WO2018229139A1 - Novel compounds for use in cancer - Google Patents

Novel compounds for use in cancer Download PDF

Info

Publication number
WO2018229139A1
WO2018229139A1 PCT/EP2018/065678 EP2018065678W WO2018229139A1 WO 2018229139 A1 WO2018229139 A1 WO 2018229139A1 EP 2018065678 W EP2018065678 W EP 2018065678W WO 2018229139 A1 WO2018229139 A1 WO 2018229139A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
substituents
compound
formula
partially unsaturated
Prior art date
Application number
PCT/EP2018/065678
Other languages
French (fr)
Inventor
Xabier Aguirre Ena
Julen Oyarzabal Santamarina
Felipe PRÓSPER CARDOSO
Maria Obdulia Rabal Gracia
Edurne SAN JOSÉ ENÉRIZ
Juan Antonio SÁNCHEZ ARIAS
Amaia VILAS ZORNOZA
Original Assignee
Fundación Para La Investigación Médica Aplicada
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 Fundación Para La Investigación Médica Aplicada filed Critical Fundación Para La Investigación Médica Aplicada
Publication of WO2018229139A1 publication Critical patent/WO2018229139A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to new quinoline compounds bearing a hydroxamic acid moiety which are capable to induce cell differentiation in cancer cell lines. It also relates to pharmaceutical or veterinary compositions containing them, and to their use in medicine, in particular as anticancer agents.
  • differentiation agents tend to have less toxicity than conventional cancer treatments.
  • ATRA all-trans retinoic acid
  • APL acute promyelocytic leukemia
  • WO 2015/192981 discloses dual inhibitors of histone methyltransferase G9a and DNA methyltransferases (DNMTs) having a 4-aminoquinoline core of the following formula
  • quinoline compounds bearing a hydroxamic acid moiety which act as cancer cell differentiation inducers as demonstrated by the examples of the invention. Since "stem-like" tumor cells (i.e. cells blocked in initial differentiation stages that can self-renew and differentiate) are considered to be responsible for tumor initiation and therapy resistance in several cancers, the compounds of the invention, as differentiation inducers, represent a promising approach for the treatment and/or prevention of cancer.
  • a first aspect of the invention relates to a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts
  • X is a biradical selected from -NR a -, -0-, -CR b R c -and
  • B is a known ring system which is attached to the quinoline ring through the N atom, and which comprises a 3- to 7-membered saturated or partially unsaturated heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; wherein the ring system B is optionally substituted with: a) one Cy 1 ; and/or
  • Cy 1 and Cy 2 are optionally substituted with one or more substituents independently selected from R d , and Z 2 optionally substituted with one or more substituents R d ;
  • L is a biradical selected from Cy 3 and Z 3 optionally substituted with one or more substituents R d and/or one or more Cy 4 ; wherein Cy 3 and Cy 4 are independently optionally substituted with:
  • L is attached to X through Cy 3 or Z 3 ; and L is attached to -C(0)NHOH through either Cy 3 or Z 3 , or alternatively, through any of the substituents of Cy 3 or Z 3 ;
  • Ri is selected from the group consisting of R , Cy 7 , halogen, -N0 2 , -CN, -OR e , -OC(0)R e , -OC(0)OR e' , -OC(0)NR e' R e' , -NR e R e , -NR e C(0)R e' , -NR e C(0)OR e' , -NR e C(0)NR e R e , -NR e S(0) 2 R e' , -NR e S0 2 NR e R e , -SR e' , -S(0)R e' , -S(0)OR e' , -S0 2 R e' , -S0 2 (OR e ),
  • R 2 is selected from the group consisting of R a , halogen, -N0 2 , -CN, -OR a , -OC(0)R a , -OC(0)OR a' , -OC(0)NR a' R a' , -NR a R a , -NR a C(0)R a' , -NR a C(0)OR a' , -NR a C(0)NR a R a , -NR a S(0) 2 R a , -NR a S0 2 NR a R a , -SR a , -S(0)R a , -S(0)OR a' , -S0 2 R a , -S0 2 (OR a ), -S0 2 NR a R a , -SC(0)NR a' R a' , -C(0)R a' , -C(0)OR a' , -C(0)NR a R
  • R 3 is selected from the group consisting of R 9 , -OR 9 , -NR a R 9 , and -NR a COR 9 ; each R a is independently H or R a ; each R a is independently selected from the group consisting of (CrC 6 )alkyl,
  • R b and R c are independently selected from the group consisting of H, halogen,
  • (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 2 -C 6 )hydrocarbon chain and Cy 9 are independently optionally substituted with one or more halogen atoms; or alternatively
  • R b and R c together with the carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; wherein the ring system is optionally substituted with:
  • each R d is independently selected from halogen, -N0 2 , -CN, -OR 3' , -OC(Y)R a' ,
  • Y is O, S, or NR a ; each R e is independently H or R e ; each R e is independently selected from the group consisting of (CrC 6 )alkyl,
  • each R e is optionally substituted with one or more substituents R a or R d ; each R is independently selected from the group consisting of (CrC 6 )alkyl,
  • each R 9 is independently Cy 14 , or Z 9 optionally substituted with one or more substituents R d and/or one Cy 15 ; wherein Cy 14 or Cy 15 are independently optionally substituted with:
  • Cy 16 and Cy 17 is optionally substituted with one or more substituents independently selected from R d , and Z 10 optionally substituted with one or more substituents R d ; and Cy 7 is a known ring system selected from the group consisting of:
  • Cy 7 is optionally substituted with:
  • Cy 18 or Cy 19 are optionally substituted with one or more substituents independently selected from R d
  • Z 12 optionally substituted with one or more substituents R d
  • Z 1 -Z 12 are independently selected from the group consisting of (CrCi 2 )alkyl
  • any carbon atom of Z 1 -Z 12 is optionally a spiro atom substituted with R h and R', and R h and R', together with the spiro carbon atom, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
  • a second aspect of the invention relates to a pharmaceutical or veterinary composition which comprises a therapeutically effective amount of a compound of formula (I) as defined above, or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of its pharmaceutically or veterinary acceptable salt, together with one or more
  • a third aspect of the invention relates to a compound of formula (I) or a pharmaceutical or veterinary composition as previously defined, for use in the treatment and/or prevention of cancer, in particular in cell differentiation therapy.
  • PG Protective group
  • Protective group refers to a group of atoms that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity.
  • substituted with one or more means that a group can be substituted with one or more, preferably with 1 , 2, 3 or 4 substituents, provided that this group has enough positions susceptible of being substituted.
  • carbocyclic ring system refers to a known ring system wherein all the ring members contain carbon atoms.
  • heterocyclic ring system refers to a known ring system wherein one or more of the ring members, preferably 1 , 2, 3, or 4 ring members, are selected from NH, N, O, and S, where chemically possible. The remaining ring members of the heterocyclic ring are independently selected from C, CH, CH 2 , O, N, NH, and S. Unless otherwise specified, the "heterocyclic” ring system may be attached to the rest of the molecule through a C or a N atom of the ring system. Both the carbocyclic and heterocyclic rings can be saturated, partially unsaturated, or aromatic and may be unsubstituted or substituted as described herein, being the substituents placed on any available position.
  • a ring member of a carbocyclic ring that is CH or CH 2 or in a ring member of a heterocyclic ring that is CH, CH 2 or NH one or more of the H atoms of these ring members may be substituted by another moiety as herein disclosed.
  • heteromatic ring refers to a known aromatic ring system, wherein one or more of the ring members, preferably 1 , 2, 3, or 4 ring members, are selected from NH, N, O, and S, where chemically possible.
  • the remaining ring members of the heteroaromatic ring are independently selected from C, CH, O, N, NH, and S.
  • the heteroaromatic ring may be unsubstituted or substituted as described herein, being the substituents placed on any available position.
  • a ring member of the heteroaromatic ring which is CH or NH the H atom may be substituted by another moiety, as herein disclosed.
  • any of the substituents "Cy” or “Z” may be attached to the rest of the molecule through any available position.
  • the present invention also includes the tautomeric forms of the compounds of formula (I).
  • tautomeric isomers means isomers, the structures of which differ in the position of an atom, generally a hydrogen atom, and of one or more multiple bonds, and which are capable of easily and reversibly changing from one to another.
  • the tautomers are used indistinctly in the present application.
  • a hydroxyphenyl group has to be considered equivalent to its tautomeric form: cyclohexa-2,4-dienone. All tautomers are to be considered equivalent for the purposes of the invention.
  • known ring system refers to a ring system which is chemically feasible and is known in the art and so intends to exclude those ring systems that are not chemically possible.
  • one or two members of the rings are optionally C(O) and/or C(NH) and/or C[N(Ci- C 4 )alkyl].
  • (CrC n )alkyl refers to a saturated branched or linear hydrocarbon chain which contains from 1 to n carbon atoms and only single bonds.
  • the term (C 2 -C n )alkenyl refers to an unsaturated branched or linear hydrocarbon chain which comprises from 2 to n carbon atoms and at least one or more double bonds.
  • the term (C 2 -C n )alkynyl refers to a saturated branched or linear hydrocarbon chain which comprises from 2 to n carbon atoms and at least one or more triple bonds.
  • the (C 2 -C n )hydrocarbon chain having one or more double bonds and one or more triple bonds is a branched or linear hydrocarbon chain which contains from 2 to n carbon atoms.
  • a halogen substituent means fluoro, chloro, bromo or iodo.
  • room temperature is 20-25 °C.
  • salts of the compounds of the invention there is no limitation on the type of salt of the compounds of the invention that can be used, provided that these are pharmaceutically or veterinary acceptable when they are used for therapeutic purposes.
  • pharmaceutically or veterinary acceptable salts embraces salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases.
  • preparation of pharmaceutically or veterinary acceptable salts of the compounds of formula (I) can be carried out by methods known in the art. For instance, they can be prepared from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods.
  • such salts are, for example, prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate pharmaceutically or veterinary acceptable base or acid in water or in an organic solvent or in a mixture of them.
  • the compounds of formula (I) and their salts may differ in some physical properties but they are equivalent for the purposes of the present invention.
  • the compounds of the invention may be in crystalline form either as free solvation compounds or as solvates (e.g. hydrates) and it is intended that both forms are within the scope of the present invention.
  • Methods of solvation are generally known within the art.
  • the solvated forms with pharmaceutically, cosmetically or veterinary acceptable solvents such as water, ethanol and the like are equivalent to the unsolvated form for the purposes of the invention.
  • stereoisomer refers to all isomers of individual compounds that differ only in the orientation of their atoms in space.
  • stereoisomer includes mirror image isomers (enantiomers), mixtures of mirror image isomers (racemates, racemic mixtures), geometric (cis/trans or syn/anti or E/Z) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers).
  • the present invention relates to each of these
  • Diastereoisomers and enantiomers can be separated by conventional techniques such as chromatography or fractional crystallization.
  • Optical isomers can be resolved by conventional techniques of optical resolution to give optically pure isomers. This resolution can be carried out on any chiral synthetic intermediates or on compounds of the invention.
  • Optically pure isomers can also be individually obtained using enantiospecific synthesis.
  • stereoisomers or mixtures of stereoisomers either of any of the compounds of formula (I) or of any of their pharmaceutically acceptable salts are always contemplated even if they are not specifically mentioned.
  • the invention relates to a compound of formula (I) as previously described, wherein X is a biradical -NR a - (i.e. a compound (IA)).
  • R a in (IA) is H, (CrC 6 )alkyl optionally substituted with one or more halogen atoms, or a 3- to 7-membered saturated or partially unsaturated carbocyclic monocyclic ring optionally substituted with one or more halogen atoms. Even more particularly, R a in (IA) is H or (CrC 6 )alkyl optionally substituted with one or more halogen atoms; even more particularly is H or -CH 3 ; and even more particularly H.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein X is a biradical
  • ring B is a known ring system comprising a 3- to 7-membered saturated or partially unsaturated heterocyclic monocyclic ring optionally substituted as previously defined. More particularly, ring B is optionally substituted with: a) one or more substituents R d , and/or b) one or more substituents Z 1 optionally substituted as previously defined. Even more particularly, Z 1 in ring B is (CrC 6 )alkyl optionally substituted with one or more substituents R d .
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein X is a biradical -O- (i.e. a compound (IC)). In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein X is a biradical -CR b R c - (i.e. a compound (ID)).
  • R b and R c in (IC) are independently H, (CrC 6 )alkyl optionally substituted with one or more halogen atoms, or R b and R c together with the carbon atom to which they are attached, form a 3- to 7-membered saturated or partially unsaturated carbocyclic monocyclic ring optionally substituted with one or more halogen atoms.
  • R b and R c in (ID) are independently H or (CrC 6 )alkyl optionally substituted with one or more halogen atoms; even more particularly are H or -CH 3 ; and even both R b and R c are H.
  • the biradical L is attached to X through Cy 3 or Z 3 ; and L is attached to -C(0)NHOH through either Cy 3 or Z 3 , or alternatively, through any of the possible substituents of Cy 3 or Z 3 .
  • L-C(0)NHOH moieties when L is attached to -C(0)NHOH through Cy 3 include, without limitation:
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein L is a biradical selected from the list consisting of:
  • Z 3 is unsubstituted or substituted with one or more substituents R d and/or one or more additional Cy 4 ;
  • Cy 3 and each Cy 4 , wherein Cy 4 either forms part of the linker or is a substituent of Z 3 , are unsubstituted or independently substituted with:
  • each Cy 5 and Cy 6 are independently optionally substituted with one or more substituents independently selected from R d , and additional Z 5 optionally substituted with one or more
  • the invention relates to a compound of formula (I) as previously described, wherein L is a moiety of formula (XVIII):
  • n and n are independently a value selected from 0 to 2;
  • p is a value selected from 0 to 1 ;
  • R j and R k are independently selected from the group consisting of H, halogen, and (CrC 3 )alkyl optionally substituted with one or more halogen atoms; or alternatively R j and R k , together with the spiro carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
  • Cy 20 is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy 20 is optionally substituted with one or more substituents selected from halogen and (CrC 3 )alkyl optionally substituted with one or more halogen atoms,
  • Cy 21 is a known ring system selected from the group consisting of phenyl; 3- to 7- membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 5- or 6-membered heteroaromatic ring; and Cy 21 is optionally substituted with one or more substituents selected from halogen and (CrC 3 )alkyl optionally substituted with one or more halogen atoms.
  • the invention relates to a compound of formula (I) as previously described wherein the moiety -LCONHOH is selected from the group consisting of the following moieties:
  • the invention relates to a compound of formula (I) as previously described wherein the moiety -LCONHOH is selected from the group consisting of the following moieties:
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R-i is selected from the group consisting of R , Cy 7 , -OR e , -NR e R e , -NR e C(0)R e' , -NR e S(0) 2 R e' , -S0 2 NR e R e , and -C(0)NR e R e ; wherein Cy 7 is optionally substituted as previously defined.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R-i is Cy 7 optionally substituted as previously defined. More particularly, Cy 7 is a known ring system selected from the group consisting of:
  • Ri is Cy 7 optionally substituted as previously defined, wherein Cy 7 is a known ring system selected from the group consisting of (i), (ii), and (iii), as defined above.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R-i is Cy 7 optionally substituted as previously defined and is attached to the quinoline through a carbon atom.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R-i is selected from the group consisting of the following moieties:
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R-i is methyl.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R 2 is selected from halogen, -CN and -OR a , more particularly, R 2 is selected from halogen and -OR a ; even more particularly, R 2 is -OR a ; wherein R a is (CrC 6 )alkyl optionally substituted with one or more halogen atoms, and even more particularly R 2 is -OCH 3 .
  • the invention relates to a compound of formula (I) as previously described, wherein R 2 is methyl. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R 3 is methyl.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R 3 is selected from the group consisting of -OR 9 , and -NR a R 9 . More particularly, R 3 is -OR 9 . Even more particularly, R 9 is Z 9 optionally substituted as previously defined. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R 3 is methoxy.
  • the invention relates to a compound of formula (I) as previously described, wherein in R 3 , R 9 contains at least one N atom. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R 3 is a moiety of formula (XIX):
  • Cy 22 is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy 22 is optionally substituted with one or more substituents selected from halogen and (CrC 3 )alkyl optionally substituted with one or more halogen atoms, X 1 and X 2 are independently H or halogen, and r is a value selected from 0 to 6.
  • R 3 is a moiety of formula (XIX) wherein Cy 22 is a 3- to 7-membered saturated heterocyclic monocyclic ring or a 3- to 7-membered saturated carbocyclic or heterocyclic monocyclic ring, which is spiro-fused to a 3- to 7-membered saturated carbocyclic or heterocyclic monocyclic ring, and Cy 22 is optionally substituted as previously defined, X 1 and X 2 are H, and r is a value selected from 0 to 6.
  • the invention in another embodiment, optionally in combination with one or more features of the various embodiments described above or below, relates to a compound of formula (I) as previously described, wherein R 3 is selected from the group consisting of the following moieties:
  • the compound of formula (I) is selected from the group consisting of:
  • R' is an hydroxamic acid protective group, more particularly an hydroxamic acid protective group selected from the group consisting of tetrahydro-2H-pyran-2-yloxy (THP), benzyl, 1 -naphthylmethyl and dimethyloxybenzyl (DMB); R 4 is hydrogen, and R1-R3, X and L are as previously defined.
  • THP tetrahydro-2H-pyran-2-yloxy
  • DMB dimethyloxybenzyl
  • hydroxy protective groups include those where the hydroxy group is either acylated or alkylated such as benzyl, and trityl ethers as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers and allyl ethers.
  • the hydroxamic acid protective group is THP, the deprotection is carried out in acidic medium, for example with HCI, in a suitable solvent such as dioxane, ethyl acetate or methanol.
  • a compound of formula (II) may be obtained from a compound of formula (IV) wherein R 4 is H by reaction with a hydroxylamine of formula (III), wherein R' is an hydroxamic acid protective group as defined above, more particularly an hydroxamic acid protective group selected from the group consisting of tetrahydro-2H-pyran-2-yloxy (THP), benzyl,
  • 1 -naphthylmethyl and dimethyloxybenzyl DMB
  • This conversion can be carried out in the presence of an activating agent such as 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC.HCI) and Hydroxybenzotriazole (HOBt), preferably in the presence of a base, such as N-methylmorpholine (NMM) or diisopropylethylamine (DIEA), in a suitable solvent, such as dichloromethane, chloroform or dimethylformamide, at a temperature comprised from room temperature to the temperature of the boiling point of the solvent, preferably at room temperature.
  • an activating agent such as 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC.HCI) and Hydroxybenzotriazole (HOBt)
  • a base such as N-methylmorpholine (NMM) or di
  • a compound of formula (IV) wherein R 4 is H can be obtained from a compound of formula (IV) wherein R 4 is a carboxy protective group such as (CrC 6 )alkyl, benzyl, p- methoxyphenyl, trimethylsilyl, or [2-(Trimethylsilyl)-ethoxy]methyl (SEM).
  • the deprotection reaction can be carried out by standard methods well-known in the art as described for example in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Chemistry (Wiley, 3rd ed. 1999, Chapter 5, pp. 369-451 ).
  • the carboxy protective group is (CrC 6 )alkyl
  • the deprotection is carried out in basic medium, for example with LiOH in a suitable solvent such as tetrahydrofuran, methanol, water or mixtures thereof.
  • a compound of formula (I) which is a compound of formula (IAa) or a compound formula (IBb) can be obtained from a compound of formula (V) by reacting it with a compound of formula (VI) or a compound of formula (VII), respectively, as shown in the scheme below:
  • X is a halogen atom, preferably CI
  • Q is -LCONHOH or a group capable of being converted to a group -LCONHOH
  • R1-R3, R a , and ring B are as previously defined.
  • the reactions for obtaining a compound of formula (IAa) or a compound of formula (IBb) are carried out optionally in the presence of p-toluenesulfonic acid (PTSA), in a suitable solvent, such as tert-butanol at a suitable temperature, preferably heating at a temperature around 100-120 °C.
  • PTSA p-toluenesulfonic acid
  • the reactions for obtaining a compound of formula (lAa) or a compound of formula (IBb) can be carried out in the presence of a palladium catalyst, such as e.g. Tris(dibenzylideneacetone)dipalladium(0) (Pd 2 (dba) 3 ), an organophosphorus compound, such as e.g.
  • Biphenyl-2-yl-dicyclohexyl-phosphane (2,2'-bis(diphenylphosphino)-1 ,1 '- binaphthyl) (BINAP) or 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos)
  • BINAP 2,2'-bis(diphenylphosphino)-1 ,1 '- binaphthyl)
  • Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene
  • a base such as e.g. Cs 2 C0 3 , sodium tert-butoxide or K 3 P0 4 .
  • the reaction is performed in a suitable solvent, such as e.g. dimethyl ether (DME), toluene or dioxane, at a suitable temperature, preferably heating.
  • DME dimethyl ether
  • a compound of formula (I) which is a compound of formula (ICc) can be obtained from a compound of formula (V), which is firstly converted into a compound of formula (VIII) and then subsequently reacted with a compound of formula (IX) as shown in the scheme below:
  • X is a halogen atom, preferably CI
  • Q is -LCONHOH or a group capable of being converted to a group -LCONHOH
  • LG is a leaving group, such as a methanesulfonate (Ms)
  • R R 3 are as previously defined.
  • the first conversion is carried out with a boronic derivative such as 4,4,5,5-tetramethyl-2- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,3,2-dioxaborolane, in the presence of a palladium catalyst, such as e.g.
  • a suitable solvent such as e.g. dioxane
  • the second conversion is carried out in the presence of a base, such as Cs 2 C0 3 , in a suitable solvent such as N,N-dimethylformamide (DMF) preferably heating at a temperature around 80-120 °C.
  • a base such as Cs 2 C0 3
  • a suitable solvent such as N,N-dimethylformamide (DMF) preferably heating at a temperature around 80-120 °C.
  • a compound of formula (V) can be directly converted into a compound of formula (ICc) by reaction with a compound of formula (IX') HO-Q, optionally in the presence of p-toluenesulfonic acid (PTSA) or a base such as NaH in DMF at a suitable temperature, preferably heating at a temperature around 100-120 °C (PTSA) or 20°C (NaH).
  • PTSA p-toluenesulfonic acid
  • NaH sodium bicarbonate
  • a compound of formula (I) which is a compound of formula (IDd) can be obtained by reacting a compound of formula (V) with a boronic derivative of formula (XX), as shown the scheme below:
  • X is a halogen atom, preferably CI
  • Q is -LCONHOH or a group capable of being converted to a group -LCONHOH
  • each R is H, (CrC 6 )alkyl or, alternatively, two R groups together with the B atom to which they are attached may form a cycle
  • R1-R3, R b and R c are as previously defined.
  • R b or R c being hydrogen
  • this conversion is carried out in the presence of a palladium catalyst, such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh 3 ) 4 ) and KOAc or K 2 C0 3 , in a suitable solvent, such as e.g. dioxane optionally mixed with water, at a suitable temperature, preferably heating at a temperature around 100-120 °C.
  • a palladium catalyst such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh 3 ) 4 ) and KOAc or K 2 C0 3
  • a suitable solvent such as e.g. dioxane optionally mixed with water
  • Q when Q is other than -LCONHOH, it is a group capable of being converted to a group -LCONHOH.
  • Q may be a group -LCO2R" wherein R" is H or a carboxylic acid protective group that is converted into an hydroxamic acid as decribed above. Further conversions of a Q moiety other than -LCONHOH into a Q moiety which is -LCONHOH are exemplified in the examples below.
  • a compound of formula (V) can be obtained from a quinoline of formula (XII) which is firstly converted into a compound of formula (XI). This compound is then reacted with a compound of formula (XIV) to give a compound of formula (X) which is converted into a compound (V) as shown in the scheme below:
  • X is a halogen atom, preferably CI, and RrR 3 are as previously defined.
  • the reduction of the compound of formula (XII) into a compound of formula (XI) is carried out by hydrogenation, e.g. in the presence of Pd/C in a suitable solvent such as methanol, whereas the conversion of a compound of formula (XI) into a compound of formula (X) is carried out in the presence of a halogenating agent, such as e.g. POCI 3 , at a suitable temperature, preferably heating.
  • a halogenating agent such as e.g. POCI 3
  • the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with a boronic derivative of formula RiB(OR) 2 (XV), wherein R-i is R or Cy 7 ; and R is H, (CrC 6 )alkyl or, alternatively, two R groups together with the B atom to which they are attached may form a cycle, in the presence of a palladium catalyst, such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh 3 ) 4 ) and a base, such as e.g. K 2 C0 3 or Na 2 C0 3 , in a suitable solvent, such as e.g.
  • a palladium catalyst such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh 3 ) 4
  • a base such as e.g. K 2 C0 3 or Na 2 C0 3
  • dioxane optionally mixed with water, at a suitable temperature, preferably heating, particularly at about 100-120 °C.
  • this conversion may be carried out with a stannate derivative in the presence of a palladium catalyst, such as e.g. Bis(triphenylphosphine)- palladium(ll) dichloride (Pd(PPh 3 )CI 2 ) in a suitable solvent, such as e.g.
  • a palladium catalyst such as e.g. Bis(triphenylphosphine)- palladium(ll) dichloride (Pd(PPh 3 )CI 2 ) in a suitable solvent, such as e.g.
  • dimethylformamide at a suitable temperature, preferably heating.
  • the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with an alcohol of formula R e OH (XVI) in the same conditions described above when Ri is R or Cy 7 .
  • R-i is -NR e R e
  • the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with an amine of formula HNR e R e (XVII) in the same conditions described above when R-i is R or Cy 7 .
  • the reactions described above can be carried out in a different order.
  • the above described reactions carried out on intermediates already containing substituents Ri-R 3 can also be performed on analogue intermediates containing one or more precursors of substituents R1-R3, which are subsequently transformed into groups
  • Compounds of formula (I) may also be converted into other compounds of formula (I) by reactions well known in the art.
  • the compounds of formulas (III), (VI), (VII), (IX), ( ⁇ '), (XI), (XIV), (XV), (XVI), (XVII) and (XX) are commercially available or can be obtained by conventional synthetic processes.
  • the present invention also relates to a pharmaceutical or veterinary composition
  • a pharmaceutical or veterinary composition comprising an effective amount of a compound of formula (I) as defined above, or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer either of the compound of formula (I) or of their pharmaceutically or veterinary acceptable salts, together with pharmaceutically or veterinary acceptable excipients or carriers.
  • terapéuticaally effective amount refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disease which is addressed.
  • the specific dose of the compound of the invention to obtain a therapeutic benefit may vary depending on the particular circumstances of the individual patient including, among others, the size, weight, age and sex of the patient, the nature and stage of the disease, the aggressiveness of the disease, and the route of administration. For example, a dose of from about 0.01 to about 300 mg/kg may be used.
  • pharmaceutically or veterinary acceptable excipients or carriers refers to pharmaceutically or veterinary acceptable materials, compositions or vehicles. Each component must be pharmaceutically or veterinary acceptable in the sense of being compatible with the other ingredients of the pharmaceutical or veterinary composition. It must also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity or other problems or complications commensurate with a reasonable benefit/risk ratio. The election of the pharmaceutical or veterinary formulation will depend upon the nature of the active compound and its route of administration. Any route of administration may be used, for example oral, parenteral and topical administration.
  • the pharmaceutical or veterinary composition may be formulated for oral administration and may contain one or more physiologically compatible carriers or excipients, in solid or liquid form. These preparations may contain conventional ingredients such as binding agents, fillers, lubricants, and acceptable wetting agents.
  • the pharmaceutical or veterinary composition may be formulated for parenteral administration in combination with conventional injectable liquid carriers, such as water or suitable alcohols.
  • Conventional pharmaceutical or veterinary excipients for injection such as stabilizing agents, solubilizing agents, and buffers, may be included in such
  • compositions which pharmaceutical or veterinary compositions may be injected intramuscularly, intraperitoneally, or intravenously.
  • the pharmaceutical composition may be formulated for topical administration.
  • Formulations include creams, lotions, gels, powders, solutions and patches wherein the compound is dispersed or dissolved in suitable excipients.
  • the pharmaceutical compositions may be in any form, including, among others, tablets, pellets, capsules, aqueous or oily solutions, suspensions, emulsions, or dry powdered forms suitable for reconstitution with water or other suitable liquid medium before use, for immediate or retarded release.
  • the appropriate excipients and/or carriers, and their amounts, can readily be determined by those skilled in the art according to the type of formulation being prepared.
  • the compounds of the invention having a quinoline core, bearing a hydroxamic acid moiety, and being substituted as previously defined have the ability of induce cancer cell differentiation. Therefore, they may be used in cell differentiation therapy.
  • Cell differentitiaton therapy consists of forcing malignant cells to undergo terminal differentiation and become normal cells. This way the maturation process is resumed and tumor phenotypes are eliminated.
  • the invention relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use as a medicament.
  • the invention also relates to an anticancer agent, in particular a cancer cell differentiation inducing agent (or cancer cell differentiation inducer) comprising a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts, wherein the compound of formula (I) is as previously defined.
  • a cancer cell differentiation inducing agent or cancer cell differentiation inducer
  • a compound of formula (I) or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts, wherein the compound of formula (I) is as previously defined.
  • a compound is considered to have the ability of inducing cancer cell differentiation when in a cell differentiation / Annexin-V staining assay such as the one illustrated in the examples gives a percentage of cells with CD1 1 b marker at the tested concentration (at 1/4 of its established Gl 50 ) of at least 10%.
  • the invention relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use in the treatment and/or prevention of cancer; in particular, by cell differentiation therapy (i.e., by inducing cell differentiation in cancer cells).
  • cell differentiation therapy i.e., by inducing cell differentiation in cancer cells.
  • this aspect of the invention relates to the use of a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for the manufacture of a medicament for the treatment and/or prevention of cancer, in particular by cell differentiation therapy.
  • the invention also relates to an anticancer agent, in particular a cancer cell differentiation inducing agent (or cancer cell differentiation inducer) comprising a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts, wherein the compound of formula (I) is as previously defined, which is an inhibitor of cell proliferation.
  • a cancer cell differentiation inducing agent or cancer cell differentiation inducer
  • the compound of formula (I) is as previously defined, which is an inhibitor of cell proliferation.
  • the invention also relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use in the treatment and/or prevention of cancer; in particular, by cell differentiation therapy (i.e., by inducing cell differentiation in cancer cells) and inhibition of cell proliferation.
  • cell differentiation therapy i.e., by inducing cell differentiation in cancer cells
  • This embodiment of the invention relates to the use of a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for the manufacture of a medicament for the treatment and/or prevention of cancer, in particular by cell differentiation therapy and inhibition of cell proliferation.
  • a compound is considered to be an inhibitor of cell proliferation when in a cell proliferation assay such as the one illustrated in the examples gives a GI 5 o below or equal to 10 ⁇ .
  • treatment of the disease refers to stopping or delaying of the disease progress, when the drug is used in the subject exhibiting symptoms of disease onset.
  • prevention refers to stopping or delaying of symptoms of disease onset, when the drug is used in the subject exhibiting no symptoms of disease onset but having high risk of disease onset.
  • the cancer is selected from the group consisting of Non-Hodgkin's Lymphoma, Hodgkin's disease, hepatopancreatic tumor, biliar tumor, gastrointestinal tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, CNS tumor, melanoma, prostate cancer, renal cancer, small-cell lung cancer, non small- cell lung cancer, acute myeloid leukemia (AML), acute lymphoblastic leukemia, and multiple myeloma.
  • Non-Hodgkin's Lymphoma Non-Hodgkin's Lymphoma
  • Hodgkin's disease hepatopancreatic tumor
  • biliar tumor hepatopancreatic tumor
  • gastrointestinal tumor bladder cancer
  • breast cancer cervical cancer
  • colorectal cancer CNS tumor
  • melanoma prostate cancer
  • renal cancer small-cell lung cancer, non small- cell lung cancer, acute myeloid leukemia (AML), acute lymphoblastic leukemia, and multiple mye
  • the HPLC measurement was performed using Gilson 281 from 233 pump (binary), an autosampler, and a UV detector. The fractions were detected by LC-MS.
  • the MS detector was configured with an electrospray ionization source. The source temperature was maintained at 300-350 °C.
  • Method 19 Reverse phase HPLC was carried out on Luna C18 (100 x 30 mm; 5 ⁇ ). Solvent A: water; Solvent B: acetonitrile. Gradient: At room temperature, 10% of B to 40% of B within 9.5 min at 20 mL/min; then 40% B at 20 mL/min over 2 min.
  • Method 20 Reverse phase HPLC was carried out on Luna C18 (100 x 30 mm; 4 ⁇ ). Solvent A: water with 0.075% trifluoroacetic acid; Solvent B: acetonitrile with 0.075% trifluoroacetic acid. Gradient: At room temperature, 20% of B to 40% of B within 6 min at 25 mL/min; then 40% of B at 25 mL/min over 2 min
  • Method 26 Reverse phase HPLC was carried out on Waters Xbridge (150 x 25 mm; 5 ⁇ ). Solvent A: water with 1 mM NH 4 HC0 3 ; Solvent B: acetonitrile. Gradient: At room temperature, 20% of B to 50% of B within 12 min at 20 mL/min; then 100% B at 20 mL/min over 3 min.
  • Method 27 Reverse phase HPLC was carried out on Waters Xbridge (150 x 25 mm; 5 ⁇ ). Solvent A: water with 10 mM NH 4 HC0 3 ; Solvent B: acetonitrile. Gradient: At room temperature, 15% of B to 50% of B within 12 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
  • HPLC-analysis was performed using a Shimadzu LC-20AB or LC-20AD with a Luna- C18(2) column (2.0 X 50 mm, 5 ⁇ ) at 40 °C and UV detection.
  • Solvent A water with 0.056% TFA
  • Solvent B acetonitrile with 0.056% TFA.
  • Gradient After 0.1 minutes at the initial condition of 90% A and 10% B, solvent B was increased to 80% over 4 minutes, maintained at 80% for 0.9 minutes, then a linear gradient to initial conditions was applied for 0.02 minutes and maintained for 0.58 minutes to re-equilibrate the column, giving a cycle time of 5.50 minutes.
  • Flow rate was 0.8 mL/min from 0.01 to 4.90 minutes, increased to 1 .2 mL/min in 0.03 minutes and maintained until the end of the run.
  • Solvent A water with 0.056% TFA
  • Solvent B acetonitrile with 0.056% TFA.
  • Gradient After 0.01 minutes at the initial condition of 100% A, solvent B was increased to 60% over 4 minutes, maintained at 60% for 0.8 minutes, then a linear gradient to initial conditions was applied for 0.02 minutes and maintained for 0.68 minutes to re-equilibrate the column, giving a cycle time of 5.90 minutes.
  • Flow rate was 0.8 mL/min from 0.01 to 5.21 minutes, increased to 1.2 mL/min in 0.02 minutes and maintained until the end of the run.
  • R-06b-2 (9.3 g, 83 mmol) in DMF (100 mL) was added Cs 2 C0 3 (32.4 g, 99 mmol) and iodoethane (15.5 g, 99 mmol) and the mixture was stirred at 20 °C for 5 hours. Then, the mixture was filtered and the filtrated was diluted with water and extracted with MTBE. The combined organic layers were washed with brine, dried over anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure to give R-06b-3 (9.5 g, 82%) as yellow oil. This compound was used in the next step without further characterization.
  • reagent R-06d methyl 4-[[4-(aminomethyl)-1 -piperidyllmethyllbenzoate
  • R-06d-1 methyl 4-formylbenzoate
  • reagent R-06d was obtained, as a white solid (44% yield, 2 steps).
  • ynthesis of reagent R-06e methyl 4-(4-amino-1 -piperidyl)benzoate
  • F is a hydrocarbon chain, a carbocycle, heterocycle, an aryl or heteroaryl
  • R 3 is 0(CrC 6 )alkyl
  • a hydrocarbon chain or a chlorine atom and R, R a , R b and R c are independently a hydrocarbon chain, which optionally contains nitrogen atoms.
  • n and m are 0 or 1
  • Cyi is a carbocycle, heterocycle, carbocyclic spiro, aryl or heteroaryl
  • R 2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl and R a , R b , R c and R d are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
  • intermediate 1-1 1 a 4-[[[6-methoxy-2-(5-methyl-2-furyl)-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminolmethyll-N-tetrahydropyran-2-yloxy-cvclohexanecarboxamide A mixture of intermediate 1-1 Oe (100 mg, 192 ⁇ ), O-tetrahydropyran-2-yl
  • n m and p are 0 or 1
  • Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl
  • R 2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl
  • a hydrocarbon chain or a chlorine atom R a , R b , R c and R d are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
  • X is a chlorine or bromine atom
  • n, m and p are 0 or 1
  • Cyi is a carbocycle, heterocycle, aryl or heteroaryl
  • A is a heterocycle
  • PG is a protecting group
  • R 2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl, hydrocarbon or chlorine
  • R a , R b , R c , R d and R e are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
  • n m and p are 0 or 1
  • Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl
  • R 3 is 0(CrC 6 )alkyl
  • R a , R b , R c , d , R e and R f are independently hydrogen, a hydrocarbon chain, which optionally contains nitrogen atoms.
  • X is a chlorine or bromine atom
  • Z is an oxygen atom or -CH 2 -
  • n, m and p are 0 or 1
  • Cyi is a carbocycle, heterocycle, aryl or heteroaryl
  • A is a heterocycle
  • PG is a protecting group
  • R 2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl and R a , R b , R c , and R d are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
  • n m and p are 0 or 1
  • Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl
  • F is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl and R, R b , R c , and R d are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
  • intermediate l-33a ethyl 2-[4-[[[7-benzyloxy-6-methoxy-2-(5-methyl-2- furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
  • R-06a 1 .65 g, 5.48 mmol, HCI salt
  • Cs 2 C0 3 4.12 g, 13 mmol
  • BINAP 786 mg, 1.26 mmol
  • Pd 2 (dba) 3 (771 mg, 0.842 mmol) in dioxane (30 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 120 °C for 12 hours under N 2 atmosphere.
  • intermediate l-36a ethyl 2-[4-[[[7-(2-azaspiro[3.31heptan-6-yloxy)-6- methoxy-2-(5-methyl-2-furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
  • HCI/EtOAc 10 mL, 2.0M
  • n m and p are 0 or 1
  • Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl
  • R 2 is a hydrocarbon chain
  • R 3 is 0(CrC 6 )alkyl and R a , R b , R c and R d are independently hydrogen or a hydrocarbon chain.
  • n m and p are 0 or 1
  • Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl
  • F is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle
  • R 3 is 0(CrC 6 )alkyl
  • R 4 , R5, R b , R c and R d are independently hydrogen or a
  • reaction mixture was filtered and the filter was concentrated.
  • the residue was disoolved in in POCI 3 (40 mL) and malonic acid (2.15 g, 20.70 mmol) was added.
  • the mixture was stirred at 90 °C for 1 hour.
  • the solution was concentrated and poured into ice-water (20 mL) and adjusted to pH ⁇ 9 with aqueous Na 2 C0 3 .
  • the mixture was extracted with EtOAc 60 mL (x3) and the combined organic layers were washed with brine 50 mL, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure.
  • Cell proliferation was analyzed after 48 hours of in vitro treatment using the CellTiter 96 Aqueous One Solution Cell Proliferation Assay (Promega, Madison, W). This is a colorimetric method for determining the number of viable cells in proliferation
  • HL-60 cells were cultured by triplicate at a density of 1 x10 6 cells/ml in 96-well plates (100.000 cells/well, 10 ⁇ /well). Only the 60 inner wells were used to avoid any border effects.
  • HL-60 cells were cultured at a density of 1 x10 6 cells/ml in 96-well plates (100.000 cells/well, ⁇ ⁇ /well). Cells were treated daily with 1 ⁇ 4 Gl 50 of compounds for 48 hours. After treatment, cells were washed twice with phosphate-buffered saline (PBS). Then cells were stained with 15 ⁇ of APC-conjugated mouse anti-human CD1 1 b (Catalog No. 550019, BD Biosciences) and 1 ⁇ of FITC- conjugated anti-human Annexin V (Catalog No.
  • Comparative example 1 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available (1 -methyl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981.
  • the crude product was purified by prep-HPLC (Method 33 in this document) to obtain comparative example 1 (39% yield) as a yellow solid.
  • ESI-MS (M+1 ): 396.3 calc. for C 23 H 29 N 3 0 3 395.22; HPLC analytical method 1 , Rt 1.656 min.
  • Comparative example 3 was obtained by coupling 7-(3-(pyrrolidin-1 -yl)propoxy)-4-chloro- 6-methoxy-2-(2,5-dimethylfuran-3-yl)quinolin-3(4H)-one (intermediate l-05c in this document) with commercially available tert-butyl 4-(aminomethyl)piperidine-1 -carboxylate (R-07b in this document) under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 , followed by deprotection in HCI/EtOAc and then N-methylation with HCOOH, NaBH 3 CN and (HCHO)n in MeOH (60 °C for 16 hours under N 2 ).
  • Comparative example 4 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available cyclohexylmethanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981.
  • the crude product was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-100% Ethyl acetate/Petroleum ether; gradient 50 mL/min) to obtain comparative example 4 (38% yield) as a light yellow solid.
  • ESI-MS (M+1 ): 381 .3 calc. for C23H28N2O3: 380.2; HPLC analytical method 1 , Rt 2.996 min.
  • Comparative example 5 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available (1 -pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 .
  • the crude product was purified by prep-HPLC (Method 53) to obtain comparative example 5 (44% yield) as a yellow solid.
  • ESI-MS (M+1 ): 460.4 calc. for C26H29N5O3: 459.2; HPLC analytical method 1 , Rt 2.296 min.
  • Comparative example 6 was obtained by coupling 4-chloro-2-(2,5-dimethyl-3-furyl)-6,7- dimethoxy-quinoline (intermediate l-05e in this document) with commercially available (1 - pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 .
  • the crude product was purified by prep-HPLC (Method 54) to obtain comparative example 6 (56% yield) as an off-white solid.
  • ESI-MS (M+1 ): 474.4 calc. for C 27 H 3 i N 5 0 3 : 473.2; HPLC analytical method 1 , Rt 2.345 min.
  • Comparative example 7 was obtained by coupling 4-chloro-2-(2,5-dimethyl-3-furyl)-6- methoxy-7-(3-pyrrolidin-1 -ylpropoxy)quinoline (intermediate l-05c in this document) with commercially available (1 -pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 .
  • the crude product was purified by prep-HPLC (Method 29) to obtain comparative example 7 (13% yield) as an off-white solid.
  • ESI-MS M+1
  • 571.4 calc. for C 3 3H42N 6 0 3 : 570.3; HPLC analytical method 1 , Rt 2.030 min.
  • ATRA all-trans-retinoic acid
  • panobinostat LH-589
  • Table 2 shows the functional response of selected compounds on HL-60 (GI 5 o , which is concentration of compound for 50% of maximal inhibition of cell proliferation); where, Gl 50 ⁇ 10 ⁇ (+), 1 ⁇ ⁇ Gl 50 ⁇ 10 ⁇ (++), and 100 nM ⁇ Gl 50 ⁇ 1 ⁇ (+++), and Gl 50 ⁇ 100 nM (++++).
  • the compounds of the invention in Table 1 are potent inducers of cellular differentiation whereas comparative examples did not show the ability to induce cell differentiation in the same cancer cell lines. Besides, some compounds of the invention (see table 2) are also potent inhibitors of cell proliferation.

Abstract

It relates to the compounds of formula (I), or their pharmaceutically or veterinary acceptable salts, or their stereoisomers or mixtures of stereoisomers, wherein X, L,R1, R 2, and R 3 are as defined herein, which are cancer cell differentiation inducing agents. It also relates to pharmaceutical or veterinary compositions containing them, and to their use in medicine, in particular in the treatment and/or prevention of cancer, in particular by cell differentiation therapy.

Description

Novel compounds for use in cancer
This application claims the benefit of European Patent Application 17382365.9 filed June 14th, 2017
Technical Field
The present invention relates to new quinoline compounds bearing a hydroxamic acid moiety which are capable to induce cell differentiation in cancer cell lines. It also relates to pharmaceutical or veterinary compositions containing them, and to their use in medicine, in particular as anticancer agents.
Background Art The treatments of human tumors either by chemotherapy or by small molecules against specific targets of each type of neoplasm have been directed mostly to kill or eliminate the cancerous cells and in general to decrease cell proliferation.
Recent studies have showed in several types of human tumors the existence of cells or populations of cells blocked in their differentiation state, in some cases also denominated stem-like cancer cells (Campos B. Clinical Cancer Research 2010). These cell populations were often found in initial maturation or differentiation stages and have been shown to be often responsible for tumor progression, metastasis and especially for the resistance to the treatments used to eradicate the human tumors. Because of this, since 1970s, various studies have demonstrated that the strategy of inducing tumoral cells to overcome their blocked differentiation could be an elegant alternative to killing cancer cells (Yan M. Chinese Journal of Cancer 2016). The strategies or therapies of
differentiation, will aim to force the blocked tumor cells in initial maturation or
dedifferentiated states towards its conversion into normal cells by reactivating the endogenous differentiation programs and in this way eliminate the tumor phenotypes of these cells. Generally, differentiation agents tend to have less toxicity than conventional cancer treatments.
The potential for differentiation therapy to improve cure rates is exemplified by the development of all-trans retinoic acid (ATRA) for the targeted treatment of acute promyelocytic leukemia (APL) (Warrell RP Jr. N Engl J Med. 1991 ). However, other types of human tumors, solid and hematological, display resistance to ATRA-based treatment and the types of differentiation inducers in clinical use are still very limited. Hence, the identification of new therapeutic agents able to potently induce cellular differentiation has become an important focus of the human cancer research.
WO 2015/192981 discloses dual inhibitors of histone methyltransferase G9a and DNA methyltransferases (DNMTs) having a 4-aminoquinoline core of the following formula
Figure imgf000003_0001
The compounds disclosed in this PCT application showed inhibition of cell proliferation in several cancer cell lines and primary cultures. However, no data on cancer cell differentiation was reported. Since cancer is the outcome of a complex process during which normal cells acquire aberrant features that enable them to become tumorigenic and/or malignant the development of additional therapies such as cell differentiation therapy is of utmost importance. Thus, there is still a need of developing compounds which show improved activity in the treatment and/or prevention of cancer.
Summary of Invention
Inventors have found new quinoline compounds bearing a hydroxamic acid moiety which act as cancer cell differentiation inducers as demonstrated by the examples of the invention. Since "stem-like" tumor cells (i.e. cells blocked in initial differentiation stages that can self-renew and differentiate) are considered to be responsible for tumor initiation and therapy resistance in several cancers, the compounds of the invention, as differentiation inducers, represent a promising approach for the treatment and/or prevention of cancer.
Similar compounds of the prior art such as the ones described in WO2015/192981 do not show the ability to induce cell differentiation in the same cancer cell lines used for the compounds of the invention. This can be explained because mechanisms leading to differentiation are activated in a different manner than mechanisms involved in cellular proliferation.
Therefore, a first aspect of the invention relates to a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts
Figure imgf000004_0001
(I)
wherein:
X is a biradical selected from -NRa-, -0-, -CRbRc-and
Figure imgf000004_0002
wherein B is a known ring system which is attached to the quinoline ring through the N atom, and which comprises a 3- to 7-membered saturated or partially unsaturated heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; wherein the ring system B is optionally substituted with: a) one Cy1; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z1 optionally substituted with one or more substituents Rd and/or one Cy2;
wherein Cy1 and Cy2 are optionally substituted with one or more substituents independently selected from Rd, and Z2 optionally substituted with one or more substituents Rd;
L is a biradical selected from Cy3 and Z3 optionally substituted with one or more substituents Rd and/or one or more Cy4; wherein Cy3 and Cy4 are independently optionally substituted with:
a) one Cy5; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z4 optionally substituted with one or more substituents Rd and/or one Cy6; wherein Cy5 and Cy6 are independently optionally substituted with one or more substituents independently selected from Rd, and Z5 optionally substituted with one or more substituents Rd;
wherein L is attached to X through Cy3 or Z3; and L is attached to -C(0)NHOH through either Cy3 or Z3, or alternatively, through any of the substituents of Cy3 or Z3;
Ri is selected from the group consisting of R , Cy7, halogen, -N02, -CN, -ORe, -OC(0)Re , -OC(0)ORe', -OC(0)NRe'Re', -NRe Re , -NRe C(0)Re', -NRe C(0)ORe', -NRe C(0)NRe Re , -NRe S(0)2Re', -NRe S02NRe Re , -SRe', -S(0)Re', -S(0)ORe', -S02Re', -S02(ORe ),
-S02NRe Re , -SC(0)NRe'Re', -C(0)Re', -C(0)ORe, -C(0)NRe Re , -C(0)NRe ORe', and -C(0)NRe'S02Re';
R2 is selected from the group consisting of Ra, halogen, -N02, -CN, -ORa , -OC(0)Ra , -OC(0)ORa', -OC(0)NRa'Ra', -NRa Ra , -NRa C(0)Ra', -NRa C(0)ORa', -NRa C(0)NRa Ra , -NRa S(0)2Ra , -NRa S02NRa Ra , -SRa , -S(0)Ra , -S(0)ORa', -S02Ra , -S02(ORa ), -S02NRa Ra , -SC(0)NRa'Ra', -C(0)Ra', -C(0)ORa', -C(0)NRa Ra , -C(0)NRa ORa', and -C(0)NRa'S02Ra';
R3 is selected from the group consisting of R9, -OR9, -NRa R9, and -NRa COR9; each Ra is independently H or Ra; each Ra is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and Cy8, wherein each Ra is optionally substituted with one or more halogen atoms,
Rb and Rc are independently selected from the group consisting of H, halogen,
(CrC6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and a Cy9; wherein (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain and Cy9 are independently optionally substituted with one or more halogen atoms; or alternatively
Rb and Rc, together with the carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; wherein the ring system is optionally substituted with:
a) one Cy10; and/or b) one or more substituents Rd, and/or
c) one or more substituents Z6 optionally substituted with one or more substituents Rd and/or one Cy11;
wherein Cy10 and Cy11 are optionally substituted with one or more substituents independently selected from Rd, and Z7 optionally substituted with one or more substituents Rd; each Rd is independently selected from halogen, -N02, -CN, -OR3', -OC(Y)Ra',
-OC(Y)ORa', -OC(Y)NRa Ra , -NRa Ra , -NRa C(Y)Ra , -NRa C(Y)ORa , -NRa C(Y)NRa Ra , -NRaS(0)2Ra', -NRaS02NRa Ra , -SRa', -S(0)Ra', -S(0)ORa', -S02Ra', -S02(ORa ),
-S02NRa Ra , -SC(Y)NRa Ra , -C(Y)Ra', -C(Y)ORa', -C(Y)NRa Ra , -C(Y)NRaORa , and -C(0)NRa'S02Ra';
Y is O, S, or NRa ; each Re is independently H or Re; each Re is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and Cy12, wherein each Re is optionally substituted with one or more substituents Ra or Rd; each R is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, wherein each R is optionally substituted with:
a) one or more substituents Rd and/or
b) one Cy13 optionally substituted with one or more substituents independently
selected from Rd, and Z8 optionally substituted with one or more substituents Rd; each R9 is independently Cy14, or Z9 optionally substituted with one or more substituents Rd and/or one Cy15; wherein Cy14 or Cy15 are independently optionally substituted with:
a) one Cy16; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z9 optionally substituted with one or more substituents Rd and/or one Cy17;
wherein Cy16 and Cy17 is optionally substituted with one or more substituents independently selected from Rd, and Z10 optionally substituted with one or more substituents Rd; and Cy7 is a known ring system selected from the group consisting of:
(i) phenyl;
(ii) 5- or 6-membered heteroaromatic ring;
(iii) 3- to 7-membered carbocyclic or heterocyclic monocyclic ring, which is saturated or partially unsaturated;
(iv) 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring;
(v) phenyl fused to a 6- to 14-membered saturated or partially unsaturated carbocyclic or heterocyclic bicyclic ring, wherein the rings of the bicyclic ring are spiro-fused; and
(vi) 5- to 6-membered heteroaromatic ring fused to a 6- to 14-membered saturated or partially unsaturated carbocyclic or heterocyclic bicyclic ring, wherein the rings of the bicyclic ring are spiro-fused;
wherein Cy7 is optionally substituted with:
a) one Cy18, and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z11 optionally substituted with one or more substituents Rd and/or one Cy19;
wherein Cy18 or Cy19 are optionally substituted with one or more substituents independently selected from Rd, and Z12 optionally substituted with one or more substituents Rd; Z1-Z12 are independently selected from the group consisting of (CrCi2)alkyl,
(C2-Ci2)alkenyl, (C2-Ci2)alkynyl, and (C2-Ci2)hydrocarbon chain having one or more double bonds and one or more triple bonds, wherein any carbon atom of Z1-Z12 is optionally a spiro atom substituted with Rh and R', and Rh and R', together with the spiro carbon atom, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
Cy1-Cy4, and Cy12-Cy15 are independently a known ring system selected from the group consisting of phenyl; 5- or 6-membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; Cy5-Cy6, Cy8-Cy11, and Cy16-Cy19 are independently a known ring system selected from the group consisting of phenyl; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 5- or 6-membered heteroaromatic ring; wherein in the carbocyclic rings all ring members are carbon atoms; and in the heterocyclic and heteroaromatic rings one or more ring members are selected from N, O, and S; and wherein in all saturated or partially unsaturated rings one or two members of the rings are optionally C(O) and/or C(NH) and/or C[N(C C4)alkyl]. The compounds of the invention may be formulated in different types of compositions. Thus, a second aspect of the invention relates to a pharmaceutical or veterinary composition which comprises a therapeutically effective amount of a compound of formula (I) as defined above, or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of its pharmaceutically or veterinary acceptable salt, together with one or more
pharmaceutically or veterinary acceptable excipients or carriers.
Further, as mentioned above, the compounds of the invention may be used in cancer, in particular in cell differentiation therapy. Accordingly, a third aspect of the invention relates to a compound of formula (I) or a pharmaceutical or veterinary composition as previously defined, for use in the treatment and/or prevention of cancer, in particular in cell differentiation therapy.
Detailed description of the invention
All terms as used herein in this application, unless otherwise stated, shall be understood in their ordinary meaning as known in the art. Other more specific definitions for certain terms as used in the present application are as set forth below and are intended to apply uniformly through-out the specification and claims unless an otherwise expressly set out definition provides a broader definition.
"Protective group" (PG) refers to a group of atoms that when attached to a reactive group in a molecule masks, reduces or prevents that reactivity. The expression "substituted with one or more" means that a group can be substituted with one or more, preferably with 1 , 2, 3 or 4 substituents, provided that this group has enough positions susceptible of being substituted. The term "carbocyclic" ring system refers to a known ring system wherein all the ring members contain carbon atoms. The term "heterocyclic" ring system refers to a known ring system wherein one or more of the ring members, preferably 1 , 2, 3, or 4 ring members, are selected from NH, N, O, and S, where chemically possible. The remaining ring members of the heterocyclic ring are independently selected from C, CH, CH2, O, N, NH, and S. Unless otherwise specified, the "heterocyclic" ring system may be attached to the rest of the molecule through a C or a N atom of the ring system. Both the carbocyclic and heterocyclic rings can be saturated, partially unsaturated, or aromatic and may be unsubstituted or substituted as described herein, being the substituents placed on any available position. Thus, in a ring member of a carbocyclic ring that is CH or CH2 or in a ring member of a heterocyclic ring that is CH, CH2 or NH, one or more of the H atoms of these ring members may be substituted by another moiety as herein disclosed.
For the purposes of the present invention, in "fused" rings the fusion occurs through one bond which is common to two adjoining rings; in "bridged-fused" rings the fusion occurs through a sequence of atoms (bridgehead) which is common to two rings; and in "spiro- fused" rings, the fusion occurs through only one atom (spiro atom), preferably a carbon atom, which is common to two adjoining rings (including bridged rings). The term "heteroaromatic" ring refers to a known aromatic ring system, wherein one or more of the ring members, preferably 1 , 2, 3, or 4 ring members, are selected from NH, N, O, and S, where chemically possible. The remaining ring members of the heteroaromatic ring are independently selected from C, CH, O, N, NH, and S. The heteroaromatic ring may be unsubstituted or substituted as described herein, being the substituents placed on any available position. Thus, in a ring member of the heteroaromatic ring which is CH or NH the H atom may be substituted by another moiety, as herein disclosed.
In the present invention any of the substituents "Cy" or "Z" may be attached to the rest of the molecule through any available position.
The present invention also includes the tautomeric forms of the compounds of formula (I). The term "tautomeric isomers" means isomers, the structures of which differ in the position of an atom, generally a hydrogen atom, and of one or more multiple bonds, and which are capable of easily and reversibly changing from one to another. The tautomers are used indistinctly in the present application. Thus, as an example, a hydroxyphenyl group has to be considered equivalent to its tautomeric form: cyclohexa-2,4-dienone. All tautomers are to be considered equivalent for the purposes of the invention. The term "known ring system" as used herein refers to a ring system which is chemically feasible and is known in the art and so intends to exclude those ring systems that are not chemically possible. For the purposes of the present invention, in all saturated or partially unsaturated rings, one or two members of the rings are optionally C(O) and/or C(NH) and/or C[N(Ci- C4)alkyl].
The term (CrCn)alkyl refers to a saturated branched or linear hydrocarbon chain which contains from 1 to n carbon atoms and only single bonds. The term (C2-Cn)alkenyl refers to an unsaturated branched or linear hydrocarbon chain which comprises from 2 to n carbon atoms and at least one or more double bonds. The term (C2-Cn)alkynyl refers to a saturated branched or linear hydrocarbon chain which comprises from 2 to n carbon atoms and at least one or more triple bonds. For the purposes of the invention, the (C2-Cn)hydrocarbon chain having one or more double bonds and one or more triple bonds is a branched or linear hydrocarbon chain which contains from 2 to n carbon atoms.
In the (d-Cn)alkyl chains, (C2-Cn)alkenyl chains, (C2-Cn)alkynyl chains, and
(C2-Cn)hydrocarbon chains having one or more double bonds and one or more triple bonds, is is contemplated that any carbon of the chain is optionally a spiro atom substituted as defined herein.
A halogen substituent means fluoro, chloro, bromo or iodo. For the purposes of the invention, room temperature is 20-25 °C.
In the embodiments of the invention referring to the compounds of formula (I), where the substitution or unsubstitution of a certain group is not specified, e.g. either because it is not indicated a certain substitution for that group or that the group is unsubstituted, it has to be understood that the possible substitution of this group is the one as in the definition of the formula (I). The same applies when in specific group is said to be "optionally substituted" without specifically indicating the substitution.
There is no limitation on the type of salt of the compounds of the invention that can be used, provided that these are pharmaceutically or veterinary acceptable when they are used for therapeutic purposes. The term "pharmaceutically or veterinary acceptable salts", embraces salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. The preparation of pharmaceutically or veterinary acceptable salts of the compounds of formula (I) can be carried out by methods known in the art. For instance, they can be prepared from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. Generally, such salts are, for example, prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate pharmaceutically or veterinary acceptable base or acid in water or in an organic solvent or in a mixture of them. The compounds of formula (I) and their salts may differ in some physical properties but they are equivalent for the purposes of the present invention.
The compounds of the invention may be in crystalline form either as free solvation compounds or as solvates (e.g. hydrates) and it is intended that both forms are within the scope of the present invention. Methods of solvation are generally known within the art. In general, the solvated forms with pharmaceutically, cosmetically or veterinary acceptable solvents such as water, ethanol and the like are equivalent to the unsolvated form for the purposes of the invention.
Some compounds of the invention can have chiral centres that can give rise to various stereoisomers. As used herein, the term "stereoisomer" refers to all isomers of individual compounds that differ only in the orientation of their atoms in space. The term
stereoisomer includes mirror image isomers (enantiomers), mixtures of mirror image isomers (racemates, racemic mixtures), geometric (cis/trans or syn/anti or E/Z) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers). The present invention relates to each of these
stereoisomers and also mixtures thereof.
Diastereoisomers and enantiomers can be separated by conventional techniques such as chromatography or fractional crystallization. Optical isomers can be resolved by conventional techniques of optical resolution to give optically pure isomers. This resolution can be carried out on any chiral synthetic intermediates or on compounds of the invention. Optically pure isomers can also be individually obtained using enantiospecific synthesis.
In all embodiments of the invention referring to the compounds of formula (I), the pharmaceutically, cosmetically or veterinary acceptable salts thereof and the
stereoisomers or mixtures of stereoisomers, either of any of the compounds of formula (I) or of any of their pharmaceutically acceptable salts are always contemplated even if they are not specifically mentioned.
Figure imgf000012_0001
(ΙΑ) (IB) (IC) (ID)
In one embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein X is a biradical -NRa- (i.e. a compound (IA)).
More particularly, Ra in (IA) is H, (CrC6)alkyl optionally substituted with one or more halogen atoms, or a 3- to 7-membered saturated or partially unsaturated carbocyclic monocyclic ring optionally substituted with one or more halogen atoms. Even more particularly, Ra in (IA) is H or (CrC6)alkyl optionally substituted with one or more halogen atoms; even more particularly is H or -CH3; and even more particularly H.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein X is a biradical
Figure imgf000012_0002
(i.e. a compound (IB)). More particularly, ring B is a known ring system comprising a 3- to 7-membered saturated or partially unsaturated heterocyclic monocyclic ring optionally substituted as previously defined. More particularly, ring B is optionally substituted with: a) one or more substituents Rd, and/or b) one or more substituents Z1 optionally substituted as previously defined. Even more particularly, Z1 in ring B is (CrC6)alkyl optionally substituted with one or more substituents Rd.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein X is a biradical -O- (i.e. a compound (IC)). In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein X is a biradical -CRbRc- (i.e. a compound (ID)).
More particularly, Rb and Rc in (IC) are independently H, (CrC6)alkyl optionally substituted with one or more halogen atoms, or Rb and Rc together with the carbon atom to which they are attached, form a 3- to 7-membered saturated or partially unsaturated carbocyclic monocyclic ring optionally substituted with one or more halogen atoms. Even more particularly, Rb and Rc in (ID) are independently H or (CrC6)alkyl optionally substituted with one or more halogen atoms; even more particularly are H or -CH3; and even both Rb and Rc are H.
In the compounds of formula (I), the biradical L is attached to X through Cy3 or Z3; and L is attached to -C(0)NHOH through either Cy3 or Z3, or alternatively, through any of the possible substituents of Cy3 or Z3. Thus, some illustrative examples of L-C(0)NHOH moieties when L is attached to -C(0)NHOH through Cy3 include, without limitation:
Figure imgf000013_0001
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein L is a biradical selected from the list consisting of:
1 ) -Cy3-;
2) -Cy3-Cy5-;
3) -Cy3-Cy5-Z5-;
4) -Cy3-Z4-;
5) -Cy3-Z4-Cy6-; 6) -Cy3-Z4-Cy6-Z5-;
7) -Z3-;
8) -Z3-Cy4-;
9) -Z3-Cy4-Cy5-;
10) -Z3-Cy4-Cy5-Z5-;
1 1 ) -Z3-Cy4-Z4-;
12) -Z3-Cy4-Z4-Cy6-; and
13) -Z3-Cy4-Z4-Cy6-Z5-;
wherein:
the left part of the biradicals 1 ) to 13) as drawn above is attached to X, and the right part is attached to -C(0)NHOH;
Z3 is unsubstituted or substituted with one or more substituents Rd and/or one or more additional Cy4;
Cy3 and each Cy4, wherein Cy4 either forms part of the linker or is a substituent of Z3, are unsubstituted or independently substituted with:
a) one additional Cy5; and/or
b) one or more substituents Rd, and/or
c) one additional Z4 optionally substituted with one or more substituents Rd and/or one additional Cy6;
each Cy5 and Cy6, either when forming part of the linker or as substituents, are independently optionally substituted with one or more substituents independently selected from Rd, and additional Z5 optionally substituted with one or more
substituents Rd.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein L is a moiety of formula (XVIII):
Figure imgf000014_0001
(XVIII)
wherein:
m and n are independently a value selected from 0 to 2;
p is a value selected from 0 to 1 ;
Rj and Rk are independently selected from the group consisting of H, halogen, and (CrC3)alkyl optionally substituted with one or more halogen atoms; or alternatively Rj and Rk, together with the spiro carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
Cy20 is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy20 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms,
Cy21 is a known ring system selected from the group consisting of phenyl; 3- to 7- membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 5- or 6-membered heteroaromatic ring; and Cy21 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms.
In the compound of formula (I), the moiety of formula (XVIII) is attached to X
through -(CRjRk)m, and is attached to -C(0)NHOH through (Cy21)p.
For example, when L is a moiety of formula (XVIII) as defined above wherein m, n, p = 0, then L is attached to -C(0)NHOH through Cy20 (corresponding to Cy3 in the definition of the compounds of formula (I) as indicated above). When L is a moiety of formula (XVIII) as defined above wherein m is 0 and one or both of n and p are other than 0, then L is attached to -C(0)NHOH through a substituent of the cycle Cy20, which is and embodiment of Cy3 in the definition of the compounds of formula (I) as indicated above. When L is a moiety of formula (XVIII) as defined above wherein m is other than 0, then L is attached to -C(0)NHOH through a substituent of the moiety -(CRjRk)m-, e.g. through Cy20 when n, p = 0, which is and embodiment of Z3 in the definition of the compounds of formula (I) as indicated above).
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described wherein the moiety -LCONHOH is selected from the group consisting of the following moieties:
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000017_0002
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described wherein the moiety -LCONHOH is selected from the group consisting of the following moieties:
Figure imgf000017_0003
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R-i is selected from the group consisting of R , Cy7, -ORe, -NRe Re , -NRe C(0)Re', -NRe S(0)2Re', -S02NRe Re , and -C(0)NRe Re ; wherein Cy7 is optionally substituted as previously defined.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R-i is Cy7 optionally substituted as previously defined. More particularly, Cy7 is a known ring system selected from the group consisting of:
(i) phenyl;
(ii) 5- or 6-membered heteroaromatic ring;
(iii) 3- to 7-membered carbocyclic or heterocyclic monocyclic ring, which is saturated or partially unsaturated;
(iv) 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to
7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring;
and even more particulaly, Ri is Cy7 optionally substituted as previously defined, wherein Cy7 is a known ring system selected from the group consisting of (i), (ii), and (iii), as defined above.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R-i is Cy7 optionally substituted as previously defined and is attached to the quinoline through a carbon atom.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R-i is selected from the group consisting of the following moieties:
Figure imgf000018_0001
Figure imgf000019_0001
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R-i is methyl.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R2 is selected from halogen, -CN and -ORa , more particularly, R2 is selected from halogen and -ORa ; even more particularly, R2 is -ORa; wherein Ra is (CrC6)alkyl optionally substituted with one or more halogen atoms, and even more particularly R2 is -OCH3.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R2 is methyl. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R3 is methyl.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R3 is selected from the group consisting of -OR9, and -NRa R9. More particularly, R3 is -OR9. Even more particularly, R9 is Z9 optionally substituted as previously defined. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R3 is methoxy.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein in R3, R9 contains at least one N atom. In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R3 is a moiety of formula (XIX):
Cy22-
Figure imgf000020_0001
(XIX)
wherein
Cy22 is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy22 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms, X1 and X2 are independently H or halogen, and r is a value selected from 0 to 6.
More particularly, R3 is a moiety of formula (XIX) wherein Cy22 is a 3- to 7-membered saturated heterocyclic monocyclic ring or a 3- to 7-membered saturated carbocyclic or heterocyclic monocyclic ring, which is spiro-fused to a 3- to 7-membered saturated carbocyclic or heterocyclic monocyclic ring, and Cy22 is optionally substituted as previously defined, X1 and X2 are H, and r is a value selected from 0 to 6.
In another embodiment, optionally in combination with one or more features of the various embodiments described above or below, the invention relates to a compound of formula (I) as previously described, wherein R3 is selected from the group consisting of the following moieties:
Figure imgf000020_0002
Figure imgf000021_0001
In another embodiment of the invention, the compound of formula (I) is selected from the group consisting of:
Figure imgf000021_0002
1-04 1-05 1-06
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
-17 2-18 2-19
Figure imgf000025_0001
Figure imgf000026_0001
3-13 3-14 3-15
Figure imgf000027_0001
-22 3-23 3-24
Figure imgf000028_0001
-01 10-01 10-02 Regarding the compounds indicated as "cis isomer" or "trans isomer", an aleatory absolute configuration of the cis or trans isomers is shown. In the examples it is clearly indicated which of the isomers is concerned in relative terms by differentiating
unambiguously between cis and trans isomers by their physical and/or spectroscopic properties.
Processes for the preparation of compounds of formula (I) are also part of the invention as well as intermediates used in these processes. Generally, compounds of formula (I) as defined above may be obtained by reacting a compound of formula (II) as shown in the scheme below:
Figure imgf000029_0001
Scheme 1
wherein R' is an hydroxamic acid protective group, more particularly an hydroxamic acid protective group selected from the group consisting of tetrahydro-2H-pyran-2-yloxy (THP), benzyl, 1 -naphthylmethyl and dimethyloxybenzyl (DMB); R4 is hydrogen, and R1-R3, X and L are as previously defined.
The removal of the protective group of the hydroxamic acid is carried out by standard methods well-known in the art as described for example in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Chemistry (Wiley, 3rd ed. 1999, Chapter 2, pp. 17- 200). Representative hydroxy protective groups include those where the hydroxy group is either acylated or alkylated such as benzyl, and trityl ethers as well as alkyl ethers, tetrahydropyranyl ethers, trialkylsilyl ethers and allyl ethers. When the hydroxamic acid protective group is THP, the deprotection is carried out in acidic medium, for example with HCI, in a suitable solvent such as dioxane, ethyl acetate or methanol.
A compound of formula (II) may be obtained from a compound of formula (IV) wherein R4 is H by reaction with a hydroxylamine of formula (III), wherein R' is an hydroxamic acid protective group as defined above, more particularly an hydroxamic acid protective group selected from the group consisting of tetrahydro-2H-pyran-2-yloxy (THP), benzyl,
1 -naphthylmethyl and dimethyloxybenzyl (DMB). This conversion can be carried out in the presence of an activating agent such as 1 -ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC.HCI) and Hydroxybenzotriazole (HOBt), preferably in the presence of a base, such as N-methylmorpholine (NMM) or diisopropylethylamine (DIEA), in a suitable solvent, such as dichloromethane, chloroform or dimethylformamide, at a temperature comprised from room temperature to the temperature of the boiling point of the solvent, preferably at room temperature.
A compound of formula (IV) wherein R4 is H can be obtained from a compound of formula (IV) wherein R4 is a carboxy protective group such as (CrC6)alkyl, benzyl, p- methoxyphenyl, trimethylsilyl, or [2-(Trimethylsilyl)-ethoxy]methyl (SEM). The deprotection reaction can be carried out by standard methods well-known in the art as described for example in T. W. Green and P. G. M. Wuts, Protective Groups in Organic Chemistry (Wiley, 3rd ed. 1999, Chapter 5, pp. 369-451 ). When the carboxy protective group is (CrC6)alkyl, the deprotection is carried out in basic medium, for example with LiOH in a suitable solvent such as tetrahydrofuran, methanol, water or mixtures thereof.
Moreover, a compound of formula (I) which is a compound of formula (IAa) or a compound formula (IBb) can be obtained from a compound of formula (V) by reacting it with a compound of formula (VI) or a compound of formula (VII), respectively, as shown in the scheme below:
Figure imgf000030_0001
(IBb)
Scheme 2
wherein X is a halogen atom, preferably CI, Q is -LCONHOH or a group capable of being converted to a group -LCONHOH, and R1-R3, Ra , and ring B are as previously defined.
In both cases the reactions for obtaining a compound of formula (IAa) or a compound of formula (IBb) are carried out optionally in the presence of p-toluenesulfonic acid (PTSA), in a suitable solvent, such as tert-butanol at a suitable temperature, preferably heating at a temperature around 100-120 °C. Alternatively, the reactions for obtaining a compound of formula (lAa) or a compound of formula (IBb) can be carried out in the presence of a palladium catalyst, such as e.g. Tris(dibenzylideneacetone)dipalladium(0) (Pd2(dba)3), an organophosphorus compound, such as e.g. Biphenyl-2-yl-dicyclohexyl-phosphane, (2,2'-bis(diphenylphosphino)-1 ,1 '- binaphthyl) (BINAP) or 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene (Xantphos), and a base, such as e.g. Cs2C03, sodium tert-butoxide or K3P04. The reaction is performed in a suitable solvent, such as e.g. dimethyl ether (DME), toluene or dioxane, at a suitable temperature, preferably heating. A compound of formula (I) which is a compound of formula (ICc) can be obtained from a compound of formula (V), which is firstly converted into a compound of formula (VIII) and then subsequently reacted with a compound of formula (IX) as shown in the scheme below:
Figure imgf000031_0001
(V) (VIII) (ICc) Scheme 3
wherein X is a halogen atom, preferably CI, Q is -LCONHOH or a group capable of being converted to a group -LCONHOH, LG is a leaving group, such as a methanesulfonate (Ms), and R R3 are as previously defined. The first conversion is carried out with a boronic derivative such as 4,4,5,5-tetramethyl-2- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-1 ,3,2-dioxaborolane, in the presence of a palladium catalyst, such as e.g. [1 ,1 '-Bis(diphenylphosphino)ferrocene]palladium(ll) dichloride (Pd(dppf)CI2) and KOAc, in a suitable solvent, such as e.g. dioxane, at a suitable temperature, preferably heating at a temperature around 100-120 °C; and then by reacting the intermediate obtained with hydrogen peroxide in a suitable solvent, such as dichloromethane.
The second conversion is carried out in the presence of a base, such as Cs2C03, in a suitable solvent such as N,N-dimethylformamide (DMF) preferably heating at a temperature around 80-120 °C.
Alternatively, a compound of formula (V) can be directly converted into a compound of formula (ICc) by reaction with a compound of formula (IX') HO-Q, optionally in the presence of p-toluenesulfonic acid (PTSA) or a base such as NaH in DMF at a suitable temperature, preferably heating at a temperature around 100-120 °C (PTSA) or 20°C (NaH).
A compound of formula (I) which is a compound of formula (IDd) can be obtained by reacting a compound of formula (V) with a boronic derivative of formula (XX), as shown the scheme below:
Figure imgf000032_0001
Scheme 4
wherein X is a halogen atom, preferably CI, Q is -LCONHOH or a group capable of being converted to a group -LCONHOH, each R is H, (CrC6)alkyl or, alternatively, two R groups together with the B atom to which they are attached may form a cycle, and R1-R3, Rb and Rc are as previously defined.
For either Rb or Rc being hydrogen, this conversion is carried out in the presence of a palladium catalyst, such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh3)4) and KOAc or K2C03, in a suitable solvent, such as e.g. dioxane optionally mixed with water, at a suitable temperature, preferably heating at a temperature around 100-120 °C.
In the compounds (lAa), (IBb), (ICc) and (IDd) above, when Q is other than -LCONHOH, it is a group capable of being converted to a group -LCONHOH. For example, Q may be a group -LCO2R" wherein R" is H or a carboxylic acid protective group that is converted into an hydroxamic acid as decribed above. Further conversions of a Q moiety other than -LCONHOH into a Q moiety which is -LCONHOH are exemplified in the examples below.
A compound of formula (V) can be obtained from a quinoline of formula (XII) which is firstly converted into a compound of formula (XI). This compound is then reacted with a compound of formula (XIV) to give a compound of formula (X) which is converted into a compound (V) as shown in the scheme below:
Figure imgf000033_0001
Figure imgf000033_0002
Scheme 5
wherein X is a halogen atom, preferably CI, and RrR3 are as previously defined.
The reduction of the compound of formula (XII) into a compound of formula (XI) is carried out by hydrogenation, e.g. in the presence of Pd/C in a suitable solvent such as methanol, whereas the conversion of a compound of formula (XI) into a compound of formula (X) is carried out in the presence of a halogenating agent, such as e.g. POCI3, at a suitable temperature, preferably heating.
When Ri is R or Cy7, the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with a boronic derivative of formula RiB(OR)2 (XV), wherein R-i is R or Cy7; and R is H, (CrC6)alkyl or, alternatively, two R groups together with the B atom to which they are attached may form a cycle, in the presence of a palladium catalyst, such as e.g. Tetrakis(triphenylphosphine)-palladium(0) (Pd(PPh3)4) and a base, such as e.g. K2C03 or Na2C03, in a suitable solvent, such as e.g. dioxane optionally mixed with water, at a suitable temperature, preferably heating, particularly at about 100-120 °C. Alternatively, this conversion may be carried out with a stannate derivative in the presence of a palladium catalyst, such as e.g. Bis(triphenylphosphine)- palladium(ll) dichloride (Pd(PPh3)CI2) in a suitable solvent, such as e.g.
dimethylformamide, at a suitable temperature, preferably heating.
When Ri is -ORe, the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with an alcohol of formula ReOH (XVI) in the same conditions described above when Ri is R or Cy7. When R-i is -NRe Re , the conversion of a compound of formula (X) into a compound of formula (V) may be carried out with an amine of formula HNRe Re (XVII) in the same conditions described above when R-i is R or Cy7. Alternatively, the reactions described above can be carried out in a different order. Thus, for example the above described reactions carried out on intermediates already containing substituents Ri-R3 can also be performed on analogue intermediates containing one or more precursors of substituents R1-R3, which are subsequently transformed into groups
Compounds of formula (I) may also be converted into other compounds of formula (I) by reactions well known in the art. The compounds of formulas (III), (VI), (VII), (IX), (ΙΧ'), (XI), (XIV), (XV), (XVI), (XVII) and (XX) are commercially available or can be obtained by conventional synthetic processes.
The present invention also relates to a pharmaceutical or veterinary composition comprising an effective amount of a compound of formula (I) as defined above, or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer either of the compound of formula (I) or of their pharmaceutically or veterinary acceptable salts, together with pharmaceutically or veterinary acceptable excipients or carriers.
The expression "therapeutically effective amount" as used herein, refers to the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disease which is addressed. The specific dose of the compound of the invention to obtain a therapeutic benefit may vary depending on the particular circumstances of the individual patient including, among others, the size, weight, age and sex of the patient, the nature and stage of the disease, the aggressiveness of the disease, and the route of administration. For example, a dose of from about 0.01 to about 300 mg/kg may be used.
The expression "pharmaceutically or veterinary acceptable excipients or carriers" refers to pharmaceutically or veterinary acceptable materials, compositions or vehicles. Each component must be pharmaceutically or veterinary acceptable in the sense of being compatible with the other ingredients of the pharmaceutical or veterinary composition. It must also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity or other problems or complications commensurate with a reasonable benefit/risk ratio. The election of the pharmaceutical or veterinary formulation will depend upon the nature of the active compound and its route of administration. Any route of administration may be used, for example oral, parenteral and topical administration. For example, the pharmaceutical or veterinary composition may be formulated for oral administration and may contain one or more physiologically compatible carriers or excipients, in solid or liquid form. These preparations may contain conventional ingredients such as binding agents, fillers, lubricants, and acceptable wetting agents. The pharmaceutical or veterinary composition may be formulated for parenteral administration in combination with conventional injectable liquid carriers, such as water or suitable alcohols. Conventional pharmaceutical or veterinary excipients for injection, such as stabilizing agents, solubilizing agents, and buffers, may be included in such
compositions. These pharmaceutical or veterinary compositions may be injected intramuscularly, intraperitoneally, or intravenously.
The pharmaceutical composition may be formulated for topical administration.
Formulations include creams, lotions, gels, powders, solutions and patches wherein the compound is dispersed or dissolved in suitable excipients. The pharmaceutical compositions may be in any form, including, among others, tablets, pellets, capsules, aqueous or oily solutions, suspensions, emulsions, or dry powdered forms suitable for reconstitution with water or other suitable liquid medium before use, for immediate or retarded release. The appropriate excipients and/or carriers, and their amounts, can readily be determined by those skilled in the art according to the type of formulation being prepared.
As mentioned above, the compounds of the invention having a quinoline core, bearing a hydroxamic acid moiety, and being substituted as previously defined have the ability of induce cancer cell differentiation. Therefore, they may be used in cell differentiation therapy. Cell differentitiaton therapy consists of forcing malignant cells to undergo terminal differentiation and become normal cells. This way the maturation process is resumed and tumor phenotypes are eliminated. Thus, the invention relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use as a medicament.
Further, the invention also relates to an anticancer agent, in particular a cancer cell differentiation inducing agent (or cancer cell differentiation inducer) comprising a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts, wherein the compound of formula (I) is as previously defined.
For the purposes of the invention, a compound is considered to have the ability of inducing cancer cell differentiation when in a cell differentiation / Annexin-V staining assay such as the one illustrated in the examples gives a percentage of cells with CD1 1 b marker at the tested concentration (at 1/4 of its established Gl50) of at least 10%.
Moreover, the invention relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use in the treatment and/or prevention of cancer; in particular, by cell differentiation therapy (i.e., by inducing cell differentiation in cancer cells).
Thus, this aspect of the invention relates to the use of a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for the manufacture of a medicament for the treatment and/or prevention of cancer, in particular by cell differentiation therapy.
It may also be formulated as a method for the treatment and/or prevention of cancer; in particular, by cell differentiation therapy, comprising administering an effective amount of the previously defined compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, and one or more
pharmaceutically or veterinary acceptable excipients or carriers, in a subject in need thereof, including a human.
Further, some compounds of the invention are also inhibitors of cell proliferation. Thus, in one embodiment of the invention, optionally in combination with one or more features of the various embodiments described above or below, the invention also relates to an anticancer agent, in particular a cancer cell differentiation inducing agent (or cancer cell differentiation inducer) comprising a compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts, wherein the compound of formula (I) is as previously defined, which is an inhibitor of cell proliferation. Therefore, the invention also relates to a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for use in the treatment and/or prevention of cancer; in particular, by cell differentiation therapy (i.e., by inducing cell differentiation in cancer cells) and inhibition of cell proliferation.
This embodiment of the invention relates to the use of a compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, for the manufacture of a medicament for the treatment and/or prevention of cancer, in particular by cell differentiation therapy and inhibition of cell proliferation.
It may also be formulated as a method for the treatment and/or prevention of cancer; in particular, by cell differentiation therapy and inhibition of cell proliferation, comprising administering an effective amount of the previously defined compound of formula (I) or a pharmaceutical composition comprising the compound of formula (I) as defined above, and one or more pharmaceutically or veterinary acceptable excipients or carriers, in a subject in need thereof, including a human. For the purposes of the invention, a compound is considered to be an inhibitor of cell proliferation when in a cell proliferation assay such as the one illustrated in the examples gives a GI5o below or equal to 10μΜ.
For the purposes of the invention, the term "treatment" of the disease refers to stopping or delaying of the disease progress, when the drug is used in the subject exhibiting symptoms of disease onset. The term "prevention" refers to stopping or delaying of symptoms of disease onset, when the drug is used in the subject exhibiting no symptoms of disease onset but having high risk of disease onset. In one embodiment, optionally in combination with one or more features of the various embodiments described above or below, the cancer is selected from the group consisting of Non-Hodgkin's Lymphoma, Hodgkin's disease, hepatopancreatic tumor, biliar tumor, gastrointestinal tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, CNS tumor, melanoma, prostate cancer, renal cancer, small-cell lung cancer, non small- cell lung cancer, acute myeloid leukemia (AML), acute lymphoblastic leukemia, and multiple myeloma.
Throughout the description and claims the word "comprise" and variations of the word, are not intended to exclude other technical features, additives, components, or steps.
Furthermore, the word "comprise" encompasses the case of "consisting of". Additional objects, advantages and features of the invention will become apparent to those skilled in the art upon examination of the description or may be learned by practice of the invention. The following examples are provided by way of illustration, and they are not intended to be limiting of the present invention. Furthermore, the present invention covers all possible combinations of particular and preferred embodiments described herein. Examples General Procedure for Preparative HPLC purification method:
The HPLC measurement was performed using Gilson 281 from 233 pump (binary), an autosampler, and a UV detector. The fractions were detected by LC-MS. The MS detector was configured with an electrospray ionization source. The source temperature was maintained at 300-350 °C.
HPLC Methods (purification methods):
General conditions for methods 1 -18, 21 -25, 28-54: Reverse phase HPLC was carried out on Synergi Poar-RP (250 x 50 mm; 5 μηι) (for method 17) or on Luna C18 (100 X 30 mm; 5 μηι) (for methods 1 -16, 18, 21 -25, 28-54). Solvent A: water with trifluoroacetic acid (0.1 % for methods 1 -16, 21 -25, 28-54 and 0.075% for methods 17 and 18; Solvent B:
acetonitrile.
Method Gradient (at room temperature)
1 30% of B to 55% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
2 20% of B to 45% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
3 10% of B to 35% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
4 20% of B to 50% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 5 min.
5 15% of B to 45% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 5 min.
6 20% of B to 60% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
7 16% of B to 46% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 3 min.
8 10% of B to 40% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
9 15% of B to 45% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
10 5% of B to 40% of B within 12 min at 20 mL/min; then 100% B at 20 mL/min over 2 min.
1 1 20% of B to 50% of B within 12 min at 20 mL/min; then 100% B at 20 mL/min over 2 min.
12 5% of B to 30% of B within 12 min at 20 mL/min; then 100% B at 20 mL/min over 2 min.
13 20% of B to 40% of B within 10 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
14 20% of B to 50% of B within 10 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
15 30% of B to 60% of B within 10 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
16 15% of B to 45% of B within 10.5 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
17 15% of B to 45% of B within 3 min at 80 mL/min.
18 5% of B to 38% of B within 5 min at 25 mL/min.
21 20% of B to 30% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
22 15% of B to 40% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
23 30% of B to 55% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
24 15% of B to 35% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
25 15% of B to 40% of B within 12 min at 20 mL/min then 100% B at 20 mL/min over 2 min.
28 30% of B to 55% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
29 15% of B to 45% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
30 20% of B to 35% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
31 20% of B to 55% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
32 20% of B to 50% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
33 3% of B to 36% of B within 20 min at 80 mL/min; then 100% B at 80 mL/min over 15 min.
34 18% of B to 48% of B within 10.5 min at 25 mL/min; then 100% B at 25 mL/min over 3 min.
35 10% of B to 40% of B within 10 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
36 20% of B to 45% of B within 10 min at 25 mL/min; then 100% B at 25 mL/min over 2 min. Method Gradient (at room temperature)
37 20% of B to 40% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
38 25% of B to 55% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
39 10% of B to 50% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
40 30% of B to 60% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
41 15% of B to 40% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
42 35% of B to 50% of B within 12 min at 25 mL/min then 100% B at 25 mL/min over 3 min.
43 25% of B to 45% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
44 20% of B to 35% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 3 min.
45 10% of B to 30% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
46 30% of B to 50% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
47 25% of B to 55% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
48 25% of B to 50% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
49 25% of B to 40% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
50 30% of B to 45% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
51 40% of B to 80% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
52 30% of B to 70% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
53 10% of B to 60% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
54 20% of B to 70% of B within 10 min at 25 mL/min then 100% B at 25 mL/min over 2 min.
Method 19: Reverse phase HPLC was carried out on Luna C18 (100 x 30 mm; 5 μηη). Solvent A: water; Solvent B: acetonitrile. Gradient: At room temperature, 10% of B to 40% of B within 9.5 min at 20 mL/min; then 40% B at 20 mL/min over 2 min.
Method 20: Reverse phase HPLC was carried out on Luna C18 (100 x 30 mm; 4 μηι). Solvent A: water with 0.075% trifluoroacetic acid; Solvent B: acetonitrile with 0.075% trifluoroacetic acid. Gradient: At room temperature, 20% of B to 40% of B within 6 min at 25 mL/min; then 40% of B at 25 mL/min over 2 min
Method 26: Reverse phase HPLC was carried out on Waters Xbridge (150 x 25 mm; 5 μηη). Solvent A: water with 1 mM NH4HC03; Solvent B: acetonitrile. Gradient: At room temperature, 20% of B to 50% of B within 12 min at 20 mL/min; then 100% B at 20 mL/min over 3 min.
Method 27: Reverse phase HPLC was carried out on Waters Xbridge (150 x 25 mm; 5 μηη). Solvent A: water with 10 mM NH4HC03; Solvent B: acetonitrile. Gradient: At room temperature, 15% of B to 50% of B within 12 min at 25 mL/min; then 100% B at 25 mL/min over 2 min.
General procedure for HPLC analysis:
HPLC-analysis was performed using a Shimadzu LC-20AB or LC-20AD with a Luna- C18(2) column (2.0 X 50 mm, 5 μπι) at 40 °C and UV detection.
Method 1
Solvent A: water with 0.056% TFA; Solvent B: acetonitrile with 0.056% TFA. Gradient: After 0.1 minutes at the initial condition of 90% A and 10% B, solvent B was increased to 80% over 4 minutes, maintained at 80% for 0.9 minutes, then a linear gradient to initial conditions was applied for 0.02 minutes and maintained for 0.58 minutes to re-equilibrate the column, giving a cycle time of 5.50 minutes. Flow rate was 0.8 mL/min from 0.01 to 4.90 minutes, increased to 1 .2 mL/min in 0.03 minutes and maintained until the end of the run.
Method 2
Solvent A: water with 0.056% TFA; Solvent B: acetonitrile with 0.056% TFA. Gradient: After 0.01 minutes at the initial condition of 100% A, solvent B was increased to 60% over 4 minutes, maintained at 60% for 0.8 minutes, then a linear gradient to initial conditions was applied for 0.02 minutes and maintained for 0.68 minutes to re-equilibrate the column, giving a cycle time of 5.90 minutes. Flow rate was 0.8 mL/min from 0.01 to 5.21 minutes, increased to 1.2 mL/min in 0.02 minutes and maintained until the end of the run.
The following abbreviations have been used in the examples:
AcOH: acetic acid, BINAP: 2,2'bis(diphenylphospinio)-1 ,1 '-binaphthyl; BOC: tert- butoxycarbonyl; calc: calculated; DCM: dichloromethane; DEAD: diethylazodicarboxylate; DIEA: N,N-Diisopropiletilamina; DME: 1 ,2-dimethoxyethane; DMF: dimethylformamide; DMSO: dimethylsulfoxide; EDC: 1 -ethyl-3-(3- dimethylamino-propyl)carbodiimide; eq: equivalent; ESI-MS: electrospray ionization mass spectrometry; EtOAc: ethyl acetate; EtOH: ethanol; HOBt: Hydroxybenzo-triazole; HPLC: High-performance liquid
chromatography; KHMDS: Potassium bis(trimethylsilyl)amide; MeOH: methanol; MTBE: tert-butyl methyl ether; MW: microwaves; NaOAc: sodium acetate; NMM: N-methyl morpholine; PhNTf2: N-Phenyl-bis(trifluoromethanesulfonimide); rt: room temperature; Rt: Retention time; t-BuOK: potassium tert-butoxide; t-BuONa: sodium tert-butoxide; TEA: triethylamine; TFA: trifluoroacetic acid; THF: tetrahydrofuran; THP: tetrahydropyran; TLC: thin layer chromatography; xantphos: 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene. Synthesis of reagent R-06b: ethyl 3-[[4-(aminomethyl)-1 -piperidyllmethyllcvclobutane- carboxylate
Figure imgf000040_0001
Conditions: a) KOH (4.0 eq), EtOH/H20 (1 :1 ), 80 °C for 16 h; b) Cs2C03 (1.2 eq), iodoethane (1.2 eq), DMF, 20 °C for 5 h; c) BH3 THF (1.0 eq), THF; d) oxalyl chloride (2.0 eq), DMSO (4.0 eq), TEA (10.0 eq), DCM, -78 °C; e) NaBH3CN (1.2 eq), AcOH (1.1 eq), tert-butyl N-(4-piperidylmethyl)- carbamate (1.1 eq), MeOH, 20 °C for 3 h; f) HCI/EtOAc (2.0 M), 20 °C for 1 h.
Preparation of R-06b-2: 3-methylenecvclobutanecarboxylic acid
To a solution of commercially available 3-methylenecyclobutanecarbonitrile (R-06b-1 , 10 g, 107 mmol) in EtOH/H20 (1 :1 , 200 ml.) was added KOH (24.1 g, 430 mmol) and the mixture was stirred at 80 °C for 16 hours. Then, EtOH was removed under reduced pressure and then the solution was cooled to below 10 °C and acidified with 1 M HCI to pH 1 . The mixture was extracted with EtOAc and the combined organic layer was dried over anhydrous Na2S04, filtered and concentrated to obtain R-06b-2 (9.3 g, 77%) as yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06b-3: ethyl 3-methylenecvclobutanecarboxylate
To a solution of R-06b-2 (9.3 g, 83 mmol) in DMF (100 mL) was added Cs2C03 (32.4 g, 99 mmol) and iodoethane (15.5 g, 99 mmol) and the mixture was stirred at 20 °C for 5 hours. Then, the mixture was filtered and the filtrated was diluted with water and extracted with MTBE. The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated under reduced pressure to give R-06b-3 (9.5 g, 82%) as yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06b-4: ethyl 3-(hvdroxymethyl)cvclobutanecarboxylate
To a solution of R-06b-3 (9.5 g, 68 mmol) in anhydrous THF (100 mL) at -10 °C was added dropwise BH3-THF (1 M, 68 mL) and the resulting reaction mixture was allowed to warm to 20 °C and stir for 2 hours. Then, the reaction mixture was cooled to - 20 °C. Methanol (50 mL) was added and the solution was stirred for 15 minutes followed by the addition of 10% aqueous NaOH (1 .36 g, 34 mmol) and H202 (1 1.52 g, 9.77 mL, 30% purity) sequentially. The resultant reaction mixture was stirred at 20 °C for 2 hours. Then, the mixture was warmed to room temperature and a saturated aqueous NH4CI solution was added. The mixture was extracted with DCM and the combined organic layer was washed with brine, dried over anhydrous Na2S04, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 2/1 ) to obtain pure R-06b-4 (6.2 g, 58%) as light yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06b-5: ethyl 3-formylcvclobutanecarboxylate
To a solution of oxalyl dichloride (9.3 g, 73 mmol) in DCM (5 mL) was added DMSO (1 1.46 g, 146 mmol) as a solution in DCM (2 mL) slowly at -78 °C. The resultant solution was stirred for 30 minutes and then a solution of R-06b-4 (5.80 g, 36 mmol) in DCM (5 mL) was added dropwise. The mixture was stirred at -78 °C for 2 hours. Then TEA (37.1 g, 366 mmol) was added to the mixture and the reaction solution was stirred at -78 °C for 30 minutes. Then, the mixture was warmed to room temperature and a saturated aqueous NH4CI solution was added. The mixture was extracted with DCM and the combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 5/1 to 2/1 ) to obtain R-06b-5 (2.1 g, 37%) as yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06b-6: ethyl 3-[[4-[(tert-butoxycarbonylamino)methyll-1 -piperidyllmethyll- cyclobutanecarboxylate
To a solution of R-06b-5 (2.1 g, 13 mmol) in MeOH (20 mL) was added NaBH3CN (1 .01 g, 16 mmol), AcOH (847 mg, 14 mmol) and tert-butyl N-(4-piperidylmethyl)carbamate (3.03 g, 14 mmol) and the mixture was stirred at 20 °C for 3 hours. Then, the reaction was quenched with water and MeOH was removed under reduce pressure. The residue was extracted with EtOAc and the combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 2/1 to 0/1 ) to obtain R-06b-6 (1 .5 g, 31 %) as a white solid. This compound was used in the next step without further characterization.
Preparation of reagent R-06b: ethyl 3-[[4-(aminomethyl)-1 -piperidyllmethyllcvclobutane- carboxylate
A solution of R-06b-6 (300 mg, 846 mol) in HCI/EtOAc (10 mL, 2.0 M) was stirred at 20 °C for 1 hour. Then, the solution was concentrated and the residue was diluted with a saturated aqueous NaHC03 solution (pH=8). The solution was extracted with EtOAc and the combined organic layers were dried over anhydrous Na2S04, filtered and concentrated to give R-06b (180 mg, 83%) as a light yellow solid. ESI-MS (M+1 ): 255.2 calc. for
Figure imgf000042_0001
Conditions: a) K2C03 (2.0 eq), tert-butyl N-(4-piperidylmethyl)carbamate (1.0 eq), DMSO, 90 °C for 16 h; b) HCI/EtOAc (2.0 M), 20 °C for 1 h.
Preparation of R-06c-2: methyl 4-[4-[(tert-butoxycarbonylamino)methyll-1 -piperidyll- benzoate
To a solution of commercially available R-06c-1 (500 mg, 3.24 mmol) in DMSO (35 mL) was added K2C03 (897 mg, 6.49 mmol) and tert-butyl N-(4-piperidylmethyl)carbamate (695 mg, 3.24 mmol) and the mixture was stirred at 90 °C for 16 hours. Then, the mixture was filtered and the filtrate was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 5/1 to 2/1 ) to obtain pure R-06c-2 (430 mg, 38%) as a white solid. ESI-MS (M+1 ): 349.2 calc. for Ci9H28N204:
348.2.
Preparation of reagent R-06c: methyl 4-[4-(aminomethyl)-1 -piperidyllbenzoate
A solution of R-06c-2 (430 mg, 1.23 mmol) in HCI/EtOAc (15 mL, 2.0 M) was stirred at 20 °C for 1 hour. Then, the mixture was concentrated to give a residue. The residue was diluted with MeOH, alkalined with a saturated aqueous solution of NaHC03, filtered, and concentrated to give R-06c (230 mg, 75%) as a white solid. ESI-MS (M+1 ): 249.3 calc. for
Figure imgf000043_0001
Figure imgf000043_0002
Synthesis of reagent R-06d: methyl 4-[[4-(aminomethyl)-1 -piperidyllmethyllbenzoate
Conditions: a) NaBH3CN (2.2 eq), AcOH (1.1 eq), tert-butyl N-(4-piperidylmethyl)carbamate (1.1 eq), MeOH, 80 °C for 3 h; b) HCI/EtOAc (2.0 M), 20 °C for 1 h.
Preparation of reagent R-06d: methyl 4-[[4-(aminomethyl)-1 -piperidyllmethyllbenzoate Following the same process for reagent R-06b starting from R06b-5, but using R-06d-1 (methyl 4-formylbenzoate) instead of R06b-5, reagent R-06d was obtained, as a white solid (44% yield, 2 steps). ESI-MS (M+1 ): 263.3 calc. for Ci5H22N202: 262.2. ynthesis of reagent R-06e: methyl 4-(4-amino-1 -piperidyl)benzoate
Figure imgf000044_0001
Conditions: a) K2C03 (2.5 eq), DMSO, 90 °C for 16 h; b) HCI/EtOAc (2.0 M), 25 °C for 16 h.
Preparation of reagent R-06e: methyl 4-(4-amino-1 -piperidyl)benzoate
Following the same process for reagent R-06c starting from R06c-1 , but using R-06e-2 (methyl 4-fluorobenzoate) and R-06e-1 (tert-butyl N-(4-piperidyl)carbamate) instead of R06c-1 and tert-butyl N-(4-piperidylmethyl)carbamate, reagent R-06e was obtained (21 % yield, 2 steps). ESI-MS (M+1 ): 235.1 calc. for Ci3H18N202: 234.2. Synthesis of reagent R-06f: ethyl 4-[4-(aminomethyl)phenyllcvclohexanecarboxylate
Figure imgf000044_0002
Figure imgf000044_0003
Conditions: a) pyridine (1.1 eq), trifluoromethylsulfonyl trifluoromethanesulfonate (1.1 eq), toluene, 40 °C for 3.5 h; b) (4-formylphenyl)boronic acid (0.8 eq), Pd/C, PPh3 (0.1 eq), DME/H20 (2:1 ), 80 °C for 16 h; c) NH2OH HCI (1.0 eq), NaOAc (0.8 eq), EtOH/H20 (2:1 ), 20 °C for 16 h; d) tert- butoxycarbonyl tert-butyl carbonate (1.05 eq), TEA (1.5 eq), Pd/C, H2 (50 Psi, 344.7 KPa), EtOH, 20 °C for 16 h; e) HCI/EtOAc (2.0 M), 20 °C for 16 h.
Preparation of R-06f-2: ethyl 4-(trifluoromethylsulfonyloxy)cvclohex-3-ene-1 -carboxylate To a solution of pyridine (2.56 g, 32.31 mmol) in toluene (35 mL) was added
trifluoromethylsulfonyl trifluoromethanesulfonate (9.12 g, 32.31 mmol) at 20 °C and the mixture was stirred for 30 minutes. Then R-06f-1 (5 g, 29.38 mmol) was added and the mixture was heated to 40 °C and stirred for 3.5 hours. Then, the reaction mixture was cooled to 20 °C and quenched with water. The mixture was extracted with EtOAc and the combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 5/1 ) to obtain R-06f-2 (2.7 g, 30%) as a yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06f-3: ethyl 4-(4-formylphenyl)cvclohex-3-ene-1 -carboxylate A mixture of (4-formylphenyl)boronic acid (1 g, 6.67 mmol), R-06f-2 (2.42 g, 8.00 mmol), Pd/C (100 mg, 10% purity) and PPh3 (157 mg, 600 mol) in DME (50 mL) and H20 (25 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 °C for 16 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 20/1 to 5/1 ) to obtain R-06f-3 (950 mg, 55%) as a yellow solid. ESI- MS (M+1 ): 259.2 calc. for Ci6H1803: 258.2.
Preparation of R-06f-4: ethyl 4-[4-[(E)-hvdroxyiminomethyllphenyllcvclohex-3-ene-1 - carboxylate
To a solution of R-06f-3 (950 mg, 3.68 mmol) in EtOH (5 mL) and H20 (2.5 mL) was added NH2OH-HCI (256 mg, 3.68 mmol) and NaOAc (241 mg, 2.94 mmol) and the mixture was stirred at 20 °C for 16 hours. Then, the reaction mixture was concentrated under reduced pressure to remove EtOH and the residue was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 10/1 to 5/1 ) to obtain R-06f-4 (980 mg, 97%) as a yellow solid. ESI-MS (M+1 ): 274.2 calc. for
Figure imgf000045_0001
Preparation of R-06f-5: ethyl 4-[4-[(tert-butoxycarbonylamino)methyllphenyll- cyclohexanecarboxylate
To a solution of tert-butoxycarbonyl tert-butyl carbonate (822 mg, 3.76 mmol) in EtOH (20 mL) was added R-06f-4 (980 mg, 3.59 mmol), TEA (544 mg, 5.4 mmol) and Pd/C (10% purity, 100 mg) under N2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (50 Psi, 344.7 KPa) at 20 °C for 16 hours. Then, the mixture was filtered and the filtrate was concentrated to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 5/1 to 2/1 ) to obtain R-06f-5 (950 mg, 73%) as yellow oil. ESI-MS (M+1 ): 362.2 calc. for
Figure imgf000045_0002
Preparation of R-06f: ethyl 4-[4-(aminomethyl)phenyllcvclohexanecarboxylate
A solution of R-06f-5 (950 mg, 2.63 mmol) in HCI/EtOAc (20 mL, 2.0 M) was stirred at 20 °C for 16 hours. Then, the mixture was concentrated to give R-06f (350 mg, 51 %) as light yellow oil. ESI-MS (M+1 ): 262.2 calc. for Ci6H23N02: 261 .2.
Synthesis of reagent R-06g: methyl 4-[4-(aminomethyl)phenyllbenzoate
Figure imgf000046_0001
Figure imgf000046_0002
R-06g-5 R-06g
Conditions: a) K2C03 (2.0 eq), Pd(PPh3)4 (0.1 eq), 1 ,4-dioxane/H20 (10:1 ), 100 °C for 16 h; b) NH2OH HCI (1.0 eq), NaOAc (0.8 eq), EtOH, 25 °C for 16 h; c) BOC20 (1.0 eq), Pd/c, EtOH, H2 (50 Psi, 344.7 KPa), 20 °C for 16 h; d) HCI/EtOAc (2.0 M), 20 °C for 5 h.
Preparation of R-06g-3: methyl 4-(4-formylphenyl)benzoate
A mixture of R-06g-1 (2.00 g, 13.34 mmol), R-06g-2 (2.87 g, 13.34 mmol), K2C03 (3.69 g, 26.68 mmol) and Pd(PPh3)4 (1.54 g, 1.33 mmol) in 1 ,4-dioxane/H20 (10:1 , 1 10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 16 hours under N2 atmosphere. Then, the mixture was cooled to 20 °C and concentrated in reduced pressure at 40 °C to give a residue. The residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give a crude product. The residue was purified by column chromatography (Si02, DCM/MeOH = 1/0 to 1/1 ) to obtain R-06g-3 (1 .20 g, 37%) as a yellow solid. ESI-MS (M+1 ): 241.1 calc. for d5H1203: 240.1 .
Preparation of R-06g-4: methyl 4-[4-[(E)-hvdroxyiminomethyllphenyllbenzoate
A mixture of R-06g-3 (1 .00 g, 4.16 mmol), NH2OH-HCI (289 mg, 4.16 mmol) and NaOAc
(273 mg, 3.33 mmol) in EtOH (40 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 16 hours under N2 atmosphere. Then, the mixture was cooled to 20 °C and concentrated in reduced pressure at 40 °C to give a residue. The residue was poured into water and extracted with DCM. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give R-06g-4 (500 mg, 47%) as a yellow solid. ESI-MS (M+1 ): 256.1 calc. for Ci5H13N03: 255.1 .
Preparation of R-06g: methyl 4-[4-(aminomethyl)phenyllbenzoate
Following the same process for reagent R-06f starting from R06f-4, but using R-06g-4 instead of R06f-4, reagent R-06g was obtained as a white solid (53% yield, 2 steps). ESI- MS (M+1 ): 242.1 calc. for Ci5H15N02: 241 .1 .
Synthesis of reagent R-06h: methyl 4-[(4-amino-1 -piperidyl)methyllbenzoate
Figure imgf000047_0001
R-06h-3
Conditions: a) NaBH3CN (3.0 eq), AcOH (1.05 eq), MeOH, 70 °C for 2 h; b) HCI/EtOAc (2.0 M), 20 °C for 5 h. Preparation of R-06h: methyl 4-[(4-amino-1 -piperidyl)methyllbenzoate
Following the same process for reagent R-06b starting from R06b-5, but using R-06h-1 (methyl 4-formylbenzoate) and R-06h-2 (tert-butyl N-(4-piperidyl)carbamate) instead of R06b-5 and tert-butyl N-(4-piperidylmethyl)carbamate, reagent R-06h was obtained as a white solid (24% yield, 2 steps). ESI-MS (M+1 ): 249.1 calc. for C14H20N2O2: 248.2.
Synthesis of reagent R-06i: methyl 4-[4-(aminomethyl)cvclohexyllbenzoate
Figure imgf000047_0002
R-06i-5 R-06i
Conditions: a) toluenesulfonyl isocyanide (1.3 eq), t-BuOK (2.4 eq), DME/EtOH, 0 °C for 1 h, then 20 °C for 2 h; b) TFA, DCM, 20 °C for 16 h; c) KHMDS (1.3 eq), THF, -78 °C, then PhNTf2 (1.3 eq), 20 °C for 8 h; d) (4-methoxycarbonylphenyl)boronic acid (0.8 eq), Pd(PPh3)4 (0.08 eq), K2C03 (1.6 eq), 1 ,4-dioxane/H20 (10:1 ), 90 °C for 1 h; e) HCI, Pd(OH)2/C, MeOH, H2 (50 Psi, 344.7 KPa), 30 °C for 16 h. Preparation of R-06i-2: 1 ,4-dioxaspiro[4.5ldecane-8-carbonitrile
To a cooled (-10 °C) suspension of R-06i-1 (10.00 g, 64.03 mmol) and toluenesulfonyl isocyanide (16.15 g, 83.24 mmol) in 1 ,2-dimethoxyethane (200 mL) containing EtOH (6.5 mL) was added portionwise t-BuOK (17.24 g, 153.67 mmol) maintaining the temperature below 5 °C. Then the reaction was stirred for 1 hour at 0 °C and 2 hours at 20 °C. The mixture was concentrated to give a solid. The residue was diluted with water and saturated brine. The mixture was filtered through Celite and extracted with MTBE. The combined extracts were washed with brine, dried over Na2S04, filtered and concentrated to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 5/1 to 2/1 ) to obtain R-06i-2 (7.00 g, 65%) as colorless oil. This compound was used in the next step without further characterization.
Preparation of R-06i-3: 4-oxocyclohexanecarbonitrile
A solution of R-06i-2 (7.00 g, 41.87 mmol) in TFA (50 mL) and DCM (50 mL) was stirred at 20 °C for 16 hours. Then, the mixture was concentrated to give the crude product. The crude product was purify by column chromatography (Si02, Petroleum ether/Ethyl acetate = 10/1 to 5/1 ) to obtain R-06i-3 (4.50 g, 87%) as yellow oil. This compound was used in the next step without further characterization.
Preparation of R-06i-4: (4-cvanocvclohexen-1 -yl) trifluoromethanesulfonate
To a stirred solution of R-06i-3 (4.00 g, 32.48 mmol) and KHMDS (1 .0 M, 42.87 mL, 42.87 mmol) in THF (200 mL) under N2 at -78 °C was added PhNTf2 (14.85 g, 41 .57 mmol) and the reaction mixture was stirred at 20 °C for 8 hours. Then, the mixture was cooled to 20 °C and concentrated in reduced pressure at 40 °C. The residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 50/1 to 1 :1 ) to give R-06i- 4 (3.00 g, 36%) as a colorless oil. ESI-MS (M+1 ): 256.1 calc. for C8H8F3N03S: 255.0.
Preparation of R-06i-5: methyl 4-(4-cvanocvclohexen-1 -yl)benzoate
A mixture of R-06i-4 (2.50 g, 9.80 mmol), (4-methoxycarbonylphenyl)boronic acid (1.47 g, 8.16 mmol), Pd(PPh3)4 (943 mg, 816 mol) and K2C03 (2.26 g, 16.33 mmol) in 1 ,4- dioxane/H20 (10:1 , 55 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 1 hour under N2 atmosphere. Then, the mixture was cooled to 20 °C and concentrated in reduced pressure at 40 °C. The residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give a crude product. The crude product was purified by column chromatography (Si02,
Petroleum ether/Ethyl acetate = 40/1 to 2/1 ) to give R-06i-5 (2.00 g, 99%) as a yellow solid. ESI-MS (M+1 ): 242.1 calc. for Ci5H15N02: 241 .1 . Preparation of R-06i: methyl 4-[4-(aminomethyl)cvclohexyllbenzoate
To a solution of R-06i-5 (1.50 g, 6.22 mmol) and HCI (1 .23 mL, 36% purity) in MeOH (20 mL) was added Pd(OH)2/C (10%, 500 mg) under H2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (50 Psi, 344.7 KPa) at 30 °C for 16 hours. Then, the reaction mixture was concentrated in vacuum to give a residue. The crude product was purified by prep-HPLC (Method 26) to give R-06i (200 mg, 13%) as a white solid. ESI-MS (M+1 ): 248.2 calc. for Ci5H21N02: 247.2.
Preparation of reagent R-07d: benzyl 4-(1 -aminocvclopropyl)piperidine-1 -carboxylate To a solution of benzyl 4-cyanopiperidine-1 -carboxylate (10 g, 40.94 mmol) and Ti(i-PrO)4 (7.56 g, 26.61 mmol, 7.85 mL) in Et20 (100 mL) was added EtMgBr (3 M, 40.94 mL) dropwise at -78 °C. After addition, the mixture was stirred at 20 °C for 1 .5 hours, and then BF3-Et20 (1 1 .62 g, 81.88 mmol, 10.10 mL) was added dropwise at -78 °C. The resulting mixture was stirred at 20 °C for 16 hours. Then, the reaction mixture was poured into 100 mL saturated NaHC03 aqueous solution and extracted with EtOAc. The organic layers was dried over Na2S04 and concentrated in vacuo to give a residue. The residue was purified by prep-HPLC (Method 33) to afford pure reagent R-07d (5 g, 44%) as a yellow oil. ESI-MS (M+1 ): 275.2 calc. for C16H22N2O2: 274.2.
Synthetic route 1
Figure imgf000049_0001
Conditions: a) R-01 (1.3 eq), PPh3 (2.0 eq), DEAD (2.0 eq) in THF, rt for 5 h; b) Pd/C in MeOH, H2 atmosphere, rt for 3-24 h; c) malonic acid (1.1-2.0 eq) in POCI3, rt, then overnight at 90-95 °C; d) K2CO3 (2.0 eq), Pd(PPh3)4 (0.1 eq), R-02 (1.1 eq), 1 ,4-dioxane/H20 (10:1 ), 110 °C for 3 h under
MW; e) R-03 (1.0 eq), Cs2C03 (2.0 eq), Pd2(dba)3 (0.1 eq), BINAP (0.1 eq), 1 ,4-dioxane, 120 °C for 16 h; f) R-04 (1.5 eq), Cs2C03 (1.0 eq), DMF, 70 °C for 16 h.
In the scheme above F is a hydrocarbon chain, a carbocycle, heterocycle, an aryl or heteroaryl, R3 is 0(CrC6)alkyl, a hydrocarbon chain or a chlorine atom and R, Ra, Rb and Rc are independently a hydrocarbon chain, which optionally contains nitrogen atoms. Preparation of intermediate l-02a: 1 -[3-(2-methoxy-5-nitro-phenoxy)propyll-pyrrolidine To a solution of commercially available 2-methoxy-5-nitro-phenol: 1-01 a (19.6 g, 0.12 mol) in THF (200 mL), PPh3 (61 g, 0.23 mol), commercially available 3-pyrrolidin-1 -yl-propan-1 - ol (R-01 a, 15 g, 0.16 mol) and DEAD (40 g, 0.23 mol) were added at 0 °C and the solution was stirred at room temperature for 5 hours. The reaction mixture was concentrated and extracted with AcOEt. The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated to give the crude product which was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 1/0 to 3/1 ) to give intermediate \-02a (14 g, 44%) as a yellow solid. ESI-MS (M+1 ): 281 calc. for Ci4H20N2O4: 280.1 .
Preparation of intermediate l-03a: 4-methoxy-3-(3-pyrrolidin-1 -ylpropoxy)aniline
To a solution of intermediate \-02a (14 g, 0.05 mol) in MeOH (200 mL) was added Pd/C (3 g). The solution was stirred at room temperature for 3 hours, in H2 atmosphere. The solution was filtrated and concentrated to give intermediate l-03a (12 g, 96%) as a yellow oil. ESI-MS (M+1 ): 251 calc. for Ci4H22N202: 250.1.
Preparation of intermediate l-04a: 2,4-dichloro-6-methoxy-7-(3-pyrrolidin-1 - ylpropoxy)quinoline
To a solution of intermediate l-03a (12.4 g, 0.049 mol) in POCI3 (200 mL) was added commercially available malonic acid (5.67 g, 0.055 mol) at room temperature. After stirring at room temperature for 4 hours, the solution was heated at 90 °C overnight. The solution was concentrated and poured into ice-water, then extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated to give intermediate l-04a (10 g, 57%) as a pale yellow solid. ESI-MS (M+1 ): 355 calc. for Ci7H20CI2N2O2: 354.1 .
Following the same synthetic route for intermediate l-04a and using the same reagents and intermediates unless otherwise indicated in the table below, the following
intermediates were obtained:
Figure imgf000050_0001
Preparation of intermediate l-04c: tert-butyl 4-[(2,4-dichloro-6-methoxy-7- quinolvDoxymethyllpiperidine-l -carboxylate To a solution of intermediate l-08a (400 mg, 1 .64 mmol) in DMF (20 mL) were added Cs2C03 (534 mg, 1.64 mmol) and commercially available tert-butyl 4- (methylsulfonyloxymethyl)piperidine-l -carboxylate (R-04a, 721 mg, 2.46 mmol) at 20 °C under N2 and the mixture was stirred at 70 °C for 16 hours. Then, the mixture was concentrated under reduced pressure at 40 °C. The resulting residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated under vacuum to give l-04c (500 mg, 69%) as a yellow solid. ESI-MS (M+1 ): 441.2 calc. for C2i H26Cl2N204: 440.1 .
Preparation of intermediate l-05a: 4-chloro-6-methoxy-2-methyl-7-(3-pyrrolidin-1 - ylpropoxy)quinoline
To a solution of intermediate l-04a (1 .6 g, 4.52 mmol) in 1 ,4-dioxane/H20 (10:1 , 1 1 mL) was added K2C03 (1 .25 g, 9.04 mmol), Pd(PPh3)4 (0.53 g, 0.452 mmol) and methylboronic acid (R-02a, 0.30 g, 4.98 mmol) and the solution was heated to 1 10 °C for 3 hours under Microwave. Then the mixture was concentrated and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 .filtered and concentrated to give the crude product which was purified by column chromatography (Si02, DCM/MeOH = 1 :0 to 4:1 ) to give pure intermediate l-05a (0.18 g, 12%) as a yellow solid. ESI-MS (M+1 ): 335 calc. for Ci8H23CIN202: 334.1 .
Following the same synthetic route for intermediate l-05a and using the same reagents and intermediates unless otherwise indicated in the table below, the following
intermediates were obtained:
Intermediate
Yield [M+1]+ Intermediate/reagent
I -05
l-04a / 4,4,5,5-tetramethyl-2-(5-methyl-2-furyl)-1 ,3,2- l-05b 59% 401 .2
dioxaborolane (R-02b)
l-04a / 2-(2,5-dimethyl-3-furyl)-4,4,5,5-tetramethyl-1 ,3,2- l-05c 68% 415.2
dioxaborolane (R-02c)
l-04b / 4,4,5,5-tetramethyl-2-(5-methyl-2-furyl)-1 ,3,2- l-05d 93% 304.1
dioxaborolane (R-02b)
l-04b / 2-(2,5-dimethyl-3-furyl)-4,4,5,5-tetramethyl-1 ,3,2- l-05e 65% 318.1
dioxaborolane (R-02c)
l-04b / 2-(cyclohexen-1-yl)-4,4,5,5-tetramethyl-1 ,3,2- l-05f 68% 304.1
dioxaborolane (R-02d)
|-04b / 4,4,5,5-tetramethyl-2-phenyl-1 ,3,2-dioxaborolane (R- l-05g 52% 300.1
02e)
l-04c / 4,4,5,5-tetramethyl-2-(5-methyl-2-furyl)-1 ,3,2- l-05h 30% 487.2
dioxaborolane (R-02b)
l-05i 22% 238.1 l-04b / methylboronic acid (R-02a)
l-04b / 4,4,5,5-tetramethyl-2-(5-methyl-2-thienyl)-1 ,3,2- l-05j 46% 320.0
dioxaborolane (R-02f) Intermediate
Yield [M+1]+ Intermediate/reagent I -05
l-04b / 2-methyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- l-05k 85% 303.1
yl)-1 H-pyrrole (R-02g)
l-04d / 4,4,5,5-tetramethyl-2-(5-methyl-2-furyl)-1 ,3,2-
I-05I 50% 288.1
dioxaborolane (R-02b)
|-04e / 4,4,5,5-tetramethyl-2-(5-methyl-2-furyl)-1 ,3,2- l-05m 85% 308.0
dioxaborolane (R-02b)
Preparation of intermediate l-06a: 4-chloro-6,7-dimethoxy-2-(1 -piperidyl)quinoline
A mixture of 2,4-dichloro-6,7-dimethoxy-quinoline (l-04b, 1 g, 3.87 mmol), piperidine (R- 03a, 330 mg, 3.87 mmol), Cs2C03 (2.52 g, 7.75 mmol), Pd2(dba)3 (354.79 mg, 387.45 μηιοΙ) and BINAP (241 mg, 387.45 μηιοΙ) in 1 ,4-dioxane (100 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 °C for 16 hours under N2 atmosphere. Then, the mixture was concentrated in reduced pressure at 45 °C to remove the solvent. The residue was poured into water and the aqueous phase was extracted with ethyl acetate. The combined organic phase was concentrated in reduced pressure at 45 °C and l-06a (300 mg, 25%) was obtained as a yellow solid. ESI-MS (M+1 ): 307.1 calc. for Ci6H19CIN202: 306.1 .
Preparation of intermediate l-07a: 5-amino-2-methoxy-phenol
To a solution of commercially available 2-methoxy-5-nitro-phenol (1-01 a, 40 g, 236.5 mmol) in MeOH (300 mL) was added Pd/C (3 g) under Ar. The suspension was degassed under vacuum and purged with H2 several times. The reaction mixture was stirred under H2 (40 Psi, 275.8 KPa) at room temperature for 24 hours. Then, the mixture was filtered and the filtrate was concentrated to give intermediate l-07a (25 g, 76%) as a yellow solid. ESI-MS (M+1 ): 140.1 calc. for C7H9NO2: 139.1 .
Preparation of intermediate l-08a: 2,4-dichloro-6-methoxy-quinolin-7-ol
To a mixture of intermediate l-07a (4.91 g, 35.29 mmol) and malonic acid (7.34 g, 70.57 mmol) was added POCI3 (70 mL) in one portion at room temperature under N2 atmosphere. The mixture was stirred at room temperature for 10 minutes, then heated at 95 °C and stirred for 12 hours. Then, the mixture was cooled to room temperature and concentrated under reduced pressure at 60 °C to remove POCI3. The residue was poured into water and stirred for 20 minutes. The aqueous phase was extracted with EtOAc. The organic phase was separated, washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (PE/EtOAc = 2/1 ) to afford intermediate l-08a (2.10 g, 24%). ESI-MS (M+1 ): 244.0 calc. for C23H27CIN2O3: 242.9.
Synthetic route 2
Figure imgf000053_0001
c)
Conditions: a) R-05 (1.2 eq), Pd2(dba)3 (0.1 eq), BINAP (0.2 eq), Cs2C03 (2.0 eq), 1 ,4-dioxane, 120 °C for 12 h; b) LiOH H20 (3.0 eq), THF/MeOH/H20 (3: 1 :1 ), 25 °C for 12 h; c) O-tetrahydropyran-2- yl hydroxylamine (2.0 eq), HOBt (1 .2-1.5 eq), EDC HCI (1 .2-1.5 eq), DIEA (2.0 eq), DMF, 20-25 °C for 2-12 h; d) HCI/EtOAc (2.0 M), 20 °C for 3 h.
In the scheme above n and m are 0 or 1 , Cyi is a carbocycle, heterocycle, carbocyclic spiro, aryl or heteroaryl, R2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl and Ra, Rb, Rc and Rd are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
Preparation of intermediate l-09a: ethyl 2-[4-[[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyllphenyllacetate
A mixture of ethyl 2-[4-(aminomethyl)phenyl]acetate (R-05a, 76 mg, 395 μηηοΙ), intermediate l-05d (100 mg, 329 μηιοΙ), Pd2(dba)3 (30 mg, 33 μηιοΙ), BINAP (41 mg, 66 μηιοΙ) and Cs2C03 (214 mg, 658 μmol) in 1 ,4-dioxane (5.00 mL) was degassed and purged with N2 for 3 times. Then, the mixture was stirred at 120 °C for 12 hours. The reaction mixture was filtrated and the filtrate was concentrated under vacuum to give intermediate l-09a (130 mg, 85%) as a yellow solid. ESI-MS (M+1 ): 461.3 calc. for C27H28N205: 460.2.
Preparation of intermediate 1-10a: 2-[4-[[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyllphenyllacetic acid
A mixture of intermediate l-09a (130 mg, 282 μηιοΙ) and LiOH-H20 (35 mg, 846 μηιοΙ) in THF/MeOH/H20 (3:1 :1 , 5.0 mL) was degassed, purged with N2 for 3 times, and stirred at 25 °C for 12 hours under N2 atmosphere. Then, the reaction mixture was washed with EtOAc (5 mL x 2) and extracted with water (5 mL x 2). The aqueous phase was quenched by 2.0 M HCI to adjust pH to 4 and the yellow solid was precipitated. The yellow solid was collected and dried to give intermediate 1-10a (80 mg, 65%) as a yellow solid. ESI-MS (M+1 ): 433.3 calc. for C25H24N205: 432.2.
Preparation of compound 1 -01 : 2-[4-[[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyllphenyllethanehydroxamic acid
A mixture of intermediate 1-10a (100 mg, 231 μηιοΙ), 0-(tetrahydro-2H-pyran-2-yl) hydroxylamine hydrochloride (71 mg, 462 μηιοΙ), HOBt (47 mg, 347 μηιοΙ), EDC-HCI (66 mg, 347 μηηοΙ) and DIEA (60 mg, 462 μηηοΙ) in DMF (5.00 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 25 °C for 12 hours under N2
atmosphere. Then, the reaction mixture was concentrated under vacuum to give a residue. The residue was purified by prep-HPLC (Method 1 ) to obtain pure compound 1 - 01 (42.4 mg, 40%) as a yellow solid. ESI-MS (M+1 ): 448.3 calc. for C25H25N305: 447.2. HPLC analytical method 1 , Rt = 2.02 min. Following the same synthetic route for compound 1 -01 from l-05d and R-05a but using the intermediates and reagents indicated below, the following compounds were obtained:
Figure imgf000054_0001
Preparation of intermediate 1-1 1 a: 4-[[[6-methoxy-2-(5-methyl-2-furyl)-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminolmethyll-N-tetrahydropyran-2-yloxy-cvclohexanecarboxamide A mixture of intermediate 1-1 Oe (100 mg, 192 μηηοΙ), O-tetrahydropyran-2-yl
hydroxylamine (45 mg, 383 μηιοΙ), HOBt (31 mg, 230 μηιοΙ), EDC-HCI (44 mg, 230 μηιοΙ) and DIEA (50 mg, 383 μηηοΙ) in DMF (3 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 20 °C for 2 hours under N2 atmosphere. Then, the mixture was concentrated in reduced pressure at 40 °C. The residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum. The crude product was purified by prep-HPLC (Method 4) to give pure intermediate 1-1 1 a (60 mg, 50%) as a yellow solid. ESI-MS (M+1 ): 621 .4 calc. for C35H48N406: 620.4. Preparation of compound 1 -05: 4-[[[6-methoxy-2-(5-methyl-2-furyl)-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminolmethyllcvclohexanecarbohvdroxamic acid
A mixture of intermediate 1-1 1 a (60 mg, 97 μηηοΙ) in HCI/EtOAc (10 mL, 2.0 M) was stirred at 20 °C for 3 hours. Then, the mixture was concentrated in reduced pressure at 40 °C to give crude product. The crude product was purified by prep-HPLC (Method 5) to give pure compound 1 -05 (9.60 mg, 18%) as a yellow solid. ESI-MS (M+1 ): 537.5 calc. for C30H40N4O5: 536.3. HPLC analytical method 2, Rt = 3.02 min. Following the same synthetic route for compound 1 -05 from l-05b and R-05b and using the intermediates and reagents indicated below, the following compounds were obtained:
Method Rt
Yield
Method Prep- AnalyAnalyIntermediates and
Example 4 [M+1]+
HPLC tical tical reagents
steps
HPLC HPLC
l-05d / ethyl 4- aminocyclo-
1-06 27 1 % 426.2 1 2.669
hexanecarboxylate (R-05d)
l-05d / ethyl 4- aminocyclohexa-
1-07 27 1 % 426.2 1 2.130
necarboxylate (R- 05d)
l-05e / methyl 4- (aminomethyl)-
1-08 14 4% 454.3 1 2.251
cyclohexanecarboxyl ate (R-05f) l-05e / methyl 4- (aminomethyl)-
1-09 14 1 % 454.3 1 2.284
cyclohexanecarboxyl ate (R-05f) l-05d / methyl 2- aminospiro[3.3]-
1-10 31 7% 438.3 1 2.220
heptane-6- carboxylate (R-05e) l-05d / methyl 4-
1-12 35 22% 420.2 1 1.687 aminobenzoate (R- 05h)
36 for
corresponding
intermediate 1-1 1 l-05d / methyl 2-
1-13 (compound 1-13 21 % 422.1 1 1.593 aminopyrimidine-5- was isolated after carboxylate (R-05i) remove solvent
under vacuum)
l-05d / methyl 3- (aminomethyl)cyclob
1-14 37 1 1 % 412.2 1 1.762
utanecarboxylate (R- 05j)
Figure imgf000056_0001
2
Conditions: a) R-06 (1.5 eq), Cs2C03 (2.0 eq), BINAP (0.1 eq), Pd2(dba)3 (0.1 eq), 1 ,4-dioxane, 120 °C for 16; b ) Pd/C, MeOH, H2 (15 Psi, 103.4 KPa), 20 °C for 16 h; c) LiOH H20 (1.5 - 3.0 eq),
THF/MeOH/H20 (5:1 :3 or 1 :3:1 ), 20-25 °C for 12-16 h; d) O-tetrahydropyran-2-ylhydroxylamine (1.5 eq), HOBt (1.5 eq), EDC HCI (1.5 eq), DIEA (3.0 eq), DMF, 20 °C for 16 h; e) HCI/MeOH (2.0 M), 25 °C for 12 h. In the scheme above n, m and p are 0 or 1 , Cyi and Cy2 are a carbocycle, heterocycle, aryl or heteroaryl, R2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl, a hydrocarbon chain or a chlorine atom and Ra, Rb, Rc and Rd are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
Preparation of intermediate l-12a: ethyl 2-r4-rrr2-(2,5-dimethyl-3-furyl)-6-methoxy-7-(3- PVrrolidin-1 -ylpropoxy)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
A mixture of l-05c (200 mg, 482 μηηοΙ), commercially available ethyl 2-[4-(aminomethyl)-1 - piperidyl]pyrimidine-5-carboxylate (R-06a, 191 mg, 723 μηηοΙ), Cs2C03 (314 mg, 964 μηιοΙ), BINAP (30 mg, 48 μηιοΙ) and Pd2(dba)3 (44 mg, 48 μηιοΙ) in 1 ,4-dioxane (10 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 120 °C for 16 hours under N2 atmosphere. Then, the mixture was concentrated in reduced pressure at 40 °C to give a residue. The residue was poured into water and extracted with ethyl acetate. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give a residue. The residue was purified by prep-HPLC (Method 6) to give intermediate 1-12a (120 mg, 39%) as a yellow solid. ESI- MS (M+1 ): 643.4 calc. for C36H46N605: 642.4.
Preparation of intermediate 1-13a: 2-r4-rrr2-(2.5-dimethyl-3-furyl)-6-methoxy-7-(3- pyrrolidin-1 -ylpropoxy)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylic acid To a solution of intermediate 1-12a (100 mg, 155 μηιοΙ) in THF/MeOH/H20 (5:1 :3, 9 mL) was added LiOH-H20 (10 mg, 233 μηιοΙ) and the mixture was stirred at 25 °C for 12 hours. Then, the mixture was concentrated in reduced pressure at 40 °C to give a residue. The residue was adjusted pH to 3-4 with 3.0 M HCI. The yellow solid was precipitated and collected to give the desired intermediate 1-13a (90 mg, 94%) as a yellow solid. ESI-MS (M+1 ): 615.4 calc. for C34H42N605: 614.3.
Preparation of intermediate 1-131: 5-[4-[[(2-cvclohexyl-6,7-dimethoxy-4- quinolyl)aminolmethyll-1 -piperidyllpyrimidine-2-carboxylic acid
To a solution of intermediate 1-121 (300 mg, 564 μιτιοΙ) in MeOH (20 mL) was added Pd/C (10%, 30 mg) under H2 atmosphere and the suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (15 Psi, 103.4 KPa) at 20 °C for 16 hours. Then, the mixture was filtered and concentrated in vacuum to give intermediate ethyl 5-[4- [[(2-cyclohexyl-6,7-dimethoxy-4-quinolyl)amino]methyl]-1 -piperidyl]pyrimidine-2- carboxylate (200 mg, 66%) as a yellow solid. A mixture of this intermediate (200 mg, 374 μηιοΙ) and LiOH-H20 (47 mg, 1.12 mmol), in MeOH/THF/H20 (1 :3.1 , 5 mL), was stirred at 20 °C for 16 hours. Then, the organic solvent was removed by evaporation and the residue was diluted with H20 and extracted with ethyl acetate. The mixture was acidified to pH = 3-4 with 1 N HCI and filtered. The filter cake was concentrated to dryness to give intermediate 1-131 (120 mg, 63%) as a yellow solid. ESI-MS (M+1 ): 506.3 calc. for
C28H35N504: 505.3.
Preparation of intermediate l-14a: 2-[4-[[[2-(2,5-dimethyl-3-furyl)-6-methoxy-7-(3- PVrrolidin-1 -ylpropoxy)-4-quinolyllaminolmethyll-1 -piperidyll-N-tetrahvdropyran-2-yloxy- pyrimidine-5-carboxamide
A mixture of intermediate 1-13a (60 mg, 98 μηηοΙ), O-tetrahydropyran-2-ylhydroxylamine (17 mg, 146 μηιοΙ), DIEA (38 mg, 293 μηιοΙ), HOBt (20 mg, 146 μηιοΙ) and EDC-HCI (28 mg, 146 μηηοΙ) in DMF (10 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at 20 °C for 16 hours under N2 atmosphere. Then, the mixture was concentrated in reduced pressure at 40 °C to give a residue. The residue was purified by prep-HPLC (Method 2) to give intermediate l-14a (35 mg, 50%) as a white solid. ESI-MS (M+1 ): 714.4 calc. for C39H51N706: 713.4. Preparation of compound 2-01 : 2-[4-[[[2-(2,5-dimethyl-3-fui^l)-6-methoxy-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
A solution of intermediate l-14a (35 mg, 49 μηιοΙ) in HCI/MeOH (10 mL, 2.0 M) was degassed and purged with N2 for 3 times. Then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. The reaction mixture was concentrated under vacuum to give a residue. The residue was purified by prep-HPLC (Method 7) to give compound 2-01 (6.5 mg, 19%) as a yellow solid. ESI-MS (M+1 ): 630.4 calc. for C34H43N7O5: 629.3. HPLC analytical method 1 , Rt = 2.04 min. Following the same synthetic route for compound 2-01 from l-05c and R-06a and using the intermediates and reagents indicated below, the following compounds were obtained:
Method Rt
Method Prep- AnalyAnaly¬
Example Yield [M+1]+ Intermediates and reagents
HPLC tical tical
HPLC HPLC
l-05b / ethyl 2-[4-
(aminomethyl)-l-pipe-
2-02 2 6% 616.3 1 2.01
ridyl] pyrim id i ne-5-carboxylate
(R-06a)
l-05d / ethyl 3-[[4- (aminomethyl)-l-piperi-
2-03 8 3% 509.4 1 1.77
dyl]methyl]cyclobutanecarbo xylate (R-06b)
l-05b / methyl 4-[4-
2-04 9 6% 614.4 1 1.797 (aminomethyl)-l-pipe- ridyl]benzoate (R-06c) l-05b / methyl 4-[[4- (aminomethyl)-l-
2-05 10 7% 628.3 1 1.443
piperidyl]methyl]benzoate (R- 06d)
l-05b / methyl 4-(4-amino-1-
2-06 1 1 4% 600.4 1 1.862
piperidyl)benzoate (R-06e) l-05b / ethyl 4-[4- (aminomethyl)phenyl]-
2-07 2 2% 613.4 1 2.100
cyclohexanecarboxylate (R- 06f)
l-05b / ethyl 4-[4- (aminomethyl)phenyl]-
2-08 2 1 % 613.3 1 2.040
cyclohexanecarboxylate (R- 06f)
l-05b / methyl 4-[4-
2-09 2 8% 607.3 1 2.009 (aminomethyl)phenyl]- benzoate (R-06g) l-05b / methyl 4-[(4-amino-1-
2-10 12 2% 614.4 1 1.428 piperidyl)-methyl]benzoate
(R-06h)
l-05b / m-ethyl 4-[4-
2-1 1 13 6% 613.4 1 2.127 (aminomethyl)cyclo- hexyl]benzoate (R-06i) Method Rt
Method Prep- AnalyAnaly¬
Example Yield [M+1]+ Intermediates and reagents
HPLC tical tical
HPLC HPLC
l-05e / ethyl 2-[4- (aminomethyl)-l-pipe-
2-12 14 3% 533.3 1 2.288
ridyl]pyrimidine-5-carboxylate (R-06a)
l-05f / ethyl 2-[4- (aminomethyl)-l-pipe- ridyl]pyrimidine-5-carboxylate (R-06a). Here, the obtained intermediate 1-12 ethyl 2-[4- [[[2-(cyclohexen-1-yl)-6,7- dimethoxy-4- quinolyl]amino]methyl]-1- piperidyl]-pyrimidine-5- carboxylate before being
2-13 15 2% 521 .3 1 2.351 converted into 1-13 was firstly hydrogenated (Pd/C, H2 (15 Psi (103.4 KPa), at 20 °C for 16 hours)) to yield ethyl 2-[4- [[(2-cyclohexyl-6,7- dimethoxy-4-quinolyl)- amino]methyl]-1- piperidyl]pyrimidine-5- carboxylate. Then, the same synthetic route as for 2-01 was followed.
l-05g / ethyl 2-[4- (aminomethyl)-l-pipe-
2-14 16 9% 515.2 1 2.157
ridyl]pyrimidine-5-carboxylate (R-06a)
l-05h / ethyl 2-[4- (aminomethyl)-l-pipe-
2-15 30 3% 602.4 1 1.959
ridyl]pyrimidine-5-carboxylate (R-06a)
l-05i / ethyl 2-[4- (aminomethyl)-l-pipe-
2-16 29 1 % 453.3 1 1.826
ridyl]pyrimidine-5-carboxylate (R-06a)
l-05e / methyl 4-[4-
2-17 35 1 % 531 .3 1 1.972 (aminomethyl)-l-pipe- ridyl]benzoate (R-06c) l-05j / ethyl 2-[4- (aminomethyl)-l-pipe-
2-18 38 31 % 535.3 1 2.186
ridyl]pyrimidine-5-carboxylate (R-06a)
38 for
corresponding
intermediate I- l-05k / ethyl 2-[4- 14 (compound
(aminomethyl)-l-pipe-
2-19 2-19 was 7% 518.3 1 2.132
ridyl]pyrimidine-5-carboxylate isolated after
(R-06a)
remove
solvent under
vacuum) Method Rt
Method Prep- AnalyAnaly¬
Example Yield [M+1]+ Intermediates and reagents
HPLC tical tical
HPLC HPLC
l-05d / methyl 4-[4-
2-20 39 21 % 517.4 1 1.912 (aminomethyl)-l-pipe- ridyl]benzoate (R-06c)
1-051 / ethyl 2-[4- (aminomethyl)-l-pipe-
2-21 40 18% 503.3 1 2.272
ridyl]pyrimidine-5-carboxylate (R-06a)
l-05m / ethyl 2-[4- (aminomethyl)-l-pipe-
2-22 15 14% 523.4 1 2.199
ridyl]pyrimidine-5-carboxylate (R-06a)
Synthetic route 4
Figure imgf000060_0001
3B or 5 1-19
Conditions: a) t-BuONa (3.0 eq), xantphos (0.2 eq), Pd2(dba)3 (0.2 eq), R-07 (3.0 eq), toluene, 1 10 °C for 2 h under MW; b) Cs2C03 (3.0 eq), BINAP (0.3 eq), Pd2(dba)3 (0.1 eq), R-07 (2.0 eq), 1 ,4- dioxane, 1 10 °C for 1 1 h under MW; c) Pd/C, H2 (20 Psi, 137.9 KPa), MeOH, 20 °C, 16 h; d)
HCI/1 ,4-dioxane (4.0 M), 1 ,4-dioxane, rt for 3 h; e) K2C03 (3.0 eq), R-08 (1.5 eq), acetonitrile, rt for 3 h; f) AcOH (0.2 eq), NaBH3CN (1.5 eq), R-09 (1.3 eq), MeOH, 60 °C for 12 h; g) ZnCI2/diethyl ether (1.0 M), NaBH3CN (2.2 eq), MeOH, 20 °C for 30 minutes, then R-10 (1.1 eq), 1-14, MeOH, 40 °C for 15.5 h; h) LiOH H20 (5.0 eq), THF/MeOH/H20 (10:1 :3), rt overnight; i) O-tetrahydropyran-2- yl hydroxylamine (1.5 eq), HOBt (2.0 eq), EDC HCI (2.0 eq), NMM (3.0 eq), DMF, rt overnight; j) HCI/1 ,4-dioxane (4.0 M), rt for 1 h; k) O-tetrahydropyran-2-yl hydrochloride (2.0 eq), HOBt (1.5 eq), EDC HCI (1.5 eq), DIEA (2.0 eq), DMF, rt overnight. I) bis(trichloromethyl) carbonate, DIEA, DCM, 0 °C, 1 h; then NH2OH HCI, 75 °C, 16 h.
In the scheme above X is a chlorine or bromine atom, n, m and p are 0 or 1 , Cyi is a carbocycle, heterocycle, aryl or heteroaryl, A is a heterocycle, PG is a protecting group, R2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl, hydrocarbon or chlorine, and Ra, Rb, Rc , Rd and Re are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
Preparation of intermediate 1-15a: tert-butyl 4-[[6-methoxy-2-methyl-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminolpiperidine-1 -carboxylate
To a solution of intermediate l-05a (33.5 mg, 0.1 mmol) in toluene (10 mL) was added t- BuONa (25.8 mg, 0.3 mmol), xantphos (12.4 mg, 0.02 mmol), Pd2(dba)3 (18.3 mg, 0.02 mmol) and commercially available tert-butyl 4-aminopiperidine-1 -carboxylate (R-07a, 60 mg, 0.3 mmol) and the solution was heated to 1 10 °C for 2 hours under Microwave. The reaction mixture was concentrated and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2S04 and concentrated to give the crude product which was purified by prep-HPLC (Method 17) to give pure intermediate 1-15a (20 mg, 40%) as yellow solid. ESI-MS (M+1 ): 499 calc. for C28H42N404: 498.3.
Preparation of intermediate 1-15c: tert-butyl 4-[[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyllpiperidine-1 -carboxylate
To a solution of intermediate l-05d (400 mg, 1.32 mmol) in 1 ,4-dioxane (5 mL) was added Cs2C03 (1 .3 g, 3.9 mmol), BINAP (244 mg, 0.39 mmol), Pd2(dba)3 (126 mg, 0.132 mmol) and commercially available tert-butyl 4-(aminomethyl)piperidine-1 -carboxylate (R-07b, 594 mg, 2.64 mmol) and the mixture was heated to 1 10 °C for 1 1 hours under Microwave. Then, the mixture was quenched with water and extracted with EtOAc. The organic layer was washed with brine, dried over anhydrous Na2S04, filtered and concentrated to give the crude product which was purified by column chromatography to obtain pure intermediate 1-15c (280 mg, 44%) as a yellow solid. ESI-MS (M+1 ): 482.2 calc. for C27H35N305: 481 .3.
Preparation of intermediate 1-16a: 6-methoxy-2-methyl-N-(4-piperidyl)-7-(3-pyrrolidin-1 - ylpropoxy)quinolin-4-amine
To a solution of intermediate 1-15a (200 mg, 0.417 mmol) in 1 ,4-dioxane (30 mL) was added HCI/1 ,4-dioxane (5 mL, 4.0 M) and the solution was stirred at room temperature for 3 hours. The reaction mixture was concentrated to give the desired intermediate 1-16a (120 mg, 75%) as white solid. ESI-MS (M+1 ): 399 calc. for C23H34N402: 398.3. Following the same synthetic route for compound 1-16a from 1-15 or from l-05a and R-07a and using the intermediates and reagents indicated below, the following compounds were obtained:
Figure imgf000062_0001
Preparation of intermediate 1-16g: 6,7-dimethoxy-2-(5-methyl-2-furyl)-N-[1 -(4- piperidyl)cvclopropyHquinolin-4-amine
To a solution of intermediate 1-15g (prepared from l-05d and R-07d following the same method as used for 1-15c; 700 mg, 1.29 mmol) in MeOH (20 mL) was added Pd/C (10%, 0.02 g) under N2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (20 Psi, 137.9 KPa) at 20 °C for 16 hours. Then, the reaction mixture was filtered and concentrated in vacuo to give 1-16g (600 mg, crude) as a yellow solid. ESI-MS (M+1 ): 408.3 calc. for C24H29N3O3: 407.2. Preparation of intermediate 1-17a: ethyl 2-[4-[[6-methoxy-2-methyl-7-(3-pyrrolidin-1 - ylpropoxy)-4-quinolyllaminol-1 -piperidyllpyrimidine-5-carboxylate
To a solution of intermediate 1-16a (120 mg, 0.3 mmol) in acetonitrile (15 mL) was added K2C03 (138 mg, 1 mmol) and commercially available ethyl 2-chloropyrimidine-5- carboxylate (R-08a, 88 mg, 0.45 mmol) and the solution was stirred at room temperature for 3 hours. Then, the mixture was concentrated to give intermediate 1-17a (0.15 g, 91 %) as a yellow solid. ESI-MS (M+1 ): 549 calc. for C3oH4oN604: 548.3. Preparation of intermediate 1-17e: ethyl 4-[[4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminol-1 -piperidyllmethyllbenzoate
A mixture of commercially available ethyl 4-formylbenzoate (R-09a, 126 mg, 707 μηηοΙ), intermediate 1-16d (200 mg, 544 μηιοΙ), AcOH (6 mg, 109 mol) and NaBH3CN (51 mg, 816 mol) in MeOH (5.00 mL) was degassed and purged with N2 for 3 times. Then, the mixture was stirred at 60 °C for 12 hours under N2 atmosphere. The reaction mixture was filtrated and the filtrate was concentrated under vacuum to give a residue. The residue was purified by prep-HPLC (Method 2) to give intermediate 1-17e (120 mg, 42%) as a yellow solid. ESI-MS (M+1 ): 530.3 calc. for C31 H35N3O5: 529.3. Preparation of intermediate 1-17h: ethyl 4-r4-rri6-methoxy-2-(5-methyl-2-furyl)-7-(3- PVrrolidin-1 -ylpropoxy)-4-quinolyllaminolmethyll-1 -piperidyllcvclohexanecarboxylate
To a solution of ZnCI2/diethyl ether (1 .0 M, 20 uL) was added NaBH3CN (52 mg, 827 mol) in MeOH (10 mL) and the mixture was stirred at 20 °C for 30 minutes. Then, a mixture of commercially available ethyl 4-oxocyclohexanecarboxylate (R-10a, 70 mg, 413 Mmol) and intermediate 1-16e (180 mg, 376 Mmol) in MeOH (40 mL) was added and the resulting mixture was heated at 40 °C for 15.5 hours. Then, the reaction mixture was quenched with water, filtered and the solvente was removed under vacum to give a residue. The residue was extracted with a solution of DCM/MeOH (3:1 ). The combined organic layers were washed with brine, dried over anhydrous Na2S0 , filtered and concentrated under reduced pressure to give intermediate 1-17h (155 mg, 65%) as a white solid. ESI-MS (M+1 ): 633.3 calc. for C37H52N405: 632.4.
Preparation of intermediate 1-18a: 2-[4-[[6-methoxy-2-methyl-7-(3-pyrrolidin-1 -ylpropoxy)- 4-quinolyllaminol-1 -piperidyllpyrimidine-5-carboxylic acid
To a solution of intermediate 1-17a (0.2 g, 0.365 mmol) in THF/MeOH/H20 (10:1 :3, 10 mL) was added LiOH-H20 (78 mg, 1.82 mmol) and the resulting mixture was stirred at room temperature overnight. Then, the mixture was diluted with water and adjusted pH to 2-3. The mixture was extracted with ethyl acetate and the combined organic phase was concentrated to give intermediate 1-18a (0.15 g, 79%). ESI-MS (M+1 ): 521 calc. for
Figure imgf000064_0001
Preparation of intermediate 1-19a: 2-[4-[[6-methoxy-2-methyl-7-(3-pyrrolidin-1 -ylpropoxy)- 4-quinolyllaminol-1 -piperidyll-N-tetrahydropyran-2-yloxy-pyrimidine-5-carboxamide
To a solution of intermediate 1-18a (104 mg, 0.2 mmol) in DMF (10 mL) was added EDC-HCI (69 mg, 0.4 mmol), HOBt (54 mg, 0.4 mmol), O-tetrahydropyran-2-yl hydroxylamine (35 mg, 0.3 mmol) and NMM (61 mg, 0.6 mmol) and the mixture was stirred at room temperature overnight. Then, the mixture was diluted with EtOAc and washed with brine. The organic phase was dried over anhydrous Na2S04, filtered and concentrated to give the crude product which was purified by prep-HPLC (Method 18) to give intermediate 1-19a (60 mg, 48%). ESI-MS (M+1 ): 620 calc. for C33H45N7O5: 619.3.
Preparation of compound 3-01 : 2-[4-[[6-methoxy-2-methyl-7-(3-pyrrolidin-1 -ylpropoxy)-4- quinolyllaminol-1 -piperidvHpyrimidine-5-carbohvdroxamic acid
A solution of intermediate 1-19a (60 mg, 0.097 mmol) in HCI/1 ,4-dioxane (4.0 M, 10 mL) was stirred at room temperature for 1 hour. Then, the reaction mixture was concentrated to give the desired compound 3-01 (31 mg, 61 %). ESI-MS (M+1 ): 536.3 calc. for C28H37N704: 535.3. HPLC analytical method 2, Rt = 2.80 min
Following the same synthetic route for compound 3-01 from 1-17a or from 1-16a and R-08a and using the intermediates and reagents indicated below, the following compounds were obtained:
Method Rt
Method Yield
AnalyAnaly¬
Example Prep- 3 or 4 [M+1]+ Intermediates and reagents tical tical
HPLC steps
HPLC HPLC
1-16b / ethyl 2-chloropyrimidine-5-
3-02 19 13% 550.3 1 2.85 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16c / ethyl 2-chloropyrimidine-5-
3-03 20 4% 519.2 1 2.37 carboxylate (R-08a); reaction
conditions as for 1-17a
3-08 21 3% 620.4 1 1.464 l-17h
1-16c / methyl 3-oxocyclobutane-
3-09 22 6% 495.4 1 1.644 carboxylate (R-10b) reaction
conditions as for 1-17h
l-16f / ethyl 2-chloropyrimidine-5-
3-10 28 4% 545.3 1 2.356 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16g / ethyl 2-chloropyrimidine-5-
3-1 1 32 3% 545.3 1 2.425 carboxylate (R-08a); reaction
conditions as for 1-17a Method Rt
Method Yield
AnalyAnaly¬
Example Prep- 3 or 4 [M+1]+ Intermediates and reagents tical tical
HPLC steps
HPLC HPLC
1-16c / methyl 6-chloropyridine-3-
3-13 41 1 1 % 518.3 1 1.553 carboxylate (R-08b); reaction
conditions as for 1-17a
1-16c / methyl 2-(2-chloropyrimidin-5-
3-14 14 6% 533.3 1 1.796 yl)acetate (R-08c); reaction conditions as for 1-17a
2.141 1-16h / ethyl 2-chloropyrimidine-5-
3-15 42 14% 559.3 1 and carboxylate (R-08a); reaction
2.172 conditions as for 1-17a
1-16i / ethyl 2-chloropyrimidine-5-
3-17 35 19% 503.3 1 1.570 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16k / ethyl 2-chloropyrimidine-5-
3-19 28 29% 545.3 1 2.135 carboxylate (R-08a); reaction
conditions as for 1-17a
43 for
corres¬
1.862 1-161 / ethyl 2-chloropyrimidine-5- ponding
3-20 50% 517.2 1 and carboxylate (R-08a); reaction
interme1.955 conditions as for 1-17a
diate
1-19
1-16m / ethyl 2-chloropyrimidine-5-
3-21 44 47% 505.3 1 1.842 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16o / ethyl 2-chloropyrimidine-5-
3-23 14 17% 600.4 1 1.849 carboxylate (R-08a); reaction
conditions as for 1-17a
Preparation of compound 3-04: 2-[4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminol-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
A mixture of intermediate 1-18d (70 mg, 143 μηηοΙ), 0-(tetrahydro-2H-pyran-2-yl) hydroxylamine hydrochloride (44 mg, 286 μηιοΙ), EDC-HCI (41 mg, 214 μηιοΙ), DIEA (37 mg, 286 μηηοΙ) and HOBt (29 mg, 214 μηηοΙ) in DMF (5.00 mL) was degassed and purged with N2 for 3 times, and the mixture was stirred at room temperature overnight under N2 atmosphere. Then, the reaction mixture was concentrated under vacuum to give a residue. The residue was purified by prep-HPLC (Method 23) to obtain pure compound 3- 04 (41 mg, 57%) as a yellow solid. ESI-MS (M+1 ): 505.3 calc. for C26H28N605: 504.2. HPLC analytical method 1 , Rt = 2.12 min.
Following the same synthetic route for compound 3-04 from 1-17d or from 1-16d and R-08a and using the intermediates and reagents indicated below, the following compounds were obtained: Method Rt
Method Yield
AnalyAnaly¬
Example Prep- 2 or 3 [M+1]+ Intermediate
tical tical
HPLC steps
HPLC HPLC
3-05 2 14% 517.4 1 1.61 l-17e
1-16c / ethyl 4-formylbenzoate (R-
3-06 24 15% 531 .4 1 1.74
09a); reaction conditions as for 1-17e
1-16c / ethyl 5-formylthiophene-2-
3-07 25 3% 537.2 1 1.76 carboxylate (R-09b); reaction
conditions as for 1-17e
1-16c / ethyl 2- (bromomethyl)pyrimidine-5-
3-16 45 14% 533.3 1 1.363
carboxylate (R-08d); reaction conditions as for 1-17a
1-16j / ethyl 2-chloropyrimidine-5-
3-18 43 21 % 503.3 1 1.898 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16n / ethyl 2-chloropyrimidine-5-
3-22 41 13% 600.4 1 1.594 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16p / ethyl 2-chloropyrimidine-5-
3-24 46 33% 545.4 1 2.362 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16q / ethyl 2-chloropyrimidine-5-
3-25 47 45% 545.4 1 2.333 carboxylate (R-08a); reaction
conditions as for 1-17a
1-16r / ethyl 2-chloropyrimidine-5-
5-01 29 2% 533.3 1 1.858 carboxylate (R-08a); reaction
conditions as for 1-17a
Preparation of compound 3-12: 4-[[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyllpiperidine-1 -carbohvdroxamic acid
To a solution of bis(trichloromethyl) carbonate (186 mg, 0.629 mmol) in 1 ,2- dichloroethane (10 mL) was added a solution of 1-16c (200 mg, 0.524 mmol) and DIEA (338 mg, 2.62 mmol) in 1 ,2-dichloroethane (20 mL) at 0 °C. After addition, the mixture was stirred at 0 °C for 1 hour. Then DIEA (338 mg, 2.62 mmol) was added, followed
immediately by NH2OH-HCI (182 mg, 2.62 mmol) and the resulting mixture was stirred at 75 °C for 16 hours. The reaction mixture was quenched with 50 mL H20 and extracted with DCM (30 mL x 3). The organic layers were dried over Na2S04 and concentrated in vacuo to give crude product. The crude product was purified by prep-HPLC (Method 22) to afford pure compound 3-12 (49 mg, 21 %) as a white solid. ESI-MS (M+1 ): 441 .3 calc. for C23H28N405: 440.2. HPLC analytical method 1 , Rt = 2.087 min. Synthetic route 5
Figure imgf000067_0001
1-21 I-22
Figure imgf000067_0002
4
Conditions: a) R-06 (1.5 eq), Pd2(dba)3 (0.1 eq), BINAP (0.1 eq), Cs2C03 (2.0 eq), 1 ,4-dioxane, 120 °C for 16h; b) LiOH H20 (1.5 eq), THF/H20 (2:1 ), 15 °C for 16 h; c) O-tetrahydropyran-2-yl hydroxylamine (2.0 eq), HOBt (2.0 eq), EDC HCI (2.0 eq), DIEA (3.0 eq), DMF, 15 °C for 16 h; d) HCI/EtOAc (1.0 M), 15 °C for 16.
In the scheme above n, m and p are 0 or 1 , Cyi and Cy2 are a carbocycle, heterocycle, aryl or heteroaryl, R3 is 0(CrC6)alkyl and Ra, Rb, Rc, d, Re and Rf are independently hydrogen, a hydrocarbon chain, which optionally contains nitrogen atoms. Preparation of intermediate l-20a: ethyl 2-[4-[[[6,7-dimethoxy-2-(1 -piperidyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
A mixture of 4-chloro-6,7-dimethoxy-2-(1 -piperidyl)quinoline (l-06a, 250 mg, 815 μηηοΙ), ethyl 2-[4-(aminomethyl)-1 -piperidyl]pyrimidine-5-carboxylate (R-06a, 323 mg, 1.22 mmol), Pd2(dba)3 (74.6 mg, 81 mol), BINAP (50.7 mg, 81 mol) and Cs2C03 (531 mg, 1 .6 mmol) in 1 ,4-dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 °C for 16 hours under N2 atmosphere. Then, the reaction mixture was concentrated in vacuo to give a residue. The residue was purified by prep- TLC (DCM:MeOH = 10:1 ) to obtain l-20a (270 mg, 62%) as a yellow solid. ESI-MS (M+1 ): 535.3 calc. for C29H38N604: 534.3.
Preparation of intermediate 1-21 a: 2-[4-[[[6,7-dimethoxy-2-(1 -piperidyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylic acid To a solution of l-20a (270 mg, 505 μηιοΙ) in THF (10 mL) and H20 (5 mL) was added LiOH-H20 (31 .8 mg, 757 μηηοΙ) and the mixture was stirred at 15 °C for 16 hours. Then, the reaction mixture was adjusted to pH = 5 with 2.0 M HCI solution. Then the mixture was concentrated in vacuo to give 1-21 a (300 mg, 99% crude) as a yellow oil. ESI-MS (M+1 ): 507.3 calc. for C27H34N604: 506.3.
Preparation of intermediate l-22a: 2-[4-[[[6,7-dimethoxy-2-(1 -piperidyl)-4- quinolyllaminolmethyll-1 -piperidyll-N-tetrahydropyran-2-yloxy-pyrimidine-5-carboxamide A mixture of 1-21 a (300 mg, 592 umol), O-tetrahydropyran-2- ylhydroxylamine (138 mg, 1 .18 mmol), EDC HCI (227 mg, 1.18 mmol), HOBt (160 mg, 1.18 mmol) and DIEA (229 mg, 1 .78 mmol) in DMF (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 15 °C for 16 hours under N2 atmosphere. Then, the reaction mixture was quenched with water and concentrated in vacuo to give a residue. The residue was dissolved in 20 mL MeCN and filtered. The filtrate was concentrated in vacuo to give l-22a (500 mg, 99% crude) as a brown oil. ESI-MS (M+1 ): 606.4 calc. for
C32H43N705: 605.3.
Preparation of compound 4-01 : 2-[4-[[[6,7-dimethoxy-2-(1 -piperidyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
A solution of l-22a (0.3 g, 495 mol) in HCI/EtOAc (1 .0 M, 10 mL) was stirred at 15 °C for 16 hours. Then, the reaction mixture was concentrated in vacuo to give a residue. The residue was purified by prep-HPLC (Method 29) to obtain 4-01 (65.6 mg, 25%) as a white solid. ESI-MS (M+1 ): 522.4 calc. for C27H35N704: 521.3. HPLC analytical method 1 , Rt = 2.235 min
Synthetic route 6
Figure imgf000069_0001
Conditions: a) R-11 (1.5 eq), NaH (1.5 eq), DMF, 20 °C for 1 h; then I-05 (1.0 eq), 50 °C for 16 h; b) R-12 (2.0 eq), 9-BBN (4.0 eq), THF, 80 °C for 3 h; then I-05 (1.0 eq), Pd2(dba)3 (0.2 eq), X-Phos (0.4 eq) and K2C03 (3.0 eq), 1 ,4-dioxane/H20 (5:1 ), 95 °C for 12 h; c) HCI/EtOAc (1.0 M), 20 °C for 16 h; d) R-08 (1.5 eq), K2C03 (3.0 eq), CH3CN, 40 °C for 16 h; e) LiOH H20 (2.0 eq), THF/H20 (2:1 ), 20 °C for 16 h; f) O-tetrahydropyran-2-yl hydroxylamine (2.0 eq), HOBt (2.0 eq), EDCI (2.0 eq), DIEA (3.0 eq), DMF, 25 °C for 12 h; g) HCI (2.0 M), CH3CN, 25 °C for 10 min.
In the scheme above X is a chlorine or bromine atom, Z is an oxygen atom or -CH2-, n, m and p are 0 or 1 , Cyi is a carbocycle, heterocycle, aryl or heteroaryl, A is a heterocycle, PG is a protecting group, R2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl and Ra, Rb, Rc, and Rd are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms. Preparation of intermediate l-23a: tert-butyl 4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolylloxymethyllpiperidine-1 -carboxylate
To a solution of commercially available tert-butyl 4-(hydroxymethyl)piperidine-1 - carboxylate (R-11 a, 531 mg, 2.47 mmol) in DMF (20 mL) was added NaH (99 mg, 2.47 mmol, 60% purity) at 20 °C. After 1 hour, intermediate l-05d (500 mg, 1 .65 mmol) was added and the mixture was stirred at 50 °C for 16 hours. Then, the reaction mixture was quenched with water and concentrated under vacuum to give a residue which was purified by column chromatography (Si02, Petroleum ether/Ethyl acetate = 20:1 to 1 :1 ) to obtain pure intermediate l-23a (500 mg, crude) as a yellow solid. ESI-MS (M+1 ): 483.3 calc. for C27H34N206: 482.2 Preparation of intermediate l-23b: tert-butyl 4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolyllmethyllpiperidine-1 -carboxylate
Tert-butyl 4-methylenepiperidine-1 -carboxylate (R-12a, 260 mg, 1 .32 mmol) was treated with 9-BBN (0.5 M, 5.27 mL) in THF (5.00 mL), and the mixture was heated to reflux at 80 °C for 3 hours. The resulting mixture was transferred into a stirred mixture of intermediate l-05d (0.2 g, 0.658 mmol), Pd2(dba)3 (120 mg, 0.131 mmol), XPhos (125 mg, 0.263 mmol) and K2C03 (273 mg, 1.98 mmol) in dioxane (10.0 mL) and water (2 mL) and the resulting mixture was stirred at 95 °C for 12 hours under N2. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0-40% Ethyl acetate/Petroleum ether; gradient 100 mL/min) to afford intermediate l-23b (0.2 g, 65%) as a yellow oil. ESI- MS (M+1 ): 467.3 calc. for C27H34N205: 466.3
Preparation of intermediate l-24a: 6,7-dimethoxy-2-(5-methyl-2-furyl)-4-(4- piperidylmethoxy)quinoline
A solution of intermediate l-23a (200 mg, 0.414 mmol) in HCI/EtOAc (1 .0 M, 10 mL) was stirred at 20 °C for 16 hours. Then, the reaction mixture was concentrated under vacuum to give intermediate l-24a (200 mg, crude) which was used into the next step without further purification as a red solid. ESI-MS (M+1 ): 383.3 calc. for C22H26N204: 382.2
Preparation of intermediate l-25a: ethyl 2-[4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolylloxymethyll-1 -piperidyllpyrimidine-5-carboxylate
To a solution of intermediate l-24a (200 mg, 0.477 mmol) in CH3CN (20 mL) was added K2C03 (198 mg, 1 .43 mmol) and ethyl 2-chloropyrimidine-5-carboxylate (R-08a, 134 mg, 0.716 mmol) and the mixture was stirred at 40 °C for 16 hours. Then, the reaction mixture was filtered and concentrated under vacuum to give intermediate l-25a (200 mg, crude) which was used into the next step without further purification as a yellow solid. ESI-MS (M+1 ): 533.4 calc. for C29H32N206: 532.2 Preparation of intermediate l-26a: 2-Γ4-ΓΓ6 J-dimethoxy-2-(5-methyl-2-furyl)-4- quinolylloxymethyll-l -piperidyllpyrimidine-5-carboxylic acid
To a solution of intermediate l-25a (200 mg, 0.375 mmol) in THF (20 mL) and H20 (10 mL) was added LiOH-H20 (31 mg, 0.751 mmol) and the mixture was stirred at 20 °C for 16 hours. Then, the reaction mixture was adjusted pH to 6 with 0.5 M aqueous HCI. The solution was concentrated under vaccum and freeze-drying to give crude product which was purified by prep-HPLC (Method 36) to afford pure intermediate l-26a (40 mg, 79 μη-ιοΙ, 21 %) as a yellow solid. ESI-MS (M+1 ): 505.3 calc. for C27H28N406: 504.2. Preparation of intermediate l-27a: 2-[4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolylloxymethyll-1 -piperidyll-N-tetrahydropyran-2-yloxy-pyrimidine-5-carboxamide
A mixture of intermediate l-26a (40 mg, 79 μηηοΙ), O-tetrahydropyran-2- ylhydroxylamine (19 mg, 0.158 mmol), HOBt (21 mg, 0.158 mmol), EDCI (30 mg, 0.158 mmol) and DIEA (31 mg, 0.237 mmol) in DMF (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. Then, the reaction mixture was quenched with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure to intermediate l-27a (50 mg, crude) as a light yellow oil. ESI-MS (M+1 ): 604.4 calc. for C32H37N507: 603.3.
Preparation of compound 6-01 : 2-[4-[[6,7-dimethoxy-2-(5-methyl-2-furyl)-4- quinolylloxymethyll-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
To a solution of intermediate l-27a (50 mg, 83 μηηοΙ) in CH3CN (5 mL) was added aqueous HCI (2.0 M, 124.24 μΙ_) and the mixture was stirred at 25 °C for 10 minutes. Then, the solvent was removed to give a residue, which was purified by prep-HPLC (Method 37) to give compound 6-01 (13.8 mg, 32%) as a light yellow solid. ESI-MS (M+1 ): 520.0 calc. for C27H29N5O6: 519.2. HPLC analytical method 1 , Rt = 1.813 min. Following the same synthetic route for compound 6-01 from l-23a and R-08a and using the intermediates and reagents indicate below, the following compunds were obtained:
Figure imgf000071_0001
Synthetic route 7
Figure imgf000072_0001
Figure imgf000072_0002
Figure imgf000072_0003
Conditions: a) TEA (2.0 eq), DCM, ethyl 3-chloro-3-oxo-propanoate (1.1 eq), 0 °C; then 25 °C for 12 h; b) LiOH H20 (1.5-5.0 eq), THF/H20 (2:1 ) or THF/H20/MeOH (3:3:2), 25 °C for 12-16 h; c) POCI3, 90 °C, 2 h; d) R-02 (1.0 eq), K2C03 (2.0 eq), Pd(PPh3)4 (0.1 eq), dioxane/H20 (3:1 ), 100 °C for 12 h; e) R-06 (1.3 eq), Cs2C03 (3.0 eq), BINAP (0.3 eq), Pd2(dba)3 (0.2 eq), dioxane, 120 °C for 12 h; f) Pd/C, H2 (30 Psi, 206.9 KPa), EtOAc, 25 °C for 2 h; g) R-13 (1.2 eq), Cs2C03 (2.0 eq), DMF, 100 °C for 12 h; h) HCI/EtOAc (2.0 M), 25 °C for 1 h; i) (HCHO)n (3.0 eq), NaBH(OAc)3 (3.0 eq), HCOOH (1.0 eq), MeOH, 70 °C for 12h; j) O-tetrahydropyran-2-yl hydroxylamine (2.0 eq), HOBt (2.0 eq), EDCI (2.0 eq), DIEA (5.0 eq), DMF, 25 °C for 12.
In the scheme above n, m and p are 0 or 1 , Cyi and Cy 2 are a carbocycle, heterocycle, aryl or heteroaryl, F is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl and R, Rb, Rc, and Rd are independently hydrogen or a hydrocarbon chain, which optionally contains nitrogen atoms.
Preparation of intermediate l-29a: ethyl 3-(3-benzyloxy-4-methoxy-anilino)-3-oxo- propanoate To the mixture of commercially available 3-benzyloxy-4-methoxy-aniline (l-28a, 35 g, 0.153 mol) and TEA (30.87 g, 0.306 mol) in DCM (1 L) was added drop wise ethyl 3- chloro-3-oxo-propanoate (25.245 g, 0.168 mol) at 0 °C. The mixture was stirred at 25 °C for 12 hours, poured into water (2 L) and extracted with DCM 2 times. The combined organic phase was dried with Na2S04, filtered and concentrated to dryness to afford l-29a (40 g, 76.3%). ESI-MS (M+1 ): 344.2 calc. for C19H21 NO5: 343.1 .
Preparation of intermediate l-30a: 3-(3-benzyloxy-4-methoxy-anilino)-3-oxo-propanoic acid
To a mixture of intermediate l-29a (20.40 g, 59.41 mmol) in THF (100 mL), MeOH (100 mL) and H20 (67 mL) was added LiOH«H20 (3.74 g, 89.12 mmol) in one portion at 25 °C. The mixture was stirred at 25 °C for 16 hours. Then, organic solvent was removed by rotary evaporation under vacuum at 45 °C. The residue was poured into ice-water (w/w = 1/1 ) (200 mL) and stirred for 10 min. The resulting slurry was filtered and the filter cake was dried under vacuum to afford l-30a (19.30 g, 61.21 mmol, crude) as a white solid. ESI-MS (M+1 ): 316.2 calc. for C17H17NO5: 315.1 .
Preparation of intermediate 1-31 a: 7-benzyloxy-2,4-dichloro-6-methoxy-quinoline
Intermediate l-30a (7.00 g, 22.20 mmol) was suspended in POCI3 (68.08 g, 443.99 mmol) in a 500 mL single-necked round bottom flask. The mixture was stirred at 90 °C for 2 hours under N2. Then, the reaction mixture was cooled to 25 °C and concentrated to remove POCI3. The residue was further purified by silica gel column chromatography (Si02, eluent gradient PE:EtOAc = 50:1 to 10:1 ) to give 1-31 a (2.50 g, 34%). ESI-MS (M+1 ): 334.2 calc. for C17H13CI2NO2: 333.0
Preparation of intermediate l-32a: 7-benzyloxy-4-chloro-6-methoxy-2-(5-methyl-2- furyQquinoline
A mixture of intermediate 1-31 a (1 .8 g, 5.39 mmol), 4,4,5,5-tetramethyl-2-(5-methyl-2- furyl)-1 ,3,2-dioxaborolane (R-02b, 1 .12 g, 5.39 mmol), K2C03 (1 .49 g, 10.77 mmol) and Pd(PPh3)4 (622 mg, 0.538 mmol) in dioxane (15 mL) and water (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 12 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-25% Ethyl acetate/Petroleum ether; gradient 100 mL/min ) to afford intermediate l-32a (1 .8 g, 88%) as a light yellow solid. ESI-MS (M+1 ): 380.1 calc. for C22H18CINO3: 379.1
Preparation of intermediate l-33a: ethyl 2-[4-[[[7-benzyloxy-6-methoxy-2-(5-methyl-2- furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate A mixture of intermediate l-32a (1 .6 g, 4.21 mmol), ethyl 2-[4-(aminomethyl)-1 - piperidyl]pyrimidine-5-carboxylate (R-06a, 1 .65 g, 5.48 mmol, HCI salt), Cs2C03 (4.12 g, 13 mmol), BINAP (786 mg, 1.26 mmol) and Pd2(dba)3 (771 mg, 0.842 mmol) in dioxane (30 mL) was degassed and purged with N2for 3 times, and then the mixture was stirred at 120 °C for 12 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-80% Ethyl acetate/Petroleum ether; gradient 100 mL/min) to afford intermediate l-33a (1.2 g, 47%) as a light yellow solid. ESI- MS (M+1 ): 608.3 calc. for CssHs/NsOs: 607.3
Preparation of intermediate l-34a: ethyl 2-[4-[[[7-hvdroxy-6-methoxy-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
To a solution of nintermediate l-33a (1.2 g, 1.97 mmol) in EtOAc (30 mL) was added Pd/C (10%, 0.5 g) under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H2 (30 Psi, 206.9 KPa) at 25 °C for 2 hours. Then, the reaction mixture was filtered and the filter was concentrated to afford
intermediate l-34a (0.9 g, 88%) as a yellow solid. ESI-MS (M+1 ): 518.2 calc. for
C28H31 N5O5: 517.2 Preparation of intermediate l-35a: tert-butyl 6-[[4-[[1 -(5-ethoxycarbonylpyrimidin-2-yl)-4- piperidyllmethylaminol-6-methoxy-2-(5-methyl-2-furyl)-7-quinolylloxyl-2- azaspiro[3.31heptane-2-carboxylate
To a solution of intermediate l-34a (0.3 g, 0.579 mmol) and tert-butyl 6-methylsulfonyloxy- 2-azaspiro[3.3]heptane-2-carboxylate (R-13a, 203 mg, 0.696 mmol) in DMF (10 mL) was added Cs2C03 (378 mg, 1.16 mmol) and the mixture was stirred at 100 °C for 12 hours. Then, the mixture was poured into ice-water (w/w = 1/1 ) (10 mL) and the aqueous phase was extracted with DCM (10 mL*3). The combined organic phase was washed with brine (30 mL), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0-5% MeOH/DCM ether; gradient 50 mL/min) to afford intermediate I- 35a (0.37 g, 89%) as a light yellow solid. ESI-MS (M+1 ): 713.4 calc. for C39H48N6O7: 712.4
Preparation of intermediate l-36a: ethyl 2-[4-[[[7-(2-azaspiro[3.31heptan-6-yloxy)-6- methoxy-2-(5-methyl-2-furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate A mixture of intermediate l-35a (370 mg, 0.519 mmol) in HCI/EtOAc (10 mL, 2.0M) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. Then, the solvent was removed and the residue was purified by flash chromatography (C18 Flash Column, Eluent of 20-35% ACN/water; gradient 50 mL/min ) to afford intermediate l-36a (0.1 g, 31 %) as a yellow solid. ESI-MS (M+1 ): 613.3 calc. for C34H40N6O5: 612.3
Preparation of intermediate l-37a: ethyl 2-[4-[[[6-methoxy-7-[(2-methyl-2- azaspiro[3.3lheptan-6-yl)oxyl-2-(5-methyl-2-furyl)-4-quinolyllaminolmethyll-1 - piperidyllpyrimidine-5-carboxylate
A mixture of intermediate l-36a (0.1 g, 0.163 mmol), (HCHO)n (44 mg, 0.490 mmol), NaBH(OAc)3 (104 mg, 0.490 mmol) and HCOOH (7.8 mg, 0.163 mmol) in MeOH (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 70 °C for 12 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by prep-HPLC (Method 48) to afford intermediate l-37a (30 mg, 29%) as a light yellow solid. ESI-MS (M+1 ): 627.4 calc. for C35H42N6O5: 626.3 Preparation of intermediate l-38a: 2-[4-[[[6-methoxy-7-[(2-methyl-2-azaspiro[3.31heptan-6- yl)oxyl-2-(5-methyl-2-fui^l)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylic acid
To a solution of intermediate l-37a (30 mg, 48 μηηοΙ) in THF (20 mL) and water (10 mL) was added LiOH-H20 (10 mg, 0.239 mmol) and the mixture was stirred at 25 °C for 12 hours. Then, the mixture was adjusted to pH~5 with HCI (2 M) and extracted with ethyl acetate (5 mL*3). The combined organic phase was washed with brine (10 mL), dried with anhydrous Na2S04, filtered and concentrated in vacuum to afford intermediate l-38a (30 mg, crude) as a yellow solid. ESI-MS (M+1 ): 599.3 calc. for C33H38N6O5: 598.3 Preparation of compound 8-01 : 2-[4-[[[6-methoxy-7-[(2-methyl-2-azaspiro[3.31heptan-6- yl)oxyl-2-(5-methyl-2-furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5- carbohydroxamic acid
A mixture of intermediate l-38a (30 mg, 50 μηηοΙ), O-tetrahydropyran-2- ylhydroxylamine (12 mg, 100 mmol), HOBt (14 mg, 0.100 mmol), EDCI (19 mg, 0.100 mmol) and DIEA (32 mg, 0.250) in DMF (8 mL) was degassed and purged with N2 for 3 times. Then, the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. Then, the mixture was poured into 0.5 M aqueous HCI (0.5 mL) and concentrated in vacuum. The residue was purified by prep-HPLC (Method 49) to afford compound 8-01 (9.9 mg, 31 %) as a yellow solid. ESI-MS (M+1 ): 614.2 calc. for C33H39N7O5: 613.3. HPLC analytical method 1 , Rt = 1 .837 min.
Synthetic route 8
Figure imgf000076_0001
Conditions: a) POCI3, malonic acid, 90 °C for 12 h; b) R-02 (1.2 eq), K2C03 (2.0 eq), Pd(PPh3)4 (0.1 eq), dioxane/H20 (3:1 ), 100 °C for 12 h; c) R-06 (1.3 eq), Cs2C03 (3.0 eq), BINAP (0.3 eq), Pd2(dba)3 (0.2 eq), dixane, 120 °C for 12 h; d) LiOH H20 (5.0 eq), THF/H20 (2:1 ), 25 °C for 12 h; e) O-tetrahydropyran-2-yl hydroxylamine (2.0 eq), HOBt (2.0 eq), EDCI (2.0 eq), DIEA (5.0 eq), DMF, 25 °C for 12.
In the scheme above n, m and p are 0 or 1 , Cyi and Cy2 are a carbocycle, heterocycle, aryl or heteroaryl, R2 is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl and Ra, Rb, Rc and Rd are independently hydrogen or a hydrocarbon chain.
Preparation of intermediate l-40a: 2,4-dichloro-6-methoxy-7-methyl-quinoline
To a solution of commercially available 4-methoxy-3-methyl-aniline (l-39a, 0.5 g, 3.64 mmol) in POCI3 (10 mL) was added malonic acid (569 mg, 5.47 mmol) and the mixture was stirred at 90 °C for 12 hours. Then, the solution was concentrated and poured into ice-water (20ml_) and adjusted to pH~9 with aqueous Na2C03. The mixture was extracted with EtOAc (x3) and the combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0-10% Ethyl acetate/Petroleum ether; gradient 100 mL/min) to afford intermediate l-40a (0.28 g, 32%) as a yellow solid. ESI-MS (M+1 ): 242.1 calc. for
C11 H9CI2NO: 241.0
Preparation of intermediate 1-41 a: 4-chloro-6-methoxy-7-methyl-2-(5-methyl-2- furvQquinoline
A mixture of intermediate l-40a (0.28 g, 1.16 mmol), 4,4,5,5-tetramethyl-2-(5-methyl-2- furyl)-1 ,3,2-dioxaborolane (R-02b, 289 mg, 1 .39 mmol), K2C03 (320 mg, 2.31 mmol) and Pd(PPh3)4 (134 mg, 0.1 15 mmol) in dioxane (15 mL) and water (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 12 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0-20% Ethyl acetate/Petroleum ether; gradient 50 mL/min) to afford intermediate 1-41 a (0.12 g, 36%) as a yellow solid. ESI-MS (M+1 ): 288.1 calc. for C16H 14CINO2: 287.1
Preparation of intermediate l-42a: ethyl 2-[4-[[[6-methoxy-7-methyl-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate
A mixture of intermediate 1-41 a (0.12 g, 0.417 mmol), ethyl 2-[4-(aminomethyl)-1 - piperidyl]pyrimidine-5-carboxylate (R-06a, 143 mg, 0.542 mmol), Cs2C03 (408 mg, 1.25 mmol), BINAP (78 mg, 0.125 mmol) and Pd2(dba)3 (76 mg, 83 mol) in dioxane (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 °C for 12 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0-100% Ethyl acetate/Petroleum ether;
gradient 50 mL/min) to afford intermediate l-42a (0.1 1 g, 51 %) as a light yellow solid. ESI- MS (M+1 ): 516.2 calc. for C29H33N5O4: 515.3
Preparation of intermediate l-43a: 2-[4-[[[6-methoxy-7-methyl-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylic acid
To a solution of intermediate l-42a (1 10 mg, 0.213 mmol) in THF (8 mL) and water (4 mL) was added LiOH-H20 (45 mg, 1 .07 mmol) and the mixture was stirred at 25 °C for 12 hours. Then, the mixture was adjusted to pH~5 with aqueous HCI (2 M) and filtered to afford intermediate l-43a (80 mg, 77%) as a light yellow solid. ESI-MS (M+1 ): 488.3 calc. for C27H29N5O4: 487.2
Preparation of compound 9-01 : 2-[4-[[[6-methoxy-7-methyl-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
A mixture of intermediate l-43a (80 mg, 0.164 mmol), O-tetrahydropyran-2- ylhydroxylamine (38 mg, 0.328 mmol), HOBt (44 mg, 0.328 mmol), EDCI (63 mg, 0.328 mmol) and DIEA (106 mg, 0.820 mmol) in DMF (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. Then, the solution was poured into aqueous HCI (1 mL, 0.5 M) and concentrated in vacuum. The residue was purified by prep-HPLC (Method 52) to afford compound 9-01 (44.2 mg, 54%) as a white solid. ESI-MS (M+1 ): 503.3 calc. for C27H30N6O4: 502.2. HPLC analytical method 1 , Rt = 2.206. Synthetic route 9
Figure imgf000078_0001
Figure imgf000078_0002
Conditions: a) SOCI2, MeOH, 80 °C for 2 h; b) Pd/C, H2 (50 Psi, 344.7 KPa), EtOAC, 25 °C for 2 h; c) POCI3, malonic acid, 90 °C for 1 h; d), R-02 (1.0 eq), K2C03 (2.0 eq), Pd(PPh3)4 (0.1 eq), dioxane, 100 °C for 12 h; then LiOH H20 (2.0 eq), 100 °C for 2 h; e) TEA, diphenylphosphoryl azide, toluene, 20 °C for 2 h; f) t-BuOH, toluene, 90 °C for 2 h; g) NaH (3.0 eq), CH3I (3.0 eq), DMF, 20 °C for 2 h; h) HCI/EtOAc (4.0 M), 20-25 °C for 1 h; i) (HCHO)n (2.4 eq), AcOH (2.5 eq), NaBH3CN (2.4 eq), 70 °C for 12 h; j) R-06 (1.3 eq), Pd2(dba)3 (0.2 eq), BINAP (0.3 eq), Cs2C03 (3.0 eq), dioxane, 120 °C for 12 h; k) LiOH H20 (4.5-5.0 eq), THF or THF/H20 (2:1 ), 25 °C for 12 h; I) O-tetrahydropyran-2-yl hydroxylamine (2.0 eq), HOBt (2.0 eq), EDCI (2.0 eq), DIEA (5.0 eq), DMF, 25 °C for 12 h.
In the scheme above n, m and p are 0 or 1 , Cyi and Cy2 are a carbocycle, heterocycle, aryl or heteroaryl, F is a hydrocarbon chain, aryl, heteroaryl, carbocycle or heterocycle, R3 is 0(CrC6)alkyl and R4, R5, Rb, Rc and Rd are independently hydrogen or a
hydrocarbon chain.
Preparation of intermediate l-45a: methyl 2-methoxy-5-nitro-benzoate
To a solution of 2-methoxy-5-nitro-benzoic acid (l-44a, 4.5 g, 22.83 mmol) in MeOH (50 mL) was added SOCI2 (8.15 g, 68.48 mmol) and the mixture was stirred at 80 °C for 2 hours. Then, the mixture was cooled to 20 °C and concentrated under reduced pressure at 50 °C. The residue was poured into aqueous NaHC03 (50 mL) and the aqueous phase was extracted with ethyl acetate (x3). The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to afford intermediate l-45a (4.6 g, 95%) as a white solid. ESI-MS (M+1 ): 212.0 calc. for C9H9NO5: 21 1 .1 Preparation of intermediate l-46a: methyl 2,4-dichloro-6-methoxy-quinoline-7-carboxylate To a solution of intermediate l-45a (4.6 g, 21 .78 mmol) in EtOAc (60 mL) was added Pd/C (2 g, 5% purity) and the mixture was stirred at 25 °C for 2 hours under H2 (50 Psi, 344.7 KPa). Then, the reaction mixture was filtered and the filter was concentrated. The residue was disoolved in in POCI3 (40 mL) and malonic acid (2.15 g, 20.70 mmol) was added. The mixture was stirred at 90 °C for 1 hour. Then, the solution was concentrated and poured into ice-water (20 mL) and adjusted to pH~9 with aqueous Na2C03. The mixture was extracted with EtOAc 60 mL (x3) and the combined organic layers were washed with brine 50 mL, dried over Na2S04, filtered and concentrated under reduced pressure. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0-30% Ethyl acetate/Petroleum ether; gradient 100 mL/min) to afford intermediate l-46a (0.5 g, 12%) as a white solid. ESI-MS (M+1 ): 285.9 calc. for
C12H9CI2NO3: 285.0
Preparation of intermediate l-47a: 4-chloro-6-methoxy-2-(5-methyl-2-furyl)quinoline-7- carboxylic acid
A mixture of intermediate l-46a (0.5 g, 1.75 mmol), 4,4,5,5-tetramethyl-2-(5-methyl-2- furyl)-1 ,3,2-dioxaborolane (R-02b, 363 mg, 1 .75 mmol), K2C03 (483 mg, 3.50 mmol) and Pd(PPh3)4 (202 mg, 0.175 mmol) in dioxane (6 mL) and water (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 12 hours under N2 atmosphere. Then, LiOH-H20 (147 mg, 3.50 mmol) was added and the mixture was stirred at 100 °C for 2 hours. Then, the mixture was poured into 1 M HCI (10 mL) and filtered to obtain intermediate l-47a (0.7 g, crude) as a light yellow solid. ESI-MS (M+1 ): 318.0 calc. for C16H12CINO4: 317.1
Preparation of intermediate l-48a: 4-chloro-6-methoxy-2-(5-methyl-2-furyl)quinoline-7- carbonyl azide
To a solution of intermediate l-47a (0.6 g, 1 .89 mmol) in toluene (10 mL) was added TEA (573 mg, 5.67 mmol) and diphenylphosphoryl azide_(780 mg, 2.83 mmol) and the mixture was stirred at 20 °C for 2 hours. Then, the reaction mixture was quenched by addition aqueous NaHC03 (10 mL) and extracted with DCM twice. The combined organic layers were washed with brine, dried over Na2S04, filtered and concentrated under reduced pressure to give intermediate l-48a (0.7 g, crude) as a light yellow solid. ESI-MS (M+1 ): 343.0 calc. for C16H11 CIN4O3: 342.1
Preparation of intermediate l-49a: tert-butyl N-[4-chloro-6-methoxy-2-(5-methyl-2-furyl)-7- quinolyllcarbamate To a solution of intermediate l-48a (0.7 g, 2.04 mmol) in toluene (5 mL) was added t- BuOH (2.27 g, 30.6 mmol) and the mixture was stirred at 90 °C for 12 hours. Then, the solvent was removed and the residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-30% Ethyl acetate/Petroleum ether; gradient 50 mL/min) to give intermediate l-49a (0.3 g, 38%) as a yellow solid. ESI- MS (M+1 ): 389.0 calc. for C20H21 CIN2O4: 388.1
Preparation of intermediate l-50a: tert-butyl N-[4-chloro-6-methoxy-2-(5-methyl-2-furyl)-7- quinolyl]-N-methyl-carbamate
To a solution of intermediate l-49a (380 mg, 0.977 mmol) in DMF (10 mL) was added NaH (1 17 mg, 2.93 mmol) and CH3I (416 mg, 2.93 mmol) and the mixture was stirred at 20 °C for 2 hours. Then, the residue was poured into aqueous NH4CI (w/w = 1/1 ) (10 mL) and the aqueous phase was extracted with ethyl acetate twice. The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by flash silica gel chromatography (ISCO®; 4 g
SepaFlash® Silica Flash Column, Eluent of 0-10% Ethyl acetate/Petroleum ether;
gradient 50 mL/min) to give Intermediate l-50a (240 mg, 61 %) as a light yellow solid. ESI- MS (M+1 ): 403.0 calc. for C21 H23CIN2O4: 402.1 Preparation of intermediate l-50b: 4-chloro-6-methoxy-N,N-dimethyl-2-(5-methyl-2- furyl)quinolin-7-amine
A mixture of intermediate l-49a (0.25 g, 0.642 mmol) in HCI/EtOAc (10 mL, 4.0 M) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 20 °C for 1 hour under N2 atmosphere. Then, the solvent was removed and the residue was dissolved in MeOH (5 mL). To this solution was added (HCHO)n (141 mg, 1 .57 mmol), AcOH (157 mg, 2.61 mmol) and NaBH3CN (99 mg, 1.57 mmol) and the mixture was stirred at 70 °C for 12 hours. Then, the solvent was removed and water (10mL) was added. The aqueous phase was extracted with ethyl acetate twice and the combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by prep-HPLC (Method 50) to give intermediate l-50b (0.1 g, 60%) as an orange solid. ESI-MS (M+1 ): 317.2 calc. for C17H17CIN2O2: 316.1
Preparation of intermediate 1-51 a: ethyl 2-[4-[[[7-[tert-butoxycarbonyl(methyl)aminol-6- methoxy-2-(5-methyl-2-furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylate A mixture of intermediate l-50a (0.22 g, 0.546 mmol), ethyl 2-[4-(aminomethyl)-1 - piperidyl]pyrimidine- 5-carboxylate (R-06a, 220 mg, 0.710 mmol), Pd2(dba)3 (100 mg, 0.109 mmol), BINAP (102 mg, 0.164 mmol) and Cs2C03 (534 mg, 1 .64 mmol) in dioxane (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 °C for 16 hours under N2 atmosphere. Then, the mixture was filtered and the filtrate was concentrated. The residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-100% Ethyl acetate/Petroleum ether; gradient 50 mL/min) to give intermediate 1-51 a (0.3 g, 87%) as a light yellow solid. ESI-MS (M+1 ): 631 .3 calc. for C34H42N6O6: 630.3
Following the same synthetic route for intermediate 1-51 a and using the same reagents, the following intermediates were obtained:
Figure imgf000081_0001
Preparation of intermediate l-52a: 2-[4-[[[6-methoxy-7-(methylamino)-2-(5-methyl-2-furyl)- 4-quinolyl]amino]methyl]-1 -piperidyl]pyrimidine-5-carboxylic acid
A solution of intermediate 1-51 a (0.1 g, 0.158 mmol) in HCI/EtOAc (10 ml_, 4.0 M) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 1 hour under N2 atmosphere. Then, the solvent was removed and the residue was dissolved in THF (6 mL) and water (3 ml_); LiOH.H20 (30 mg, 0.705 mmol) was added and the mixture was stirred at 25 °C for 12 hours. Then, the mixture was adjusted to pH~5 with aqueous HCI (2 M) and extracted with ethyl acetate (x3). The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give intermediate l-52a (70 mg, 99%) as a yellow solid. ESI-MS (M+1 ): 503.3 calc. for C27H30N6O4: 502.2
Preparation of intermediate l-52b: 2-[4-[[[7-(dimethylamino)-6-methoxy-2-(5-methyl-2- furyl)-4-quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carboxylic acid
To a solution of intermediate 1-51 b (80 mg, 0.147 mmol) in THF (6 mL) and water (3 mL) was added LiOH-H20 (31 mg, 0.734 mmol) and the mixture was stirred at 25 °C for 12 hours. Then, the mixture was adjusted to pH~5 with aqueous HCI (2 M) and extracted with ethyl acetate (x3). The combined organic phase was washed with brine, dried with anhydrous Na2S04, filtered and concentrated in vacuum to give intermediate l-52b (70 mg, 92%) as a yellow solid. ESI-MS (M+1 ): 517.1 calc. for C28H32N6O4: 516.3
Preparation of compound 10-01 : 2-[4-[[[6-methoxy-7-(methylamino)-2-(5-methyl-2-furyl)-4- quinolyllaminolmethyll-1 -piperidyllpyrimidine-5-carbohvdroxamic acid
A mixture of intermediate l-52a (70 mg, 0.139 mmol), O-tetrahydropyran-2- ylhydroxylamine (33 mg,0.279 mmol), HOBt (38 mg, 0.279 mmol), EDCI (53 mg, 278 mmol) and DIEA (90 mg, 0.696 mmol) in DMF (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 12 hours under N2 atmosphere. Then, the mixture was poured into aqueous HCI (1 ml_, 0.5 M) and concentrated in vacuum. The residue was purified by prep-HPLC (Method 47) to give compound 10-01 (33.2 mg, 44%) as a yellow solid. ESI-MS (M+1 ): 518.4 calc. for C27H31 N7O4: 517.3. HPLC analytical method 1 , Rt = 2.241 min.
Following the same synthetic route for compound 10-01 and using the same reagents, the following compounds were obtained:
Figure imgf000082_0001
Biological Tests
Cell Proliferation Assay
Cell proliferation was analyzed after 48 hours of in vitro treatment using the CellTiter 96 Aqueous One Solution Cell Proliferation Assay (Promega, Madison, W). This is a colorimetric method for determining the number of viable cells in proliferation For the assay, HL-60 cells were cultured by triplicate at a density of 1 x106 cells/ml in 96-well plates (100.000 cells/well, 10ΟμΙ/well). Only the 60 inner wells were used to avoid any border effects.
After 48 hours of treatment, plates were centrifuged at 800g for 10 minutes and medium was removed. Then, cells were incubated with 100μΙ / well of medium and 20ul / well of CellTiter 96 Aqueous One Solution reagent. After 1 -3 hours of incubation at 37°C, absorbance was measured at 490nm in a 96-well plate reader. The background absorbance was measured in wells with only cell line medium and solution reagent. Data was calculated as a percentage of total absorbance of treated cell / absorbance of non treated cells. Gl50 values were then calculated using GraphPad Prism.
Cell Differentiation / Annexin-V Assay
Myeloid differentiation induction was confirmed by CD1 1 b surface marker expression and apoptosis was measured by Annexin-V staining. HL-60 cells were cultured at a density of 1 x106 cells/ml in 96-well plates (100.000 cells/well, Ι ΟΟμΙ/well). Cells were treated daily with ¼ Gl50 of compounds for 48 hours. After treatment, cells were washed twice with phosphate-buffered saline (PBS). Then cells were stained with 15 μΙ of APC-conjugated mouse anti-human CD1 1 b (Catalog No. 550019, BD Biosciences) and 1 μΙ of FITC- conjugated anti-human Annexin V (Catalog No. 556419, BD Biosciences) for 15 min at room temperature in the dark. Cells were washed twice PBS and analyzed by flow cytometry with a minimum acquisition of 10.000 events. In this assay three comparative compounds encompassed in the Markush formula disclosed in WO2015192981 were also tested. The formulas of these examples are shown below:
Figure imgf000083_0001
Comparative example Comparative example 2 Comparative example 3
Figure imgf000083_0002
Comparative Comparative Comparative Comparative example 4 example 5 example 6 example 7 Comparative example 2 corresponds to compound 3-07 of WO2015192981.
Comparative example 1 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available (1 -methyl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981. The crude product was purified by prep-HPLC (Method 33 in this document) to obtain comparative example 1 (39% yield) as a yellow solid. ESI-MS (M+1 ): 396.3 calc. for C23H29N303 395.22; HPLC analytical method 1 , Rt = 1.656 min.
Comparative example 3 was obtained by coupling 7-(3-(pyrrolidin-1 -yl)propoxy)-4-chloro- 6-methoxy-2-(2,5-dimethylfuran-3-yl)quinolin-3(4H)-one (intermediate l-05c in this document) with commercially available tert-butyl 4-(aminomethyl)piperidine-1 -carboxylate (R-07b in this document) under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 , followed by deprotection in HCI/EtOAc and then N-methylation with HCOOH, NaBH3CN and (HCHO)n in MeOH (60 °C for 16 hours under N2). The crude product was purified by prep-HPLC (Method 12 in this document) to obtain comparative example 3 (12% yield) as a yellow solid. ESI-MS (M+1 ): 507.3 calc. for C30H42N4O3 506.33; HPLC analytical method 1 , Rt = 1 .55 min.
Comparative example 4 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available cyclohexylmethanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981. The crude product was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-100% Ethyl acetate/Petroleum ether; gradient 50 mL/min) to obtain comparative example 4 (38% yield) as a light yellow solid. ESI-MS (M+1 ): 381 .3 calc. for C23H28N2O3: 380.2; HPLC analytical method 1 , Rt = 2.996 min.
Comparative example 5 was obtained by coupling 4-chloro-6,7-dimethoxy-2-(5- methylfuran-2-yl)quinoline (intermediate l-05d in this document) with commercially available (1 -pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 . The crude product was purified by prep-HPLC (Method 53) to obtain comparative example 5 (44% yield) as a yellow solid. ESI-MS (M+1 ): 460.4 calc. for C26H29N5O3: 459.2; HPLC analytical method 1 , Rt = 2.296 min.
Comparative example 6 was obtained by coupling 4-chloro-2-(2,5-dimethyl-3-furyl)-6,7- dimethoxy-quinoline (intermediate l-05e in this document) with commercially available (1 - pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 . The crude product was purified by prep-HPLC (Method 54) to obtain comparative example 6 (56% yield) as an off-white solid. ESI-MS (M+1 ): 474.4 calc. for C27H3i N503: 473.2; HPLC analytical method 1 , Rt = 2.345 min.
Comparative example 7 was obtained by coupling 4-chloro-2-(2,5-dimethyl-3-furyl)-6- methoxy-7-(3-pyrrolidin-1 -ylpropoxy)quinoline (intermediate l-05c in this document) with commercially available (1 -pyrimidin-2-yl-4-piperidyl)methanamine under the same reaction conditions as described for the preparation of compound 3-01 of WO2015192981 . The crude product was purified by prep-HPLC (Method 29) to obtain comparative example 7 (13% yield) as an off-white solid. ESI-MS (M+1 ): 571.4 calc. for C33H42N603: 570.3; HPLC analytical method 1 , Rt = 2.030 min.
Table 1 shows the functional response of selected compounds on established cell line HL- 60 (AML - Accute myeloid leukemia) expressed as the percentage of cells with CD1 1 b marker at the tested concentration (at 1/4 of its established GI5o); where "N.E" means neglibible effect (i.e. CD1 1 b% <5%); 10% <= CD1 1 b% <= 20% (+); 20% < CD1 1 b% <= 40% (++) and CD1 1 b% > 40% (+++). For comparison purposes, the corresponding effect of well-established reference compounds ATRA (all-trans-retinoic acid), and panobinostat (LBH-589) are given.
Figure imgf000085_0002
Figure imgf000085_0001
Table 1
Table 2 shows the functional response of selected compounds on HL-60 (GI5o, which is concentration of compound for 50% of maximal inhibition of cell proliferation); where, Gl50 ≥10μΜ (+), 1 μΜ < Gl50 < 10 μΜ (++), and 100 nM < Gl50 < 1 μΜ (+++), and Gl50 < 100 nM (++++).
Example G o HL-60
2-01 ++
2-02 ++
2-12 ++
3-03 ++
3-06 ++
2-10 ++ Example G o HL-60
2-14 ++
2-18 ++
2-21 +++
2-22 ++
3-15 ++
3-19 ++
3-20 ++
3-25 ++
4-01 ++
5-01 ++
9-01 +++
10-01 ++
10-02 +++
Table 2
The compounds of the invention in Table 1 are potent inducers of cellular differentiation whereas comparative examples did not show the ability to induce cell differentiation in the same cancer cell lines. Besides, some compounds of the invention (see table 2) are also potent inhibitors of cell proliferation.
Citation List
- Campos B. et al., "Differentiation Therapy Exerts Antitumor Effects on Stem-Like Glioma Cells", Clinical Cancer Research 2010, 16, 2715-2728.
- Yan M. et al.: "Differentiation therapy: a promising strategy for cancer treatment', Chinese Journal of Cancer 2016, 35:3) - Warrell RP Jr. et al., "Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid)", N Engl J Med. 1991 ), 324(20): 1385-93
- WO 2015/192981 - T. W. Green and P. G. M. Wuts, Protective Groups in Organic Chemistry (Wiley, 3rd ed. 1999, Chapter 2, pp. 17-200)
- T.W. Green and P. G. M. Wuts, Protective Groups in Organic Chemistry (Wiley, 3rd ed. 1999, Chapter 5, pp. 369-451 )

Claims

Claims
1 . A compound of formula (I), or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of any of its pharmaceutically or veterinary acceptable salts
Figure imgf000087_0001
(I)
wherein:
X is a biradical selected from -NRa-, -0-, -CRDRc-and
Figure imgf000087_0002
wherein B is a known ring system which is attached to the quinoline ring through the N atom, and which comprises a 3- to 7-membered saturated or partially unsaturated heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; wherein the ring system B is optionally substituted with: a) one Cy1; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z1 optionally substituted with one or more substituents Rd and/or one Cy2;
wherein Cy1 and Cy2 are optionally substituted with one or more substituents independently selected from Rd, and Z2 optionally substituted with one or more substituents Rd;
L is a biradical selected from Cy3 and Z3 optionally substituted with one or more substituents Rd and/or one or more Cy4; wherein Cy3 and Cy4 are independently optionally substituted with:
a) one Cy5; and/or
b) one or more substituents Rd, and/or c) one or more substituents Z4 optionally substituted with one or more substituents Rd and/or one Cy6;
wherein Cy5 and Cy6 are independently optionally substituted with one or more substituents independently selected from Rd, and Z5 optionally substituted with one or more substituents Rd;
wherein L is attached to X through Cy3 or Z3; and L is attached to -C(0)NHOH through either Cy3 or Z3, or alternatively, through any of the substituents of Cy3 or Z3;
Ri is selected from the group consisting of R , Cy7, halogen, -N02, -CN, -ORe, -OC(0)Re , -OC(0)ORe', -OC(0)NRe'Re', -NRe Re , -NRe C(0)Re', -NRe C(0)ORe', -NRe C(0)NRe Re , -NRe S(0)2Re', -NRe S02NRe Re , -SRe', -S(0)Re', -S(0)ORe', -S02Re', -S02(ORe ),
-S02NRe Re , -SC(0)NRe'Re', -C(0)Re', -C(0)ORe, -C(0)NRe Re , -C(0)NRe ORe', and -C(0)NRe'S02Re'; R2 is selected from the group consisting of Ra, halogen, -N02, -CN, -OR3', -OC(0)Ra', -OC(0)ORa', -OC(0)NRa Ra', -NRa Ra , -NRa C(0)Ra', -NRa C(0)ORa', -NRa C(0)NRa Ra , -NRa'S(0)2Ra', -NRa S02NRa Ra , -SRa', -S(0)Ra', -S(0)ORa', -S02Ra', -S02(ORa ),
-S02NRa Ra , -SC(0)NRa'Ra', -C(0)Ra', -C(0)ORa', -C(0)NRa Ra , -C(0)NRa ORa', and -C(0)NRa'S02Ra';
R3 is selected from the group consisting of R9, -OR9, -NRa R9, and -NRa COR9; each Ra is independently H or Ra; each Ra is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and Cy8, wherein each Ra is optionally substituted with one or more halogen atoms, Rb and Rc are independently selected from the group consisting of H , halogen,
(CrC6)alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and a Cy9; wherein (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain and Cy9 are independently optionally substituted with one or more halogen atoms; or alternatively
Rb and Rc, together with the carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring, which is optionally fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or
heterocyclic monocyclic ring; wherein the ring system is optionally substituted with:
a) one Cy10; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z6 optionally substituted with one or more substituents Rd and/or one Cy11;
wherein Cy10 and Cy11 are optionally substituted with one or more substituents independently selected from Rd, and Z7 optionally substituted with one or more substituents Rd; each Rd is independently selected from halogen, -N02, -CN, -OR3', -OC(Y)Ra',
-OC(Y)ORa', -OC(Y)NRa Ra , -NRa Ra , -NRa C(Y)Ra , -NRa C(Y)ORa , -NRa C(Y)NRa Ra , -NRaS(0)2Ra', -NRaS02NRa Ra , -SRa', -S(0)Ra', -S(0)ORa', -S02Ra', -S02(ORa ),
-S02NRa Ra , -SC(Y)NRa Ra , -C(Y)Ra', -C(Y)ORa', -C(Y)NRa Ra , -C(Y)NRaORa , and -C(0)NRa'S02Ra';
Y is O, S, or NRa ; each Re is independently H or Re; each Re is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, and Cy12, wherein each Re is optionally substituted with one or more substituents Ra or Rd; each R is independently selected from the group consisting of (CrC6)alkyl,
(C2-C6)alkenyl, (C2-C6)alkynyl, (C2-C6)hydrocarbon chain having one or more double bonds and one or more triple bonds, wherein each R is optionally substituted with:
a) one or more substituents Rd and/or
b) one Cy13 optionally substituted with one or more substituents independently
selected from Rd, and Z8 optionally substituted with one or more substituents Rd; each R9 is independently Cy14, or Z9 optionally substituted with one or more substituents Rd and/or one Cy15; wherein Cy14 or Cy15 are independently optionally substituted with:
a) one Cy16; and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z9 optionally substituted with one or more substituents Rd and/or one Cy17; wherein Cy16 and Cy17 is optionally substituted with one or more substituents independently selected from Rd, and Z10 optionally substituted with one or more substituents Rd; and Cy7 is a known ring system selected from the group consisting of:
(i) phenyl;
(ii) 5- or 6-membered heteroaromatic ring;
(iii) 3- to 7-membered carbocyclic or heterocyclic monocyclic ring, which is saturated or partially unsaturated;
(iv) 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring;
(v) phenyl fused to a 6- to 14-membered saturated or partially unsaturated carbocyclic or heterocyclic bicyclic ring, wherein the rings of the bicyclic ring are spiro-fused; and
(vi) 5- to 6-membered heteroaromatic ring fused to a 6- to 14-membered saturated or partially unsaturated carbocyclic or heterocyclic bicyclic ring, wherein the rings of the bicyclic ring are spiro-fused;
wherein Cy7 is optionally substituted with:
a) one Cy18, and/or
b) one or more substituents Rd, and/or
c) one or more substituents Z11 optionally substituted with one or more substituents Rd and/or one Cy19;
wherein Cy18 or Cy19 are optionally substituted with one or more substituents independently selected from Rd, and Z12 optionally substituted with one or more substituents Rd;
Z1-Z12 are independently selected from the group consisting of (CrCi2)alkyl,
(C2-Ci2)alkenyl, (C2-Ci2)alkynyl, and (C2-Ci2)hydrocarbon chain having one or more double bonds and one or more triple bonds, wherein any carbon atom of Z1-Z12 is optionally a spiro atom substituted with Rh and R', and Rh and R', together with the spiro carbon atom, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
Cy1-Cy4, and Cy12-Cy15 are independently a known ring system selected from the group consisting of phenyl; 5- or 6-membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring;
Cy5-Cy6, Cy8-Cy11, and Cy16-Cy19 are independently a known ring system selected from the group consisting of phenyl; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 5- or 6-membered heteroaromatic ring; wherein in the carbocyclic rings all ring members are carbon atoms; and in the
heterocyclic and heteroaromatic rings one or more ring members are selected from N, O, and S; and wherein in all saturated or partially unsaturated rings one or two members of the rings are optionally C(O) and/or C(NH) and/or C[N(C C4)alkyl].
2. The compound of formula (I) according to claim 1 , wherein X is a biradical selected from -NRa-, -0-.
3. The compound of formula (I) according to any of the claims 1 -2, wherein L is a moiety of formula (XVIII):
Figure imgf000091_0001
(XVIII)
wherein: m and n are independently a value selected from 0 to 2; p is a value selected from 0 to 1 ;
Rj and Rk are independently selected from the group consisting of H, halogen, and
(CrC3)alkyl optionally substituted with one or more halogen atoms; or alternatively
R and Rk, together with the spiro carbon atom to which they are attached, form a known ring system comprising a 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring;
Cy is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy20 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms,
Cy21 is a known ring system selected from the group consisting of phenyl; 3- to 7- membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 5- or 6-membered heteroaromatic ring; and Cy21 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms.
4. The compound of formula (I) according to any of the claims 1 -3, wherein R-i is Cy7 optionally substituted as defined in claim 1.
5. The compound of formula (I) according to claim 4, wherein Cy7 is a known ring system selected from the group consisting of:
(i) phenyl;
(ii) 5- or 6-membered heteroaromatic ring;
(iii) 3- to 7-membered carbocyclic or heterocyclic monocyclic ring, which is saturated or partially unsaturated;
wherein Cy7 is optionally substituted as defined in claim 1.
6. The compound of formula (I) according to any of the claims 1 -5, wherein R2 is selected from halogen, -CN and -ORa .
7. The compound of formula (I) according to claim 6, wherein R2 is -ORa.
8. The compound of formula (I) according to any of the claims 1 -7, wherein R3 is selected from the group consisting of -OR9, and -NRa R9.
9. The compound of formula (I) according to claim 8, wherein R3 is -OR9.
10. The compound of formula (I) according to claim 9, wherein R9 is Z9 optionally substituted as defined in claim 1.
1 1 . The compound of formula (I) according to any of the claims 1 -10, wherein in R3, R9 contains at least one N atom.
12. The compound of formula (I) according to any of the claims 1 -1 1 , wherein R3 is a moiety of formula (XIX):
Cy22-
Figure imgf000093_0001
(XIX)
wherein:
Cy22 is a known ring system selected from the group consisting of phenyl; 5- or 6- membered heteroaromatic ring; 3- to 7-membered saturated or partially unsaturated carbocyclic or heterocyclic monocyclic ring; and 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring, which is fused, bridged-fused or spiro-fused to a 3- to 7-membered saturated or partially unsaturated or aromatic carbocyclic or heterocyclic monocyclic ring; and Cy22 is optionally substituted with one or more substituents selected from halogen and (CrC3)alkyl optionally substituted with one or more halogen atoms,
X1 and X2 are independently H or halogen, and r is a value selected from 0 to 6.
13. A pharmaceutical or veterinary composition which comprises a therapeutically effective amount of a compound of formula (I) as defined in any of the claims 1 -12, or a pharmaceutically or veterinary acceptable salt thereof, or any stereoisomer or mixtures of stereoisomers, either of the compound of formula (I) or of its pharmaceutically or veterinary acceptable salt, together with one or more pharmaceutically or veterinary acceptable excipients or carriers.
14. A compound of formula (I) according to any of the claims 1 -12 or a pharmaceutical or veterinary composition according to claim 13, for use in the treatment and/or prevention of cancer.
15. The compound of formula (I) or a pharmaceutical composition for use according to claim 14, wherein the cancer is selected from the group consisting of Non-Hodgkin's Lymphoma, Hodgkin's disease, hepatopancreatic tumor, biliar tumor, gastrointestinal tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, CNS tumor, melanoma, prostate cancer, renal cancer, small-cell lung cancer, non small-cell lung cancer, acute myeloid leukemia (AML), acute lymphoblastic leukemia, and multiple myeloma.
PCT/EP2018/065678 2017-06-14 2018-06-13 Novel compounds for use in cancer WO2018229139A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17382365 2017-06-14
EP17382365.9 2017-06-14

Publications (1)

Publication Number Publication Date
WO2018229139A1 true WO2018229139A1 (en) 2018-12-20

Family

ID=59078010

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/065678 WO2018229139A1 (en) 2017-06-14 2018-06-13 Novel compounds for use in cancer

Country Status (1)

Country Link
WO (1) WO2018229139A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022523105A (en) * 2019-02-01 2022-04-21 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー ENPP1 Inhibitors and Methods to Modulate Immune Responses
CN114751858A (en) * 2022-04-20 2022-07-15 沈阳药科大学 Quinolinyl-containing tranexamic acid derivative, preparation and application thereof
US11701371B2 (en) 2017-09-08 2023-07-18 The Board Of Trustees Of The Leland Stanford Junior University ENPP1 inhibitors and their use for the treatment of cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005053610A2 (en) * 2003-11-26 2005-06-16 Aton Pharma, Inc. Diamine and iminodiacetic acid hydroxamic acid derivatives
WO2015192981A1 (en) 2014-06-16 2015-12-23 Fundación Para La Investigación Médica Aplicada Novel compounds as dual inhibitors of histone methyltransferases and dna methyltransferases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005053610A2 (en) * 2003-11-26 2005-06-16 Aton Pharma, Inc. Diamine and iminodiacetic acid hydroxamic acid derivatives
WO2015192981A1 (en) 2014-06-16 2015-12-23 Fundación Para La Investigación Médica Aplicada Novel compounds as dual inhibitors of histone methyltransferases and dna methyltransferases

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
CAMPOS B, CLINICAL CANCER RESEARCH, 2010
CAMPOS B. ET AL.: "Differentiation Therapy Exerts Antitumor Effects on Stem-Like Glioma Cells", CLINICAL CANCER RESEARCH, vol. 16, 2010, pages 2715 - 2728, XP055049309, DOI: doi:10.1158/1078-0432.CCR-09-1800
SERGIO VALENTE ET AL: "Novel Cinnamyl Hydroxyamides and 2-Aminoanilides as Histone Deacetylase Inhibitors: Apoptotic Induction and Cytodifferentiation Activity", CHEMMEDCHEM, vol. 6, no. 4, 4 April 2011 (2011-04-04), pages 698 - 712, XP055427149, ISSN: 1860-7179, DOI: 10.1002/cmdc.201000535 *
T. W. GREEN; P. G. M. WUTS: "Protective Groups in Organic Chemistry", 1999, WILEY, article "Chapter 2", pages: 17 - 200
T. W. GREEN; P. G. M. WUTS: "Protective Groups in Organic Chemistry", 1999, WILEY, pages: 369 - 451
T.W. GREEN; P. G. M. WUTS: "Protective Groups in Organic Chemistry", 1999, WILEY, pages: 369 - 451
WARRELL RP JR. ET AL.: "Differentiation therapy of acute promyelocytic leukemia with tretinoin (all-trans-retinoic acid", N ENGL J MED., vol. 324, no. 20, 1991, pages 1385 - 1393
WARRELL RP JR., N ENGL J MED., 1991
YAN M, CHINESE JOURNAL OF CANCER, 2016
YAN M. ET AL.: "Differentiation therapy: a promising strategy for cancer treatmenf", CHINESE JOURNAL OF CANCER, vol. 35, 2016, pages 3, XP055431765, DOI: doi:10.1186/s40880-015-0059-x

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11701371B2 (en) 2017-09-08 2023-07-18 The Board Of Trustees Of The Leland Stanford Junior University ENPP1 inhibitors and their use for the treatment of cancer
US11707471B2 (en) 2017-09-08 2023-07-25 The Board Of Trustees Of The Leland Stanford Junior University ENPP1 inhibitors and their use for the treatment of cancer
JP2022523105A (en) * 2019-02-01 2022-04-21 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー ENPP1 Inhibitors and Methods to Modulate Immune Responses
CN114751858A (en) * 2022-04-20 2022-07-15 沈阳药科大学 Quinolinyl-containing tranexamic acid derivative, preparation and application thereof
CN114751858B (en) * 2022-04-20 2023-09-12 沈阳药科大学 Quinolinyl-containing tranexamic acid derivative and preparation and application thereof

Similar Documents

Publication Publication Date Title
TWI798446B (en) Compounds as nuclear transport modulators and uses thereof
TWI551595B (en) 2,4-disubstituted benzene-1,5-diamine derivatives and their use, Its preparation of pharmaceutical compositions and pharmaceutical compositions
CN112204029B (en) Therapeutic compounds
US9890168B2 (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
WO2021169963A1 (en) Aromatic compound and use thereof in preparing antineoplastic drugs
JP2022518591A (en) Heterocyclic compound benzopyridone and its use
WO2022105855A1 (en) Kras g12d inhibitors
CA3035860A1 (en) Pyridine compound
WO2016070816A1 (en) Pyrimidine or pyridine compounds, preparation method therefor and pharmaceutical uses thereof
EP2964223A1 (en) Compounds inhibiting leucine-rich repeat kinase enzyme activity
WO2018229139A1 (en) Novel compounds for use in cancer
TW201838984A (en) Jak1 selective inhibitors
JP2023524361A (en) Heterocyclic compounds that inhibit TYK2 activity
CN113024544A (en) Cyano-containing heterocyclic compound and application thereof
WO2016140501A1 (en) Pyridine n-oxide for enhancer of zeste homolog 2 inhibitors
JP7039579B2 (en) 4-Anilino-quinoline compound as an anticancer agent
CN114585621B (en) Imidazoquinoline amine derivative, pharmaceutical composition and application thereof
WO2018137655A1 (en) Pyrrolo-pyridines n-oxide derivative, preparation method therefor, and application thereof
TW202017928A (en) Novel tricyclic compounds
WO2019223548A1 (en) 3-oxazolinone compound, preparation method therefor and pharmaceutical application thereof
JP7110335B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
WO2022037365A1 (en) Double-target inhibitor targeting fgfr and hdac, preparation method therefor and application thereof, pharmaceutical composition and medicament
WO2019149128A1 (en) 5-chloro-2,4-pyrimidine derivative used as anti-tumor drug
JP2005529079A (en) Compounds and methods for treating cell proliferative disorders
KR20200012971A (en) Carboxylic Acid Derivatives of Pyridoquinazoline Useful as Protein Kinase Inhibitors

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: 18729460

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18729460

Country of ref document: EP

Kind code of ref document: A1