrogen atom, is prepared as a series of analogues in which various combinations of hydrogen atoms are replaced by deuterium atoms, so that some, most or all of these hydrogen atoms have been replaced by deuterium atoms.
  • Half-life determinations enable favourable and accurate determination of the extent of the extent to which the improve-ment in resistance to oxidative metabolism has improved. In this way, it is deter-mined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type.
  • Deuterium-hydrogen exchange in a compound of the formula I can also be used to achieve a favourable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites.
  • a toxic metabolite arises through oxidative carbon-hydrogen (C-H) bond cleavage
  • C-H oxidative carbon-hydrogen
  • the deuterated analogue will greatly diminish or eliminate production of the unwanted metabolite, even if the particular oxidation is not a rate- determining step.
  • Further information on the state of the art with respect to deuterium-hydrogen exchange may be found, for example in Hanzlik et al., J. Org. Chem. 55, 3992-3997, 1990, Reider et al., J.
  • the invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically acceptable derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants.
  • compositions can be administered in the form of dosage units which comprise a predetermined amount of active ingredient per dosage unit.
  • a unit can comprise, for example, 0.5 mg to 1 g, prefer- ably 1 mg to 700 mg, particularly preferably 5 mg to 100 mg, of a com- pound according to the invention, depending on the condition treated, the method of administration and the age, weight and condition of the patient, or pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active ingredient per dosage unit.
  • Preferred dosage unit formulations are those which comprise a daily dose or part-dose, as indicated above, or a corresponding fraction thereof of an active ingredient.
  • pharmaceutical formulations of this type can be prepared using a process which is generally known in the pharmaceutical art.
  • compositions can be adapted for administration via any desired suitable method, for example by oral (including buccal or sublin- gual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods.
  • oral including buccal or sublin- gual
  • rectal nasal
  • topical including buccal, sublingual or transdermal
  • vaginal or parenteral including subcutaneous, intramuscular, intravenous or intradermal
  • parenteral including subcutaneous, intramuscular, intravenous or intradermal
  • compositions adapted for oral administration can be administered as separate units, such as, for example, capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or foam foods; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.
  • the active-ingredient component can be combined with an oral, non-toxic and pharmaceutically acceptable inert excipient, such as, for example, ethanol, glycerol, water and the like.
  • Powders are prepared by comminuting the compound to a suitable fine size and mixing it with a pharmaceutical excipient comminuted in a similar manner, such as, for example, an edible carbohydrate, such as, for example, starch or mannitol.
  • a pharmaceutical excipient comminuted in a similar manner, such as, for example, an edible carbohydrate, such as, for example, starch or mannitol.
  • a flavour, preservative, dispersant and dye may likewise be present.
  • Capsules are produced by preparing a powder mixture as described above and filling shaped gelatine shells therewith.
  • Glidants and lubricants such as, for example, highly disperse silicic acid, talc, magnesium stearate, cal- cium stearate or polyethylene glycol in solid form, can be added to the powder mixture before the filling operation.
  • a disintegrant or solubiliser such as, for example, agar-agar, calcium carbonate or sodium carbonate, may likewise be added in order to improve the availability of the medica- ment after the capsule has been taken.
  • suitable binders include starch, gelatine, natural sugars, such as, for example, glucose or beta-lactose, sweeteners made from maize, natural and synthetic rubber, such as, for example, acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • the lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • the disintegrants include, without being restricted thereto, starch, methylcellulose, agar, bentonite, xanthan gum and the like.
  • the tablets are formulated by, for example, preparing a powder mixture, granulating or dry-pressing the mixture, adding a lubricant and a disinteg- rant and pressing the entire mixture to give tablets.
  • a powder mixture is prepared by mixing the compound comminuted in a suitable manner with a diluent or a base, as described above, and optionally with a binder, such as, for example, carboxymethylcellulose, an alginate, gelatine or polyvinyl- pyrrolidone, a dissolution retardant, such as, for example, paraffin, an ab- sorption accelerator, such as, for example, a quaternary salt, and/or an absorbant, such as, for example, bentonite, kaolin or dicalcium phosphate.
  • a binder such as, for example, carboxymethylcellulose, an alginate, gelatine or polyvinyl- pyrrolidone
  • a dissolution retardant such as, for example, paraffin
  • an ab- sorption accelerator such as, for example, a quaternary salt
  • an absorbant such as, for example, bentonite, kaolin or dicalcium phosphate.
  • the powder mixture can be granulated by wetting it with a binder, such as, for example, syrup, starch paste, acadia mucilage or solutions of cellulose or polymer materials and pressing it through a sieve.
  • a binder such as, for example, syrup, starch paste, acadia mucilage or solutions of cellulose or polymer materials
  • the powder mixture can be run through a tabletting machine, giving lumps of non-uniform shape, which are broken up to form granules.
  • the granules can be lubricated by addition of stearic acid, a stearate salt, talc or mineral oil in order to prevent sticking to the tablet casting moulds. The lubricated mixture is then pressed to give tablets.
  • the compounds according to the invention can also be combined with a free-flowing inert excipient and then pressed directly to give tablets without carrying out the granulation or dry-pressing steps.
  • a transparent or opaque protective layer consisting of a shellac sealing layer, a layer of sugar or polymer material and a gloss layer of wax may be present. Dyes can be added to these coatings in order to be able to differentiate between different dosage units.
  • Oral liquids such as, for example, solution, syrups and elixirs, can be pre- pared in the form of dosage units so that a given quantity comprises a pre- specified amount of the compound.
  • Syrups can be prepared by dissolving the compound in an aqueous solution with a suitable flavour, while elixirs are prepared using a non-toxic alcoholic vehicle.
  • Suspensions can be for- mulated by dispersion of the compound in a non-toxic vehicle.
  • Solubilisers and emulsifiers such as, for example, ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavour additives, such as, for example, peppermint oil or natural sweeteners or saccharin, or other artificial sweeteners and the like, can likewise be added.
  • the dosage unit formulations for oral administration can, if desired, be en- capsulated in microcapsules.
  • the formulation can also be prepared in such a way that the release is extended or retarded, such as, for example, by coating or embedding of particulate material in polymers, wax and the like.
  • the compounds of the formula I and pharmaceutically salts, tautomers and stereoisomers thereof can also be administered in the form of liposome delivery systems, such as, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • liposomes can be formed from various phospholipids, such as, for example, cholesterol, stearylamine or phosphatidylcholines.
  • the compounds of the formula I and the salts, tautomers and stereoisomers thereof can also be delivered using monoclonal antibodies as individual carriers to which the compound molecules are coupled.
  • the compounds can also be coupled to soluble polymers as targeted medicament carriers.
  • Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxy- ethylaspartamidophenol or polyethylene oxide polylysine, substituted by palmitoyl radicals.
  • the compounds may furthermore be coupled to a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-epsilon-capro- lactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihy- droxypyrans, polycyanoacrylates and crosslinked or amphipathic block co- polymers of hydrogels.
  • a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-epsilon-capro- lactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihy- droxypyrans, polycyanoacrylates and crosslinked or amphipathic block co- polymers of hydrogels.
  • compositions adapted for transdermal administration can be administered as independent plasters for extended, close contact with the epidermis of the recipient.
  • the active ingredient can be delivered from the plaster by iontophoresis.
  • Pharmaceutical compounds adapted for topical administration can be for- mulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • the formulations are preferably applied as topical ointment or cream.
  • the active ingredient can be employed either with a paraffinic or a water-miscible cream base.
  • the active ingredient can be formulated to give a cream with an oil-in-water cream base or a water-in-oil base.
  • compositions adapted for topical application to the eye include eye drops, in which the active ingredient is dissolved or suspended in a suitable carrier, in particular an aqueous solvent.
  • compositions adapted for topical application in the mouth encompass lozenges, pastilles and mouthwashes.
  • compositions adapted for rectal administration can be ad- ministered in the form of suppositories or enemas.
  • compositions adapted for nasal administration in which the carrier substance is a solid comprise a coarse powder having a particle size, for example, in the range 20-500 microns, which is administered in the manner in which snuff is taken, i.e. by rapid inhalation via the nasal passages from a container containing the powder held close to the nose.
  • suitable formulations for administration as nasal spray or nose drops with a liquid as carrier substance encompass active-ingredient solutions in water or oil.
  • compositions adapted for administration by inhalation en- compass finely particulate dusts or mists, which can be generated by vari- ous types of pressurised dispensers with aerosols, nebulisers or insuffla- tors.
  • compositions adapted for vaginal administration can be administered as pessaries, tampons, creams, gels, pastes, foams or spray formulations.
  • compositions adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions comprising antioxi- dants, buffers, bacteriostatics and solutes, by means of which the formula- tion is rendered isotonic with the blood of the recipient to be treated; and aqueous and non-aqueous sterile suspensions, which may comprise sus- pension media and thickeners.
  • the formulations can be administered in single-dose or multidose containers, for example sealed ampoules and vials, and stored in freeze-dried (lyophilised) state, so that only the addition of the sterile carrier liquid, for example water for injection purposes, imme- diately before use is necessary.
  • Injection solutions and suspensions pre- pared in accordance with the recipe can be prepared from sterile powders, granules and tablets.
  • the formulations may also comprise other agents usual in the art with respect to the particular type of formulation; thus, for example, for- mulations which are suitable for oral administration may comprise flavours.
  • a therapeutically effective amount of a compound of the formula I depends on a number of factors, including, for example, the age and weight of the animal, the precise condition that requires treatment, and its severity, the nature of the formulation and the method of administration, and is ultimate- ly determined by the treating doctor or vet.
  • an effective amount of a compound according to the invention is generally in the range from 0.1 to 100 mg/kg of body weight of the recipient (mammal) per day and particu- larly typically in the range from 1 to 10 mg/kg of body weight per day.
  • the actual amount per day for an adult mammal weighing 70 kg is usually between 70 and 700 mg, where this amount can be administered as a single dose per day or usually in a series of part-doses (such as, for exam- ple, two, three, four, five or six) per day, so that the total daily dose is the same.
  • An effective amount of a salt or solvate or of a physiologically func- tional derivative thereof can be determined as the fraction of the effective amount of the compound according to the invention perse. It can be assumed that similar doses are suitable for the treatment of other condi- tions mentioned above.
  • a combined treatment of this type can be achieved with the aid of simulta- neous, consecutive or separate dispensing of the individual components of the treatment.
  • Combination products of this type employ the compounds according to the invention.
  • the invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active ingredient.
  • the invention also relates to a set (kit) consisting of separate packs of
  • the set comprises suitable containers, such as boxes, individual bottles, bags or ampoules.
  • the set may, for example, comprise separate am- poules, each containing an effective amount of a compound of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and an effective amount of a further medicament active ingredient in dis- solved or lyophilised form.
  • Treating means an alleviation, in whole or in part, of symptoms associated with a disorder or disease, or slowing, or halting of further progression or worsening of those symptoms, or prevention or prophylaxis of the disease or disorder in a subject at risk for developing the disease or disorder.
  • ⁇ ективное ⁇ ество in connection with a compound of formula (I) can mean an amount capable of alleviating, in whole or in part, symptoms associated with a disorder or disease, or slowing or halting further progression or worsening of those symptoms, or preventing or providing prophylaxis for the disease or disorder in a subject having or at risk for developing a disease disclosed herein, such as inflammatory conditions, immunological conditions, cancer or metabolic conditions.
  • the present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the use for the treatment of diseases in which the degradation and/or modulation of MetAP-2 plays a role.
  • the present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the use for the degradation and/or modulation of MetAP-2.
  • the present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for use for the treatment and control of diseases.
  • tumour cells pathological neovascularisation (or angiogenesis), which promotes the growth of solid tumours, neovascularisation in the eye (diabetic retinopathy, age-induced macular degeneration and the like) and inflammation (psoriasis, rheumatoid arthritis and the like), and proliferative diseases of the mesangial cells.
  • pathological neovascularisation or angiogenesis
  • angiogenesis which promotes the growth of solid tumours
  • neovascularisation in the eye diabetic retinopathy, age-induced macular degeneration and the like
  • inflammation psoriasis, rheumatoid arthritis and the like
  • proliferative diseases of the mesangial cells proliferative diseases of the mesangial cells.
  • the invention relates to compounds for use of the formula I according to claim 1 and pharmaceutically acceptable salts, solvates, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the treatment and/or prevention of tumours, tumour metastases, proliferative diseases of the mesangial cells, haemangioma, proliferative retinopathy, rheumatoid arthritis, atherosclerotic neovascularisation, psoriasis, ocular neovascularisation, osteoporosis, diabetes and obesity, lymphoid leu- kaemia, lymphoma, malaria and prostate hypertrophy.
  • the invention relates to compounds for use where the tumour disease is selected from the group of the squamous epithelium, of the bladder, of the stomach, of the kidneys, of head and neck, of the oesophagus, of the cervix, of the thyroid, of the intestine, of the liver, of the brain, of the prostate, of the urogenital tract, of the lymphatic system, of the stomach, of the larynx, of the lung, of the skin, monocytic leukaemia, lung adenocarcinoma, small-cell lung carcinoma, pancreatic cancer, glioblastoma, breast carcinoma, acute myeloid leukaemia, chronic myeloid leukaemia, acute lymphatic leukaemia, chronic lymphatic leukaemia, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma.
  • the present invention encompasses the use of the compounds of the for- mula I and/or physiologically acceptable salts and solvates thereof for the preparation of a medicament for the treatment or prevention of tumours, tumour diseases and/or tumour metastases.
  • the tumour disease is preferably selected from the group tumour of the squamous epithelium, the bladder, the stomach, the kidneys, of head and neck, the oesophagus, the cervix, the thyroid, the intestine, the liver, the brain, the prostate, the urogenital tract, the lymphatic system, the stomach, the larynx, the lung, the skin, monocytic leukaemia, lung adenocarcinoma, small-cell lung carcinoma, pancreatic cancer, glioblastoma, breast carcinoma, acute myeloid leukaemia, chronic myeloid leukaemia, acute lymphatic leukaemia, chronic lymphatic leukaemia, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma.
  • a disease of this type in which angiogenesis is involved is an eye disease, such as retina vascularisation, diabetic retinopathy, age-induced macular degeneration and the like.
  • the angiogenic disease is preferably selected from the group diabetic retinopathy, arthritis, cancer, psoriasis, Kaposi's sarcoma, haeman- gioma, myocardial angiogenesis, atherosclerotic plaque neovascularisation, angiogenic eye diseases, choroidal neovascularisation, retrolental fibroplasia, macular degeneration, corneal transplant rejection, rubeosis iridis, neuroscular glaucoma, Oster Webber syndrome.
  • the proliferative disease of the mesangial cells is preferably selected from the group glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, throm- botic microangiopathy syndrome, transplant rejection, glomerulopathy.
  • the inflammatory disease is preferably selected from the group inflammatory bowel disease, arthritis, atherosclersosis, asthma, allergies, inflammatory kidney diseases, multiple sclerosis, chronic obstructive pulmo- nary disease, inflammatory skin diseases, pardontai diseases, psoriasis, T-cell-promoted immune disease.
  • the inflammatory bowel disease is preferably selected from the group ulcerative colitis, Crohn’s disease, non-specific colitis.
  • the T-cell-promoted immune disease is preferably selected from the group allergic encephalomyelitis, allergic neuritis, transplant rejection, graft-versus- host reaction, myocarditis, thyroiditis, nephritis, systemic lupus erythematosus, insulin-dependent diabetes mellitus.
  • the arthritis disease is preferably selected from the group rheumatoid arthritis, osteoarthritis, Caplan's syndrome, Felty's syndrome, Sjogren's syndrome, spondylitis ankylosans, Still’s disease, chondrocalcinosis, metabolic arthritis, rheumatic fever, Reiter’s disease, Wissler's syndrome.
  • the inflammatory kidney disease is preferably selected from the group glomerulonephritis, glomerular injury, nephrotic syndrome, interstitial nephritis, lupus nephritis, Goodpasture’s syndrome, Wegener’s granulomatosis, renal vasculitis, IgA nephropathy, idiopatic glomerular disease.
  • the inflammatory skin disease is preferably selected from the group psoriasis, atopic dermatitis, contact sensitivity, acne.
  • a therapeutically effective amount of a compound according to the invention is administered to a sick mammal in need of such treatment.
  • the therapeutic amount varies according to the specific disease and can be determined by the person skilled in the art without undue effort.
  • the present invention also encompasses the use compounds of the for- mula I and/or physiologically acceptable salts and solvates thereof for the preparation of a medicament for the treatment or prevention of retinal vas- cularisation.
  • a therapeutically effective amount of a compound according to the invention is administered to a sick mammal in need of such treatment.
  • the therapeutic amount varies according to the specific disease and can be determined by the person skilled in the art without undue effort.
  • anticancer agent relates to any agent which is administered to a patient with cancer for the purposes of treating the can- cer.
  • the anti-cancer treatment defined above may be applied as a monotherapy or may involve, in addition to the herein disclosed compounds of formula I, conventional surgery or radiotherapy or medicinal therapy.
  • Such medicinal therapy e.g. a chemotherapy or a targeted therapy, may include one or more, but preferably one, of the following anti-tumor agents:
  • Alkylating agents such as altretamine, bendamustine, busulfan, carmustine, chlorambucil, chlormethine, cyclophosphamide, dacarbazine, ifosfamide, improsulfan, tosilate, lomustine, melphalan, mitobronitol, mitolactol, nimustine, ranimustine, temozolomide, thiotepa, treosulfan, mechloretamine, carboguone; apaziguone, fotemustine, glufosfamide, palifosfamide, pipobroman, trofosfamide, uramustine, TH-302 4 , VAL-083 4 ;
  • Platinum Compounds such as carboplatin, cisplatin, eptaplatin, m iriplatine hydrate, oxaliplatin, lobaplatin, nedaplatin, picoplatin, satraplatin; lobaplatin, nedaplatin, picoplatin, satraplatin;
  • DNA altering agents such as amrubicin, bisantrene, decitabine, mitoxantrone, procarbazine, trabectedin, clofarabine; amsacrine, brostallicin, pixantrone, laromustine 1 3 ;
  • Topoisomerase Inhibitors such as etoposide, irinotecan, razoxane, sobuzoxane, teniposide, topotecan; amonafide, belotecan, elliptinium acetate, voreloxin;
  • Microtubule modifiers such as cabazitaxel, docetaxel, eribulin, ixabepilone, paclitaxel, vinblastine, vincristine, vinorelbine, vindesine, vinflunine; fosbretabulin, tesetaxel;
  • Antimetabolites such as asparaginase 3 , azacitidine, calcium levofolinate, capecitabine, cladribine, cytarabine, enocitabine, floxuridine, fludarabine, fluorouracil, gemcitabine, mercaptopurine, methotrexate, nelarabine, pemetrexed, pralatrexate, azathioprine, thioguanine, carmofur; doxifluridine, elacytarabine, raltitrexed, sapacitabine, tegafur 2 3 , trimetrexate; Anticancer antibiotics such as bleomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, levamisole, miltefosine, mitomycin C, romidepsin, streptozocin, valrubicin, zinostatin, zorubicin, daunu
  • Hormones/Antagonists such as abarelix, abiraterone, bicalutamide, buserelin, calusterone, chlorotrianisene, degarelix, dexamethasone, estradiol, fluocortolone fluoxymesterone, flutamide, fulvestrant, goserelin, histrelin, leuprorelin, megestrol, mitotane, nafarelin, nandrolone, nilutamide, octreotide, prednisolone, raloxifene, tamoxifen, thyrotropin alfa, toremifene, trilostane, triptorelin, diethylstilbestrol; acolbifene, danazol, deslorelin, epitiostanol, orteronel, enzalutamide 1 3 ;
  • Aromatase inhibitors such as aminoglutethimide, anastrozole, exemestane, fadrozole, letrozole, testolactone; formestane;
  • Small molecule kinase inhibitors such as crizotinib, dasatinib, erlotinib, imatinib, lapatinib, nilotinib, pazopanib, regorafenib, ruxolitinib, sorafenib, sunitinib, vandetanib, vemurafenib, bosutinib, gefitinib, axitinib; afatinib, alisertib, dabrafenib, dacomitinib, dinaciclib, dovitinib, enzastaurin, nintedanib, lenvatinib, linifanib, linsitinib, masitinib, midostaurin, motesanib, neratinib, orantinib, perifosine, ponatinib, radotinib, rigo
  • Photosensitizers such as methoxsalen 3 ; porfimer sodium, talaporfin, temoporfin; Antibodies such as avelumab, alemtuzumab, besilesomab, brentuximab vedotin, cetuximab, denosumab, ipilimumab, ofatumumab, panitumumab, rituximab, tositumomab, trastuzumab, bevacizumab, pertuzumab 2 3 ; catumaxomab, elotuzumab, epratuzumab, farletuzumab, mogamulizumab, necitumumab, nimotuzumab, obinutuzumab, ocaratuzumab, oregovomab, ramucirumab, rilotumumab, silt
  • Cytokines such as aldesleukin, interferon alfa 2 , interferon alfa2a 3 , interferon alfa2b 2 3 ; celmoleukin, tasonermin, teceleukin, oprelvekin 1 3 , recombinant interferon beta-1 a 4 ;
  • Drug Conjugates such as denileukin diftitox, ibritumomab tiuxetan, iobenguane 1123, prednimustine, trastuzumab emtansine, estramustine, gemtuzumab, ozogamicin, aflibercept; cintredekin besudotox, edotreotide, inotuzumab ozogamicin, naptumomab estafenatox, oportuzumab monatox, technetium (99mTc) arcitumomab 1 ’ 3 , vintafolide 1 3 ;
  • Vaccines such as sipuleucel 3 ; vitespen 3 , emepepimut-S 3 , oncoVAX 4 , rindopepimut 3 , troVax 4 , MGN-1601 4 , MGN-1703 4 ;
  • FIG. 1 MetAP2 expression levels (in %, normalized to y-Tubulin) in A549 or HT1080 cells at various concentrations of compounds and at time points 24 hours (black bars, filled or dotted pattern) and 48 hours (white bars filled or dotted pattern) after treatment.
  • A Treatment with compound A2 of WO 2020/152067 A1 (filled bars) or compound A1 of WO 2020/152067 A1 (bars with dotted pattern).
  • FIG. 2 MetAPI expression levels (in %, normalized to y-Tubulin) in A549 or HT1080 cells at various concentrations of compound B2 of the present invention (filled bars) or compound A56 of WO 2020/152067 A1 (bars with dotted pattern) and at time points 24 hours (black bars, filled or dotted pattern) and 48 hours (white bars filled or dotted pattern).
  • Cell lysates were analyzed via Simple Western and DMSO treated samples were set to 100% MetAPI protein expression.
  • EXAMPLES The preferred exemplary compounds B1 and B2 shown below were synthesized as described in the following Examples 1 and 2, respectively.
  • reaction mixture was than filtered over through Celite, all volatile incredients were removed under reduced pressure and the crude product was purified by column chromatography to provide 3-[4-(12-hydroxy- 4,7,10-trioxa-1-azadodecan-1-yl)-1-oxo-2,3-dihydro-1H-isoindol-2- yl]piperidine-2,6-dione (920 mg, 2.11 mmol, 57% yield) as a colourless oil.
  • reaction mixture was than filtered over through Celite, all volatile incredients were removed under reduced pressure and the crude product was purified by column chromatography to provide tert-butyl 4-(14-hydroxy-3,6,9,12- tetraoxatetradecan-1-yl)piperazine-1-carboxylate (3.78 g, 9.30 mmol, 71% yield) as a colourless oil.
  • the reaction was quenched by the addition of saturated NaHCO3-proof (20 mL), the phases were seperated and the water phase was extracted with DCM (3 x 20 mL).
  • the combinded organic layers were dried over Na 2 SO 4 , the solvents were removed under reduced pressure and the crude product was used directly in the next step.
  • Jess loading was 0.2 mg/ml per sample.
  • Simple Western analysis was performed on a Jess system (ProteinSimple, 004-650) using a 12-230 kDa Separation Module (ProteinSimple, SM-W004) according to the manufacturer’s instructions. Based on primary antibodies against ⁇ -Tubulin (4 ⁇ g/ml; Sigma, T6557) and MetAP2 (1 ⁇ g/ml; CST, 12547) the anti-mouse NIR and the anti- rabbit detection module were used (ProteinSimple, DM-009 and DM-001).
  • the anti-rabbit NIR and the anti-mouse detection module were used (ProteinSimple, DM-007 and DM-002). Data analysis was accomplished using Compass Software Version 5.0.0 (ProteinSimple). The amount of MetAP2 or MetAP1 degradation was determined by quantifying the peak area of MetAP2 or MetAP1 and normalization to the respective ⁇ -Tubulin peak area. Subsequently, DMSO control was set to 100% MetAP2 or MetAP1 protein expression and the MetAP2 or MetAP1 expression of each treatment condition was calculated accordingly.
  • Results were plotted in bar graphs against compound concentration using GraphPad Prism Version 8.2.1 (Fig.1 and Fig.2).
  • Results Treatment with the compounds A1 and A2 of WO 2020/152067 A1 led to a dose-dependent MetAP2 degradation in two cell lines, A549 and HT1080 (Fig. 1A).
  • Compound A56 of WO 2020/152067 A1 was found to have a stronger dose-dependent effect on MetAP2 degradation in these cell lines than compounds A1 and A2 of WO 2020/152067 A1 (Fig.1B).
  • Compound B2 of the present invention was found to have an even stronger effect on MetAP2 degradation than compound A56 of WO 2020/152067 A1 (Fig.1B).
  • No MetAP2 degradation was seen for control compounds C1 and C2 (Fig.1C).
  • MetAP1 was not degraded upon treatment with compound B2 (Fig.2).
  • Example A Injection vials A solution of 100 g of an active ingredient of the formula I and 5 g of disodium hydrogenphosphate in 3 l of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contains 5 mg of active ingredient.
  • Example B Suppositories A mixture of 20 g of an active ingredient of the formula I with 100 g of soya lecithin and 1400 g of cocoa butter is melted, poured into moulds and allowed to cool. Each suppository contains 20 mg of active ingredient.
  • Example C Solution A solution is prepared from 1 g of an active ingredient of the formula I, 9.38 g of NaH2PO4 ⁇ 2 H2O, 28.48 g of Na2HPO4 ⁇ 12 H2O and 0.1 g of benzalkonium chloride in 940 ml of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 l and sterilised by irradiation. This solution can be used in the form of eye drops.
  • Example D Ointment 500 mg of an active ingredient of the formula I are mixed with 99.5 g of Vaseline under aseptic conditions.
  • Example E Tablets A mixture of 1 kg of active ingredient of the formula I, 4 kg of lactose, 1.2 kg of potato starch, 0.2 kg of talc and 0.1 kg of magnesium stearate is pressed in a conventional manner to give tablets in such a way that each tablet contains 10 mg of active ingredient.
  • Example F Dragees Tablets are pressed analogously to Example E and subsequently coated in a conventional manner with a coating of sucrose, potato starch, talc, traga- canth and dye.
  • Example G Capsules 2 kg of active ingredient of the formula I are introduced into hard gelatine capsules in a conventional manner in such a way that each capsule con- tains 20 mg of the active ingredient.
  • Example H Ampoules A solution of 1 kg of active ingredient of the formula I in 60 l of bidistilled water is sterile filtered, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Diabetes (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Les composés de formule (I) Q1-Q2-Q3 dans laquelle Q1, Q2 et Q3 ont les significations indiquées dans la revendication 1, dégradent les protéines cibles, et peuvent être utilisés, entre autres, pour le traitement de maladies telles que le cancer, la sclérose en plaques, les maladies cardiovasculaires, les lésions du système nerveux central et différentes formes d'inflammation.
PCT/EP2023/051464 2022-01-26 2023-01-23 Dérivés hétérocycliques WO2023144053A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22153540 2022-01-26
EP22153540.4 2022-01-26

Publications (1)

Publication Number Publication Date
WO2023144053A1 true WO2023144053A1 (fr) 2023-08-03

Family

ID=80121891

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/051464 WO2023144053A1 (fr) 2022-01-26 2023-01-23 Dérivés hétérocycliques

Country Status (1)

Country Link
WO (1) WO2023144053A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002020740A2 (fr) 2000-09-08 2002-03-14 California Institute Of Technology Medicament chimere de ciblage de proteolyse
WO2012048775A1 (fr) 2010-10-13 2012-04-19 Merck Patent Gmbh Pyrrolidinones en tant qu'inhibiteurs de metap-2
WO2013149704A1 (fr) 2012-04-04 2013-10-10 Merck Patent Gmbh Amides cycliques comme inhibiteurs de metap-2
WO2016020031A1 (fr) 2014-08-04 2016-02-11 Merck Patent Gmbh Dérivés de pyrrolidinone utiles en tant qu'inhibiteurs de la metap -2
WO2018033556A1 (fr) 2016-08-18 2018-02-22 Glaxosmithkline Intellectual Property Development Limited Nouveaux composés
WO2020152067A1 (fr) 2019-01-22 2020-07-30 Merck Patent Gmbh Dérivés hétérocycliques
EP3725771A1 (fr) * 2017-12-13 2020-10-21 Shanghaitech University Agent de dégradation de protéine alk et son application antitumorale

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002020740A2 (fr) 2000-09-08 2002-03-14 California Institute Of Technology Medicament chimere de ciblage de proteolyse
WO2012048775A1 (fr) 2010-10-13 2012-04-19 Merck Patent Gmbh Pyrrolidinones en tant qu'inhibiteurs de metap-2
WO2013149704A1 (fr) 2012-04-04 2013-10-10 Merck Patent Gmbh Amides cycliques comme inhibiteurs de metap-2
WO2016020031A1 (fr) 2014-08-04 2016-02-11 Merck Patent Gmbh Dérivés de pyrrolidinone utiles en tant qu'inhibiteurs de la metap -2
WO2018033556A1 (fr) 2016-08-18 2018-02-22 Glaxosmithkline Intellectual Property Development Limited Nouveaux composés
EP3725771A1 (fr) * 2017-12-13 2020-10-21 Shanghaitech University Agent de dégradation de protéine alk et son application antitumorale
WO2020152067A1 (fr) 2019-01-22 2020-07-30 Merck Patent Gmbh Dérivés hétérocycliques

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
ANGEW. CHEM., vol. 92, 1980, pages 129
BUCKLEY ET AL., J. AM. CHEM. SOC., vol. 134, no. 10, 2012, pages 4465 - 4468
CYRUS K ET AL., CHEMBIOCHEM., vol. 11, 2010, pages 1531 - 1534
FOSTER, ADV. DRUG RES., vol. 14, 1985, pages 1 - 40
GILLETTE ET AL., BIOCHEMISTRY, vol. 33, no. 10, 1994, pages 2927 - 2937
HANZLIK ET AL., J. ORG. CHEM., vol. 55, 1990, pages 3992 - 3997
JARMAN ET AL., CARCINOGENESIS, vol. 16, no. 4, 1993, pages 683 - 688
LI W ET AL., PLOS ONE, vol. 3, 2008, pages e1487
OTTIS ET AL., ACS CHEM. BIOL., vol. 12, no. 4, 2017, pages 892 - 898
OTTIS PHILIPP ET AL: "Proteolysis-Targeting Chimeras: Induced Protein Degradation as a Therapeutic Strategy", ACS CHEMICAL BIOLOGY, vol. 12, no. 4, 21 April 2017 (2017-04-21), pages 892 - 898, XP055819452, ISSN: 1554-8929, Retrieved from the Internet <URL:https://pubs.acs.org/doi/pdf/10.1021/acschembio.6b01068> DOI: 10.1021/acschembio.6b01068 *
REIDER ET AL., J. ORG. CHEM., vol. 52, 1987, pages 3326 - 3334
RODRIGUEZ-GONZALEZ A ET AL., ONCOGENE, vol. 27, 2008, pages 7201 - 7211
SAKAMOTO KM ET AL., PROC NATL ACAD SCI USA., vol. 98, 2001, pages 8554 - 8559
SCHNEEKLOTH JS JR. ET AL., J AM CHEM SOC., vol. 126, 2004, pages 3748 - 3754
WINTER ET AL., SCIENCE, vol. 348, 19 June 2015 (2015-06-19), pages 1376 - 1381
ZHANG D ET AL., BIOORG MED CHEM LETT., vol. 14, 2004, pages 645 - 648

Similar Documents

Publication Publication Date Title
AU2014298959B2 (en) 1,3-disubstituted cyclopentane derivatives
US11026936B2 (en) Piperidinyl-propanone derivatives
US9670142B2 (en) 1,3-diaminocyclopentane carboxamide derivatives
US20220062258A1 (en) Heterocyclic derivatives
CA3129601A1 (fr) Derives d&#39;indazolyl-isoxazole pour le traitement de maladies telles que le cancer
US10118919B2 (en) 3-substituted cyclopentylamine derivatives
WO2023144053A1 (fr) Dérivés hétérocycliques
AU2020393364A1 (en) Condensed thiophene derivatives as hypoxia inducible factor (HIF) inhibitors
AU2014308182A1 (en) 3-substituted cyclopentylamine derivatives

Legal Events

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

Ref document number: 23701482

Country of ref document: EP

Kind code of ref document: A1