Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
QUINAZOLINE DERIVATIVES AND THEIR USE AS DNA METHYLTRANSFERASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2015/040169
Kind Code:
A1
Abstract:
The present invention relates to compounds of the following formula (I): and pharmaceutically acceptable salts and solvates thereof, their methods of preparation, their use as a drug, notably in the treatment of cancer, and pharmaceutical compositions containing such compounds.

Inventors:
HALBY LUDOVIC (FR)
ARIMONDO PAOLA BARBARA (FR)
Application Number:
PCT/EP2014/070011
Publication Date:
March 26, 2015
Filing Date:
September 19, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PF MEDICAMENT (FR)
CENTRE NAT RECH SCIENT (FR)
International Classes:
C07D401/14; A61K31/517; A61P35/00; C07D401/12
Domestic Patent References:
WO2006078752A22006-07-27
WO2009049132A12009-04-16
Attorney, Agent or Firm:
REGIMBEAU (Paris Cedex 17, Paris Cedex 17, FR)
Download PDF:
Claims:
CLAIMS

A compound f the following formula (I):

or a pharmaceutically acceptable salt or solvate thereof,

wherein:

- represents a single bond or a double bond on the condition that the two bonds do not represent a double bond at the same time,

- nl and n2 represent, independently of each other, an integer comprised between 0 and 8,

- Q represents an optionally substituted aryl or an optionally substituted heterocycle,

- W represents NRo, a piperidinediyl, a piperazinediyl or a pyrrolidinediyl,

- Xi represents O or NRi,

- X2 represents O or NR2,

- X3 represents:

v y

N when 3 represents a double bond 3 ? and

V V

NR3 when 3 represents a single bond 3 ?

- X4 represents:

v y

O or NR4 when 4 represents a double bond 4 s and

v v

■ OR4 or NR4R5 when 4 represents a single bond s

- Ro represents H; CHO; C02-((Ci-C6)alkyl); or a (Ci-C6)alkyl optionally substituted with CHO, C02H or C02-((Ci-C6)alkyl),

- Ri and R2 represent, independently of each other, H or a (Ci-C6)alkyl,

- R3 and R4 represent, independently of each other, H, (Ci-C6)alkyl, aryl, heterocycle, -((Ci-C6)alkyl)-X5-aryl or -((Ci-C6)alkyl)-X5-heterocycle,

with X5 representing a bond, O or NR6 and each aryl or heterocycle moiety being optionally substituted, and - R5 and R6 represent, independently of each other, H or a (Ci-C6)alkyl.

2. The compound according to claim 1 , wherein it is a compound of the following formula (I- 1 c) or (I- Id):

or a pharmaceutically acceptable salt or solvate thereof.

3. The compound according to any one of claims 1 and 2, wherein nl and n2 represent, independently of each other, an integer comprised between 1 and 4.

4. The compound according to any one of claims 1 to 3, wherein Xi represents NH and X2 represents O. 5. The compound according to any one of claims 1 to 4, wherein W represents NRo,

the nitrogen atom being linked

6. The compound according to any one of claims 1 to 5, wherein Q represents an aryl or heterocycle optionally substituted with one or several groups selected from halogen; oxo (=0); OR„; NR12R13; C(0)R14; C02R15; OC(0)R16; C(0)NR17Ri8; NR19C(O)R20; (d- C6)alkyl optionally substituted with one or several groups selected from halogen, OR2i , NR22R23, C(0)R24, C02R25, OC(0)R26, C(0)NR27R28, and NR29C(O)R30; and aryl optionally substituted with one or several groups selected from halogen, OR31 , NR32R33, C(0)R34, C02R35, OC(0)R36, C(0)NR37R38, and NR39C(0)R4o,

with R11 to R40 representing, independently of one another, H or (Ci-C6)alkyl,

the aryl being in particular a phenyl or a naphtyl, and

the heterocycle being in particular a saturated, unsaturated or aromatic hydrocarbon monocycle or bicycle, each cycle having 5 or 6 members and 1 to 4 carbon atoms having each been replaced with a nitrogen or oxygen atom.

7. The compound according to claim 6, wherein Q represents a cycle of the following formula:

wherein:

- X11 represents N or CR41,

- X12 represents N or CR42,

- Xi3 represents N or C-NR43aR43b,

- Xi4 represents N or CR44,

- Xi 5 represents N or CR45,

- R43a and R4¾ each represent, independently of each other, H or (Ci-C6)alkyl, and in particular H,

- R41 , R42, R44 and R45 each represent, independently of each other, hydrogen; halogen;

OR„; NR12R13; C(0)R14; C02R15; OC(0)R16; C(0)NR17R18; NR19C(O)R20; (d- C6)alkyl optionally substituted with one or several groups selected from halogen, OR2i, NR22R23, C(0)R24, C02R25, OC(0)R26, C(0)NR27R28, and NR29C(O)R30; or aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR3h NR32R33, C(0)R34, C02R35, OC(0)R36, C(0)NR37R38, and

NR39C(0)R4o; notably hydrogen; halogen; ORn; NRi2Ri3; (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR2i, and NR22R23; or aryl optionally substituted with one or several groups selected from halogen, OR3i, and NR32R33, or

in the case of R44 and R45, R44 and R45 form together a chain of the following formula:

wherein:

Χΐ6 represents Ν or CR46,

Χΐ7 represents Ν or CR47,

■ Χι 8 represents Ν or CR48,

Χΐ9 represents Ν or CR49, and

R46, R47, R48 and R49 each represent, independently of one another, hydrogen; halogen; OR„; NRi2Ri3; C(0)RM; C02Ri5; OC(0)Ri6; C(0)NRi7Ri8; NRi9C(0)R2o; (Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR21 , NR22R23, C(0)R24, C02R25, OC(0)R26, C(0)NR27R28, and NR2gC(0)R3o; or aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR3i , NR32R33, C(0)R34, C02R35, OC(0)R36, C(0)NR37R38, and NR39C(O)R40; notably hydrogen; halogen; ORn; NRi2Ri3; (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR2i , and NR22R23; or aryl optionally substituted with one or several groups selected from halogen, OR3i , and NR32R33,

on the proviso that no more than three, notably two, and preferably one, of Xn , X12, X14, Xi5, Xi6, Xn, Xi8 and X19 represent N. 8. The compound according to any one of claims 1 to 7, wherein R3 and R4 represent, independently of each other, H, (Ci-C6)alkyl, aryl, heterocycle, -((Ci-C6)alkyl)-X5-aryl or - ((Ci-C6)alkyl)-X5-heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N02; ORn ; NRi2Rn; C(0)Ri4; C02Ri5; OC(0)Ri6; C(0)NRi7Ri8; NRi9C(O)R20; S(O)R50; S(0)2R5i; S(0)2NR52R53; (Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR2i , NR22R23, C(0)R24, C02R25, OC(0)R26, C(0)NR27R28, NR29C(O)R30, S(0)R54, S(0)2R55, and S(0)2NR56R57; and aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR31 , NR32R33, C(0)R34, C02R35, OC(0)R36, C(0)NR37R38, NR39C(0)R4o, S(0)R58, S(0)2R59, and S(O)2NR60R6i,

with R11 to R4o and R50 to 51 representing, independently of one another, H or (Ci-C6)alkyl, the aryl being in particular a phenyl or a naphtyl, and

the heterocycle being in particular a saturated, unsaturated or aromatic hydrocarbon monocycle or bicycle, each cycle having 5 or 6 members and 1 to 4 carbon atoms having each been replaced with a nitrogen or oxygen atom.

9. The compound according to claim 8, wherein R3 and R4 represent, independently of each other, H, (Ci-C6)alkyl, aryl, heterocycle, aryl-(Ci-Ce)alkyl, heterocycle-(Ci-C6)alkyl, - ((Ci-C6)alkyl)-NH-aryl or -((Ci-C6)alkyl)-NH-heterocycle,

each aryl or heteroaryl moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N02; ORn; NRi2Rn; C(0)Ri4; S(0)2NR52R53; (Ci- C6)alkyl optionally substituted with one or several groups selected from halogen, OR2i, NR22R23, C(0)R24, and S(0)2NR56R57; and aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR3i, NR32R33, C(0)R34, and

the aryl being in particular a phenyl, and

the heterocycle being in particular a saturated hydrocarbon monocycle or bicycle, each cycle having 5 or 6 members and 1 to 4 carbon atoms having each been replaced with a nitrogen or oxygen atom, such as piperidine, piperazine, triazinane or pyrrolidine, and in particular piperidine.

10. The compound according to any one of claims 1 to 9, wherein it is selected from the following compounds:

and the pharmaceutically acceptable salts and solvates thereof.

11. A compound according to any one of claims 1 to 10 for use as a drug. 12. A compound according to any one of claims 1 to 10 for use in the treatment of cancer.

13. A compound according to any one of claims 1 to 10 for use as a DNA methylation inhibitor, in particular as a DNA methyltransferase (DNMT) inhibitor.

14. A pharmaceutical composition comprising at least one compound of formula (I) according to any one of claims 1 to 10 and at least one pharmaceutically acceptable excipient. 15. A pharmaceutical composition comprising:

(i) at least one compound of formula (I) according to any one of claims 1 to 10, and

(ii) at least one other active ingredient, such as an anticancer agent,

as a combination product for simultaneous, separate or sequential use. 16. A method to prepare a compound of formula (I) according to claim 1 or a pharmaceutically acceptable salt or solvate thereof, in which W = NRo with Ro representing a (Ci-C6)alkyl optionally substituted with CHO, C02H or C02-((Ci-C6)alkyl), comprising: (a) reacting a compound of formula (I) according to claim 1 in which W = NH with:

- a compound of formula Ro-LG where Ro represents a (Ci-C6)alkyl optionally substituted with CHO, C02H or C02-((Ci-C6)alkyl) and LG represents a leaving group to give a compound of formula (I) in which W = NRo with Ro representing a (Ci-C6)alkyl optionally substituted with CHO, C02H or C02- ((d-C6)alkyl), - dimethylformamide (DMF) to give a compound of formula (I) in which W = NRo with Ro = CHO, or

- a compound of formula Ro-Ai where Ro represents C02-((Ci-Ce)alkyl) and Ai represents a (Ci-C6)alkoxy group or a halogen atom to give a compound of formula (I) in which W = NRo with Ro representing C02-((Ci-C6)alkyl), and

(b) optionally salifying or solvating the compound obtained in step (a) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) according to claim 1 in which W = NRo with Ro as defined above.

17. A method to prepare a compound of formula (I) according to claim 1 or a pharmaceutically acceptable salt or solvate thereof, in which W represents NRo,

comprising:

(1) reacting a compound of the following formula (II):

which Q, Xi and nl are as defined in claim 1 and Wi represents LGi, NHR8 with a compound of the following formula (III):

, defined in claim 1 and W2 represents LG2, NHRs

wherein LGi and LG2 represent, independently of each other, a leaving group and Rs represents Ro as defined in claim 1 or a N-protecting group, on the condition that:

when Wi represents LGi, then W2 represents NHR8

when Wi represents NHR8 or , then W2 represents LG2,

and, when Wi or W2 represents NHR8 with R8 representing a N-protecting group, deprotecting the nitrogen atom bearing the N-protecting group,

to give a compound of formula (I) as defined in claim 1 , and

(2) optionally salifying or solvating the compound obtained in step (1) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) as defined in claim 1.

18. A method to prepare a compound of formula (I) according to claim 1 or a pharmaceutically acceptable salt or solvate thereof, comprising:

(A) reacting a compound of the following formula (VII):

Q *6 (VII)

in which Q is as defined in claim 1 and X6 represents a halogen atom or -Xi- (CH2)ni-W-(CH2)n2-X2H with W, Xi, X2, nl and n2 as defined in claim 1 , with a compound of the following formula (VIII):

in which X3 and X4 are as defined in claim 1 and X7 represents a halogen atom or X2-(CH2)„2-W-(CH2)„i-XiH with W, X X2, nl and n2 as defined in claim 1 , on the condition that: when X6 represents a halogen atom, then X7 represents -X2-(CH2)n2-W-

when X6 represents -Xi-(CH2)ni-W-(CH2)n2-X2H, then X7 represents a halogen atom,

to give a compound of formula (I) as defined in claim 1, and

(B) optionally salifying or solvating the compound obtained in step (A) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) as defined in claim 1.

Description:
QUINAZOLINE DERIVATIVES AND THEIR USE AS DNA

METHYLTRANSFERASE INHIBITORS

The present invention relates to quinazoline derivatives useful as DNA methyltransferase (DNMT) inhibitors, notably in the treatment of cancer.

Gene expression is known to be controlled by epigenetic modifications. Methylation of deoxycytidines (dC) in the DNA was shown to play a key role in epigenetic regulation in mammals (Berger et al. Genes Dev. 2009, 23, 781; Kelly et al. Biotechnol. 2010, 28, 1069). It is the most stable epigenetic mark and occurs at CpG sites, which are regrouped in island and essentially located in promoters, repeated sequences and CpG island shores (Gros et al. Biochimie 2012, 94, 2280). Hypermethylation of promoters' CpG islands induces gene silencing while hypomethylation induces gene expression (Sharma et al. Carcinogenesis 2010, 31, 27; Esteller N. Engl. J. Med. 2008, 358, 1148).

The enzymes responsible for DNA methylation are DNA methyltransferases (DNMTs). Two families of catalytically-active DNMTs have been identified: DNMT1, responsible for DNA methylation maintenance during replication, and DNMT3A and 3B, responsible for de novo DNA methylation. DNMTs add a methyl group on the carbon-5 position of the deoxycytosine at the CpG site in the DNA by using S-adenosyl-L-methionine (AdoMet) as methyl donor (Jurkowska et al. ChemBioChem 2011, 12, 206).

Alteration of DNA methylation patterns lead to various diseases such as cancer

(Baylin and Jones Nat. Rev. Cancer 2011, 11, 726). Cancerous cells often present aberrant DNA methylation, in particular a specific hypermethylation of tumour suppressor genes is observed. Restoring their expression by specific inhibition of DNA methylation represents an attractive therapeutic strategy (Kelly et al. Biotechnol. 2010, 28, 1069; Fahy et al. Expert Opin. Ther. Pat. 2012, 22, 1427).

DNMT inhibitors can be divided into two families: nucleoside analogues and non- nucleosides. The first are the most active ones. Two of them were FDA approved: 5- azacytidine (Vidaza®) and 5-azadeoxycytidine (Dacogene®) (Gros et al. Biochimie 2012, 94, 2280). Despite their high efficiency, their poor bioavailability, their instability in physiologic media and their little selectivity restrict their use (Fahy et al. Expert Opin. Ther. Pat. 2012, 22, 1427). Non-nucleoside analogues present various structures and mechanisms of action. Many of them were shown to target the catalytic site but suffer from high toxicity, lack of specificity and weak activity. There exists thus a need for novel DNMT inhibitors.

The inventors of the present invention have thus discovered that quinazoline derivatives can be used as DNA methyltransferase (DNMT) inhibitors.

The present invention concerns thus a com ound of the following formula (I):

or a pharmaceutically acceptable salt or solvate thereof,

wherein:

- represents a single bond or a double bond on the condition that the two bonds do not represent a double bond at the same time,

- nl and n2 represent, independently of each other, an integer comprised between 0 and 8, notably between 1 and 8,

- Q represents an optionally substituted aryl or an optionally substituted heterocycle, - W represents a bond, NRo, a piperidinediyl, a piperazinediyl or a pyrrolidinediyl,

- Xi represents O or NRi,

- X 2 represents O or NR 2 ,

- X 3 represents:

v y

N when 3 represents a double bond 3 ? and

V V

■ NR 3 when 3 represents a single bond 3 ?

- X 4 represents:

v y

O or NR 4 when 4 represents a double bond 4 s and

v v

OR 4 or NR 4 R 5 when 4 represents a single bond s

- Ro represents H; CHO; C0 2 -((Ci-C 6 )alkyl); or a (Ci-C 6 )alkyl optionally substituted with CHO, C0 2 H or C0 2 -((Ci-C 6 )alkyl),

- Ri and R 2 represent, independently of each other, H or a (Ci-C 6 )alkyl, - R 3 and R4 represent, independently of each other, H, (Ci-C 6 )alkyl, aryl, heterocycle, -((Ci-C 6 )alkyl)-X 5 -aryl or -((Ci-C 6 )alkyl)-X 5 -heterocycle,

with X5 representing a bond, O or NR 6 and each aryl or heterocycle moiety being optionally substituted, and

- R 5 and R6 represent, independently of each other, H or a (Ci-C 6 )alkyl.

For the purpose of the invention, the term "pharmaceutically acceptable" is intended to mean what is useful to the preparation of a pharmaceutical composition, and what is generally safe and non-toxic, for a pharmaceutical use.

The term "pharmaceutically acceptable salt or solvate" is intended to mean, in the framework of the present invention, a salt or solvate of a compound which is pharmaceutically acceptable, as defined above, and which possesses the pharmacological activity of the corresponding compound.

The pharmaceutically acceptable salts comprise:

(1) acid addition salts formed with inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric and phosphoric acid and the like; or formed with organic acids such as acetic, benzenesulfonic, fumaric, glucoheptonic, gluconic, glutamic, glycolic, hydroxynaphtoic, 2-hydroxyethanesulfonic, lactic, maleic, malic, mandelic, methanesulfonic, muconic, 2-naphtalenesulfonic, propionic, succinic, dibenzoyl-L-tartaric, tartaric, p-toluenesulfonic, trimethylacetic, and trifluoroacetic acid and the like, and

(2) base addition salts formed when an acid proton present in the compound is either replaced by a metal ion, such as an alkali metal ion, an alkaline-earth metal ion, or an aluminium ion; or coordinated with an organic or inorganic base. Acceptable organic bases comprise diethanolamine, ethanolamine, N-methylglucamine, triethanolamine, tromethamine and the like. Acceptable inorganic bases comprise aluminium hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate and sodium hydroxide.

Acceptable solvates for the therapeutic use of the compounds of the present invention include conventional solvates such as those formed during the last step of the preparation of the compounds of the invention due to the presence of solvents. As an example, mention may be made of solvates due to the presence of water (these solvates are also called hydrates) or ethanol.

The term "(Ci-C 6 )alkyl", as used in the present invention, refers to a straight or branched saturated hydrocarbon chain containing from 1 to 6 carbon atoms including, but not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, t-butyl, n- pentyl, n-hexyl, and the like.

The term "(C2-C 6 )alkenyl", as used in the present invention, refers to a straight or branched unsaturated hydrocarbon chain containing from 2 to 6 carbon atoms and comprising at least one double bond, notably one double bond, including, but not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl and the like. It can be in particular an allyl group.

The term "aryl", as used in the present invention, refers to an aromatic hydrocarbon group comprising preferably 6 to 10 carbon atoms and comprising one or more, notably 1 or 2, fused rings, such as, for example, a phenyl or naphtyl group. Advantageously, it will be a phenyl group.

The term "aryl-(Ci-C 6 )alkyl", as used in the present invention, refers to an aryl group as defined above bound to the molecule via a (Ci-C 6 )alkyl group as defined above. In particular, the aryl-(Ci-C 6 )alkyl group is a benzyl group.

The term "(Ci-C 6 )alkyl-aryl", as used in the present invention, refers to a (Ci-

C 6 )alkyl group as defined above bound to the molecule via an aryl group as defined above. In particular, it can be a tolyl group (-PI1CH 3 ).

The term "heterocycle" as used in the present invention refers to a saturated, unsaturated or aromatic hydrocarbon monocycle or polycycle (comprising fused, bridged or spiro rings), such as a bicycle, in which one or more, advantageously 1 to 4, and more advantageously 1 or 2, carbon atoms have each been replaced with a heteroatom selected from nitrogen, oxygen and sulphur atoms, and notably being a nitrogen atom. Advantageously, the heterocycle comprises 5 to 15, notably 5 to 10 atoms in the ring(s). The ring(s) of the heterocycle has/have advantageously 5 or 6 members.

According to a particular embodiment, the heterocycle is a saturated, unsaturated or aromatic hydrocarbon monocycle or bicycle (comprising fused, bridged or spiro rings, notably fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom.

A heterocycle can be notably thiophene, furan, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazoles (1,2,3-triazole and 1,2,4-triazole), benzofuran, indole, benzothiophene, benzimidazole, indazole, benzoxazole, benzisoxazole, benzothiazole, benzisothiazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline, quinazoline, piperidine, piperazine, triazinane, morpholine, pyrrolidine, dihydropyridines, dihydropyrimidines (notably 1,2- dihydropyrimidine), dihydropyridazines, dihydropyrazines, dihydrotriazines, tetrahydropyridines, tetrahydropyrimidines, tetrahydropyridazines, tetrahydropyrazines, tetrahydrotriazines, etc.

The term "heterocycle-(Ci-C 6 )alkyl", as used in the present invention, refers to a heterocycle group as defined above bound to the molecule via a (Ci-C 6 )alkyl group as defined above.

The term "heteroaryl" as used in the present invention refers to an aromatic heterocycle as defined above.

According to a particular embodiment, the heteroaryl is an aromatic hydrocarbon monocycle or bicycle (i.e. comprising fused rings), each cycle having 5 or 6 members, notably 6 members, and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom.

A heteroaryl can be notably thiophene, furan, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazoles (1,2,3-triazole and 1,2,4-triazole), benzofuran, indole, benzothiophene, benzimidazole, indazole, benzoxazole, benzisoxazole, benzothiazole, benzisothiazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline, quinazoline, etc.

The term "heteroaryl-(Ci-C 6 )alkyl", as used in the present invention, refers to a heteroaryl group as defined above bound to the molecule via a (Ci-C 6 )alkyl group as defined above.

The term "piperidinediyl", as used in the present invention, refers to a divalent

-N N- piperidine moiety. It can be in particular W 1 or 1 .

The term "piperazinediyl", as used in the present invention, refers to a divalent

-N N- piperazine moiety. It can be in particular

The term "pyrrolidinediyl", as used in the present invention, refers to a divalent pyrrolidine moiety. It can be in particular or

An "optionally substituted" radical, as used in the present invention, refers to a radical optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; OR„; NR 12 R 13 ; C(0)R 14 ; C0 2 R 15 ; OC(0)R 16 ; C(0)NR 17 Ri8; NR 19 C(O)R 20 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 R5 3 ; (C 1 -C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , NR 29 C(O)R 30 , S(0)R 54 , S(0) 2 R 55 , and S(0) 2 NR 56 R 5 7; and aryl or aryl-(Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , NR 3 9C(0)R4o, S(0)R 58 , S(0) 2 R 59 , and

with Rii to R 4 o and R50 to 51 representing, independently of one another, H or (Ci-C6)alkyl.

An "optionally substituted" radical can be in particular a radical optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NRi 2 Ri 3 ; C(0)Ri 4 ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(O)R 20 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , and NR 29 C(O)R 30 ; and aryl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 39 C(0)R 4 o, with Rn to R 4 o as defined previously.

The term "halogen", as used in the present invention, refers to a fluorine, bromine, chlorine or iodine atom.

According to a particular embodiment of the present invention, the compound of the present invention is a compound of the following formula (I-l):

or a pharmaceutically acceptable salt or solvate thereof.

The formula (I) of the present invention comprises two bonds . According to a particular embodiment, one of them is a single bond and the other is a double bond. Thus the compound of the present invention can correspond to a compound of the following formulas (la) and (lb), preferably of the following formulas (I-la) and (I-lb): (lb), preferably

and

or a pharmaceutically acceptable salt or solvate thereof. In particular, nl can represent 0, 1, 2, 3 or 4, notably 1, 2, 3, or 4.

In particular, n2 can represent 0, 1, 2, 3 or 4, notably 1, 2, 3, or 4, such as 2 or 3.

Xi represents advantageously NH or O, in particular NH.

X 2 represents advantageously NH or O, in particular O.

According to a particular embodiment, Xi represents NRi and X 2 represents O, notably Xi represents NH and X 2 represents O. According to a first embodiment, W represents a bond, NRo, a piperidinediyl or a piperazinediyl. Advantageously, W represents NRo, a piperidinediyl or a piperazinediyl, notably NRo or a piperidinediyl.

According to a second embodiment, W represents a bond, NRo, a piperidinediyl, a piperazinediyl or a pyrrolidinediyl. Advantageously, W represents NRo, a piperidinediyl, a piperazinediyl or a pyrrolidinediyl, notably NRo, a piperidinediyl or a pyrrolidinediyl.

In these two embodiments, the piperidinediyl group can be or

-N N-

. The piperazinediyl group is in particular . The pyrrolidinediyl

group can

the nitrogen atom being linked to (CH 2 ) nl .

Ro represents notably H; CHO; or a (C 1 -C6)alkyl optionally substituted with C0 2 H or C0 2 -((C 1 -C6)alkyl) (e.g. C0 2 Me). According to a first particular embodiment, Ro represents H. According to a second particular embodiment, Ro represents CHO or C0 2 - ((Ci-C 6 )alkyl), such as CHO. According to a third particular embodiment, Ro represents a (Ci-C 6 )alkyl optionally substituted with CHO, C0 2 H or C0 2 -((Ci-C 6 )alkyl), notably with C0 2 H or C0 2 -((Ci-C 6 )alkyl) (e.g. C0 2 Me).

Q represents notably an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NR 12 R 13 ; C(0)R 14 ; C0 2 R 15 ; OC(0)R 16 ; C(0)NR 17 Ri8; NR 19 C(O)R 20 ; S(O)R 50 ; S(0) 2 R 51 ; S(0) 2 NR 52 R53; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , NR 29 C(O)R 30 , S(0)R 54 , S(0) 2 R 5 5, and S(0) 2 NR 5 6R57; and aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , NR 3 9C(0)R4o, S(0)R 58 , S(0) 2 R 59 , and S(0) 2 R6oR6i,

with Rii to R 4 o and R50 to 51 representing, independently of one another, H or (Ci-C6)alkyl. Q represents notably an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NR 12 R 13 ; C(0)Ri 4 ; CO2R15; OC(0)Ri 6 ; C(0)NRi 7 Ri8; NRi 9 C(O)R 20 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 2 3, C(0)R 2 4, CO2R25, OC(0)R 26 , C(0)NR 27 R 2 8, and NR 2 9C(O)R 30 ; and aryl optionally substituted with one or several groups selected from halogen, OR31, NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 3 9C(0)R4o,

with R 1 1 to R40 representing, independently of one another, H or (Ci-C6)alkyl.

Q represents in particular an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 23 ,

C(0)R 24 , CO2R25, OC(0)R 26 , C(0)NR 27 R 2 8, and NR 29 C(O)R 30 ; and aryl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 ,

C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 39 C(O)R 40 .

Q can also represent an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NRi 2 Ri 3 ;

C(0)Ri 4 ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(O)R 20 ; and (d-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 23 , C(0)R 24 ,

C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , and NR 29 C(O)R 30 .

Q can represent in particular an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NRi 2 Ri 3 ; and (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen,

OR 2 i andNR 22 R 23 .

Q represents particularly an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); and (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR 2 i and NR 22 R 23 .

In the definitions of Q above, the aryl is preferably a phenyl or a naphtyl, in particular a phenyl.

In the definitions of Q above, the heterocycle is notably a saturated, unsaturated or aromatic hydrocarbon monocycle or bicycle (comprising fused, bridged or spiro rings, notably fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom. The heterocycle can be notably chosen among pyrrole, imidazole, pyrazole, triazoles, indole, benzimidazole, indazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline, quinazoline, piperidine, piperazine, triazinane, pyrrolidine, dihydropyridines, dihydropyrimidines (notably 1 ,2-dihydropyrimidine), dihydropyridazines, dihydropyrazines, dihydrotriazines, tetrahydropyridines, tetrahydropyrimidines, tetrahydropyridazines, tetrahydropyrazines and tetrahydrotriazines. In particular, the heterocycle can be chosen among pyridine, pyrimidine, pyridazine, pyrazine, quinoline, isoquinoline, quinoxaline, quinazoline, piperidine, piperazine, dihydropyridines, dihydropyrimidines (notably 1,2-dihydropyrimidine), dihydropyridazines, dihydropyrazines, tetrahydropyridines, tetrahydropyrimidines, tetrahydropyridazines and tetrahydropyrazines. More particularly, the heterocycle can be chosen among quinoline, quinazoline, pyridine, pyrimidine and dihydropyrimidines (notably 1,2-dihydropyrimidine). Notably, the heterocycle can be chosen among quinoline, pyridine and dihydropyrimidines (notably 1,2-dihydropyrimidine).

In the definitions of Q above, the heterocycle is preferably an heteroaryl, such as an aromatic hydrocarbon monocycle or bicycle (i.e. comprising fused rings), each cycle having 5 or 6 members, notably 6 members, and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom. Preferably, the hereoaryl is an aromatic hydrocarbon monocycle or bicycle (i.e. comprising fused rings), each cycle having 6 members, and 1 or 2 carbon atoms having each been replaced with a heteroatom chosen among nitrogen and oxygen atoms, at least one heteroatom being a nitrogen atom and preferably all the heteroatoms being a nitrogen atom when two heteroatoms are present. The heteroaryl can be notably chosen among thiophene, furan, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazoles (1,2,3- triazole and 1,2,4-triazole), benzo furan, indole, benzothiophene, benzimidazole, indazole, benzoxazole, benzisoxazole, benzothiazole, benzisothiazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline and quinazoline. In particular, the heteroaryl can be chosen among pyridine, pyrimidine, pyridazine, pyrazine, quinoline, isoquinoline, quinoxaline, and quinazoline. Notably, the heteroaryl can be chosen among quinoline, quinazoline, pyridine and pyrimidine. In particular, it is quinoline or pyridine.

According to a preferred embodiment, Q represents a cycle of the following formula:

wherein:

- Xi i represents N or CR 41 ,

- X12 represents N or CR42,

- Xi3 represents N or C-NR43 a R43b,

- Xi4 represents N or CR44,

- Xi 5 represents N or CR45,

- R 43a and R 43b each represent, independently of each other, H or (Ci-C 6 )alkyl, and in particular H,

- R 41 , R 42 , R 44 and R 45 each represent, independently of each other, hydrogen; halogen;

N0 2 ; OR„; NR 12 Ri 3 ; C(0)R 14 ; C0 2 Ri 5 ; OC(0)R 16 ; C(0)NR 17 Ri8; NR 19 C(O)R 20 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 R53; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21, NR22R23, C(0)R24, CO2R25, OC(0)R26, C(0)NR 27 R 2 8, NR 2 9C(O)R 30 , S(0)R 54 , S(0) 2 R 55 , and S(0) 2 NR 56 R57; or aryl or aryl-(Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 31 ,

NR32R33, C(0)R 34 , CO2R35, OC(0)R 36 , C(0)NR 37 R 3 8, NR 39 C(0)R4o, S(0)R 58 , S(0) 2 R 59 , and S(O) 2 NR6 0 R6i, or

in the case of R 44 and R 45 , R 44 and R 45 form together a chain of the following formula:

- V '

' 19

Xl 7 Xl 8

wherein:

Xi6 represents N or CR46,

Xi 7 represents N or CR 47 ,

Xi 8 represents N or CR48,

Xi9 represents N or CR49, and

■ R 46 , R 47 , R 48 and R 49 each represent, independently of one another, hydrogen; halogen; N0 2 ; OR„; NR12R13; C(0)Ri 4 ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(O)R 20 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 R53; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 21 , NR 22 R 23 , C(0)R 24 , CO2R25, OC(0)R 26 , C(0)NR 27 R 2 8, NR 2 9C(O)R 30 , S(0)R 54 , S(0) 2 R 55 , and S(0) 2 NR 56 R 57 ; or aryl or aryl-(Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR31, NR32R33, C(0)R 34 , CO2R35, OC(0)R 36 , C(0)NR 37 R 3 8, NR 39 C(0)R4o, S(0)R 58 , S(0) 2 R 59 , and S(O) 2 NR6 0 R6i, on the proviso that no more than three, notably two, and preferably one, of Xn, X 12 , X 14 , Xi5, Xi6, Xn, Xi8 and X19 represent N.

In particular, none of Xn, Xi 2 , X14, X15, Xi6, Xn, Xi8 and X19 represents N.

Advantageously, R 41 , R4 2 , R44 and R 45 each represent, independently of each other, hydrogen; halogen; OR„; Ν¾ 2 ¾ 3 ; C(0)R M ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(0)R 2 o; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21, NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , and NR 29 C(O)R 30 ; or aryl or aryl-(Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 3h NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 39 C(O)R4 0 , or in the case of R 44 and R 45 , R 44 and R 45 form together a chain of the following formula:

with R 46 , R 47 , R 48 and R 49 each representing, independently of one another, hydrogen; halogen; OR„; Ν¾ 2 ¾ 3 ; C(0)R M ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(O)R 20 ; (Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , and NR 29 C(O)R 30 ; or aryl or aryl-(Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 39 C(0)R4o.

In particular, R 41 , R 42 , R 44 and R 45 each represent, independently of each other, hydrogen; halogen; OR„; Ν¾ 2 ¾ 3 ; C(0)R M ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(0)R 2 o; or (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 21 , NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 28 , and NR 29 C(O)R 30 , or

in the case of R 44 and R 45 , R 44 and R 45 form together a chain of the following formula: with R 46 , R 47 , 48 and R 49 each representing, independently of one another, hydrogen; halogen; OR„; Ν¾ 2 ¾ 3 ; C(0)R M ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri 8 ; NRi 9 C(O)R 20 ; or (Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21 , NR 22 R 23 , C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 2 8, and NR 29 C(O)R 30 .

Notably, R41 , R4 2 , R44 and R45 each represent, independently of each other, hydrogen; halogen; ORn; NRi 2 Ri 3 ; or (Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, and NR 22 R 23 , or

in the case of R44 and R45, R44 and R45 form together a chain of the following formula: with R46, R4 7 , R48 and R49 each representing, independently of one another, hydrogen; halogen; ORn; NRi 2 Ri 3 ; or (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i , and NR 22 R 23 .

In particular, R41 , R4 2 , R44 and R45 each represent, independently of each other, hydrogen; halogen; ORn; or NRi 2 Ri 3 ; and notably hydrogen, or

in the case of R44 and R45, R44 and R45 form together a chain of the following formula: with R46, R4 7 , R48 and R49 each representing, independently of one another, hydrogen; halogen; ORn; or NRi 2 Ri 3 .

According to a most preferred embodiment, Q represents one of the following

with R4 3a and R4¾ as defined above and in particular representing H, and with R41 , R4 2 and R44 to R49 as defined above, and in particular with R41 , R4 2 and R44 to R49 each representing, independently of one another, hydrogen; halogen; ORn; or NRi 2 Ri 3 . In particular, Q represents one of the following cycles:

with R43 a and R43b as defined above and in particular representing H, and with R46 to R49 each representing, independently of one another, hydrogen; halogen; ORn; or NR^Rn- Q can be for example one of the following cycles:

OMe and MeO OMe

According to a particular embodiment, X3 represents:

V Y

N when 3 represents a double bond 3 s and

v v

NR3 when 3 represents a single bond 3 s

According to another particular embodiment, X 4 represents:

v y

O when 4 represents a double bond 4 s and

v v

NR 4 R 5 when 4 represents a single bond 4 s The compound of the present invention can correspond in particular to a compound of the following formulas (Ic) and (Id), preferably of the following formulas (I-lc) and (I- Id):

and

preferably

and

or a pharmaceutically acceptable salt or solvate thereof.

More particularly, R 3 and R4 will represent, independently of each other, H, (Ci- C 6 )alkyl, aryl, heterocycle, -((Ci-C6)alkyl)-X 5 -aryl or -((Ci-C6)alkyl)-X 5 -heterocycle, each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NRi 2 Ri 3 ; C(0)Ri 4 ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRivRi8; NRi 9 C(O)R 20 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 R 53 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21, NR22R23, C(0)R24, CO2R25, OC(0)R 26 , C(0)NR 27 R 2 8, NR 2 9C(O)R 30 , S(0)R 54 , S(0) 2 R 55 , and S(0) 2 NR 56 R57; and aryl or aryl-(Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 3 NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , NR 3 9C(0)R4o, S(0)R 58 , S(0) 2 R 59 , and S(O) 2 NR6 0 R6i ,

with R 1 1 to R 40 and R 50 to 51 representing, independently of one another, H or (Ci-C 6 )alkyl.

R 3 and R 4 represent notably, independently of each other, H, (Ci-C 6 )alkyl, aryl, heterocycle, -((Ci-C 6 )alkyl)-X 5 -aryl or -((Ci-C 6 )alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NRi 2 Ri 3 ; C(0)Ri 4 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 R 53 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21, NR22R23, C(0)R 24 , S(0)R 54 , S(0) 2 R 55 , and S(0) 2 NR 5 6R57; and aryl or aryl- (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR 3 i, NR 32 R 33 , C(0)R 34 , S(0)R 58 , S(0) 2 R 59 , and S(O) 2 R6 0 R6i .

R 3 and R 4 represent notably, independently of each other, H, (Ci-C 6 )alkyl, aryl, heterocycle, -((Ci-C 6 )alkyl)-X 5 -aryl or -((Ci-C 6 )alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NRi 2 Ri 3 ; C(0)Ri 4 ; S(O)R 50 ; S(0) 2 R 5 i; S(0) 2 NR 52 53 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 21 , and NR 22 R 23 ; and aryl or aryl-(C 1 -C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 31 , and NR 32 R 33 ,.

R 3 and R 4 represent notably, independently of each other, H, (C 1 -C 6 )alkyl, aryl, heterocycle, -((C 1 -C 6 )alkyl)-X 5 -aryl or -((Ci-C 6 )alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NRi 2 Ri 3 ; C(0)Ri 4 ; S(0) 2 NR 5 2R 53 ; (Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 21 , NR22R2 3 , C(0)R24, and S(0)2NR 5 6R57; and aryl or aryl-(C 1 -C6)alkyl optionally substituted with one or several groups selected from halogen, OR 31 , NR 32 R 33 , C(0)R 34 , and

R 3 and R 4 represent notably, independently of each other, H, (C 1 -C 6 )alkyl, aryl, heterocycle, -((C 1 -C 6 )alkyl)-X 5 -aryl or -((Ci-C 6 )alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NRi 2 Ri 3 ; C(0)R M ; S(0) 2 NR 5 2R 53 ; (Ci- C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21 , and NR22R23; and aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR31 , and NR¾R 33 .

R 3 and R4 represent notably, independently of each other, H, (Ci-C6)alkyl, aryl, heterocycle, -((Ci-C6)alkyl)-X 5 -aryl or -((Ci-C6)alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn ; NR12R13; C(0)Ri 4 ; S(0) 2 NR 5 2R 5 3; (Ci- C 6 )alkyl; aryl; and aryl-(Ci-C6)alkyl.

R 3 and R4 represent notably, independently of each other, H, (Ci-C6)alkyl, aryl, heterocycle, -((Ci-C6)alkyl)-X 5 -aryl or -((Ci-C6)alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NR12R13; C(0)Ri 4 ; C0 2 Ri 5 ; OC(0)Ri 6 ; C(0)NRi 7 Ri8; NRi9C(0)R2o; (C 1 -C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21 , NR22R23, C(0)R 24 , C0 2 R 25 , OC(0)R 26 , C(0)NR 27 R 2 8, and NR2 C(0)R 3 o; and aryl optionally substituted with one or several groups selected from halogen, OR 3 NR 32 R 33 , C(0)R 34 , C0 2 R 35 , OC(0)R 36 , C(0)NR 37 R 38 , and NR 39 C(0)R4o.

R 3 and R4 represent notably, independently of each other, H, (C 1 -C6)alkyl, aryl, heterocycle, -((C 1 -C6)alkyl)-X 5 -aryl or -((Ci-C6)alkyl)-X 5 -heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; ORn; NR12R13; (C 1 -C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21 and NR22R23; and aryl optionally substituted with one or several groups selected from halogen, OR 3 i and NR¾R 33 .

R 3 and R4 represent notably, independently of each other, H, (C 1 -C6)alkyl, aryl, heterocycle, aryl-(C 1 -C6)alkyl, heterocycle-(C 1 -C6)alkyl, -((Ci-Ce)alkyl)-NH-aryl or -((Ci- C 6 )alkyl)-NH-heterocycle,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; ORn; NR12R13; (C 1 -C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR21 and NR22R23; and aryl optionally substituted with one or several groups selected from halogen, OR 3 i and NR¾R 33 .

In the definitions of R 3 and R4 above, the aryl preferably is a phenyl or a naphtyl, in particular a phenyl.

In the definitions of R 3 and R4 above, the heterocycle is notably a saturated, unsaturated or aromatic (notably aromatic) hydrocarbon monocycle or bicycle (comprising fused, bridged or spiro rings, notably fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom. The heterocycle can be a heteroaryl. The heterocycle can be notably chosen among pyrrole, imidazole, pyrazole, triazoles, indole, benzimidazole, indazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline, quinazoline, piperidine, piperazine, triazinane, pyrrolidine, dihydropyridines, dihydropyrimidines (notably 1,2-dihydropyrimidine), dihydropyridazines, dihydropyrazines, dihydrotriazines, tetrahydropyridines, tetrahydropyrimidines, tetrahydropyridazines, tetrahydropyrazines and tetrahydrotriazines. According to a first embodiment, the heterocycle is chosen among pyrrole, imidazole, pyrazole, triazoles, indole, benzimidazole, indazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline and quinazoline; notably chosen among pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline and quinazoline. According to a second embodiment, the heterocycle is chosen among piperidine, piperazine, triazinane or pyrrolidine; and in particular is piperidine.

In the definitions of R 3 and R4 above, X 5 represents in particular a bond or NR6, notably a bond or NH.

According to a preferred embodiment, R5 represents H.

According to a particular embodiment, the compounds according to the present invention are compounds of formula (I- 1 c) or (I- Id), or a pharmaceutically acceptable salt or solvate thereof,

wherein:

- nl and n2 represent, independently of each other, 1, 2, 3 or 4,

- Q represents an aryl or heteroaryl optionally substituted with one or several groups selected from halogen; oxo (=0); ORn; NRi 2 Ri 3 ; and (Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR 2 i and NR 22 R 23 . - W represents a bond, NRo, the nitrogen

atom being linked to (CH 2 ) n i; notably NRo,

the nitrogen atom being linked to (CH 2 ) n i,

- Xi represents NH,

- X 2 represents O,

- Ro represents H; CHO; or a (Ci-C6)alkyl optionally substituted with C0 2 H or C0 2 - ((Ci-C 6 )alkyl),

- R 3 and R4 represent, independently of each other, H, (Ci-C6)alkyl, aryl heterocycle, aryl-(Ci-C 6 )alkyl, heterocycle-(Ci-C 6 )alkyl, -((Ci-C 6 )alkyl)-NH-aryl or -((Ci- C6)alkyl)-NH-heterocycle, notably , H, (Ci-C 6 )alkyl, heterocycle, aryl-(Ci-C6)alkyl, or -((Ci-C 6 )alkyl)-NH-aryl,

each aryl or heterocycle moiety being optionally substituted with one or several groups selected from halogen; oxo (=0); N0 2 ; ORn; NR12R13; C(0)R 14 ; S(0) 2 NR 52 R53; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i, and NR22R23; and aryl or aryl-(Ci-C6)alkyl optionally substituted with one or several groups selected from halogen, OR31 , and NR32R33, and

- R 5 represents H,

wherein:

- the aryl is phenyl,

- the heterocycle is a saturated hydrocarbon monocycle or bicycle (comprising fused, bridged or spiro rings, notably fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom; such as piperidine, piperazine, triazinane or pyrrolidine; and in particular piperidine, and

- the heteroaryl is an aromatic hydrocarbon monocycle or bicycle (comprising fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom; such as pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline or quinazoline; in particular pyridine or quinoline. According to another particular embodiment, the compounds according to the present invention are compounds of formula (I- 1 c) or (I- Id), or a pharmaceutically acceptable salt or solvate thereof,

wherein:

- nl and n2 represent, independently of each other, 1, 2, 3 or 4,

- Q represents an aryl or heterocycle, notably a heterocycle, optionally substituted with one or several groups selected from halogen; oxo (=0); and (Ci-Ce)alkyl optionally substituted with one or several groups selected from halogen, OR 2 i and

- the nitrogen atom being linked to (CH 2 ) nl ; notably NRo or , the nitrogen atom being linked to

- Xi represents NH,

- X 2 represents O,

- Ro represents H; CHO; or a (C 1 -C 6 )alkyl optionally substituted with C0 2 H or C0 2 - ((C 1 -C 6 )alkyl),

- R 3 and R4 represent, independently of each other, H, (C 1 -C6)alkyl, aryl, heteroaryl, aryl-(Ci-C 6 )alkyl, heteroaryl-(Ci-C 6 )alkyl, -((Ci-C 6 )alkyl)-NH-aryl or -((d- C 6 )alkyl)-NH-heteroaryl,

each aryl or heteroaryl moiety being optionally substituted with one or several groups selected from halogen; ORn; NRi 2 Ri 3 ; (Ci-C 6 )alkyl optionally substituted with one or several groups selected from halogen, OR 2 i and NR 22 R 23 ; and aryl optionally substituted with one or several groups selected from halogen, OR 3 i and NR 32 R 33 , and

- R 5 represents H,

wherein:

- the aryl is phenyl,

- the heterocycle is a saturated, unsaturated or aromatic hydrocarbon monocycle or bicycle (comprising fused, bridged or spiro rings, notably fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom; such as pyridine, pyrimidine, pyridazine, pyrazine, quinoline, isoquinoline, quinoxaline, quinazoline, piperidine, piperazine, dihydropyridines, dihydropyrimidines, dihydropyridazines, dihydropyrazines, tetrahydropyridines, tetrahydropyrimidines, tetrahydropyridazines or tetrahydropyrazines; in particular quinoline, pyridine or dihydropyrimidines

(notably 1 ,2-dihydropyrimidine), and

- the heteroaryl is an aromatic hydrocarbon monocycle or bicycle (comprising fused rings), each cycle having 5 or 6 members and 1 to 4, notably 1 or 2, carbon atoms having each been replaced with a nitrogen or oxygen atom, notably a nitrogen atom; such as pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline, quinoxaline or quinazoline; in particular quinoline or pyrimidine.

The compounds of the present invention may be selected from compounds A to U described in the experimental part below and the pharmaceutically acceptable salts and solvates thereof.

The compounds of the present invention can also be selected from compounds AA to

AS described in the experimental part below and the pharmaceutically acceptable salts and solvates thereof.

The present invention relates also to a compound of formula (I) such as defined above, for use as a drug, notably intended for the treatment of cancer.

The present invention also relates to the use of a compound of formula (I) such as defined above, for the manufacture of a drug, notably intended for the treatment of cancer.

The present invention also relates to a method for the treatment of cancer comprising the administration to a person in need thereof of an effective dose of a compound of formula (I) such as defined above.

The cancer may be more particularly in this case colon cancer, breast cancer, kidney cancer, liver cancer, pancreatic cancer, prostate cancer, glioblastoma, non-small cell lung cancer, neuroblastoma, inflammatory myo fibroblastic tumor, leukemia (acute myeloid leukemia, myelodysplasia syndrome, chronic myelomonocytic leukemia), melanoma, diffuse B-cell lymphoma or anaplastic large-cell lymphoma.

The present invention relates also to a compound of formula (I) such as defined above, for use as a DNA methylation inhibitor, in particular as a DNMT inhibitor. According to the invention, the expression "DNA methylation inhibitor" and "DNMT inhibitor" refers to molecules that are able to reduce or inhibit the DNA methylation and the DNA methyltransferase activity respectively. Preferentially, the use of a DNMT inhibitor according to the invention makes it possible to suppress the activity of said DNMT.

The present invention also relates to a pharmaceutical composition comprising at least one compound of formula (I) such as defined above, and at least one pharmaceutically acceptable excipient.

The pharmaceutical compositions according to the invention may be formulated notably for oral administration or for injection, wherein said compositions are intended for mammals, including humans.

The pharmaceutical composition can be administered orally by means of tablets and gelatin capsules.

When a solid composition is prepared in the form of tablets, the main active ingredient is mixed with a pharmaceutical vehicle such as gelatin, starch, lactose, magnesium stearate, talc, gum arabic and the like. The tablets may be coated with sucrose or with other suitable materials, or they may be treated in such a way that they have a prolonged or delayed activity and they continuously release a predetermined amount of active principle.

A preparation in gelatin capsules is obtained by mixing the active ingredient with a diluent and pouring the mixture obtained into soft or hard gelatin capsules.

For administration by injection, aqueous suspensions, isotonic saline solutions or sterile and injectable solutions which contain pharmacologically compatible dispersing agents and/or wetting agents are used.

The active ingredient may be administered in unit dosage forms of administration, in mixture with standard pharmaceutical carriers, to animals or to humans. The compounds of the invention as active ingredients may be used in doses ranging between 0.01 mg and 1000 mg per day, given in a single dose once per day or administered in several doses throughout the day, for example twice a day in equal doses. The dose administered per day advantageously is between 5 mg and 500 mg, even more advantageously between 10 mg and 200 mg. It may be necessary to use doses outside these ranges as determined by the person skilled in the art. The pharmaceutical compositions according to the invention may further comprise at least one other active ingredient, such as an anticancer agent.

The present invention relates also to a pharmaceutical composition comprising: (i) at least one compound of formula (I) such as defined above, and

(ii) at least one other active ingredient, such as an anticancer agent,

as a combination product for simultaneous, separate or sequential use.

The present invention also relates to a pharmaceutical composition such as defined above for use as a drug, notably intended for the treatment of cancer. The present invention also relates to methods for the preparation of the compounds of formula (I) according to the invention.

A first method is a method to prepare a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, in which W = NRo with Ro ≠ H, comprising:

(a) reacting a compound of formula (I) in which W = NH with:

- a compound of formula Ro-LG where Ro represents a (Ci-C 6 )alkyl optionally substituted with CHO, C0 2 H or C0 2 -((Ci-C 6 )alkyl) and LG represents a leaving group to give a compound of formula (I) in which W = NRo with Ro representing a (Ci-C 6 )alkyl optionally substituted with CHO, C0 2 H or C0 2 -

((Ci-C 6 )alkyl),

- dimethylformamide (DMF) to give a compound of formula (I) in which W = NRo with Ro = CHO, or

- a compound of formula Ro-Ai where Ro represents C0 2 -((Ci-C 6 )alkyl) and Ai represents a (Ci-C 6 )alkoxy group or a halogen atom (such as CI or Br) to give a compound of formula (I) in which W = NRo with Ro representing C0 2 -((Ci- C 6 )alkyl), and

(b) optionally salifying or solvating the compound obtained in step (a) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) in which W = NRo with Ro as defined above. Step (a):

When Ro represents a (Ci-Ce) lkyl optionally substituted with CHO, CO 2 H or CO 2 - ((Ci-Ce)alkyl):

The term "leaving group", as used in the present invention, refers to a chemical group which can be easily replaced with a nucleophile during a nucleophile substitution reaction, the nucleophile being in the case of step (a) a secondary amine, i.e. a molecule carrying a group NH. Such a leaving group can be in particular a halogen atom or a sulfonate. The sulfonate is in particular a group -OSO2-R7 with R 7 representing a (Ci- C 6 )alkyl, aryl, aryl-(Ci-C6)-alkyl or (Ci-C6)-alkyl-aryl group. The sulfonate can be in particular a mesylate (CH 3 -S(0 2 )0-), a triflate (CF 3 -S(0) 2 0-) or a tosylate (/?-Me-C 6 H 4 - S(0) 2 0-).

The LG group can be in particular a halogen atom such as a bromine.

Step (a) is advantageously carried out in the presence of a base such as triethylamine.

When Ro represents a substituted (Ci-C 6 )alkyl group, the (Ci-C 6 )alkyl group will be advantageously substituted with a C0 2 -((Ci-C6)alkyl) group. This group can then be hydrolysed, notably in the presence of NaOH or KOH, to give a C0 2 H group (Ro represents then a (Ci-C6)alkyl substituted with C0 2 H). A reduction step in conditions well known to the one skilled in the art allows obtaining a CHO group (Ro represents then a (Ci-C6)alkyl substituted with CHO).

When Ro represents CHO:

The reaction is advantageously performed using DMF as solvent, notably in the presence of a base such as triethylamine.

When Ro represents C0 2 -((Ci-Ce)alkyl):

This reaction can be carried out in conditions to prepare carbamates well known to the one skilled in the art.

Step (b :

The salification or solvatation step can be carried out by methods well known to the one skilled in the art, in particular by reaction of the compound of formula (I) obtained in step (a) with a pharmaceutically acceptable acid (organic or inorganic acid), base (organic or inorganic acid) or solvent, as defined previously.

The solvent can be notably the solvent used in the last step of the preparation of the compound according to the invention, in particular the solvent used in step (a). Thus steps (a) and (b) can be carried out in a single step, without isolating intermediate compounds.

A second method is a method to prepare a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, in which W represents NRo, comprising:

(1) reacting a compound of the following formula (II):

in which Q, Xi and nl are as defined above and Wi represents LGi, NHR 8 ,

with a compound of the following formula (III):

in which X 2 , X 3 , X 4 and n2 are as defined above and W 2 represents LG 2 , NHR 8 , wherein LGi and LG 2 represent, independently of each other, a leaving group and Rs represents Ro or a N-protecting group,

on the condition that:

when Wi represents LGi, then W 2 represents NHR 8 and or , then W 2 represents LG 2 ,

and, when Wi or W 2 represents NHR 8 with R 8 representing a N-protecting group, deprotecting the nitrogen atom bearing the N-protecting group,

to give a compound of formula (I) as defined above, and

(2) optionally salifying or solvating the compound obtained in step (1) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) as defined above. Step (1):

The LGi and LG 2 groups can be in particular a halogen atom such as a bromine or chlorine.

The reaction between the compounds of formula (II) and (III) can be carried out in the presence of a base, such as K 2 CO 3 . A catalytic amount of KI can also be added to the reaction medium.

R 8 can represent in particular H or a N-protecting group, notably a N-protecting group. When Wi or W 2 represents NHR 8 with R 8 representing H or a N-protecting group, it is possible to prepare compounds of formula (I) with W = NH. The term "protecting group", as used in the present invention, refers to a chemical group which selectively blocks a reactive site in a multifunctional compound so as to allow selectively performing a chemical reaction on another unprotected reactive site.

The term "N-protecting group", as used in the present invention, refers to those groups intended to protect an amine function (notably a primary amine function) against undesirable reactions (such as a disubstitution of the primary amine function) during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, "Protective Groups In Organic Synthesis," (John Wiley & Sons, New York (1981)). An amine function protected by a N-protecting group can be a carbamate, an amide, a sulfonamide, an N-alkyl derivative, an amino acetal derivative, a N-benzyl derivative, an imine derivative, an enamine derivative or a N-heteroatom derivative. In particular, N- protecting groups include formyl; benzyl (Bn); -CO-R 9 such as acetyl (Ac), pivaloyl (Piv or Pv) or benzoyl (Bz); -CO 2 -R 9 such as tbutyloxycarbonyl (Boc), trichloroethoxycarbonyl (TROC), allyloxycarbonyl (Alloc) or benzyloxycarbonyl (Cbz or Z); -SO 2 -R 9 such as phenylsulfonyl or 2-nitrobenzenesulfonyl (Nos or Ns); and the like, with R 9 representing a (Ci-C 6 )alkyl optionally substituted with one or several halogen atoms such as F or CI; a (C 2 - C 6 )alkenyl such as an allyl; an aryl, such as a phenyl, optionally substituted with N0 2 ; or an aryl-(Ci-C6)alkyl such as a benzyl.

The step of deprotecting the nitrogen atom bearing the N-protecting group can be carried out by methods well known to the one skilled in the art, notably as disclosed in Greene, "Protective Groups In Organic Synthesis," (John Wiley & Sons, New York (1981)).

The N-protecting group will be in particular 2-nitrobenzenesulfonyl (Nos or Ns). It can be deprotected in the presence of thiophenol.

The compounds of formulas (II) and (III) are either commercially available or prepared by methods well known to the one skilled in the art, notably as illustrated in the examples below.

In particular, the compound of formula (II) can be prepared by reacting a compound of formula Q-Hal with a compound of formula HXi-(CH 2 ) n i-W 3 where:

- Q, Xi and nl are as defined above,

- Hal represents a halogen atom such as CI or Br, and

- W 3 represents a group Wi, optionally in a protected form (W 3 can represent notably OH).

This reaction can be performed optionally in the presence of a base.

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the W 3 group can be carried out to introduce the Wi function on the molecule.

When the compound of formula (III) is a compound of the following formula (IIIc):

with W 2 , X 2 , R4, R5 and n2 as defined above,

this compound can be prepared by reacting a compound of the following formula (IV):

with X 2 and n2 as defined above and W 4 representing a group W 2 , optionally in a protected form,

with an amine of formula R 4 R5NH with R 4 and R 5 as defined above.

This reaction can be performed in the presence of a base such as K 2 C0 3 or triethylamine.

The carbonyl function of the compound of formula (IV) can be activated in the form of a triazole, notably by reaction with POCl 3 and triazole (more particularly 1,2,3-triazole) preferably in the presence of a base such as triethylamine.

Thus the compound of formula (IIIc) can be prepared by:

- activating the compound of formula (IV) in the form of a triazole of the following formula (V):

with W 4 , X 2 and n2 as defined above, and

reacting the triazole of formula (V) with the amine of formula R 4 RsNH. Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the W 4 group can be carried out to introduce the W 2 function on the molecule. When the compound of formula (III) is a compound of the following formula (Hid):

with W 2 , X 2 , R 3 and n2 as defined above, and R 3 ≠ H,

this compound can be prepared by reacting a compound of formula (IV) as defined above with a compound of formula R 3 -LG 3 with R 3 as defined above and LG 3 representing a leaving group, such as a halogen atom (e.g. CI or Br).

This reaction can be carried out in the presence of a base, such as K 2 C0 3 . A catalytic amount of KI can also be added to the reaction medium.

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the W 4 group can be carried out to introduce the W 2 function on the molecule.

The compound of formula (IV) can be prepared by reacting a compound following formula (VI):

where Hal represents a halogen atom such as F,

with a compound of formula W 4 -(CH 2 ) n2 -X 2 H where W 4 , X 2 and n2 are as defined above.

This reaction can be carried out in the presence of a base such as NaH. Step (2):

The salification or solvatation step can be carried out by methods well known to the one skilled in the art, in particular by reaction of the compound of formula (I) obtained in step (1) with a pharmaceutically acceptable acid (organic or inorganic acid), base (organic or inorganic acid) or solvent, as defined previously.

The solvent can be notably the solvent used in the last step of the preparation of the compound according to the invention, in particular the solvent used in step (1).

Thus steps (1) and (2) can be carried out in a single step, without isolating intermediate compounds.

A third method is a method to prepare a compound of formula (I) or a pharmaceutically acceptable salt or solvate thereof, comprising:

(A) reacting a compound of the following formula (VII):

Q *6 (VII)

in which Q is as defined above and X 6 represents a halogen atom (e.g. CI or Br) or - Xi-(CH 2 )„i-W-(CH 2 )„ 2 -X 2 H with W, X X 2 , nl and n2 as defined above,

with a compound of the following formula (VIII):

(VIII)

in which X3 and X 4 are as defined above and X 7 represents a halogen atom (e.g. F) or -X 2 -(CH 2 ) n2 -W-(CH 2 ) n i-XiH with W, X X 2 , nl and n2 as defined above, on the condition that:

when X 6 represents a halogen atom, then X 7 represents -X 2 -(CH 2 ) n2 -W-

when X 6 represents -Xi-(CH 2 ) n i-W-(CH 2 ) n2 -X 2 H, then X 7 represents a halogen atom,

to give a compound of formula (I), and

(B) optionally salifying or solvating the compound obtained in step (A) to give a pharmaceutically acceptable salt or solvate of a compound of formula (I) as defined above. Step (A):

The reaction between the compounds of formula (VII) and (VIII) can be carried out in the presence of a base, such as K 2 CO 3 . A catalytic amount of KI can also be added to the reaction medium.

The compounds of formulas (VII) and (VIII) are either commercially available or prepared by methods well known to the one skilled in the art, notably as illustrated in the examples below.

In particular, the compound of formula (VII), when X 6 represents -Xi-(CH 2 ) n i-W- (CH 2 ) n2 -X 2 H, can be prepared by reacting a compound of formula Q-Hal with a compound of formula HXi-(CH 2 )„i-W-(CH 2 )„2-X8 where:

- Q, Xi, W, nl and n2 are as defined above,

- Hal represents a halogen atom such as CI or Br, and

- Xs represents a group X 2 H, optionally in a protected form.

This reaction can be performed optionally in the presence of a base.

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the Xs group can be carried out to introduce the X 2 H function on the molecule.

When the compound of formula (VIII) is a compound of the following formula

(VIIIc):

(VIIIc)

with X 7 , R4 and R 5 as defined above,

this compound can be prepared by reacting a compound of the following formula (IX):

(IX) with X 9 representing a group X 7 , optionally in a protected form,

with an amine of formula R 4 R 5 NH with R 4 and R 5 as defined above.

This reaction can be performed in the presence of a base such as K 2 CO 3 or triethylamine.

The carbonyl function of the compound of formula (IX) can be activated in the form of a triazole, notably by reaction with POCI 3 and triazole (more particularly 1,2,3-triazole) preferably in the presence of a base such as triethylamine.

Thus the compound of formula (VIIIc) can be prepared by:

- activating the compound of formula (IX) in the form of a triazole of the following formula (X):

with X 9 as defined above, and

- reacting the triazole of formula (X) with the amine of formula R 4 R 5 NH

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the X9 group can be carried out to introduce the X 7 group on the molecule.

When the compound of formula (VIII) is a compound of the following formula

(Vllld):

(Vllld)

with X 7 and R 3 as defined above, and R 3 ≠ H, this compound can be prepared by reacting a compound of formula (IX) as defined above with a compound of formula R3-LG3 with R3 as defined above and LG3 representing a leaving group, such as a halogen atom (e.g. CI or Br).

This reaction can be carried out in the presence of a base, such as K 2 CO 3 . A catalytic amount of KI can also be added to the reaction medium.

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out, in particular a deprotection step of the X9 group can be carried out to introduce the X 7 group on the molecule. The compound of formula (IX), when X 9 represents -X2-(CH 2 )n2-W-(CH2) n i-Xio where X 10 represents XiH optionally in a protected form, can be prepared by reacting a compound of the formula (VI) with a compound of formula HX2-(CH2)n2-W-(CH 2 )ni-Xio where W, X 2 , X 10 , nl and n2 are as defined above.

This reaction can be carried out in the presence of a base such as NaH.

Step (B):

The salification or solvatation step can be carried out by methods well known to the one skilled in the art, in particular by reaction of the compound of formula (I) obtained in step (A) with a pharmaceutically acceptable acid (organic or inorganic acid), base (organic or inorganic acid) or solvent, as defined previously.

The solvent can be notably the solvent used in the last step of the preparation of the compound according to the invention, in particular the solvent used in step (A).

Thus steps (A) and (B) can be carried out in a single step, without isolating intermediate compounds.

Further steps of protection(s), deprotection(s) and/or functionalization(s) well known to the one skilled in the art can be carried out to obtained the compounds of formula (I).

The compound according to the present invention obtained by one of the methods described above can be separated from the reaction medium by methods well known to the one skilled in the art, such as by extraction, evaporation of the solvent or by precipitation or crystallisation (followed by filtration). This compound can also be purified if necessary by methods well known to the one skilled in the art, such as by recrystallisation, by distillation, by chromatography on a column of silica gel or by high performance liquid chromatography (HPLC). The examples which follow illustrate the invention without limiting its scope in any way.

EXAMPLES

The following abbreviations have been used in the following examples.

a.a. Amino acid

AdoMet S-Adenosyl-L-methionine

BSA N, O-Bis(trimethylsilyl)acetamide

DCM Dichloromethane

DiPEA N, N-Diisopropy lethy lamine

DMAP 4-Dimethylaminopyridine

DMF Dimethylformamide

DMSO Dimethylsulfo xide

EDTA Ethylenediaminetetraacetic acid

ESI Electrospray ionisation

HEPES 4-(2-Hydroxyethyl)- 1 -piperazineethanesulfonic acid

HPLC High Performance Liquid Chromatography

HRMS High Resolution Mass Spectrometry

NMR Nuclear Magnetic Resonance

Nos 2-Nitrobenzenesulfonyl

PBS Phosphate buffered saline

PBST Phosphate buffered saline + Tween-20

RT Room temperature

SAH S-Adenosyl-L-homocysteine

SAM S-Adenosyl-L-methionine

TEA Triethylamine

TFA Trifluoroacetic acid

TLC Thin Layer Chromatography

Tris Tris(hydroxymethyl)aminomethane I. Synthesis of the compounds according to the invention

Example 1 : Compound F

16 17

a) i) SOC-2,110°C, lh. ii) Phenylpropylamine, DMF, K 2 C0 3 , RT, 2h, 85%.

4-(3-phenylpropylamino)-7-(2-chloroethoxy)quinazoline (17)

A solution of 16 (440mg; 2.01mmol) in thionylchloride (lOmL) and a catalytic amount of DMF was boiled for 30min. The solvent was removed and the crude product was dissolved in a solution of phenylpropylamine (570μί; 4.0mmol) in DMF and the mixture was stirred at room temperature for 2h. The mixture was diluted with ethylacetate and the organic phase was washed with a saturated solution of Na 2 C0 3 , brine and dried over magnesium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ethylacetate (0— 100% ethylacetate) in cyclohexane to obtain 17 as a pale brown solid (607mg; 1.70mmol; yield 85%).

1H NMR (500MHz, CDC1 3 ) δ 8.58 (s, 2H, Hal), 7.35-7. 13 (m, 7H, Ha7, Hal5, Hal3 and Hal4), 6.99 (dd, J=2.4, 9.0Hz, 1H, Ha5), 5.53 (brs, 1H, HNH), 4.22 (t, J=6.0Hz, 2H, Hbl), 3.76 (t, J=6.0Hz, 2H, Hb3), 3.70 (q, J=7.2Hz, 2H, Ha9), 2.79 (t, J=7.2Hz, 2H, Hal l), 2.28 (quint, J=6.1Hz, 2H, HalO), 2.07 (quint, J=7.0Hz, 2H, Hb2).

13 C NMR (125MHz, CDC1 3 ) δ 162,0 (Ca6), 159.1 (Ca2), 156.1 (Cal), 151.7 (Ca8), 141.7 (Cal2), 128.8 (Cal3), 128.6 (Cal4), 126.3 (Cal5), 122.0 (Ca4), 117.8 (Ca5), 109.3 (Ca3), 108.1 (Ca7), 64.7 (Cbl), 41.4 (Cb3), 41.3 (Ca9), 33.8 (Cal l), 32.1(Cb2), 30.8 (CalO). HRMS-ESI (m/z) calculated for C 20 H 23 N 3 CIO [M+H] : 356.1524; Found:

a) phthalimide potassium salt, DMF, 90°C, 3h, 98%. b) i) CH 3 NHNH 2 , ethanol, RT, 82%. ii) NosCl, TEA, DMF, RT, 6h, 96%.

7-((3-phthalimido)propyloxy)-4-((3-phenylpropyl)amino)qui nazoline (22)

To a solution of 17 (50mg; 141μιηο1) in DMF (lmL) was added phthalimide potassiums salt and the mixture was heated at 90°C for 6h. The mixture was diluted with ethylacetate and the organic phase was washed with a saturated solution of Na 2 C0 3 , brine and dried over magnesium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ethylacetate (0— »100% ethylacetate) in cyclohexane to obtain 22 as a pale yellow solid (63mg; 138μιηο1; yield 98%).

1H NMR (500MHz; CDC1 3 ) δ 8.58 (s, 1H, Hal), 7.86 (m, 2H, Hphtha), 7.74 (m, 4H, Hphtha), 7.35-7.25 (m, 6H, Ha4 and Hal3 and Hal4 and Hal5), 7.10 (d, J=2.5Hz, 1H, Ha7), 6.92 (dd, J=2.5, 9.0Hz, 1H, Ha5), 5.47 (brt, J=5.1Hz, 1H, HNH), 4.15 (t, J=6.3Hz, 2H, Hbl), 3.96 (t, J=7.0Hz, 2H, Hb3), 3.72 (q, J=6.7Hz, 2H, Ha9), 2.82 (q, J=7.3Hz, 2H, Hal 1), 2.26 (quint, J=6.3Hz, 2H, Hb2), 2.10 (quint, H=7.3Hz, 2H, HalO).

13 C NMR (125MHz, CDC1 3 ) δ 168.3 (Cphtha), 162.0 (Ca6), 159.0 (Ca2), 157.8 (Cal), 151.3 (Ca8), 141.5 (Cal2), 134.0 (Cphtha), 132.1 (Cphtha), 128.7 (Cal3 ), 128.4 (Cal4), 126.1 (Cal5), 123.3 (Cphtha), 121.8 (Ca4), 117.9 (Ca5), 109.0 (Ca3), 107.6 (Ca7), 65.7 (Cbl), 41.1 (Ca9), 35.3 (Cb3), 33.7 (Cal l), 30.7 (CalO), 28.0 (Cb2). HRMS-ESI (m/z) calculated for C28H27N4O3 [M+H] : 467.2078; Found: 467.2078.

7-((2-nitrobenzenesulfonamido)propyloxy)-4-((3-phenylprop yl)amino) quinazoline (23)

To a solution of 22 (60mg; 129μηιο1) in ethanol (2mL), was added N- methylhydrazine (200μί). After stirring at room temperature for 12h, the solvent was removed and the residue was co-evaporated with toluene until the N-methylhydrazine was completely eliminated. To the crude product was added a solution of 2-nitrobenzene sulfonyl chloride (71mg; 322mmol) and TEA (54μΙ,; 387μιηο1). The mixture was stirred at room temperature for 3h, then was diluted with ethylacetate. The organic phase was washed with saturated Na 2 C03, with brine and dried over magnesium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of methanol (0— »10% MeOH) in dichloromethane to obtain 23 as a pale yellow solid (61mg; 117μιηο1; yield 91%).

1H NMR (500MHz; CDC1 3 ) δ 8.59 (s, 1H, Hal), 8.15 (m, 1H, HNos), 7.83 (m, 1H, HNos), 7.69 (m, 2H, HNos), 7.37-7.30 (m, 3H, Ha4 and Hal3), 7.22-7.15 (m, 3H, Hal4 and Hal5), 7.10-7.05 (m, 2H, Ha7 and Ha5), 5.84 (brt, J=5.3Hz, 1H, HNH), 5.51 (brt, J=5.4Hz, 1H, HNH), 4.13 (t, J=3.4Hz, 2H, Hbl), 3.73 (q, J=6.3Hz, 2H, Ha9), 3.42 (q, J=6.5Hz, Hb3), 2.82 (q, J=6.8Hz, 2H, Hal l), 2.17-2.07 (m, 4H, HalO and Hb2).

13 C NMR (125MHz, CDC1 3 ) δ 161.5 (Ca6), 159.0 (Ca2), 155.9 (Cal), 151.3 (Ca8), 148.0 (CNos), 141.5 (Cal2), 133.6 (CNos), 133.5 (CNos ), 132.8 (CNos), 130.9 (CNos), 128.7 (Cal3 ), 128.4 (Cal4), 126.2 (Cal5), 125.4 (CNos), 122.0 (Ca4), 117.9 (Ca5), 109.2 (Ca3), 107.6 (Ca7), 66.1 (Cbl), 41.8 (Cb3), 41.2 (Ca9), 33.7 (Cal l), 30.7 (CalO), 28.9 (Cb2).

HRMS-ESI (m/z) calculated for C29H37N8O4 [M+H] + : 522.1806; Found: 522.1801.

25

Compound F

a) Ethanolamine, 125°C, 4h, quantitative yield, b) SOCl 2 , DMF cat., Flash boiling, quantitative yield, c) i) 23, K 2 C0 3 , KI, DMF, 90°C, 12h, 74%. ii) PhSH, K 2 C0 3 , DMF, RT, 24h, 91%.

4-((2-Hydroxyethyl)amino)quinoline (25)

A mixture of 4-chloroquinoline (360mg; 2.21mmol) in ethanolamine (1.5mL;

22mmol) was stirred at 1 10°C for 3h. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of methanol (0— »10% MeOH) in dichloromethane to afford 25 as a white powder (414mg; 2.20mmol; quantitative yield).

1H NMR (500MHz, CDC1 3 ) δ 8.38 (d, J=5.4Hz, IH, Hc5), 8.19 (dd, J=0.9, 8.3Hz, IH, Hc8), 7.77 (dd, J=0.9, 8.3Hz, IH, Hel l), 7.59 (ddd, J=1.3, 6.7, 8.3Hz, IH, HclO), 7.40 (ddd, J=1.3, 6.7, 8.3Hz, IH, Hc9), 7.07 (brt, J=5.2Hz, IH, HOH), 6.46 (d, J=5.4Hz, IH, Hc4), 4.83 (brt, J=5.5Hz, IH, HNHc), 3.66 (q, J=6.0Hz, 2H, Hcl), 3.35 (q, J=5.4Hz, 2H, Hc2).

13 C NMR (125MHz, CDC1 3 ) δ 151.1 (Cc5), 150.5 (Cc3), 148.8 (Cc6), 129.5 (Cc8), 129.1 (CclO), 124.2 (Cc9), 122.1 (Cel l), 1 19.3 (Cc7), 98.6 (Cc4), 59.3 (Ccl), 45.5 (Cc2).

HRMS-ESI (m/z) calculated for CnHi 3 N 2 0 [M+H] + : 189.1022; found: 189.1031. 4-((2-chloroethyl)amino)quinoline chlorhydrate (26)

25 (360mg; 1.92mmol) was solubilized in thionyl chloride (3ml). The mixture was flash boiled and the solvent was removed. Toluene was added to remove the residual thionyl chloride by co-evaporation. The residue was triturated in dichloromethane and the solid was filtrated to afford 26 as a white solid (360mg; 1.75mmol; 91%).

1H NMR (500MHz; CDC1 3 ) δ 8.59 (d, J=5.2Hz, 1H, Hc5), 8.00 (dd, J=0.7, 8.3Hz, 1H, Hc8), 7.79 (d, J=8.3Hz, 1H, Hel l), 7.65 (ddd, J=1.3, 7.9, 8.3Hz, 1H, HclO), 7.45 (ddd, J=1.3, 7.0, 8.3Hz, 1H, Hc9), 6.43 (d, J=5.3Hz, 1H, Hc4), 5.51 (brs, 1H, HNHc), 3.84 (t, J=5.8Hz, 2H, Hcl), 3.70 (q, J=5.8Hz, 2H, Hcl).

13 C NMR (125MHz, CDC1 3 ) δ 151.0 (Cc5), 148.9 (Cc3), 148.5 (Cc6), 130.0 (Cc8), 129.2 (CclO), 125.0 (Cc9), 1 19.3 (Cel l), 1 18.9 (Cc7), 99.0 (Cc4), 44.4 (Cc2), 42.6 (Ccl).

HRMS-ESI (m/z) calculated for CnHi 3 N 2 Cl [M+H] + : 207.0684; found: 207.0678. 7-(3-((2-(quinolin-4-ylamino)ethyl)amino)propyloxy)-4-((3-ph enylpropyl) amino)quinazoline (Compound F)

To a solution of 23 (50mg; 96μιηο1), K 2 C0 3 (22mg; 0.160mmol) and a catalytic amount of KI in DMF (lmL) was added 26 (40mg; 288μmol). The mixture was stirred at 65°C overnight then thiophenol (24μΙ ^ ; 240mmol) was added. The mixture was stirred for a day then diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.2% of TEA (0→80% CH 3 CN) to afford Compound F as a white powder (32mg; 0.64mmol, 67%).

1H NMR (DMSO+MeOD) δ 8.39 (m, 2H, Hal and Hc5), 8.19-8.07 (m, 3H, Hc8, HNH and Ha4), 7.76 (dd, J=0.9, 8.4Hz, 1H, Hel l), 7.58 (ddd, J=1.3, 6.7, 8.2Hz, 1H, HclO), 7.39 (ddd, J=1.3, 6.7, 8.2Hz, 1H, Hc9), 7.43-7.20 (m, 4H, Hal3 and Hal4), 7.17 (brt, J= 7.1Hz, Hal5), 7.12-7.04 (m, 3H, Ha7, HNH and Ha5), 6.47 (d, J=5.7Hz, 1H, Hc4), 4.17 (t, J=6.2Hz, 2H, Hbl), 3.52 (q, J=6.9Hz, 2H, Ha9), 3.35 (m, 2H, Hc2), 2.84 (t, J= 6.6Hz, 2H, Hcl), 2.73 (t, J=6.5Hz, 2H, Hb3), 2.67 (t, J=7.4Hz, 2H, Hal l), 2.00-1.83 (m, 4H, Hal0 and Hb2).

13 C NMR (DMSO+MeOD) δ 162.1 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8), 151.1 (Cc5), 150.4 (Cc3), 148.7 (Cc6), 142.2 (Cal2), 129.5 (Cel l), 129.1 (CclO), 128.8 (Cal3), 128.7 (Cal4), 126.2 (Cal5), 124.7 (Ca4), 124.2 (Cc9), 122.0 (Cc8), 119.3 (Cc7), 117.1 (Ca5), 109.5 (Ca3), 107.8 (Ca7), 98.6 (Cc4), 66.9 (Cbl), 48 (Ccl), 46 (Cb3), 43(Cc2), 40.6(Ca9), 33.1 (Cal l), 30.8 (CalO), 29.8 (Cb2).

HRMS-ESI (m/z) calculated for C 3 iH 35 N 6 0 [M+H] + : 507.2667; found: 507.2666.

Example 2: Compounds A, B, C, D, E, T and U

a) 3-Bocaminopropylamine or 4-Bocaminobutylamine, DMF, 125°C, 4h, 78% for n=l and 73% for n=2. b) TFA, 91% for n=l and 84% for n=2. c) NosCl, TEA, DMAP cat., DMF, RT, 6h, 66% for n=l and 71% for n=2. d) i) 17, K 2 C0 3 , KI cat., DMF, RT, 80°C, 6h, 71% for n=l and 75% for n=2. ii) PhSH, K 2 C0 3 , MeCN, RT, 24h, 85% for n=l and 75% for n=2. e) TFA, water, RT, lh. or e) i) 4-Bromobutanoic acid methyl ester, TEA, DMF, 90°C, 12h, 44% for n=l and 45% for n=2. f) 0.5N NaOH, dioxane, 18h, 78% for n=l and 79% for n=2.

4-((2-Bocaminopropyl)amino)quinoline (27)

4-((2-Bocaminobutyl)amino)quinoline (28)

A solution of N-bocaminopropane amine (2.06g, 1 1.84 mmol) or N-bocamino butane amine (2.25g, 12 mmol), 4-chloroquinoline (1.525g, 9.32 mmol) and DiPEA (1.8 mL, 10.3 mmol) in 12 mL of n-pentanol was stirred at reflux for 6 hours. The solvent was removed and the residue was diluted in DCM. The organic layer was washed with NaOH 2M (2x) and the aqueous phase was extracted with DCM (3x). The organics layers are collected and combined, dried over Na 2 S0 4 and the solvent was removed. The residue was purified by recrystallization in toluene. 27 was obtained as a brown solid (2.02g, 6.69 mmol; 72%) and 28 was obtained as a pale brown solid (2.35g; 7.44 mmol; 62%>).

1H NMR (500MHz; DMSO) δ 8.37 (d, J=5.5Hz, 1H, Hc6), 8.19 (d, J=8.6Hz, 1H,

Hc8), 7.77 (d, J=8.39Hz, 1H, Hel l), 7.60 (dd, J=1.2, 6.9Hz, 1H, HclO), 7.56 (dd, J=1.2, 6.9Hz, 1H, Hc9), 7.17 (t, J=4.9Hz, 1H, HNHBoc), 6.98 (t, J=5.4Hz, 1H, HNH), 6.43 (d, J=5.4Hz, 1H, Hc5), 3.31 (q, J=6.3Hz , 2H, Hc3), 3.09 (q, J=6.3Hz, 2H, Hcl), 1.81 (quint, J=7.1Hz , 2H, Hc2), 1.38 (s, 9H, HBoc).

1 3 C NMR (125MHz, CDC1 3 ) δ 156.1 (CBoc), 151.1 (Cc6), 150.2 (Cc4), 148.7

(Ccl2), 129.5 (CclO), 129.1 (Cel l), 124.2 (CC9), 122.0 (Cc8), 119.2 (Cc7), 98.5 (Cc5), 78.0 (CcBoc), 40.4 (Cc3), 38.2 (Ccl), 28.7 (CcBoc), 28.6 (Cc2).

HRMS-ESI (m/z) calculated for C17H23N3O2 [M+H]+: 302.1863; Found: 302.1865.

1H NMR (500MHz, DMSO) δ 8.37 (d, J=5.4Hz, 1H, Hcl3), 8.22 (d, J=8.3, 1H,

Hc8), 7.77 (d, J=8.3Hz, 1H, Hel l), 7.59 (m, 1H, HclO), 7.40 (ddd, J=l .l, 6.9, 8.9Hz, 1H, Hc9), 7.15 (brt, J=5.2Hz, 1H, HNH), 6.86 (brt, J=5.6Hz, 1H, HNH), 6.43 (d, J=5.6Hz, 1H, Hc6), 3.27 (q, J=5.6Hz, 2H, Hc4), 2.98 (q, J=6.3Hz, 2H, Hcl), 1.64 (quint, J=7.0Hz, 2H, Hc3), 1.51 (quint, J=7.0Hz, 2H, Hc2), 1.37 (s, 9H, HBoc).

1 3 C NMR (125MHz, CDC1 3 ) δ 156.1 (CBoc), 151.1 (Cc6), 150.2 (Cc4), 148.7

(Ccl2), 129.5 (CclO), 129.1 (Cel l), 124.2 (CC9), 122.0 (Cc8), 119.2 (Cc7), 98.5 (Cc5), 78.0 (CcBoc), 40.4 (Cc3), 38.2 (Ccl), 28.7 (CcBoc), 28.6 (Cc2).

HRMS-ESI (m/z) calculated for C18H26N3O2 [M+H]+: 316.2020; Found: 316.2018. 4-((2-aminoethyl)amino)quinoline (29)

4-((2-aminobutyl)amino)quinoline (30) A solution of 27 (lg; 3.32 mmol) or 28 (l .lg; 3.48 mmol) in 10 mL of trifluoroacetic acid was stirred 1 hour at room temperature. The reaction mixture was diluted in toluene and the solvents were removed. The residue was diluted in water, basified to pH = 14 with NaOH 30% and extracted with DCM. The organic layer was dried and the solvent was removed.

29 was obtained as a yellow oil (638 mg; 3.17 mmol; 96%>) and 30 was obtained as a yellow oil 710 mg; 3.30 mmol; 95%).

1H NMR (500MHz; DMSO) δ 8.38 (d, J=4.9Hz, 1H, Hc6), 8.17 (d, J=8.7Hz, 1H, Hc8), 7.76 (d, J=8.3Hz, 1H, Hel l), 7.60 (m, 1H, HclO), 7.40 (m, 1H, Hc9), 7.25 (brs, 1H, HNH), 6.71 (brs, 2H, HNH2), 6.44 (d, J=5.9Hz, 1H, Hc5), 3.32 (m, 2H, Hc3), 2.71 (m, 2H, Hcl), 1.76 (m , 2H, Hc2).

13 C NMR (125MHz, DMSO) δ 151.7 (Cc6), 150.9 (Cc4), 149.2 (Ccl2), 130.0 (CclO), 129.6 (Cel l), 124.7 (CC9), 122.6 (Cc8), 119.8 (Cc7), 99.0 (Cc5), 41.6 (Cc3), 39.0 (Ccl), 29.4 (Cc2).

HRMS-ESI m/z) calculated for Ci 2 Hi 6 N 3 [M+H] + : 202.1339; Found: 202.1343

1H NMR (500MHz; DMSO) δ 8.37 (d, J=5.2Hz, 1H, Hcl3), 8.22 (d, J=8.2Hz, 1H, Hc8), 7.76 (d, J=8.4Hz, 1H, Hel l), 7.59 (m, 1H, HclO), 7.40 (m, 1H, Hc9), 7.25 (brt, J=5.2Hz, 1H, HNH), 6.44 (d, J=5.4Hz, 1H, Hc6), 3.34 (m, 2H, Hc4), 3.18 (m, 2H, Hcl), 1.72 (m , 2H, Hc3), 1.64 (m, 2H, Hc2).

13 C NMR (125MHz, DMSO) δ 151.2 (Ccl3), 150.4 (Cc5), 148.8 (Ccl2), 129.4 (CclO), 129.1 (Cel l), 124.1 (CC9), 122.1 (Cc8), 119.3 (Cc7), 98.6 (Cc6), 50.7 (Cc4), 42.7 (Ccl), 28.5 (Cc3), 26.4 (Cc2).

HRMS-ESI (m/z) calculated for Ci 3 Hi 8 N 3 [M+H] + : 216.1495; Found: 216.1493. 4-((2-(2-nitrobenzenesulfonamido)propyl)amino)quinoline (31) 4-((2-(2-nitrobenzenesulfonamido)butyl)amino)quinoline (32)

To a solution of 29 (170 mg; 0.85 mmol) or 30 (182 mg; 0.85 mmol), triethylamine (378 μί; 2.72 mmol) and DMAP in catalytic amount in 4 mL of DMF was added 2-nosyl chloride (207 mg; 0.94 mmol). The reaction was stirred at room temperature overnight. The solvent was removed.

The residue containing crude product of 31 was suspended in DCM and one piece of methanol was added until the suspension was diluted. After 30 min at room temperature, 31 crystallized and it was filtrated and washed with DCM to give a yellow powder (225 mg; 0.58 mmol; 68%).

The residue containing crude product of 32 was purified by silica gel flash chromatography using a linear gradient of dicholomethane / methanol (0 to 10%) to give 32 (108 m 0.27 mmol; 32%) as a yellow oil.

1H NMR (500MHz; CDC1 3 ) δ 8.49 (d, J=5.4Hz, 1H, Hc6), 8.08 (dd, J=1.7, 7.7Hz,

1H, HNos), 7.97 (dd, J=1.0, 8.7Hz, 1H, Hc8), 7.83-7.78 (m, 2H, Hel l and HNos), 7.69- 7.59 (m, 3H, HclOand HNos), 7.45 (ddd, J=1.4, 7.0Hz, 1H, Hc9), 6.37 (d, J=5.4Hz, 1H, Hc5), 5.64 (brs, 1H, HNH), 3.29 (brt, J=6.1Hz, 2H, Hc3), 3.09 (q, J=6.1Hz, 2H, Hcl), 1.99 (quint, J=6.2Hz , 2H, Hc2).

1 3 C NMR (125MHz, CDC1 3 ) δ 150.6 (Cc6), 149.5 (Cc4), 148.1 (Ccl2), 148.0

(CNos), 133.7 (CNos), 133.3 (CNos), 132.8 (CNos), 130.9 (CNos), 129.5 (CclO), 129.3 (Cel l), 125.4 (CNos), 124.9 (CC9), 119.5 (Cc8), 118.8 (Cc7), 98.5 (Cc5), 78.0 (CcBoc), 40.9 (Cc3), 39.4 (Ccl), 28.1 (Cc2).

HRMS-ESI (m/z) calculated for C18H19N4O4S [M+H] + : 387.1122; Found: 387.1130.

1H NMR (500MHz; CDC1 3 ) δ 8.52 (d, J=5.4Hz, 1H, Hcl3), 8.08 (m, 1H, HNos), 7.99 (dd, J=1.0, 8.7Hz, 1H, Hc8), 7.80-7.75 (m, 2H, Hel l and HNos), 7.69-7.61 (m, 3H, HclOand HNos), 7.45 (ddd, J=1.4, 7.1Hz, 1H, Hc9), 6.37 (d, J=5.5Hz, 1H, Hc6), 5.26 (brs, 1H, HNH), 3.34 (m, 2H, Hc4), 3.21 (t, J=6.7Hz 2H, Hcl), 1.83 (m, 2H, Hc2), 1.73 (m, 2H,Hc3).

13 C NMR (125MHz, CDC1 3 ) δ 150.6 (Ccl3), 149.7 (Cc5), 148.0 (CNos), 147.9 (Ccl2), 133.6 (CNos), 133.5 (CNos), 132.8 (CNos), 130.9 (CNos), 129.5 (CclO), 129.3 (Cel l), 125.3 (CNos), 124.8 (CC9), 119.5 (Cc8), 118.6 (Cc7), 98.6 (Cc6), 43.4 (Cc4), 42.6 (Ccl), 27.3 (Cc3), 25.7 (Cc2).

HRMS-ESI (m/z) calculated for C19H21N4O4S [M+H] + : 401.1284; Found: 401.1280.

7-(3-((2-(quinolin-4-ylamino)propyl)amino)propyloxy)-4-(( 3-phenylpropyl) amino)quinazoline (Compound A)

7-(3-((2-(quinolin-4-ylamino)butyl)amino)propyloxy)-4-((3-ph enylpropyl) amino)quinazoline (Compound B)

To a solution of 17 (30mg; 85μηιο1) in DMF (lmL) was added K 2 C0 3 (35mg; 253μηιο1), a catalytic amount of KI and 31 or 32 (128μιηο1). The mixture was heated at 90°C for 6h then thiophenol (32μΙ ^ ; 320mmol) was added. The mixture was stirred at room temperature overnight then diluted with ethyl acetate. The organic phase was washed with a solution of saturated Na 2 C0 3 and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.2% of TEA (0— »80% CH 3 CN) to afford Compound A as a white powder (26mg; 5 Ιμιηοΐ; 60%) or Compound B as a white powder (25mg; 0.48mmol; 56%>).

1H NMR (CDC1 3 ) δ 8.58 (s, 1H, Hal), 8.48 (d, J=5.5Hz, 1H, Hc6), 7.96 (d, J=8.4, 1H, Hc8), 7.85 (d, J=8.4Hz, 1H, Ha4), 7.64 (brs, 1H, HNH), 7.58 (ddd, J=1.3, 6.8, 8.2Hz, 1H, HclO), 7.43-7.20 (m, 8H, Hc9, Hal5, Ha5, Hel l, Hal3 and Hal4), 7.14 (d, J=2.6Hz, 1H, Ha7), 6.34 (d, J=5.7Hz, 1H, Hc5), 5.72 (brt, J=5.2Hz, 1H, HNH), 4.19 (t, J=6.0Hz, 2H, Hbl), 3.71 (q, J=7.0Hz, 2H, Ha9), 3.45 (t, J=6.0Hz, 2H, Hc3), 3.00-2.92 (m, 4H, Hcl and Hb3), 2.80 (t, J=7.4Hz, 2H, Hal 1), 2.21-2.00 (m, 4H, HalO and Hb2), 1.98 (quint, J=6.0Hz, 2H, Hc2).

13 C NMR (CDC1 3 ) δ 161.8 (Ca6), 159.0 (Ca2), 156.0 (Cal), 151.4 (Ca8), 151.1 (Cc4), 149.8 (Cc6), 146.9 (Ccl2), 141.5 (Cal2), 135.0 (Cel l), 129.4 (CclO), 128.6 (Cal3), 128.4 (Cal4), 126.1 (Cal5), 124.6 (Ca4), 122.1 (Cc9), 120.4 (Cc8), 118.8 (Cc7), 117.5 (Ca5), 109.2 (Ca3), 107.8 (Ca7), 97.9 (Cc5), 66.6 (Cbl), 49.2 (Ccl), 47.3 (Cb3), 43.6 (Cc3), 41.1 (Ca9), 33.6 (Cal l), 30.6 (CalO), 29.3 (Cb2) 27.4 (Cc2).

HRMS-ESI (m/z) calculated for C 32 H 37 N 6 O [M+H]+: 521.3023; Found: 521.3022.

1H NMR (CDC1 3 ) δ 8.54 (s, 1H, Hal), 8.46 (d, J=5.7Hz, 1H, Hcl3), 7.94 (d, J=8.3, 1H, Hc8), 7.81 (d, J=8.3Hz, 1H, Ha4), 7.58 (ddd, J=l .l, 6.9, 8.3Hz, 1H, HclO), 7.37 (ddd, J=l .l, 6.9, 8.3Hz, 1H, Hc9), 7.33-7.17 (m, 6H, Hal5, Hel l, Hal3 and Hal4), 7.10 (d, J=2.5Hz, 1H, Ha7), 6.90 (dd, J=2.3, 9.1Hz, 1H, Ha5), 6.35 (d, J=5.6Hz, 1H, Hc6), 6.00 (brs, 1H, HNH), 5.67 (brt, J=4.9Hz, 2H, HNH), 4.14 (t, J=6.0Hz, 2H, Hbl), 3.67 (q, J=6.5Hz, 2H, Ha9), 3.31 (t, J=6.3Hz, 2H, Hc4), 2.87 (t, J=6.4Hz, Hal l), 2.80-2.72 (m, 4H, Hcl and Hb3), 2.11-2.01 (m, 4H, HalO and Hb2), 1.85 (quint, J=7.2Hz, 2H, Hc2 or Hc3), 1.71 (quint, J=7.2Hz, 2H, Hc2 or Hc3).

13 C NMR (CDC1 3 ) δ 162.0 (Ca6), 159.0 (Ca2), 155.9 (Cal), 151.4 (Ca8), 150.4 (Cc5), 150.1 (Ccl3), 147.3 (Ccl2), 141.5 (Cal2), 134.9 (Cel l), 129.3 (CclO), 128.6 (Cal3), 128.4 (Cal4), 126.1 (Cal5), 124.7 (Ca4), 122.1 (Cc9), 119.9 (Cc8), 118.6 (Cc7), 117.6 (Ca5), 109.1 (Ca3), 107.7 (Ca7), 98.5 (Cc6), 66.5 (Cbl), 49.1 (Ccl), 46.8 (Cb3), 43.1 (Cc4), 41.1 (Ca9), 33.6 (Cal l), 30.7 (CalO), 29.1 (Cb2), 27.4 (Cc2), 26.3 (Cc3).

HRMS-ESI (m/z) calculated for C 33 H 38 N 6 0 [M+H]+: 535.3180; Found:535.3172.

Methyl 4-((3-((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy)propyl) (3- (quinolin-4-ylamino)propyl)amino)butanoate (Compound U)

Methyl 4-((3-((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy)propyl) (4- (quinolin-4-ylamino)butyl)amino)butanoate (Compound C)

To a solution of Compound A (30mg; 59μηιο1) or Compound B (26mg; 49μηιο1), K 2 CO 3 (25mg; 181μηιο1), methyl 4-bromobutyrate (ΙΟμί; 87μmol) and a catalytic amount of KI was added. The mixture was stirred at 90°C overnight then diluted with ethylacetate. The organic phase was washed with a saturated solution of NaHC0 3 and brine. The organic phase was dried over Na 2 S0 4 and the solvent was removed. The residue was purified by silica gel flash chromatography using a linear gradient of methanol (0— »10% MeOH) in dichloromethane .

Compound U (Methyl ester of Compound D) was obtained as a white amorphous solid (16mg; 26μιηο1; 44%) and Compound C (Methyl ester of Compound T) was obtained as a white amor hous solid (14mg; 26μιηο1; 45%>).

1H NMR (CDC1 3 ) δ 8.52 (s, 1H, Hal), 8.40 (d, J=6.0Hz, 1H, Hc6), 7.96 (d, J=8.4, 1H, Hc8), 7.79 (d, J=8.4Hz, 1H, Ha4), 7.56 (ddd, J=1.0, 6.0, 8.4Hz, 1H, HclO), 7.43-7.20 (m, 8H, Hc9, Hal5, HNH, Hel l, Hal3 and Hal4), 7.10-7.02(m, 2H, Ha7 and HNH), 7.14 (dd, J=2.6, 9.0Hz, 1H, Ha5), 6.31 (d, J=5.7Hz, 1H, Hc5), 5.92 (m, 1H, HNH), 4.06 (t, J=6.0Hz, 2H, Hbl), 3.67 (q, J=6.3Hz, 2H, Ha9), 3.62 (s, 3H, HMe), 3.37 (q, J=6.0Hz, 2H, Hc3), 2.76 (t, J=7.4Hz, 2H, Hb3), 2.71 (t, J= 7.2Hz, 2H, Hb4), 2.65 (t, J=6.0Hz, 2H, Hcl), 2.55 (t, J=7.2Hz, 2H, Hb6), 2.35 (t, J=7.4Hz, 2H, Hal l), 2.06 (quint, J=7.4Hz, 2H, HalO), 2.01-1.80(m, 6H, Hb5, Hb2 and Hc2).

13 C NMR (CDC1 3 ) δ 173.9 (Cb7), 161.9 (Ca6), 159.1 (Ca2), 155.9 (Cal), 151.3 (Ca8), 151.2 (Cc4), 148.7 (Cc6), 145.9 (Ccl2), 141.6 (Cal2), 129.8 (Cel l), 128.6 (Cal3), 128.4 (Cal4), 127.7 (CclO), 126.0 (Cal5), 124.9 (Ca4), 122.4 (Cc9), 120.2 (Cc8), 118.4 (Cc7), 117.2 (Ca5), 109.1 (Ca3), 107.8 (Ca7), 98.0 (Cc5), 66.0 (Cbl), 53.5 (Ccl), 52.8 (Cb3), 51.6 (CMe), 50.5 (Cb4), 43.0 (Cc3), 41.1 (Ca9), 33.6 (Cal l), 31.7 (Cb6), 30.7 (CalO), 26.5 (Cb2) 25.4 (Cc2), 22.0 (Cb5).

HRMS-ESI m/z) calculated for C 3 7H 45 N 6 03 [M+H]+: 621.3548; Found: 621.3560.

1H NMR (CDC1 3 ) δ 8.50 (s, 1H, Hal), 8.47 (d, J=5.4Hz, 1H, Hcl3), 7.94 (d, J=7.4, 1H, Hc8), 7.69 (d, J=8.3Hz, 1H, Ha4), 7.56 (t, J=7.4Hz, 1H, HclO), 7.34 (t, J=7.4Hz, 1H, Hc9), 7.30-7.22 (m, 3H, Hal3 and Hel l) 7.21-7.12 (m, 3H, Hal5 and Hal4), 7.11 (d, J=2.5Hz, 1H, Ha7), 6.90 (dd, J=2.5, 8.9Hz, 1H, Ha5), 6.31 (d, J=5.3Hz, 1H, Hc6), 5.65 (m, 1H, HNH), 5.30 (m, 1H, HNH), 4.10 (t, J=6.1Hz, 2H, Hbl), 3.67-3.60 (m, 5H, Ha9 and HMe), 3.19 (q, J=7.0Hz, 2H, Hc4), 2.75 (t, J=7.4Hz, 2H, Hcl), 2.59 (t, J=6.6Hz, 2H, Hb6), 2.55 (m, 4H, Hb4 and Hb3), 2.32 (t, J=7.4Hz, 2H, Hal l), 2.03 (quint, J=7.4Hz, 2H, HalO), 1.91 (quint, J=6.3Hz, 2H, Hb5), 1.80-1.68 (m, 4H, Hb2 and Hc2), 1.56 (quint, J=7.3Hz, 2H, Hc3).

1 3 C NMR (CDC1 3 ) δ 174.1 (Cb7), 162.2 (Ca6), 159.0 (Ca2), 155.9 (Cal), 151.4

(Cc5), 150.5(Ccl3), 149.7 (Ca8), 147.9 (Ccl2), 141.6 (Cal2), 129.3 (Cel l), 129.1 (CclO), 128.6 (Cal3), 128.4 (Cal4), 126.0 (Cal5), 124.5 (Ca4), 122.1 (Cc9), 119.6 (Cc8), 118.6 (Cc7), 117.6 (Ca5), 109.0 (Ca3), 107.6 (Ca7), 98.5 (Cc6), 65.9 (Cbl), 53.5 (Ccl), 53.1 (Cb3), 51.5 (CMe), 49.9 (Cb4), 43.2 (Cc4), 41.1 (Ca9), 33.6 (Cal l), 31.7 (Cb6), 30.7 (CalO), 26.8 (Cc3), 26.6 (Cb2), 25.2 (Cc2), 22.4 (Cb5).

HRMS-ESI (m/z) calculated for C38H 4 7N 6 03 [M+H]+: 635.3704; Found: 635.3709. N-(3-((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy)propyl)- N-(3-(quinolin-4- ylamino)propyl)formamide (Compound E)

Compound E is a by-product isolated from the synthesis of Compound U and was obtained as a white amor hous solid (8mg; 15μιηο1; 25%).

1H NMR (DMSO) δ 8.42-8.35 (m, 2H, Hal and Hc6), 8.24-8.10 (m, 3H, Hc8, Ha4 and HNH), 8.08 (d, J=8.9Hz, 1H, Hb4), 7.78 (d, J=8.5Hz, Hcl 1), 7.63-7.58 (m, 1H, HclO), 7.44-7.39 (m, 1H, Hc9), 7.32-7.22 (m, 4H, Hal3 and Hal4), 7.21-7.11 (m, 2H, Hal5 and HNH), 7.10-7.02( m, 2H, Ha5 and Ha7), 6.45 (t, J=5.0Hz, 1H, Hc5), 4.09 (m, 2H,Hbl), 3.57-3.31 (m, 6H, Hc3, Hcl and Hb3), 3.30-3.22(m, 1H, Ha9a), 2.71 (t, J=7.4Hz, 2H, Hal l), 2.05-1.87 (m, 6H, Hc2, HalO and Hb2).

13 C NMR (DMSO) δ 163.5-163.4 (Cb4), 161.9-161.8 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8), 151.2 (Cc6), 150.3-150.2 (Cc4), 148.8-148.7 (Ccl2), 142.2 (Cal2), 129.5-129.4 (Cel l), 129.1 (CclO), 128.8 (Cal3), 128.7 (Cal4), 126.2 (Cal5), 124.8-124-7 (Ca4), 124.2-124.2 (Cc9), 122.2-122.0 (Cc8), 119.3 (Cc7), 117.2-117.1 (Ca5), 109.7-109.6 (Ca3), 107.9-107.8 (Ca7), 98.7-98.6 (Cc5), 66.1-65.4 (Cbl), 46.1 (Ccl), 45.9 (Cb3), 40.5 (Cc3), 39.1 (Ca9), 33.1 (Cal l), 30.8 (CalO), 27.9-27.3 (Cb2), 27.1-26.3 (Cc2).

HRMS-ESI (m/z) calculated for C 33 H 37 N 6 O2 [M+H]+: 549.2973; Found: 549.2972. 4-((3-((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy)propyl) (3-(quinolin-4- ylamino)propyl)amino)butanoic acid (Compound D)

4-((3-((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy)propyl) (4-(quinolin-4- ylamino)butyl)amino)butanoic acid (Compound T)

To a solution of Compound U (16mg; 26μιηο1) or Compound C (14mg; 22μmol) in dioxane (0.5mL) was added a 0.5N solution of NaOH (0.5mL). The mixture was stirred at room temperature for 6h then was neutralized using a 0.5N HC1 solution (0.5mL). The solvent was removed and the residue was triturated in methanol. The mixture was filtrated and the filtrate was purified by reversed phase HPLC using a linear acetonitrile gradient (0→80% CH 3 CN).

Compound D was obtained as pale yellow viscous oil (12mg; 20μηιο1; 78%) and Compound T was obtained as ale yellow viscous oil (1 lmg; 17μηιο1; 79%).

b7

D

1H NMR (DMSO+MeOD) δ 8.38 (s, IH, Hal), 8.33 (d, J=5.3Hz, IH, Hc6), 8.1 (d, J=7.7Hz, IH, Hc8), 8.13 (d, J=8.9Hz, IH, Ha4), 8.09 (t, J=5.3Hz, IH, HNH), 7.76 (dd, J=0.8, 8.3Hz, IH, Hel l), 7.58 (ddd, J=1.0, 6.0, 8.4Hz, IH, HclO), 7.39 (ddd, J=1.0, 6.0, 8.4Hz, IH, Hc9), 7.32-7.21 (m, 4H, Hal3 and Hal4), 7.22-7.12 (m, 2H, Hal5, HNH), 7.10- 7.02(m, 2H, Ha7 and Ha5), 6.37 (d, J=5.7Hz, IH, Hc5), 4.12 (t, J=6.1Hz, 2H, Hbl), 3.53 (q, J=6.3Hz, 2H, Ha9), 3.27 (q, J=5.6Hz, 2H, Hc3), 2.67 (t, J=7.5Hz, 2H, Hcl), 2.60 (t, J= 6.7Hz, 2H, Hb4), 2.56 (t, J=6.7Hz, 2H, Hb3), 2.46 (t, J=6.7Hz, 2H, Hb6), 2.22 (t, J=7.1Hz, 2H, Hal l), 1.99-1.86 (m, 4H, Hb2 and HalO), 1.81 (quint, J=7.4Hz, 2H, Hb5), 1.65 (quint, J=7.4Hz, 2H, Hc2).

1 3 C NMR (DMSO+MeOD) δ 175.1 (Cb7), 162.1 (Ca6), 159.5 (Ca2), 156.0 (Cal),

151.8 (Ca8), 151.0 (Cc6), 150.4 (Cc4), 148.6 (Ccl2), 142.2 (Cal2), 129.3 (Cel l), 129.1 (CclO), 128.8 (Cal3), 128.7 (Cal4), 126.1 (Cal5), 124.7 (Ca4), 124.2 (Cc8), 122.1 (Cc9), 119.3 (Cc7), 117.1 (Ca5), 109.5 (Ca3), 107.8 (Ca7), 98.5 (Cc5), 66.4 (Cbl), 53.4 (Ccl), 51.8 (Cb3), 50.2 (Cb4), 41.2 (Ca9), 40.6 (Cc3), 33.1 (Cal l), 32.6 (Cb6), 30.8 (CalO), 26.7 (Cb2), 25.8 (Cc2), 22.6 (Cb5).

HRMS-ESI (m/z) calculated for C36H43N6O3 [M+H]+: 607.3391; Found:607.3391.

1H NMR (DMSO+MeOD) δ 8.37 (s, IH, Hal), 8.33 (d, J=5.5Hz, IH, Hcl3), 8.19 (d, J=7.9Hz, IH, Hc8), 8.13 (d, J=8.9Hz, IH, Ha4), 8.09 (t, J=5.3Hz, IH, HNH), 7.75 (dd, J=0.9, 8.4Hz, IH, Hel l), 7.57 (ddd, J=1.2, 6.7, 8.2Hz, IH, HclO), 7.38 (ddd, J=1.2, 6.7, 8.2Hz, IH, Hc9), 7.29-7.20 (m, 4H, Hal 3 and Hal4), 7.19-7.1 1 (m, 2H, Hal 5, HNH), 7.09 (dd, J=2.5, 9.0Hz, IH, Ha5), 7.03(d, J=2.5Hz, IH, Ha7), 6.35 (d, J=5.3Hz, IH, Hc5), 4.1 1 (t, J=6.1Hz, 2H, Hbl), 3.52 (q, J=7.0Hz, 2H, Ha9), 3.19 (m, 2H, Hc4), 2.67 (t, J=7.5Hz, 2H, Hcl), 2.56 (t, J=6.7Hz, 2H, Hb6), 2.48-2.39 (m, 4H, Hb4 and Hb3), 2.22 (t, J=7.1Hz, 2H, Hal l), 1.91 (quint, J=7.4Hz, 2H, HalO), 1.86 (quint, J=6.3Hz, 2H, Hb5), 1.71-1.58 (m, 4H, Hb2 and Hc2), 1.51 (quint, J=7.6Hz, 2H, Hc3).

13 C NMR (DMSO+MeOD) δ 175.2 (Cb7), 162.0 (Ca6), 159.5 (Ca2), 156.0 (Cal), 151.7 (Cc5), 151.0 (Ccl 3), 150.4 (Ca8), 148.6 (Ccl2), 142.2 (Cal2), 129.3 (Cel l), 129.1 (Ccl O), 128.8 (Cal 3), 128.7 (Cal4), 126.1 (Cal 5), 124.7 (Ca4), 124.1 (Cc8), 122.2 (Cc9), 1 19.2 (Cc7), 1 17.2 (Ca5), 109.5 (Ca3), 107.8 (Ca7), 98.5 (Cc6), 65.4 (Cbl), 53.6 (Ccl), 53.4 (Cb3), 50.1 (CMe), 46.0 (Cb4), 42.8 (Cc4), 40.5 (Ca9), 33.2 (Cal 1), 32.7 (Cb6), 30.8 (CalO), 26.7 (Cc3), 26.1 (Cb2), 24.7 (Cc2), 22.6 (Cb5).

HRMS-ESI (m/z) calculated for [M+H]+: 621.3548; Found: 621.3545.

Example 3 : Compound G

Compound G 45 a) N-Boc-piperidine-4-methanol, NaH, DMF, 1 10°C, 3h, 67%. b) POCl 3 , triazole, TEA, MeCN, RT, 18h. c) 3-Phenylpropylamine, TEA, DMF, RT, 2h, 80% over two steps, d) TFA, RT, lh, 96%. e) 26, K 2 C0 3 , KI, DMF, 65°C, 12h, 62%.

7-0-(( V-Boc)piperidin-4-ylmethoxy)quinazolinone (43)

To a mixture of (N-Boc)piperidin-4-ylmethanol (1.12g; 5.2mmol) in DMF (2mL) at 0°C under argon was added sodium hydride (125mg, 5.2mmol). The mixture was stirred for 15min at 0°C then 42 (162mg; lmmol) was added portion wise. The mixture was stirred at 0°C for lOmin then at room temperature for lOmin, at 60°C for 15min and finally at 1 10°C for 2h. The reaction mixture was diluted with ethylacetate and washed with water and brine. The organic phase was dried over magnesium sulfate and the solvent was removed. The crude product was purified by silica gel flash chromatography using a linear gradient of ethylacetate (0— »100% AcOEt) in cyclohexane to afford 43 as a white powder (241mg; 0.67 μιηοΐ; 67%).

1H NMR (500MHz; CDC1 3 ) δ 11.50 (s, 1H, HNH), 8.19 (d, 1H, J= 8.9, Ha4), 8.7 (s, 1H, Hal), 7.12-7.07 (m, 2H, Ha7 and Ha5), 4.18 (sb, 2H, Hb4), 3.94 (d, J= 6.8, 2H, Hbl), 2.76 (m, 2H, Hb4), 2.03 (m, 1H, Hb2), 1.84 (m, 2H, Hb3), 1.47 (s, 9H, HBoc), 1.38-1.1 1 ( m, 2H, Hb3).

1 3 C NMR (125MHz, CDC1 3 ) δ 164.6 (Ca6), 162.3 (Ca2), 155.2 (CBoc),

151.5(Ca8), 144.3 (Cal), 128.3 (Ca4), 118.0 (Ca5), 116.3 (Ca3), 109.4 (Ca7), 79.9 (CBoc), 73.0 (Cbl), 47.0(Cb4), 36.3 (Cb2), 29.1(Cb3), 28.8 (CBoc).

HRMS-ESI (m/z) calculated for C 19 H 26 N 3 O 4 [M+H] + : 360.1918; found:360.1911. 4-((3-phenylpropyl)amino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy))

quinazoline (44)

To a solution of triazole (280mg; 4mmol) and POCI 3 (120μ1; 1.32mmol) in 3 mL of acetonitrile at 0°C was added TEA (560μί) dropwise. The reaction mixture was stirred at 0°C for 40min then 30min at room temperature. 43 (215mg; 0.6mmol) was added and the mixture was vigorously stirred at room temperature overnight. The reaction was followed by TLC using ethyl acetate as eluent. The mixture was refluxed for lh to reach completion. After complete consumption of the starting material, the solvent was removed and the residue was taken off with ethylacetate and washed with water and brine, and dried over sodium sulfate. The solvent was removed and the residue was solubilized in DMF (2mL). Phenylpropylamine (130μί; l .Ommol) and TEA (167μί; 1.2mmol) were added and the mixture was stirred for 3h at room temperature. The mixture was diluted with ethyl acetate and washed with water, brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ethyl acetate (0— »100% AcOEt) in cyclohexane to afford 44 as a white powder (232mg; 0.49mmol; 81%).

a9 a1 1 _ a13 1H NMR (500MHz; CDC1 3 ) δ 8.57 (s, 1H, Hal), 7.33-7.28 (m, 3H, Ha4 and Hal3), 7.25-7.20 (m, 3H, Hal5 and Hal4), 7.12 (d, J=2.6Hz, 1H, Ha7), 7.00 (dd, J=2.6, 9.5Hz, 1H, Ha5), 5.44 (brt, J=5.2, 1H, HNH), 4.17 (brs, 2H, Hb4eq), 3.92 (d, J=6.3Hz, 2H, Hbl), 3.70 (q, J=7.2Hz, 2H, Ha9), 2.79 (t, 2H, J=7.3Hz, Hal l), 2.75 (brt, J=11.2Hz, 2H, Hb4ax), 2.09 (quint, J=7.3Hz, 2H, HalO), 2.01 (m, 1H, Hb2), 1.83 (d, J=12.3Hz, 2H, Hb3eq), 1.47 (s, 9H, HBoc), 1.31 (dq, J=4.5-12.3Hz, 2H, Hb3ax).

13 C NMR (125MHz; CDC1 3 ) δ 162.9 (Ca6), 159.2 (Ca2), 156.1 (Cal), 155.0 (CBoc),151.7(Ca8), 141.7 (Cal2), 128.8 (Cal3), 128.6 (Cal4), 126.3(Cal5), 122.0(Ca4) 118.0 (Ca5), 109.1 (Ca3), 108.0 (Ca7), 79.6 (CBoc), 72.6 (Cbl), 43.7(Cb4), 41.3(Ca9), 36.1 (Cb2), 33.8 (Cal 1), 30.9 (CalO), 29.0(Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated for C19H26N3O4 [M+H] + : 477.2860; found:477.2861.

4-((3-phenylpropyl)amino)-7-0-(piperidin-4-ylmethoxy)quin azoline (45)

A mixture of 44 (220mg; 0.46mmol) in TFA was stirred for lh at room temperature. TFA was removed. The residue was diluted with dichloromethane and the organic phase was washed with saturated Na 2 C0 3 . The solvent was removed and 45 was obtained as pale blue foam (165mg; 0.44mmol; 96%).

1H NMR (DMSO) δ 9.94 (brs, 1H, HNH), 8.79 (s, 1H, Hal), 8.4 (d, J=9.3Hz, 1H, Ha4), 7.43-7.13 (m, 7H, Ha5, Ha7, Hal3, Hal4 and Hal5), 4.06 (d, J=6.2Hz, 2H, Hbl), 3.70 (q, J=7.2Hz, 2H, Ha9), 3.34 (brd, J=12.6Hz, 2H, Hb4eq), 2.94 (brt, J=11.5Hz, 2H, Hb4ax), 2.69 (t, J=7.3Hz, 2H, Hal l), 2.15 (m, 1H, Hb2), 2.00 (quint, 2H, J=7.3Hz, HalO), 1.95 (brd, 2H, Hb3eq), 1.53 (dq, J=4.0-l 5.0Hz, 2H, Hb3ax).

13 C NMR (DMSO) δ 164.5 (Ca6), 160.8(Ca2), 152.5 (Cal), 142.3(Ca8), 141.7 (Cal2), 129.3 (Cal3), 129.2 (Cal4), 127.2 (Ca4),126.7 (Cal5), 119.1 (Ca5), 107.9 (Ca3), 102.6 (Ca7), 72.9 (Cbl), 43.6(Cb4), 42.2(Ca9), 33.8 (Cb2), 33.4 (Cal l), 30.8 (CalO), 26.0(Cb3).

HRMS-ESI (m/z) calculated for C19H26N3O4 [M+H] + : 377.2336; found: 377.2303. 4-((3-phenylpropyl)amino)-7-((l-(2-(quinolin-4-ylamino)ethyl )piperidin-4- yl)methoxy)quinazoline (Compound G)

To a solution of 45 (30mg; 80μηιο1), K 2 CO 3 (22mg; 160μηιο1) and a catalytic amount of KI in DMF (lmL) was added 26 (33mg; 160μιηο1). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound G as a white powder (35mg; 64μιηο1; 80%).

1H NMR (500MHz; CDC1 3 ) δ 8.58 (s, 1H, Hal), 8.56 (d, J=5.4Hz, 1H, Hc5), 7.98 (dd, J=0.7, 8.4Hz, 1H, Hc8), 7.76 (dd, J=0.7, 8.4Hz, 1H, Hel l), 7.63 (ddd, J=1.2, 6.9, 8.2Hz, 1H, HclO), 7.46 (ddd, J=1.2, 6.9, 8.2Hz, 1H, Hc9), 7.34-7.28 (m, 3H, Ha4 and Hal3), 7.25-7.20 (m, 3H, Hal5 and Hal4), 7.13 (d, J=2.5Hz, 1H, Ha7), 7.00 (dd, J=2.6, 5.1Hz, 1H, 9.1Hz, Ha5), 6.40 (d, J=5.1Hz, 1H, Hc4), 5.96 (brt, J=4.5Hz, 1H, HNHc), 5.48 (brt, J=5.1Hz, 1H, HNHa), 3.96 (d, J=6.1Hz, 2H, Hbl), 3.70 (q, J=7.2Hz, 2H, Ha9), 3.34 (q, J=5.2Hz, 2H, Hc2), 3.00 (brd, J=12.0Hz, 2H, Hb4eq), 2.75 (m, 4H, Hal l and Hcl), 2.14 (dt, J=2.1Hz, 2H, Hb4ax), 2.09 (quint, J=7.2Hz, 2H, HalO), 1.94 (m, 1H, Hb2), 1.91 (d, J=12.3Hz, 2H, Hb3eq), 1.47 (dq, J=3.4, 12.5Hz, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 161.6 (Ca6), 158.4 (Ca2), 155.3 (Cal), 151.0 (Ca8), 150.5 (Cc5), 149.1 (Cc3), 147.8 (Cc6), 140.9 (Cal2), 129.3 (Cc8), 128.3 (CclO), 128.0 (Cal3), 127.8 (Cal4), 125.5 (Cal5), 124.0 (Cc9), 121.2 (Ca4), 118.8 (Cel l), 118.3 (Cc7), 117.2 (Ca5), 108.3 (Ca3), 107.2 (Ca7), 98.4 (Cc4), 72.0 (Cbl), 55.3(Ccl), 52.3 (Cb4), 40.5 (Ca9), 38.6 (Cc2), 35.0 (Cb2), 33.0 (Cal l), 30.1 (CalO), 28.6 (Cb3).

HRMS-ESI (m/z) calculated for ^Η 26 Ν 3 0 4 [M+H] + : 547.3180; found: 547.3171. Example 4: Compound O

Compound O was synthesized following the same procedure as for Compound G. 4-(3-phenethylamino)-7-((l-(2-(quinolin-4-ylamino)ethyl)pipe ridin-4-yl) methoxy)quinazoline (Compound O)

To a solution of 4-(2-phenethylamino)-7-(piperidin-4-ylmethoxy)quinazoline (30mg; 0.08mmol), K 2 CO 3 (23mg; 0.16mmol) and a catalytic amount of KI in DMF (1.5mL) was added 26 (40mg; 0.16mmol). The mixture was stirred at 65°C for 24 hours. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound O as a white powder (28mg; 0.05mmol; 64%).

1H NMR (500MHz; CDC1 3 ) δ 8.64 (s, IH, Hal), 8.59 (d, J=5.6Hz, IH, Hc5), 8.04 (d, J=8.2Hz, IH, Hc8), 7.80 (d, J=0.7, 8.4Hz, IH, Hel l), 7.68 (ddd, J=1.3, 6.9, 8.3Hz, IH, HclO), 7.51 (ddd, J=l .l, 6.7, 8.3Hz, IH, Hc9), 7.44 (d, J=9.28Hz, IH, Ha4), 7.38-7.33 (m, 2H, Hal2), 7.31-7.26 (m, 3H, Hal4 and Hal3), 7.18 (d, J=2.6Hz, IH, Ha7), 7.05 (dd, J=2.6, 9.0Hz, IH, Ha5), 6.43 (d, J=5.4Hz, IH, Hc4), 6.15 (brs, IH, HNHc), 5.60 (brt, J=5.7, IH, HNHa), 4.00 (d, J=6.2Hz, 2H, Hbl), 3.70 (dd, J=6.7, 12.2Hz, 2H, Ha9), 3.38 (q, J=5.2Hz, 2H, Hc2), 3.08-3.00 (m, 4H, Hb4eq and Hcl), 2.83 (t, J=6.2Hz, HalO), 2.18 (dt, J=1.3, 11.6Hz, 2H, Hb4ax), 2.09-1.89 (m, 3H, Hb2 and Hb3eq), 1.47 (dq, J=3.0-12.5Hz, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 162.2 (Ca6), 159.0 (Ca2), 156.0 (Cal), 151.7 (Ca8), 150.3 (Cc5), 150.2 (Cc3), 147.5 (Cc6), 138.9 (Cal2), 129.4 (Cc8), 129.1 (CclO), 128.9 (Cal2), 128.8 (Cal3), 126.6 (Cal4), 124.9 (Cc9), 121.8 (Ca4), 119.5 (Cel l), 118.7(Cc7), 118.1 (Ca5), 109.2 (Ca3), 107.9 (Ca7), 98.8 (Cc4), 72.6 (Cbl), 55.8(Ccl), 52.9 (Cb4), 42.1 (Ca9), 39.1 (Cc2), 35.6 (Cb2), 35.3 (CalO), 29.2 (Cb3).

HRMS-ESI (m/z) calculated for C 33 H 37 N 6 0 [M+H] + : 533.3023; found:533.3023. Example 5 : Compound P

Compound P was synthesized following the same procedure as for Compound G. 4-(3-benzylamino)-7-((l-(2-(quinolin-4-ylamino)ethyl)piperid in-4-yl)methoxy) quinazoline (Compound P)

To a solution of 4-(2-benzylamino)-7-(piperidin-4-ylmethoxy)quinazoline (30mg;

0.086mmol), K 2 CO 3 (24mg; 0.172mmol) and a catalytic amount of KI in DMF (1.5mL) was added 26 (42mg; 0.172mmol). The mixture was stirred at 65°C overnight. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound P as a white powder (30mg; 0.058mmol; 68%).

1H NMR (500MHz; CDC1 3 ) δ 8.66 (s, 1H, Hal), 8.58 (d, J=5.1Hz, 1H, Hc5), 8.00 (dd, J=0.7, 8.5Hz, 1H, Hc8), 7.79 (dd, J=0.7, 8.5Hz, 1H, Hel l), 7.61 (d, J=8.9Hz, 1H, Ha4), 7.66 (ddd, J=1.4, 6.8, 8.3Hz, 1H, HclO), 7.48 (ddd, J=1.4, 6.8, 8.3Hz, 1H, Hc9), 7.45-7.32 (m, 5H, Hal l and Hal2 and Hal3), 7.20 (d, J=2.4Hz, 1H, Ha7), 7.08 (dd, J=2.4, 1H, 9.1Hz, Ha5), 6.42 (d, J=5.3Hz, 1H, Hc4), 5.96 (brt, J=4.1, 1H, HNHc), 5.87(m, 1H, HNHa), 4.87 (d, J=5.2Hz, Ha9), 4.00 (d, J=6.1Hz, 2H, Hbl), 3.36 (q, J=5.2Hz, 2H, Hc2), 3.03 (brd, J=11.7Hz, 2H, Hb4eq), 2.81 (t, J=6.0Hz, 2H, Hal l), 2.16 (dt, J=1.2, 11.8Hz, 2H, Hb4ax), 2.01-1.89 (m, 3H, Hb2 and Hb3eq), 1.50 (dq, J=3.6-12.1Hz, 2H, Hb3ax).

1 3 C NMR (125MHz, CDC1 3 ) δ 162.3 (Ca6), 158.9 (Ca2), 156.0 (Cal), 151.8 (Ca8),

151.1 (Cc5), 149.8 (Cc3), 148.3 (Cc6), 138.2 (CalO), 129.8 (Cc8), 129.0 (CclO), 128.8 (Cal2), 128.0 (Cal l), 127.8 (Cal3), 124.6 (Cc9), 122.0 (Ca4), 119.5 (Cel l), 118.9 (Cc7), 118.1 (Ca5), 109.0 (Ca3), 107.9 (Ca7), 99.0 (Cc4 ), 72.7 (Cbl), 55.9(Ccl), 52.9 (Cb4), 45.3 (Ca9), 39.2 (Cc2), 35.7 (Cb2), 29.2 (Cb3).

HRMS-ESI (m/z) calculated for C 32 H 35 N 6 0 [M+H] + : 519.2867; Found: 519.2870. Example 6: Compound H

Compound H was synthesized following the same procedure as for Compound G. 4-propylamino-7-((l-(2-(quinolin-4-ylamino)ethyl)piperidin-4 -yl)

methoxy)quinazoline (Compound H)

To a solution of 4-propylamino-7-O-(piperidin-4-ylmethoxy)quinazoline (lOmg;

33μιηο1), K 2 CO 3 (9mg; 66μιηο1) and a catalytic amount of KI in DMF (0.5mL) was added 26 (14mg; 67μιηο1). The mixture was stirred at 65°C overnight then was diluted with ethylacetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NHs) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.2% of TEA (0→80% CH 3 CN) to afford Compound H as a white powder (12.0mg; 1

1H NMR (DMSO) δ 8.39 (d, J=5.3Hz, 1H, Hc5), 8.37 (s, 1H, Hal), 8.17-8.11 (m,

2H, Ha4 and Hc8), 8.07 (brt, J=5.3, 1H, HNHa), 7.77 (dd, J=l . l, 8.4Hz, 1H, Hel l), 7.60 (ddd, J=1.3, 6.9, 8.2Hz, 1H, HclO), 7.41 (ddd, J=1.3, 6.9, 8.2Hz, 1H, Hc9), 7.09 (dd, J=2.5, 9.0Hz, 1H, Ha5), 7.04 (m, 1H, Ha7), 6.47 (d, J=5.8Hz, 1H, Hc4), 3.96 (d, J=5.85Hz, 2H, Hbl), 3.45 (q, J=5.8Hz, 2H, Ha9), 3.40 (q, J=6.2Hz, 2H, Hc2), 3.00 (brd, J=10.1Hz, 2H, Hb4eq), 2.62 (t, J=7.1Hz, 2H,Hcl), 2.04 (t, J=10.1Hz, 2H, Hb4ax), 1.91 (m, 3H, Hb3eq and Hb2), 1.63 (sext, J=7.4Hz, 2H, HalO), 1.36 (dq, J=3.0-12.6Hz, 2H, Hb3ax), 0.91 (t, J=7.3Hz, 3H, Hal l).

13 C NMR (DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8), 151.2 (Cc5), 150.2 (Cc3), 148.2 (Cc6), 129.5 (Cel l), 129.1 (CclO), 124.7 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 119.2 (Cc7), 117.1 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6(Ccl), 53.5 (Cb4), 42.6 (Ca9), 40.6 (Cc2), 35.7 (Cb2), 29.0 (Cb3), 22.4 (CalO), 11.9 (Cal l). HRMS-ESI (m/z) calculated for C 2 8H 35 N 6 0 [M+H] : 471.2867; found: 471.2876. Example 7: Compound I

Compound I was synthesized following the same procedure as for Compound G. 4-amino-7-((l-(2-(quinolin-4-ylamino)ethyl)piperidin-4-yl)me thoxy)quinazoline (Compound I)

To a solution of 4-amino-7-(piperidin-4-ylmethoxy)quinazoline (lOmg; 0.039mmol), K 2 CO 3 (21mg; 0.078mmol) and a catalytic amount of KI in DMF (0.5mL) was added 26 (16mg, 0.078mmol). The mixture was stirred at 65°C overnight then was diluted with ethylacetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound I as a white powder (11.4mg; 0.027mmol; 68%).

1H NMR (DMSO) δ 8.39 (d, J=5.5Hz, IH, Hc5), 8.29 (s, IH, Hal), 8.15 (m, 3H,

Hc8 and HNH 2 ), 8.10 (d, J=9.0, IH, Ha4), 7.77 (dd, J=1.0Hz, 8.4Hz, IH, Hel l), 7.60 (ddd, J=1.2, 6.8, 8.2Hz, IH, HclO), 7.42 (ddd, J=1.2, 6.8, 8.2Hz, IH, Hc9), 7.08 (dd, J=2.5, 9.0Hz, IH, Ha5), 7.06-7.01 (m, 2H, Ha7 and HNH), 6.47 (d, J=5.3Hz, IH, Hc4), 3.97 (d, J=5.8Hz, 2H, Hbl), 3.41 (q, J=6.4Hz, 2H, Hc2), 3.00 (brd, J=11.0Hz, 2H, Hb4eq), 2.62 (t, J=6.2Hz, 2H,Hcl), 2.05 (t, J=11.9Hz, 2H, Hb4ax), 1.78 (m, 3H, Hb3eq and Hb2), 1.36 (dq, J=2.5, 11.9Hz, 2H, Hb3ax).

13 C NMR (DMSO) δ 162.4 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8), 151.2 (Cc5), 150.2 (Cc3), 148.2 (Cc6), 129.5 (Cel l), 129.1 (CclO), 124.7 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 119.2 (Cc7), 117.1 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6(Ccl), 53.5 (Cb4), 42.6 (Ca9), 40.6 (Cc2), 35.7 (Cb2), 29.0 (Cb3), 22.4 (CalO), 11.9 (Cal l).

HRMS-ESI (m/z) calculated for C25H29N6O [M+H] + : 429.2397; found: 429.2404. Example 8: Compound N

Compound N was synthesized following the same procedure as for Compound G. 4-([l,l'-biphenyl]-4-ylmethylamino)-7-((l-(2-(quinolin-4-yla mino)ethyl) piperidin-4-yl)methoxy)quinazoline (Compound N)

To a solution of 4-([l, -biphenyl]-4-ylmethylamino)-7-((piperidin-4-yl) methoxy)quinazoline (12mg, 28μιηο1), K 2 CO 3 (8mg, 56μmol) and a catalytic amount of KI in DMF (0.5mL) was added 26 (14mg, 56μιηο1). The mixture was stirred at 65°C overnight. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound N as a white owder (5mg, 84μιηο1, 31%).

1H NMR (500MHz; CDC1 3 ) δ 8.67 (s, 1H, Hal), 8.57 (d, J=5.3Hz, 1H, Hc5), 8.08 (d, J=8.2Hz, 1H, Hc8), 7.81 (dd, J=8.2Hz, 1H, Hel l), 7.70 (ddd, J=1.2, 6.9, 8.2Hz, 1H, HclO), 7.46 (ddd, J=l . l, 6.8, 8.2Hz, 1H, Hc9), 7.66-7.58 (m, 5H, Ha4 and Hal7 and Hal2), 7.56-7.43 (m, 4H, Hal l and Hal6), 7.38 (m, 1H, Hal7), 7.20 (d, J=2.6Hz, 1H, Ha7), 7.08 (dd, J=2.6, 9.2Hz, 1H, Ha5), 6.43 (d, J=5.3Hz, 1H, Hc4), 6.40 (brs, 1H, HNH), 5.93 (brt, J=5.4Hz, 1H, HNH), 4.92 (d, J=5.3Hz, 2H, Ha9), 4.01 (d, J=6.0Hz, 2H, Hbl), 3.70 (q, J=7.2Hz, 2H, Ha9), 3.41 (brq, J=4.3Hz, 2H, Hc2), 3.00 (brd, J=11.5Hz, 2H, Hb4eq), 2.84 (t, J=5.9Hz, 2H, Hcl), 2.20 (dt, J=2.0, 12.0Hz, 2H, Hb4ax), 2.05-1.91 (m, 3H, Hb2 and Hb3eq), 1.52 (dq, J=2.4, 12.5Hz, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 162.4 (Ca6), 159.0 (Ca2), 156.0 (Cal), 151.7 (Ca8), 150.8 (Cc3), 149.2 (Cc5), 146.1 (Cc6), 140.8 (Cal4), 140.6 (Cal3), 137.3 (CalO), 129.9 (CclO), 128.8 (Cal6), 128.5 (Cal l), 128.1 (Cc8), 127.6 (Cal2), 127.4 (Cal7), 125.2 (Cc9), 122.0 (Ca4), 119.7 (Cel l), 118.4 (Cc7), 118.2 (Ca5), 109.0 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.6 (Cbl), 55.7 (Ccl), 52.9 (Cb4), 45.0 (Ca9), 39.1 (Cc2), 35.6 (Cb2), 29.2 (Cb3).

HRMS-ESI(m/z) calculated for C 38 H 39 N 6 0 [M+H] + : 595.3180; found: 595.3172. Example 9: Compound Q

Compound Q was synthesized following the same procedure as for Compound G. 6-methyl-N 4 -(2-((7-((l-(2-(quinolin-4-ylamino)ethyl)piperidin-4-y l)methoxy) quinazolin-4-yl)amino)ethyl)pyrimidine-2,4-diamine (Compound Q)

To a solution of 6-methyl-N4-(2-(7-(piperidin-4-ylmethoxy)quinazolin-4- ylamino)ethyl)pyrimidine-2,4-diamine (95mg, 0.23mmol), K 2 CO 3 (64mg, 0.46mmol) and a catalytic amount of KI in DMF (1.5mL) was added 26 (113mg; 46μιηο1). The mixture was stirred at 65°C overnight. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0— »80% CH 3 CN to afford Compound as a white powder (46mg; 79μιηο1; 34%).

1H NMR (500MHz, DMSO) δ 8.40 (m, 2H, Hc5 and Hal), 8.20 (brt, J=8.15Hz, IH, HNH), 8.15 (d, J=7.9Hz, IH, Hc8), 8.11 (d, J=9.2Hz, IH, Ha4), 7.77 (dd, J=l .l, 8.5Hz, IH, Hel l), 7.62 (ddd, J=l .l, 7.0, 8.3Hz, IH, HclO), 7.43 (ddd, J= J=l .l, 7.0, 8.3Hz, IH, Hc9), 7.11 (dd, J=2.5Hz, 7.1Hz, IH, Ha5), 7.07 (d, J=2.5Hz, IH, Ha7), 7.04 (brt, IH, HNH), 6.48 (d, J=5.2Hz, IH, Hc4), 5.84 (brs, 2H, HNH 2 ), 5.63 (brs, IH, HNH), 3.98 (d, J=6.0Hz, 2H, Hbl), 3.65 (brq, J=5.5Hz, 2H, Ha9), 3.55-3.36 (m, 4H, HalO and Hc2), 3.01 (d, J=11.0Hz, 2H, Hb4eq), 2.64 (t, J=7.0Hz, Hcl), 2.06 (t, J=11.0Hz, 2H, Hb4ax), 2.00 (s, 3H, Hal5), 1.86-1.76 (m, 3H, Hb2 and Hb3eq), 1.38 (dq, J=2.8-12.5Hz, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 164.1 (Cal3), 163.3 (Cal4), 162.2 (Ca6), 159.6 (Ca2), 156.0 (Cal), 151.8 (Ca3), 151.1 (Cc5), 150.3 (Cc7), 148.7 (Cc6), 129.4 (Cel l), 129.1 (CclO), 124.8 (Ca4), 124.3 (Hc9), 121.9 (Cc8), 119.2 (Cc7), 117.3(Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.8 (Cal2), 98.7 (Cc4), 72.7 (Cbl), 56.6(Ccl), 53.5 (Cb4), 41.2 (a9), 40.6 (Cc2), 39.4 (CalO), 35.7 (Cb2), 29.0 (Cb3), 23.6 (Cal5).

HRMS-ESI (m/z) calculated for C32H39N10O [M+H] + : 579.3303; found: 579.3319. Example 10: Compound

a) i) 2-(Bocamino)-ethyl bromide, TEA, DMF, 65°C, 2h. ii) TFA, RT, lh. b) MOMC1, BSA, DCM, RT, 18h. c) TosCl, Me-piperidine, TEA, MeCN, 12h, RT. d) 47, TEA, DMF, RT, 2h, 23% over three steps from 45. l-(methoxymethyl)uracyl (48)

To a solution of uracyl (0.88g; 7.94 mmol) in 250 mL of dichloromethane was added N,0-bis(trimethylsilyl)acetamide (4.8 mL; 19.4 mmol). The mixture was stirred 1 hour at room temperature. To the reaction mixture was added chloromethyl methyl ether (784 μί; 10.32 mmol) and the mixture was stirred 17 hours at room temperature. The solvent was removed and the residue was purified by silica gel chromatography using the eluent cyclohexane / ethyl acetate (7/3) to give 48 (988 mg; 7.4 mmol; 93%) as a white powder.

48

1H NMR (500MHz; DMSO) δ 11.32 (s, 1H, HNH), 7.70 (d, J=8.0Hz, 1H, Hc5),

5.61 (d, J=8.0Hz, 1H, Hc4), 5.02 (s, 2H, Hc7), 3.27 (s, 3H, Hc8).

13 C NMR (125MHz, CDC1 3 ) δ 164.0 (Cc3), 151.5 (Cc6), 145.4 (Cc5), 101.9 (Cc4), 78.0 (Cc7), 56.4 (C8). HRMS-ESI (m/z) calculated for C 6 H 8 N 2 Na03 [M+Na] : 179.0427; Found: 179.0416. l-(methoxymethyl)- V 4 -(2-(4-(((4-((3-phenylpropyl)amino)quinazolin-7-yl)oxy ) methyl)piperidin-l-yl)ethyl)cytosine (Compound K)

To a solution of 45 (lOmg; 27μηιο1), TEA (30uL; 0.22mmol), in DMF (0.2mL) was added 2-(N-bocamino)ethylbromide (lOmg; 35μηιο1). The mixture was stirred at room temperature for 2.5h. The mixture was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the crude product was immediately solubilized in TFA (0.5mL). The mixture was stirred at room temperature for 0.5h. TFA was removed by vacuum. The residue was solubilized in ammonia 7N in methanol and the solvent was removed to afford crude 47 that was used without further purification.

To a solution of triazole (28mg; 0.40mmol) and POCI 3 (12μ1; 0.132mmol) in 0.3 mL of acetonitrile at 0°C was added TEA 56 μΐ ^ dropwise. The reaction mixture was stirred at 0°C for 40min then 30min at room temperature. 48 (lOmg; 60μιηο1) was added and the mixture was vigorously stirred at room temperature overnight. The solvent was removed and 0.5 mL of a solution of previously prepared 47 was added to the residue. The reaction mixture was stirred 3 h at 35°C. The mixture was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NHs) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0— »80% CH3CN) to afford Compound K (3.5mg; 60μιηο1, 23%) as a white powder.

1H NMR (500MHz; CDC1 3 ) δ 8.57 (s, 1H, Hal), 7.34-7.28 (m, 3H, Ha4 and Hal3), 7.28-7.20 (m, 4H, Hal5, Hal4 and Hc5), 7.11 (d, J=2.3Hz, 1H, Ha7), 7.00 (dd, J=2.3, 8.9Hz, 1H, Ha5), 5.79 (brt, J=4.4Hz, 1H, HNHc), 5.67 (d, J=7.3Hz, 1H, Hc4), 5.44 (brt, J=5.5, 1H, HNHa), 5.17 (s, 2H, Hc7), 4.85 (brt, J=5.1Hz, 1H, HNHc), 3.92 (d, J=5.8Hz, 2H,Hbl), 3.70 (q, J=7.0Hz, 2H, Ha9), 3.58 (q, J=4.9Hz, 2H, Hc2), 3.39 (s, 3H, Hc8), 2.94 (brd, J=10.6Hz, 2H, Hb4eq), 2.79 (t, J=7.2Hz, Hal l), 2.55 (t, J=5.7Hz, 2H, Hcl), 2.11-2.00 (m, 4H,HalO and Hb4ax), 1.95-1.79 (m, 3H, Hb2 and Hb3eq), 1.46-1.35 (m, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 163.6 (Cc3), 162.2 (Ca6), 159.0 (Ca2), 157.0 (Cc6), 156.0 (Cal), 151.6 (Ca8), 142.7 (Cc5), 141.5 (Cal2), 128.6 (Cal3), 128.4 (Cal4), 126.1 (Cal5), 121.8 (Ca4), 117.9 (Ca5), 109.0 (Ca3), 107.8 (Ca7), 95.8 (Cc4), 78.8(Cc7), 72.7 (Cbl), 56.7 (Cc8), 56.2(Ccl), 53.0 (Cb4), 41.1 (Ca9), 37.2 (Cc2), 35.6 (Cb2), 33.7 (Cal l), 30.7 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated for C31H40N7O3 [M+H] + : 558.3187; found: 558.3182.

Example 11 : Compound J

a) Ethanolamine, 125°C, 4h, quantitative yield, b) SOCl 2 , DMF, Flash boiling, quantitative yield, c) 45, K 2 C0 3 , KI, DMF, 90°C, 12h, 69%.

4-((2-Hydroxyethyl)amino)pyridine (49)

A mixture of 4-chloropyridine (500mg; 4.41mmol) in ethanolamine (2.6mL; 44mmol) was stirred at 1 10°C for 3h. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ethyl acetate (0— »100% AcOEt) in cyclohexane to afford 49 as a white powder (607mg; 4.40mmol; quantitative yield).

1H NMR (500MHz, DMSO) δ 8.00 (d, J=6.1Hz, 1H, Hc5), 6.49 (m, 3H, Hc4 and HNH), 4.77 (brs, 1H, HOH), 3.53 (t, J=6.0Hz, 2H, Hcl), 3.13 (q, J=5.9Hz, 2H, Hc2).

13 C NMR (125MHz, DMSO) δ: 154.1 (Cc3), 149.7(Cc5), 107.5 (Cc4), 59.7 (Ccl), 40.6 (Cc2). HRMS-ESI (m/z) calculated for C 7 Hi 0 N 2 NaO [M+Na] : 161.0685; found: 161.0650.

4-((2-chloroethyl)amino)quinoline chlorhydrate (50)

49 (300mg; 1.92mmol) was solubilized in thionyl chloride (2ml). The mixture was flash boiled and the solvent was removed. Toluene was added to remove the residual thionyl chloride by co-evaporation. The residue was triturated in dichloromethane and the solid was filtrated to afford 50 chlorhydrate as a white solid (360mg; 1.87mmol; 97%).

1H NMR (DMSO) δ 8.22 (brs, 1H, HNH), 8.1 1 (d, J=6.8Hz, 2H, Hc5), 6.81 (d, J=6.8Hz, 2H, Hc4), 3.76 (t, J=6.0Hz, 2H, Hcl), 3.58 (q, J=5.6Hz, 2H, Hc2).

13 C NMR (DMSO) δ: 156.6 (Cc3), 143.9(Cc5), 107.8 (Cc4), 44.1(Ccl), 43.5 (Cc2). HRMS-ESI(m/z) calculated for C 7 Hi 0 N 2 Cl [M+H] + : 157.0527; found: 157.0541. 4-(3-phenylpropylamino)-7-((l-(2-(pyridin-4-ylamino)ethyl)pi peridin-4-yl) methoxy)quinazoline (Compound J)

To a solution of 45 (lOmg; 27μιηο1), K 2 C0 3 (15mg; 109μmol) and a catalytic amount of KI in DMF (0.5mL) was added 50 (13mg, 68μιηο1). The mixture was stirred at 65°C overnight. The mixture was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— »10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound J (9.2mg, 19μιηο1, 69%>) as a white powder.

1H NMR (500MHz; CDC1 3 ) δ 8.57 (s, IH, Hal), 8.18 (dd, J=1.5, 4.8Hz, 2H, Hc5), 7.32-7.28 (m, 3H, Ha4 and Hal3), 7.25-7.20 (m, 3H, Hal5 and Hal4), 7.12 (d, J=2.5Hz, IH, Ha7), 7.00 (dd, J=2.5, 9.0Hz, IH, Ha5), 6.44 (dd, J=1.5, 4.8Hz, 2H, Hc4), 5.96 (brt, J=5.2, IH, HNHa), 4.85(brt, J=5.1Hz, IH, HNHc), 3.94 (d, J=6.0Hz, 2H,Hbl), 3.70 (q, J=6.8Hz, 2H, Ha9), 3.19 (q, J=5.5Hz, 2H, Hc2), 2.95 (brd, J=11.4Hz, 2H, Hb4eq), 2.79 (t, J=7.3Hz, Hal l), 2.62 (t, J=6.2Hz, 2H, Hcl), 2.09 (quint, J=6.8Hz, 2H,HalO), 2.05 (dt, J=2.1, 9.8Hz, 2H, Hb4ax), 1.90 (m, IH, Hb2), 1.87 (d, J=12.2Hz, 2H, Hb3eq), 1.44 (dq, J=2.9-12.2Hz, 2H, Hb3ax).

1 3 C NMR (125MHz, CDC1 3 ) δ 162.2 (Ca6), 159.0 (Ca2), 155.9 (Cal), 153.3 (Cc5),

151.5 (Ca8), 150.5 (Cc4), 149.9 (Cc3), 141.5 (Cal2), 128.6 (Cal3), 128.4 (Cal4), 126.1 (Cal5), 121.8 (Ca4), 117.9 (Ca5), 109.0 (Ca3), 107.8 (Ca7), 107.6 (Cc4), 72.7 (Cbl), 56.3(Ccl), 53.0 (Cb4), 41.1 (Ca9), 38.9 (Cc2), 35.6 (Cb2), 33.6 (Cal l), 30.7 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated for C30H37N7O [M+H] + : 497.3023; found: 497.3025.

Example 12: Compound M

a) TFA, RT, 2h, quantitative yield, b) 26, K 2 C0 3 , KI, DMF, 24h, 69%. 7-0-(piperidin-4-ylmethoxy)quinazolinone (51)

A mixture of 43 (220mg; 0.46mmol) in TFA was stirred for lh at room temperature. TFA was removed. The residue was diluted with dichloromethane and the organic phase was washed with saturated Na 2 C03. The solvent was removed and 51 was obtained as pale blue foam (155mg; 0.43mmol; 94%).

3-(2-(quinolin-4-ylamino)ethyl)-7-((l-(2-(quinolin-4-ylam ino)ethyl)

piperidin-4-yl)methoxy)quinazolinone (Compound M)

To a solution of 51 (lOOmg; 0.39mmol), K 2 C0 3 (160mg; 1.16mmol) and a catalytic amount of KI in DMF (5mL) was added 26 (187mg; 0.77mmol). The mixture was stirred at 65°C overnight and 90°C for 7 hours. The resulting mixture was filtered and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0— >10% MeOH/NH 3 ) in dichloromethane to afford Compound M as a white powder (92mg; 154μιηο1; 40%) and Compound L as a white solid (17mg; 39μιηο1; 11%).

1H NMR (500MHz; CDC1 3 ) δ 8.56 (d, J=5.5Hz, 1H, Hc5), 8.19 (d, J=8.8Hz, 1H, Ha4), 8.08 (s, 1H, Hal), 8.03 (d, J=8.5Hz, 2H, Hc8), 7.78 (d, J=8.5Hzs, 1H, Hel l), 7.65 (ddd, J=1.2, 6.7Hz, 1H, HclO), 7.47 (ddd, J=1.2, 6.7Hz, 1H, Hc9), 7.13-7.06 (m, J=2.4, 8.3Hz, 1H, Ha5 and Ha7), 6.40 (d, J=5.4Hz, 1H, Hc4), 7.05 (brs, 1H, HNH), 3.97 (d, J=2.2Hz, 2H, Hbl), 3.37 (q, J=6.5Hz, 2H, Hc2), 3.02 (brd, J=l l . lHz, 2H, Hb4eq), 2.80 (t, J=5.6Hz, 2H, Hcl), 2.15 (brt, J=11.5Hz, 2H, Hb4ax), 2.00-1.87 (m, 3H, Hb2 and Hb3eq), 1.54-1.41 (m, 2H, Hb3ax).

13 C NMR (125MHz, CDC1 3 ) δ 164.3 (Ca6), 162.4 (Ca2), 151.2 (Ca8), 150.4 (Ca3), 150.0 (Cc5), 147.1 (Cc6), 144.3 (Cal), 129.6 (Cel l), 128.8 (CclO), 127.9 (Ca4), 125.0 (Cc9), 120.0 (Cc8), 118.6 (Cc7), 117.6 (Ca5), 115.9 (Ca3), 109.0 (Ca7), 98.8 (Cc4), 72.8 (Cbl), 58.4 (Ccl), 55.8 (Cb4), 52.9 (Cc2), 39.2 (Cb2), 29.1 (Cb3).

HRMS-ESI (m/z) calculated for C 2 5H27N 5 0 2 [M+H] + : 430.5216; Found: 430.5223.

c4 H c1

M

1H NMR (500MHz; DMSO) δ 8.40 (d, J=5.3Hz, 2H, Hc5 and Hal4), 8.15 (d, J=8.2Hz, IH, Ha4), 8.14 (s, IH, Hal), 8.16 (d, J=8.8Hz, 2H, Hc8 and Hal7), 7.79 (m, 2H, Hel l and Ha20), 7.61 (m, 2H, HclO and Hal9), 7.42 (m, 2H, Hc9 and Hal 8), 7.37 (brt, J=6.2Hz, HNH), 7.14 (dd, J=2.4, 8.3Hz, IH, Ha5), 7.07 (d, J=7.07Hz, IH, Ha7), 7.05 (brt, J=5.5Hz, IH, HNH), 6.66 (d, J=5.3Hz, IH, Hal3), 6.47 (d, J=5.3Hz, IH, Hc4), 4.22 (t, J=6.0Hz, 2H, Ha9), 3.99 (d, J=2.0Hz, 2H, Hbl), 3.69 (q, J=5.8Hz, 2H, HalO), 3.41 (q, J=6.3Hz, 2H, Hc2), 3.00 (brd, J=11.3Hz, 2H, Hb4eq), 2.68-2.60 (m, 3H, Hcl), 2.07 (t, J=11.0Hz, 2H, Hb4ax), 1.85-1.72 (m, 3H, Hb2 and Hb3eq), 1.37 (m, 2H, Hb3ax).

1 3 C NMR (125MHz, DMSO) δ 163.8 (Ca6), 160.6 (Ca2), 151.2 (Cc5 and Cal4),

150.7 (Ca8), 150.2 (Cc3), 149.9 (Cal2), 149.0 (Cal), 148.8 (Cc6), 148.7 (Cal5), 129.6 (Ccl l),129.5 (Ca20), 129.2 (CclO), 129.1 (Cal9), 128.1 (Ca4), 124.5 (Cal 8), 124.3 (Cc9),

121.8 (Cc8),121.8 (Cal7), 119.3 (Cal6), 119.2 (Cc7), 117.2 (Ca5), 115.4 (Ca3), 109.2 (Ca7), 98.8 (Cal3), 98.7 (Cc4), 72.9 (Cbl), 56.6 (Ccl), 53.4 (Cb4), 44.8 (Ca9), 41.1 (Cc2), 40.6 (CalO), 35.7 (Cb2), 28.9 (Cb3).

HRMS-ESI (m/z) calculated for C 3 4H38N 5 0 2 [M+H] + : 600.3080; Found:600.3076.

Example 13: Compound S

a) 25, NaH, DMF, 110°C, 4h, 54%. b) i) POCl 3 , triazole, TEA, MeCN, RT, 18h. ii) 3- Phenylpropylamine, TEA, DMF, RT, 2h, 45%. 7-(2-(quinolin-4-ylamino)ethoxy)quinazolinone (56)

To a mixture of 25 (143mg; 760μιηο1) in DMF (3mL) at 0°C under argon was added sodium hydride (91mg; 3.81mmol). The mixture was stirred for lOmin at 0°C then 42 (250mg. 1.52mmol) was added. The mixture was stirred at 110°C for 4h. The reaction mixture was diluted with ethyl acetate and washed with water. The aqueous phase was filtered and the precipitate was dried to afford 56 as a white powder (136mg; 409 μιηοΐ; 54%).

1H NMR (500MHz; DMSO) δ 8.43 (d, J=5.3Hz, 1H, Hc5), 8.24 (dd, J=0.7, 8.5Hz, 1H, Hc8), 8.06 (s, 1H, Hal), 8.03 (d, J=8.7Hz, 1H, Ha4), 7.79 (dd, J=0.7, 8.5Hz, 1H, Hcl 1), 7.61 (ddd, J=1.2, 6.7, 8.1Hz, 1H, HclO), 7.43 (ddd, J=1.2, 6.7, 8.2Hz, 1H, Hc9), 7.37 (brt, J=6.0Hz, 1H, HNHc), 7.15 (d, J=2.6Hz, 1H, Ha7), 7.13 (dd, J=2.4, 8.9Hz, 1H, Ha5), 6.61 (d, J=5.4Hz, 1H, Hc4), 4.42 (t, J=5.5Hz, 2H, Hcl), 3.75 (q, J=5.5Hz, 2H, Hc2).

13 C NMR (125MHz, DMSO) δ 163.5 (Ca6), 160.7 (Ca2), 151.4 (Ca8), 151.2 (Cc5), 150.2 (Cc3), 148.8 (Cc6), 146.5 (Cal), 129.5 (Cel l), 129.2 (CclO), 128.0 (Ca4), 124.3 (Cc9), 122.1 (Cc8), 119.2(Cc7), 116.9 (Ca5), 116.5 (Ca3), 109.5 (Ca7), 98.9 (Cc4), 66.6 (Ccl), 42.2 (Cc2).

HRMS-ESI (m/z) calculated for C19H17N4O2 [M+H] + : 333.1346; Found: 333.1350. 4-(3-phenylpropylamino)-7-(2-(quinolin-4-ylamino)ethoxy)quin azoline

(Compound S)

To a solution of 56 (50mg; 13μmol), TEA (26mg; 26μmol) in DMF (1 mL) was added 3-phenylpropylamine (35mg; 26μmol). The mixture was stirred at room temperature overnight. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound S as a white powder (25mg; 56μιηο1; 45%>).

1H NMR (500MHz; DMSO) δ 8.44 (d, J=5.3Hz, IH, Hc5), 8.39 (s, IH, Hal), 8.25 (dd, J=0.9, 8.3Hz, IH, Hc8), 8.15 (m, lH,Ha4), 8.10 (brt, J=5.3Hz, IH, HNH), 7.79 (dd, J=0.9, 8.5Hz, IH, Hel l), 7.61 (ddd, J=1.2, 6.9, 8.0Hz, IH, HclO), 7.43 (ddd, J=1.2, 6.9, 8.0Hz, IH, Hc9), 7.36 (brt, J=5.2Hz, IH, HNHc), 7.32-7.22 (m, 4H, Hal4 and Hal3), 7.17 (m, IH, Hal5), 7.15-7.10 (m, 2H, Ha7 and Ha5), 6.62 (d, J=5.4Hz, IH, Hc4), 4.41 (t, J=5.6Hz, 2H, Hcl), 3.75 (q, J=5.4Hz, 2H, Hc2), 3.52 (q, J=5.9Hz, 2H, Ha9), 2.67 (t, J=7.8Hz, 2H, Hal l), 1.95 (quint, J=7.8Hz, 2H, HalO).

13 C NMR (125MHz, DMSO) δ 161.9 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8), 151.2 (Cc5), 150.3 (Cc3), 148.8 (Cc6), 140.9 (Cal2), 129.5 (Cc8), 129.2 (CclO), 128.8 (Cal3), 128.7 (Cal4), 126.2 (Cal5), 124.3 (Cc9), 122.1 (Ca4), 118.8 (Cel l), 119.2 (Cc7), 117.1 (Ca5), 109.7 (Ca3), 108.0 (Ca7), 98.9 (Cc4), 66.4 (Ccl), 42.3 (Cc2), 40.5 (Ca9), 33.1 (Cal l), 30.8 (CalO).

HRMS-ESI (m/z) calculated for C 2 8H 2 8N 5 0 [M+H] + : 450.2288; Found: 450.2287.

Example 14: Compound R

a) 4-(2-Chloroethyl)benzene, K 2 C0 3 , KI, DMF, 65°C, 12h, 95%. b) TFA, RT, lh, 83%. c) 26, K 2 C0 3 , KI, DMF, 65°C, 12h, 28%.

3-(3-phenylpropylamino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy))quinazoline (58)

To a solution of 43 (150mg; 42μιηο1), K 2 C0 3 (115mg; 84μmol) in DMF (1.5mL) was added l-chloro-3-phenylpropane (129mg; 84μmol). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed to afford 58 as a white powder 190mg; 40μιηο1; 95%).

58

1H NMR (500MHz; DMSO) δ 8.31 (s, 1H, Hal), 8.05 (d, J=8.4Hz, 1H, Ha4), 7.31-

7.25 (m, 2H Hal3), 7.25-7.20 (m, 2H, Hal4), 7.17 (m, 1H, Hal5), 7.14-7.08 (m, 2H, Ha7 and Ha5), 4.08-3.92 (m, 5H, HNH and Hbl and Ha9), 2.78 (m, 2H, Hb4ax), 2.74 (t, J=8.5Hz, 2H, Hal l), 2.07-1.95 (m, 3H, Hb2 and HalO), 2.10-2.00 (brd, J=l lHz, 2H, Hb3eq), 1.40 (s, 9H, HBoc), 1.26-1.13 (m, 2H, Hb3ax).

1 3 C NMR (125MHz, DMSO) δ 163.7 (Ca6), 160.2 (Ca2), 154.3 (CBoc), 150.6

(Ca8), 149.0 (Cal), 1421.4 (CBoc), 128.8 (Cal3), 128.6 (Cal4), 128.1 (Ca4), 126.3 (Cal5), 117.2 (Ca5), 115.4 (Ca3), 109.2 (Ca7), 78.9 (CBoc), 72.6 (Cbl), 46.1 (Ca9), 45.1 (Cb4), 35.6 (Cb2), 32.6 (Cal l), 30.7 (CalO), 28.6 (Cb3), 28.5 5 (CBoc).

HRMS-ESI (m/z) calculated for CzsHssNsNaC^ [M+Na] + : 500.2520; Found: 500.2516.

3-(3-phenylpropyl)-7-(piperidin-4-ylmethoxy)quinazolinone (59)

A mixture of 58 (190mg, 40μmol) in TFA was stirred for lh30 at room temperature. TFA was removed. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford 59 as a white owder (124mg, 33μιηο1, 83%>).

59 1H NMR (500MHz; CC1 3 ) δ 8.21 (d, J=8.4Hz, 1H, Ha4), 7.93 (s, 1H, Hal), 7.31- 7.28 (m, 2H Hal 3), 7.26-7.18 (m, 3H, Hal 5 and Hal 4), 7.08-7.00 (m, 2H, Ha7 and Ha5), 4.03-3.90 (m, 5H, HNH and Hbl and Ha9), 3.56 (brd, J=9.6Hz, 2H, Hb4eq), 2.96 (m, 2H, Hb4ax), 2.74 (t, J=7.5Hz, 2H, Hal l), 2.22-2.10 (m, 3H, Hb2 and HalO), 2.10-2.00 (m, 2H, Hb3eq), 1.95-1.72 (m, 2H, Hb3ax).

13 C NMR (125MHz, CC1 3 ) δ 163.2 (Ca6), 160.6 (Ca2), 150.2 (Ca8), 147.3 (Cal), 128.6 (Cal3), 128.4 (Ca4), 128.3 (Cal4), 126.2 (Cal5), 117.2 (Ca5), 115.9 (Ca3), 108.7 (Ca7), 71.6 (Cbl), 46.5 (Ca9), 43.6 (Cb4), 34.1 (Cb2), 32.7 (Cal l), 30.5 (CalO), 25.6 (Cb3).

HRMS-ESI (m/z) calculated for C23H28N3O2 [M+H] + : 378.2176; Found: 378.2173.

3-(3-phenylpropyl)-7-((l-(2-(quinolin-4-ylamino)ethyl)pip eridin-4-yl)

methoxy)quinazolinone (Compound R)

To a solution of 59 (124mg; 0.33mmol), K 2 C0 3 (91mg; 0.66mmol) and a catalytic amount of KI in DMF (1.5mL) was added 26 (80mg; 0.33mmol). The mixture was stirred at 65°C for 24 hours. The solvent was removed and the residue was purified by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound R as a white owder (50mg; 91μιηο1, 28%>).

R

1H NMR (500MHz; DMSO) δ 8.40 (d, J=5.5Hz, 1H, Hc5), 8.35 (s, 1H, Hal), 8.16

(d, 8.4Hz, 1H, Hc8), 8.05 (d, J=8.4Hz, 1H, Ha4), 7.79 (dd, J=0.7, 8.5Hz, 1H, Hel l), 7.61 (ddd, J=1.2, 6.8, 8.2Hz, 1H, HclO), 7.43 (ddd, J=1.2, 6.9, 8.2Hz, 1H, Hc9), 7.31-7.25 (m, 2H, Hal3), 7.25-7.20 (m, 3H, Hal5 and Hal4), 7.14-7.08 (m, 2H, Ha7 and Ha5), 7.04 (brt, J=4.9Hz, 1H, HNH), 6.47 (d, J=5.4Hz, 1H, Hc4), 4.03-3.95 (m, 5H, HNH and Hbl and Ha9), 3.40 (m, 2H, Hc2), 3.00 (brd, J=11.0Hz, 2H, Hb4eq), 2.68-2.60 (m, 4H, Hal l and Hcl), 2.10-1.96 (m, 4H, Hb4ax and HalO), 1.85-1.72 (m, 3H, Hb2 and Hb3eq), 1.37 (dq, J=2.6, 11.5Hz, 2H, Hb3ax). 1J C NMR (125MHz, DMSO) δ 163.7 (Ca6), 160.2 (Ca2), 151.2 (Cc5), 150.7 (Ca8), 150.2 (Cc3 ), 149.0 (Cal), 148.8 (Cc6), 141.5 (Cal2), 129.5 (Cel l), 129.1 (CclO), 128.7 (Cal3), 128.6 (Cal4), 128.1 (Ca4), 126.3 (Cal5), 124.3 (Cc9), 121.8 (Cc8), 119.2 (Cc7), 117.2 (Ca5), 115.4 (Ca3), 109.2 (Ca7), 98.7 (Cc4), 72.9 (Cbl), 56.6(Ccl), 53.4 (Cb4), 46.0 (Ca9), 40.5 (Cc2), 35.7 (Cb2), 32.6 (Cal 1), 30.7 (CalO), 29.2 (Cb3).

HRMS-ESI (m/z) calculated for C 3 4H38N 5 0 2 [M+H] + : 548.3020; Found: 548.3026.

Example 15: Compounds AA to AU

Compounds 60 to 71 were synthesized following the general procedure below from compound 43.

General rocedure for compounds 60 to 71.

To a solution of triazole (0.93mmol) and POCl 3 (0.32mmol) in 0.7 mL of acetonitrile at 0°C was added TEA (130μΕ) dropwise. The reaction mixture was stirred at 0°C for 40min then 30min at room temperature. 43 (0.14mmol) was added and the mixture was vigorously stirred at room temperature overnight. The reaction was followed by TLC using ethyl acetate as eluent. The mixture was refluxed for lh to reach completion. After complete consumption of the starting material, the solvent was removed and the residue was taken off with ethyl acetate and washed with water and brine, and dried over sodium sulfate. The solvent was removed and the residue was solubilized in DMF (0.5mL). The desired amine (0.23mmol) and TEA (39μί; 0.28mmol) were added and the mixture was stirred for 3h at room temperature. The mixture was diluted with ethyl acetate and washed with water, brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ethyl acetate (0 to 100% AcOEt) in cyclohexane to afford 60 to 71. 4-(2-(3-chlorophenyl)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline 60) (70 mg ; 0.14 mmol, quantitative):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.18 (brt, J=5.5Hz, IH, HNH), 8.10 (d, J=9.1Hz, IH, Ha4), 7.35-7.19 (m, 4H, Hal2, Hal3 et Hal4), 7.11 (dd, J=2.5, 9.1Hz, IH, Ha5), 7.06 (d, J=2.5Hz, IH, Ha7), 4.03-3.93 (m, 2H, Hb4eq), 3.99 (d, J=6.3Hz, 2H, Hbl), 3.77-3.71 (m, 2H, Ha9), 2.97 (brt, J=7.0Hz, 2H, HalO), 2.85-2.68 (m, 2H, Hb4ax), 2.02-1.93 (m, IH, Hb2), 1.81-1.74 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.20 (dq, J=4.9, 13.1Hz, 2H, Hb3ax)

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 142.7 (Cal l), 133.3 (Cal6), 130.5 (Cal5), 129.0 (Cal3), 127.9 (Cal2), 126.5 (Cal4), 124.61 (Ca4), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 42.02 (Ca9), 35.6 (Cb2), 34.5 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 497.2319 [M+H] + ; found: 497.2342.

4-(2-(2-chlorophenyl)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline (61) (54 mg, 0.11 mmol, 88%):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.17 (brt, J=5.5 Hz, IH, HNH), 8.09 (d, J=9.3 Hz, IH, Ha4), 7.36-7.26 (m, 4H, Hal2,Hal3 et Hal4), 7.11 (dd, J=2.7, 9,12 Hz, IH, Ha5), 7.06 (d, J=2.3Hz, IH, Ha7), 4.04-3.93 (m, 2H, Hb4eq), 3.99 (d, J=6.3Hz, 2H, Hbl), 3.75-3.69 (m, 2H, Ha9), 2.95 (brt, J=7.3Hz, 2H, Hal O), 2.83-2.68 (m, 2H, Hb4ax), 2.02-1.92 (m, IH, Hb2), 1.81-1.73 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.20 (dq, J=3.8, 12.3Hz, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 139.1 (Cal l), 131.14 (Cal6), 131 (Cal5), 128.7 (Cal 3), , 128.7 (Cal4), 127.7 (Cal2), 124.6 (Ca4), 1 17.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 43.3 (Cb4), 42.2 (Ca9), 35.6 (Cb2), 34.3 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 497.2319 [M+H] + ; found: 497.2325 .

4-(2-(4-chlorophenyl)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinaz ol, quantitatif) :

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.21 (brt, J=5.74 Hz, IH, HNH), 8.10 (d, J=9.20 Hz, IH, Ha4), 7.47-7.31 (m, 4H, Hal2,Hal3 et Hal4), 7.1 1 (dd, J=2.47, 8.94 Hz, IH, Ha5), 7.06 (d, J=2.65, IH, Ha7), 4.03-3.94 (m, 2H, Hb4eq), 3.98 (d, J=6.41Hz, 2H, Hbl), 3.80-3.71 (m, 2H, Ha9), 3.09 (brt, J=7.40 Hz, 2H, Hal O), 2.75-2.60 (m, 2H, Hb4ax), 2.02-1.92 (m, IH, Hb2), 1.81-1.73 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.20 (dq, J=3.94, 12.45 Hz, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 138.2 (Cal l), 131.6 (Cal4), 129.6 (Cal2), 127.7 (Cal3), 124.7 (Ca4), 1 17.3 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 43.1 (Cb4), 42.4 (Ca9), 35.6 (Cb2), 32.8 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 497.2319 [M+H] + ; found: 497.2318.

4-(2-(4-sulfonamidophenyl)ethylamino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy)) quinazoline (63) (53 mg, 98 μηιοΐ, 70%): a15

1H NMR (500 MHz ; DMSO) δ 8.42 (s, IH, Hal), 8.21 (brt, J=5.8 Hz, IH, HNH), 8.10 (d, J=9.1 Hz, IH, Ha4), 7.77-7.73 (m, 2H, Hal3), 7.47-7.43 (m, 2H, Hal2), 7.29 (brs, 2H, Hal5), 7.12 (dd, J=2.6, 9.1 Hz, IH, Ha5), 7.07 (d, J=2.5Hz, IH, Ha7), 4.02-3.94 (m, 2H, Hb4eq), 3.99 (d, J=6.5Hz, 2H, Hbl), 3.76 (q, J=6.5Hz, 2H, Ha9), 3.04 (brt, J=7.4 Hz, 2H, HalO), 2.84-2.68 (m, 2H, Hb4ax), 2.02-1.92 (m, IH, Hb2), 1.81-1.74 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.13 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 144.3 (Cal l), 142.6 (Cal4), 129.6 (Cal2), 126.2 (Cal3), 124.6 (Ca4), 117.3 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 42 (Ca9), 35.6 (Cb2), 34.7 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 542.2437 [M+H] + ; found: 542.2445 .

4-(2-(4-nitrophenyl)ethylamino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy)) quinazoline (64 (68 mg, 132 μηιοΐ, 93%):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.20 (brt, J=5.6 Hz, IH, HNH), 8.18-8.14 (m, 2H, Hal3), 8.12 (d, J=9.2 Hz, IH, Ha4), 7.56-7.52 (m, 2H, Hal2), 7.11 (dd, J=2.5, 9.0 Hz, IH, Ha5), 7.06 (d, J=2.5Hz, IH, Ha7), 4.06-3.96 (m, 2H, Hb4eq), 3.98 (d, J=6.3Hz, 2H, Hbl), 3.74-3.67 (m, 2H, Ha9), 2.94-2.88 (m, 2H, HalO), 2.88-2.72 (m, 2H, Hb4ax), 2.85 (q, J=6.69 Hz, 1H, Hal5) 2.02-1.94 (m, 1H, Hb2), 1.81-1.75 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.27-1.13 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 146.5 (Cal l), 137.4 (Cal4), 130.6 (Cal2), 124.7 (Ca4), 123.8 (Cal3), 117.2 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 43.2 (Cb4), 42.5 (Ca9), 35.6 (Cb2), 34.7 (CalO), 28.8 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 508.2559 [M+H] + ; found : 508.2565.

4-(2-(4-isopropylphenyl)ethylamino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy)) quinazoline (65) (44 mg, 87 μηιοΐ, 62%):

a16

1H NMR (500 MHz ; DMSO) δ 8.41 (s, 1H, Hal), 8.20 (brt, J=5.6 Hz, 1H, HNH), 8.10 (d, J=9.1 Hz, 1H, Ha4), 7.22-7.14 (m, 4H, Hal2,Hal3), 7.11 (dd, J=2.6, 9.1 Hz, 1H, Ha5), 7.06 (d, J=2.6Hz, 1H, Ha7), 4.0-3.96 (m, 2H, Hb4eq), 3.98 (d, J=6.4Hz, 2H, Hbl), 3.80 (q, J=5.9Hz, 2H, Ha9), 3.11 (brt, J=7.2Hz, 2H, HalO), 2.85-2.67 (m, 2H, Hb4ax), 2.03- 1.91 (m, 1H, Hb2), 1.81-1.73 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.13 (m, 8H, Hb3ax et Hal6).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.7 (Ca8), 146.5 (Cal4), 137.3 (Cal l), 129.6 (Cal2), 129 (Cal3), 124.7 (Ca4), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 43.2 (Cb4), 42.2 (Ca9), 35.6 (Cb2), 34.8 (CalO), 33.5 (Cal5), 28.8 (Cb3), 28.6 (CBoc), 24.4 (Cal6).

HRMS-ESI (m/z) calculated: 505.3178 [M+H] + ; found: 505.3182.

4-(2-(4-methoxyphenyl)ethylamino)-7-(0-(( V-Boc)piperidin-4-ylmethoxy)) quinazoline (66) (49 mg, 101 μηιοΐ, 71%): a15

1H NMR (500 MHz ; DMSO) δ 8.42 (s, IH, Hal), 8.16 (brt, J=5.7 Hz, IH, HNH), 8.10 (d, J=9.2 Hz, IH, Ha4), 7.20-7.15 (m, 2H, Hal2), 6.88-6.83 (m, 2H, Hal3 ), 7.1 1 (dd, J=2.6, 9.1 Hz, IH, Ha5), 7.06 (d, J=2.5Hz, IH, Ha7), 4.06-3.94 (m, 2H, Hb4eq), 3.99 (d, J=6.3 Hz, 2H, Hbl), 3.72 (s, 3H, Hal5), 3.71-3.64 (m, 2H, Ha9), 2.88(brt, J=7.1 Hz, 2H, HalO), 2.84-2.67 (m, 2H, Hb4ax), 2.04-1.93 (m, IH, Hb2), 1.83-1.74 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.12 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 158.1 (Cal4), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 144.3 (Cal l), 130.2 (Cal2), 124.6 (Ca4), 117.2 (Ca5), 114.2 (Cal3), 109.6 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 55.4 (Cal5), 43.4(Cb4), 42.7 (Ca9), 35.6 (Cb2), 34.2 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 493.2814 [M+H] + ; found: 493.2825.

4-(2-(4-aminophenyl)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline (67) (67 mg, 0.14 mmol, quantitative):

1H NMR (500 MHz ; DMSO) δ 8.40 (s, IH, Hal), 8.14 (brt, J=5.5 Hz, IH, HNH), 8.12 (d, J=9.3 Hz, IH, Ha4), 7.10 (dd, J=2.6, 9.6 Hz, IH, Ha5), 7.05 (d, J=2.5Hz, IH, Ha7), 6.93-6.88 (m, 2H, Hal2), 6.52-6.47 (m, 2H, Hal3 )4.85 (s, 2H, Hal5), 4.06-3.92 (m, 2H, Hb4eq), 3.99 (d, J=6.5Hz, 2H, Hbl), 3.67-3.57 (m, 2H, Ha9), 2.75(brt, J=7.5 Hz, 2H, HalO), 2.84-2.68 (m, 2H, Hb4ax), 2.03-1.92 (m, IH, Hb2), 1.82-1.73 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.14 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162 (Ca6), 159.4 (Ca2), 156.1 (Cal), 154.3 (CBoc), 151.8 (Ca8), 147.2 (Cal4), 129.6 (Cal2), 129.3 (Cal l), 114.4 (Cal3), 124.7 (Ca4), 117.1 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 43.1 (Cb4), 42.9 (Ca9), 35.6 (Cb2), 34.4 (CalO), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 478.2818 [M+H] + ; found: 478.2831.

4-(2-(quinolinylamino)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline 68) (52 mg, 98 μηιοΐ, 70%):

1H NMR (500 MHz ; DMSO) δ 8.50 (s, IH, Hal), 8.42 (d, J= 5.3Hz, IH, Hal3), 8.16 (dd, J=0.7, 8.2 Hz, IH, Hal8), 8.10 (d, J=9.2Hz, IH, Ha4), 7.79 (dd, J=0.9, 8.5Hz, IH, Hal5), 7.61(ddd, J=0.9, 6.9, 9.3Hz, IH, Hal6), 7.45-7.38 (m, 2H, Hal7 and HNH), 7.14 (dd, J=2.5, 9.2 Hz, IH, Ha5), 7.09 (d, J=2.6Hz, IH, Ha7), 6.69(d, J=5.5Hz, IH, Hal2), 4.01-3.94 (m, 4H, Hbl and Hb4eq), 3.81 (q, J=6.3Hz, 2H, Ha9), 3.58 (q, J=6.2Hz, 2H, HalO), 2.03-1.93 (m, IH, Hb2), 1.83-1.73 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.15 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.7 (Ca2), 156.0 (Cal), 154.3 (CBoc), 151.8 (Ca8), 151.2 (Cal3), 150.3 (Cal l), 148.9 (Cal4), 129.5 (Cal5), 129.1 (Cal6), 124.7 (Ca4), 124.4 (Cal7), 121.9 (Cal8), 119.3 (Cal9), 117.4 (Ca5), 109.6 (Ca3), 108.0 (Ca7), 80.0 (CBoc), 72.4 (Cbl), 42.1 (CalO), 39.3 (Ca9), 35.6 (Cb2), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 529.2922 [M+H] + ; found: 529.2928.

4-(2-(naphtylamino)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline (69) (59 mg, 112 μηιοΐ, 79%):

1H NMR (500 MHz ; DMSO) δ 8.51 (s, IH, Hal), 8.42 (brt, J=5.1 Hz, IH, HNH), 8.14 (d, J=9.2 Hz, IH, Hal 9), 8.09 (d, J=7.8Hz, IH, Ha4), 7.76 (dd, J=1.8, 6.9Hz, IH, Hal5), 7.46-7.35 (m, 2H, Hal7 and Hal8), 7.31 (t, J=7.9Hz, IH, Hal3), 7.15 (dd, J=2.5, 9.1 Hz, IH, Ha5), 7.11 (d, J=8.1Hz, IH, Hal4), 7.08 (d, J=2.6Hz, IH, Ha7), 6.71(d, J=7.5Hz, IH, Hal2), 6.45 (brt, J=5.1Hz, HNH), 4.05-3.91 (m, 4H, Hbl and Hb4eq), 3.85 (q, J=6.3Hz, 2H, Ha9), 3.51 (brt, J=6.1Hz, 2H, HalO), 2.87-2.67 (m, 2H, Hb4ax), 1.98-1.92 (m, IH, Hb2), 1.81-1.72 (m, 2H, Hb3eq), 1.25-1.15 (m, 2H, Hb3ax).

13, C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.8 (Ca2), 155.9 (Cal), 154.3 (Ca8), 151.5 (Cal l), 144.3 (Cal l), 134.5 (Ca21), 128.4 (Cal5), 127.6 (Cal3), 126.1 (Cal7), 124.8 (Cal9), 124.7 (Ca4), 124.5 (Cal 8), 123.3 (Ca20), 121.8 (Ca4), 117.4 (Ca5), 115.8 (Cal4), 109.5 (Ca3), 107.8 (Ca7), 103.3 (Cal2), 79.0 (CBoc), 72.1 (Cbl), 46.2 (Cb4), 43.2 (CalO), 39.4 (Ca9), 35.6 (Cb2), 28.7 (Cb3), 28.6 (CBoc).

HRMS-ESI (m/z) calculated: 528.2969 [M+H] ; found: 528.3002.

4-(2-(phenylamino)ethylamino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline 70) (50 mg, 105 μηιοΐ, 75%):

1H NMR (500 MHz ; DMSO) δ 8.42 (s, IH, Hal), 8.21 (brt, J=5.6 Hz, IH, HNH), 8.09 (d, J=9.0Hz, IH, Ha4), 7.15 (dd, J=2.7, 9.1 Hz, IH, Ha5), 7.11-7.05 (m, 3H, Ha7 and Hal 3), 6.64(dd, J=0.9, 8.6Hz, 2H, Hal2), 6.53 (dt, J=0.9, 7.3Hz, 2H, Hal4), 5.78 (brt, J=5.9Hz, HNH), 4.06-3.91 (m, 4H, Hbl and Hb4eq), 3.67 (q, J=6.5Hz, 2H, Ha9), 3.30 (brt, J=6.51Hz, 2H, Hal O), 2.85-2.67 (m, 2H, Hb4ax), 2.03-1.93 (m, 1H, Hb2), 1.82-1.75 (m, 2H, Hb3eq), 1.41 (s, 9H, HBoc), 1.26-1.15 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.6 (Ca2), 156.0 (Cal), 154.3 (CBoc), 151.8 (Ca8), 149,1 (Cal l), 129.4 (Cal3), 124.7 (Ca4), 1 17.3 (Ca5), 1 16.0 (Cal4), 1 12.4 (Cal2), 109.6 (Ca3), 108.0 (Ca7), 79.0 (CBoc), 72.3 (Cbl), 42.3 (CalO), 40.2 (Ca9), 35.6 (Cb2), 28.7 (Cb3), 28.56 (CBoc).

HRMS-ESI (m/z) calculated: 478.2813 [M+H] + ; found: 478.2819.

4-(( V-(l-benzylpiperidin-4-yl))amino)-7-(0-(( V-Boc) piperidin-4-ylmethoxy)) quinazoline 71) (60 mg, 1 13 μηιοΐ, 81%):

1H NMR (500 MHz ; DMSO) δ 8.36 (s, 1H, Hal), 8.28 (brs, 1H, HNH), 8.21 (d, J=9.2Hz, 1H, Ha4), 7.73 (d, J=7.6 Hz, 1H, Ha7), 7.37-7.22 (m, 5H, Hal4, Hal5 and Hal6), 7.10 (dd, J=2.6, 9.0Hz, 1H, Ha5) , 7.04 (d, J=2.7Hz, 1H, Ha7), 4.16- 4.10 (m, 1H, Ha9), 4.06-3.86 (m, 3H, Hb l and Hb4eq), 3.49 (s, 2H, Hal2), 2.86 (brd, J=1 1.8Hz, 2H, Hal leq), 2.81-2.67 (m, 2H, Hb4ax), 2.06 (dd, J=1.7, 1 1.8Hz, 2H, Hal lax), 2.02-1.92 (m, 1H, Hb2), 1.90 (brd, J=12.4Hz, 2H, HalOeq), 1.78 (brd, J=13.4Hz, 2H, Hb3eq), 1.64 (dd, J=3.6, 12.4Hz, 2H, HalOax), 1.41 (s, 9H, HBoc), 1.19 (dd, J=4.6, 13.4Hz, 2H, Hb3ax)

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 158.8 (Ca2), 156.0 (Cal), 154.3 (CBoc), 151.9 (Ca8), 139,2 (Cal 3), 129.1 (Cal4), 128.6 (Cal 5), 127.3 (Cal5), 124.97 (Ca4), 1 17.4 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 78.9 (CBoc), 72.3 (Cbl), 62.6 (Cal2), 52.8 (Cal l), 48.2(Ca9), 35.7 (Cb2), 31.7 (CalO), 28.7 (Cb3), 28.5 (CBoc).

HRMS-ESI (m/z) calculated: 532.3282 [M+H] + ; found: 532.3296.

Compounds 72 to 83 were synthesized following the general procedure below from compounds 60 to 71 respectively.

60 to 71 72 to 83

A mixture of the desired compound 60 to 71 in TFA was stirred for lh at room temperature. TFA was removed. The residue was diluted with dichloromethane and the organic phase was washed with saturated Na 2 C03. The solvent was removed and gave respectively compound 72 to 83.

4-(2-(3-chlorophenyl)ethylamino)-7-0-(piperidin-4-ylmetho xy)quinazoline mg, 0.14 mmol quantitative) from 60 (0.14mmol):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.21 (brt, J=5.3 Hz, IH, HNH),

8.12 (d, J=9.1 Hz, IH, Ha4), 7.35-7.20 (m, 4H, Hal2, Hal3 et Hal4), 7.11 (dd, J=2.6, 9,0 Hz, IH, Ha5), 7.07 (d, J=2.54Hz, IH, Ha7), 3.96 (d, J=6.4Hz, 2H, Hbl), 3.77-3.71 (m, 2H, Ha9), 3.11-3.06 (m, 2H, Hb4eq) 2.97 (brt, J=7.1Hz, 2H, HalO), 2.68-2.60 (m, 2H, Hb4ax), 2.00-1.89 (m, IH, Hb2), 1.83-1.74 (m, 2H, Hb3eq), 1.30 (dq, J=2.9, 12.2Hz, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8),

142.7 (Cal l), 133.3 (Cal6), 130.6 (Cal5), 129.0 (Cal3), 127.9 (Cal2), 126.6 (Cal4) 124.7 (Ca4), 117.2 (Ca5), 109.6 (Ca3), 108 (Ca7), 72.7 (Cbl), 45.1 (Cb4), 42 (Ca9), 34.9 (Cb2), 34.5 (CalO), 28.5 (cb3).

HRMS-ESI (m/z) calculated: 397.1795 [M+H] + ; found: 397.1799.

4-(2-(2-chlorophenyl)ethylamino)-7-0-(piperidin-4-ylmethoxy) quinazoline (73)

(52 mg, 0.13 mmol, 93%) from 61 (0.1 lmmol):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.19 (brt, J=5.6 Hz, IH, HNH), 8.11 (d, J=9.1Hz, IH, Ha4), 7.37-7.22 (m, 4H, Hal2, Hal3 et Hal4), 7.10 (dd, J=2.7, 9.9 Hz, IH, Ha5), 7.06 (d, J=2.5Hz, IH, Ha7), 3.95 (d, J=6.4Hz, 2H, Hbl), 3.75-3.69 (m, 2H, Ha9), 3.07-3.01 (m, 2H, Hb4eq) 2.95 (brt, J=7.2 Hz, 2H, HalO), 2.62-2.54 (m, 2H, Hb4ax), 1.97-1.87 (m, IH, Hb2), 1.82-1.72 (m, 2H, Hb3eq), 1.32-1.20 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8), 139.1 (Cal l), 131.1 (Cal6), 131 (Cal5), 128.7 (Cal3), 128.7 (Cal4), 126.6 (Cal2) 126.6 (Ca4), 117.2 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 72.9 (Cbl), 45.5 (Cb4), 42.2 (Ca9), 34.6 (Cb2), 34.3 (CalO), 29.1 (cb3).

HRMS-ESI (m/z) calculated: 397.1795 [M+H] + ; found: 397.1791.

4-(2-(4-chlorophenyl)ethylamino)-7-0-(piperidin-4-ylmethoxy) quinazoline (74) (50 mg, 0.13 mmol 93%) from 62 (0.14mmol):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.23 (m, IH, HNH), 8.11 (d, J=9.12 Hz, IH, Ha4), 7.37-7.22 (m, 4H, Hal2, Hal3 et Hal4), 7.13-7.08 (m, IH, Ha5), 7.08-7.03 (m, IH, Ha7), 3.95 (d, J=5.95 Hz, 2H, Hbl), 3.75-3.69 (m, 2H, Ha9), 3.07-3.01 (m, 2H, Hb4eq) 2.95 (brt, J=7.20 Hz, 2H, HalO), 2.62-2.54 (m, 2H, Hb4ax), 1.97-1.87 (m, IH, Hb2), 1.82-1.72 (m, 2H, Hb3eq), 1.32-1.20 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8), 139.1 (Cal l), 131.1 (Cal6), 131 (Cal5), 128.7 (Cal3), 128.7 (Cal4), 126.6 (Cal2) 126.6 (Ca4), 117.2 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 72.9 (Cbl), 45.5 (Cb4), 42.2 (Ca9), 34.6 (Cb2), 34.3 (CalO), 29.1 (cb3)

HRMS-ESI (m/z) calculated: 397.1795 [M+H] + ; found: 397.1794.

4-(2-(4-sulfonamidophenyl)ethylamino)-7-0-(piperidin-4-ylmet hoxy)

quinazoline (75) (33 mg, 75 μηιοΐ, 77%) from 63 (98μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.42 (s, IH, Hal), 8.21 (brt, J=5.45 Hz, IH, HNH), 8.10 (d, J=9.11 Hz, IH, Ha4), 7.77-7.72 (m, 2H, Hal3), 7.47-7.42 (m, 2H, Hal2), 7.37-7.21 (brs, 2H, Hal5), 7.11 (dd, J=2.38, 9.03 Hz, IH, Ha5), 7.05 (d, J=2.55 Hz, IH, Ha7), 3.94 (d, J=6.47 Hz, 2H, Hbl), 3.79-3.72 (m, 2H, Ha9), 3.04 (brt, J=7.15 Hz, 2H, HalO), 2.99-2.92 (m, 2H, Hb4eq), 2.63-2.52 (m, 2H, Hb4ax), 1.93-1.79 (m, IH, Hb2), 1.76-1.66 (m, 2H, Hb3eq), 1.26-1.12 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8), 144.3 (Cal l), 142.6 (Cal4), 129.6 (Cal2), 126.2 (Cal3), 124.6 (Ca4), 117.3 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 72.3 (Cbl), 43.2 (Cb4), 42 (Ca9), 35.6 (Cb2), 34.7 (CalO), 28.7 (Cb3).

HRMS-ESI (m/z) calculated: 397.1795 [M+H] + ; found: 397.1794.

4-(2-(4-nitrophenyl)ethylamino)-7-0-(piperidin-4-ylmethoxy)q uinazoline (76) (51 mg, 132 μιηοΐ, quantitative) from 64 (132μιηο1):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.19 (brt, J=5.40 Hz, IH, HNH), 8.18-8.14 (m, 2H, Hal3), 8.08 (d, J=9.18 Hz, IH, Ha4), 7.57-7.52 (m, 2H, Hal2), 7.11 (dd, J=2.56, 9.09 Hz, IH, Ha5), 7.06 (d, J=2.56 Hz, IH, Ha7), 3.94 (d, J=6.44 Hz, 2H, Hbl), 3.83-3.76 (m, 2H, Ha9), 3.11 (brt, J=7.20 Hz, 2H, HalO), 3.03-2.97 (m, 2H, Hb4eq), 2.61- 2.53 (m, 2H, Hb4ax), 1.94-1.83 (m, IH, Hb2), 1.77-1.69 (m, 2H, Hb3eq), 1.26-1.16 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8), 146.5 (Cal l), 137.4 (Cal4), 130.6 (Cal2), 124.7 (Ca4), 123.8 (Cal3), 117.2 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 72.3 (Cbl), 43.2 (Cb4), 42.5 (Ca9), 35.6 (Cb2), 34.7 (CalO), 28.8 (Cb3).

HRMS-ESI (m/z) calculated: 408.2035 [M+H] + ; found: 408.2024.

4-(2-(4-isopropylphenyl)ethylamino)-7-0-(piperidin-4-ylmetho xy)quinazoline (77) (34 mg, 80 μηιοΐ, 92%) from 65 (87 μηιοΐ):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.20 (brt, J=5.67 Hz, IH, HNH), 8.12 (d, J=9.17 Hz, IH, Ha4), 7.20-7.13 (m, 4H, Hal2,Hal3), 7.11 (dd, J=2.59, 9.03 Hz, IH, Ha5), 7.06 (d, J=2.73, IH, Ha7), 3.95 (d, J=6.39Hz, 2H, Hbl), 3.73-3.66 (m, 2H, Ha9), 3.05-2.97 (m, 2H, Hb4eq), 2.91 (brt, J=7.21 Hz, 2H, HalO), 2.58-2.53 (m, 2H, Hb4ax), 1.95-1.84 (m, 1H, Hb2), 1.78-1.70 (m, 2H, Hb3eq), 1.29-1.10 (m, 8H, Hb3ax et Hal6).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.7 (Ca8), 146.5 (Cal4), 137.3 (Cal l), 129.6 (Cal2), 129 (Cal3), 124.7 (Ca4), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 72.3 (Cbl), 43.2 (Cb4), 42.2 (Ca9), 35.6 (Cb2), 34.8 (CalO), 33.5 (Cal5), 28.8 (Cb3), 24.4 (Cal6).

HRMS-ESI (m/z) calculated: 405.2654 [M+H] + ; found: 405.2659.

4-(2-(4-aminophenyl)ethylamino)-7-0-(piperidin-4-ylmethoxy)q uinazoline (78) (39.0 mg, 101 μηιοΐ, quantitative) from 66 (101 μηιοΐ):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, 1H, Hal), 8.16 (brt, J=5.40 Hz, 1H, HNH), 8.11 (d, J=9.20 Hz, 1H, Ha4), 7.20-7.15 (m, 2H, Hal2), 7.11 (dd, J=2.58, 9.12 Hz, 1H, Ha5), 7.05 (d, J=2.58, 1H, Ha7), 6.88-6.83 (m, 2H, Hal3), 3.94 (d, J=6.38Hz, 2H, Hbl), 3.72 (s, 3H, Hal5) 3.71-3.65 (m, 2H, Ha9), 3.01-2.92 (m, 2H, Hb4eq), 2.88 (brt, J=7.30 Hz, 2H, HalO), 2.57-2.52 (m, 2H, Hb4ax), 1.92-1.82 (m, 1H, Hb2), 1.76-1.68 (m, 2H, Hb3eq), 1.26-1.14 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.4 (Ca2), 158.1 (Cal4), 156.1 (Cal), 151.8 (Ca8), 144.3 (Cal l), 130.2 (Cal2), 124.6 (Ca4), 117.2 (Ca5), 114.2 (Cal3), 109.6 (Ca3), 107.9 (Ca7), 72.3 (Cbl), 55.4 (Cal5), 42.7 (Ca9), 35.6 (Cb2), 34.2 (CalO), 28.7 (Cb3).

HRMS-ESI (m/z) calculated: 393.2290 [M+H] + ; found: 393.2297.

4-(2-(4-aminophenyl)ethylamino)-7-0-(piperidin-4-ylmethoxy)q uinazoline (79) (34 mg, 90 mmol, 64%) from 67 (0.14 mmol):

1H NMR (500 MHz ; DMSO) δ 8.40 (s, IH, Hal), 8.13 (brt, J=5.75 Hz, IH, HNH), 8.11 (d, J=9.58 Hz, IH, Ha4), 7.11 (dd, J=2.49, 9.10 Hz, IH, Ha5), 7.04 (d, J=2.63, IH, Ha7), 6.94-6.86 (m, 2H, Hal2), 6.52-6.46 (m, 2H, Hal3), 4.85 (s, 2H, Hal5), 3.94 (d, J=6.31 Hz, 2H, Hbl), 3.66-3.59 (m, 2H, Ha9), 3.02-2.94 (m, 2H, Hb4eq), 2.76 (brt, J=7.34 Hz, 2H, HalO), 2.58-2.52 (m, 2H, Hb4ax), 1.94-1.83 (m, IH, Hb2), 1.76-1.68 (m, 2H, Hb3eq), 1.26-1.15 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8), 147.2 (Cal4), 129.6 (Cal2), 129.3 (Cal l), 114.4 (Cal3), 124.7 (Ca4), 117.1 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 72.3 (Cbl), 43.1 (Cb4), 42.9 (Ca9), 35.6 (Cb2), 34.4 (CalO), 28.7

(Cb3).

HRMS-ESI (m/z) calculated: 378.2293 [M+H] ; found: 378.2285.

4-(2-(phenylamino)ethylamino)-7-0-(piperidin-4-ylmethoxy)qui nazoline

mg 5.3 μηιοΐ, 23%) from 68 (lOmg, 23.3μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.49 (s, IH, Hal), 8.43 (brt, J=5.7 Hz, IH, HNH), 8.42 (d, J= 5.3Hz, IH, Hal3), 8.17 (d, J=6.2 Hz, IH, Hal 8), 8.12 (d, J=9.2Hz, IH, Ha4), 7.78 (d, J=8.3 Hz, IH, Hal5), 7.60(t, J=7.2Hz, IH, HclO), 7.42 (t, J=7.4Hz, IH, Hal7), 7.13 (dd, J=2.5, 9.0Hz, IH, Ha5), 7.09 (d, J=2.5Hz, IH, Ha7), 6.82 (brt,J=5.1Hz, IH, HNH), 6.70(d, J=5.4Hz, IH, Hal2), 3.99 (d, J=5.7Hz, 2H, Hbl), 3.82 (q, J=6.1Hz, 2H, Ha9), 3.58 (q, J=6.2Hz, 2H, HalO), 3.00 (brd, 2H, Hb4eq), 2.06 (t, J=10.5Hz, 2H, Hb4ax), 1.865-1.74 (m, 3H, Hb3eq and Hb2), 1.443-1.32 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.3 (Ca6), 159.8 (Ca2), 156.0 (Cal), 151.8 (Ca8), 151.2 (Cal3), 150.3 (Cal l), 148.7 (Cal4), 129.5 (Cal5), 129.2 (Cal6), 124.7 (Ca4), 124.4 (Cal7),127.3 (Cal3), 122.0 (Cal 8), 121.8 (Ca4), 119.3 (Cal9), 117.5 (Ca5), 109.5 (Ca3), 108.0 (Ca7), 98.5 (Cal2), 72.7 (Cbl), 53.5 (Cb4), 42.1 (CalO), 39.3 (Ca9), 35.7 (Cb2), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 429.2393 [M+H] + ; found: 429.2394.

4-(2-(naphtylamino)ethylamino)-7-0-(piperidin-4-ylmethoxy)qu inazoline (81)

(47 mg, 110 ηιοΐ, 91%) from 69 (121 μηιοΐ):

1H NMR (500 MHz ; DMSO) δ 8.50 (s, IH, Hal), 8.38 (brt, J=5.05 Hz, IH, HNH), 8.12 (d, J=9.16 Hz, IH, Hal9), 8.11 (d, J=7.6Hz, IH, Ha4), 7.76 (dd, J=1.6, 7.1Hz, IH, Hal5), 7.47-7.38 (m, 2H, Hal7 and Hal8), 7.31 (t, J=7.9Hz, IH, Hal3), 7.15 (dd, J=2.8, 9.1 Hz, IH, Ha5), 7.11 (d, J=8.4Hz, IH, Hal4), 7.08 (d, J=2.5Hz, IH, Ha7), 6.71(d, J=7.6Hz, IH, H12), 6.46 (brt, J=5.3Hz, IH, HNH), 3.96 (d, J=6.8Hz, 2H, Hbl), 3.85 (q, J=6.8Hz, 2H, Ha9), 3.51 (brt, J=6.8Hz, 2H, HalO), 3.07-3.00 (m, 2H, Hb4eq), 2.62-2.54 (m, 2H, Hb4ax), 1.97-1.96 (m, IH, Hb2), 1.81-1.71 (m, 2H, Hb3eq), 1.31-1.20 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.3 (Ca6), 159.8 (Ca2), 156.0 (Cal), 151.9 (Ca8),

151.8 (Cal l), 144.3 (Cal l), 134.5 (Ca21), 128.4 (Cal5), 127.3 (Cal3), 126.1 (Cal7), 124.7 (Cal9), 124.5 (Ca4), 124.4 (Cal 8), 123.3 (Ca20), 121.9 (Ca4), 117.4 (Ca5), 115.8 (Cal4), 109.5 (Ca3), 108.0 (Ca7), 103.2 (Cal2), 72.9 (Cbl), 45.6 (Cb4), 43.2 (CalO), 39.7 (Ca9), 35.8 (Cb2), 29.2 (Cb3).

HRMS-ESI (m/z) calculated: 428.2445 [M+H] + ; found: 428.2610.

4-(2-(phenylamino)ethylamino)-7-0-(piperidin-4-ylmethoxy)qui nazoline (82)

(38mg, 101 μηιοΐ, 96%) from 70 (105μιηο1):

1H NMR (500 MHz ; DMSO) δ 8.43 (s, IH, Hal), 8.21 (brt, J=5.8 Hz, IH, HNH), 8.11 (d, J=9.2Hz, IH, Ha4), 7.12 (dd, J=2.6, 9.2 Hz, IH, Ha5), 7.11-7.05 (m, 3H, Ha7 and Hal3), 6.64 (dd, J=0.9, 8.6Hz, 2H, Hal2), 6.53 (dt, J=0.9, 7.2Hz, 2H, Hal4), 5.78 (brt, J=6.1Hz, HNH), 3.96 (d, J=6.4Hz 2H, Hbl), 3.68 (q, J=6.5Hz, 2H, Ha9), 3.31 (brt, J=6.5Hz, 2H, HalO), 3.08-2.97 (m, 2H, Hb4eq), 2.62-2.547 (m, 2H, Hb4ax), 1.97-1.86 (m, IH, Hb2), 1.81-1.72 (m, 2H, Hb3eq), 1.32-1.20 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.6 (Ca2), 156.0 (Cal), 151.8 (Ca8), 149,1 (Cal l), 129.4 (Cal3), 124.7 (Ca4), 117.3 (Ca5), 116.1 (Cal4), 112.4 (Cal2), 109.5 (Ca3), 108.0 (Ca7), 72.9 (Cbl), 45.6 (Cb4), 42.3 (CalO), 40.1 (Ca9), 35.8 (Cb2), 29.2 (Cb3).

HRMS-ESI (m/z) calculated: 378.2289 [M+H] + ; found: 378.2280.

4-(( V-(l-benzylpiperidin-4-yl))amino)-7-0-(piperidin-4-ylmethoxy ) quinazoline (83) (42 m 97μηιο1, 86%) from 71 (105μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.37 (s, IH, Hal), 8.28 (brs, IH, HNH), 8.22 (d, J=9.2Hz, IH, Ha4), 7.74 (d, J=7.6 Hz, IH, Ha7), 7.37-7.24 (m, 4H, Hal4 and Hal5), 7.28- 7.23 (m, IH, Hal6), 7.09 (dd, J=2.6, 9.1Hz, IH, Ha5), 7.04 (d, J=2.6Hz, IH, Ha7), 4.20- 4.10 (m, IH, Ha9), 3.95 (d, J=6.4Hz, 3H, Hbl), 3.49 (s, 2H, Hal2), 3.01 (brd, J=12.2Hz, 2H, Hb4eq), 2.87 (brd, J=11.7Hz, 2H, Hal leq), 2.58 (m, 2H, Hb4ax), 2.06 (dt, J=1.5, 11.7Hz, 2H, Hal lax), 1.94-1.85 (m, 3H, Hb2 and HalOeq), 1.78 (brd, J=10.5Hz, 2H, Hb3eq), 1.65 (ddd, J=3.7, 11.7Hz, 2H, HalOax), 1.23 (m, 2H, Hb3ax)

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 158.8 (Ca2), 156.0 (Cal), 151.9 (Ca8), 139,2 (Cal3), 129.1 (Cal4), 128.6 (Cal5), 127.3 (Cal5), 124.9 (Ca4), 117.0 (Ca5), 109.4 (Ca3), 107.9 (Ca7), 72.9 (Cbl), 62.6 (Cal2), 52.8 (Cal l), 48.2(Ca9), 45.7 (Cb4), 35.9 (Cb2), 31.7 (CalO), 29.4 (Cb3).

HRMS-ESI (m/z) calculated: 432.2758 [M+H] + ; found: 432.2753.

Compounds AA to AL were synthesized following the general procedure from com ounds 72 to 83 respectively.

To a solution of 0.1 M of compound 72 to 83 K 2 CO 3 (2eq) and a catalytic amount of KI in DMF was added 26 (2eq). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0→10% MeOH/NHs) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compounds AA to AL.

4-(2-(3-chlorophenyl)ethylamino)-7-((l-(2-(quinolin-4-yla mino)ethyl) piperidin- 4-yl)methoxy)quinazoline (AA) (5.0 mg, 8.8 μιηοΐ, 35%) from 72 (25 μιηοΐ):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, 1H, Hal), 8.39 (d, J= 5.37Hz, 1H, Hc5) 8.18 (brt, J=5.35 Hz, 1H, HNH), 8.15 (d, J=8.40 Hz, 1H, Hc8), 8.10 (d, J=9.03 Hz, 1H, Ha4), 7.77 (dd, J=0.89, 8.40 Hz, 1H, Hel l), 7.60(m, 1H, HclO), 7.42 (m, 1H, Hc9), 7.36- 7.18 (m, 4H, Hal2, Hal3 et Hal4), 7.1 1 (dd, J=2.6, 9,21 Hz, 1H, Ha5), 7.06 (d, J=2.53, 1H, Ha7), 7.03 (brt, J=5.42 Hz, 1H, HNH), 6.47 (d, J=5.35 Hz, 1H, Hc4), , 3.98 (d, J=5.87Hz, 2H, Hbl), 3.79-3.69 (m, 2H, Ha9), 3.44-3.36(m, 2H, Hc2), 3.00 (m, 2H, Hb4eq), 2.97 (brt, J=7.13 Hz, 2H, HalO), 2.63 (t, J=6.69 Hz, 2H, Hcl), 2.06 (m, 2H, Hb4ax), 1.85-1.72(m, 3H, Hb2 et Hb3eq), 1.45-1.30 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 142.7 (Cal l), 133.3 (Cal6), 130.6 (Cal5), 129.5 (Cel l), 129.1 (CclO), 129.0 (Cal3), 127.9 (Cal2), 126.5 (Cal4), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.6 (Cb4), 42.1 (Ca9), 40.4(Hc2), , 34.5 (Cb2), 34.5 (CalO), 29 (Cb3)

HRMS-ESI (m/z) calculated: 567.2639 [M+H] + ; found: 567.2644.

4-(2-(2-chlorophenyl)ethylamino)-7-((l-(2-(quinolin-4-yla mino)ethyl) piperidin- 4-yl)methoxy)quinazoline (AB) (4.0 mg ; 7.1 μηιοΐ, 29%) from 73 (25μηιο1): a14

1H NMR (500 MHz ; DMSO) δ 8.40 (s, IH, Hal), 8.39 (d, J= 5.8 Hz, IH, Hc5) 8.16 (brt, J=5.06 Hz, IH, HNH), 8.15 (d, J=8.1 Hz, IH, Hc8), 8.10 (d, J=9.03 Hz, IH, Ha4), 7.77 (dd, J=0.9, 8.3 Hz, IH, Hel l), 7.60 (m, IH, HclO), 7.42 (m, IH, Hc9), 7.36-7.24 (m, 4H, Hal2, Hal3 et Hal4), 7.11 (dd, J=2.47, 9,12 Hz, IH, Ha5), 7.05 (d, J=2.61, IH, Ha7), 7.03 (brt, J=5.1 Hz, IH, HNH), 6.47 (d, J=5.3 Hz, IH, Hc4), 3.98 (d, J=5.7Hz, 2H, Hbl), 3.77-3.67 (m, 2H, Ha9), 3.44-3.36(m, 2H, Hc2), 3.04-2.97 (m, 2H, Hb4eq), 2.94 (brt, J=7.1 Hz, 2H, HalO), 2.63 (t, J=6.8 Hz, 2H, Hcl), 2.06 (m, 2H, Hb4ax), 1.84-1.74(m, 3H, Hb2 et Hb3eq), 1.44-1.30 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 139.1 (Cal l), 131.14 (Cal6), 131(Cal5), 129.5 (Cel l), 129.1 (CclO), 128.7 (Cal3), 128.7 (Cal4), 127.9 (Cal2), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 42.2 (Ca9), 40.5(Hc2), 35.7 (Cb2), 34.53 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 567.2639 [M+H] + ; found: 567.2641.

4-(2-(4-chlorophenyl)ethylamino)-7-((l-(2-(quinolin-4-yla mino)ethyl) piperidin- 4-yl)methoxy)quinazoline (AC) (3.0 mg, 5.3 μηιοΐ, 21%) from 74 (25 μηιοΐ):

1H NMR (500 MHz ; DMSO) δ 8.40 (s, IH, Hal), 8.40 (d, J= 5.3Hz, IH, Hc5), 8.21 (brt, J=5.6 Hz, IH, HNH), 8.16 (dd, J=0.9, 8.2 Hz, IH, Hc8), 8.10 (d, J=9.3 Hz, IH, Ha4), 7.78 (dd, J=l . l, 8.5 Hz, IH, Hel l), 7.61(m, IH, HclO), 7.46-7.22 (m, 5H, Hc9, Hal2 and Hal3), 7.11 (dd, J=2.6, 9.1 Hz, IH, Ha5), 7.06 (d, J=2.5Hz, IH, Ha7), 7.03 (brt, J=5.3 Hz, IH, HNH), 6.48 (d, J=5.5 Hz, IH, Hc4), 3.99 (d, J=5.9Hz, 2H, Hbl), 3.80-3.73 (m, 2H, Ha9), 3.41 (q, J=6.5Hz, 2H, Hc2), 3.05-2.97 (m, 2H, Hb4eq), 3.09 (brt, J=6.9 Hz, 2H, HalO), 2.64 (t, J=6.9 Hz, 2H, Hcl), 2.11-2.02 (m, 2H, Hb4ax), 1.85-1.76(m, 3H, Hb2 et Hb3eq), 1.38 (dq, J=2.5, 12.2 Hz, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 137.5 (Cal l), 131.6 (Cal4), 129.6(Cal2), 129.5 (Cel l), 129.1 (CclO), 127.7 (Cal3), 124.7 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 46.1 (Ca9), 40.6(Hc2), 35.7 (Cb2), 32.8 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 567.2639 [M+H] + ; found: 567.2635.

4-(2-(4-sulfonamidophenyl)ethylamino)-7-((l-(2-(quinolin- 4-ylamino)ethyl) piperidin-4-yl)methoxy)quinazoline (AD) (11 mg, 18 μηιοΐ, 52%) from 75 (34μηιο1): a15

1H NMR (500 MHz ; DMSO) δ 8.43 (s, 1H, Hal), 8.40 (d, J= 5.3Hz, 1H, Hc5), 8.21 (brt, J=5.5 Hz, 1H, HNH), 8.16 (dd, J=0.8, 8.8 Hz, 1H, Hc8), 8.11 (d, J=9.1 Hz, 1H, Ha4), 7.78 (dd, J=l .l, 8.4 Hz, 1H, Hel l), 7.76-7.72 (m, 2H, Hal3), 7.61(m, 1H, HclO), 7.46-7.40 (m, 5H, Hc9, Hal2, Hal5), 7.12 (dd, J=2.6, 9.0 Hz, 1H, Ha5), 7.07 (d, J=2.6 Hz, 1H, Ha7), 7.04 (brt, J=5.5 Hz, 1H, HNH), 6.48 (d, J=5.46 Hz, 1H, Hc4), 3.99 (d, J=5.9Hz, 2H, Hbl), 3.79-3.73 (m, 2H, Ha9), 3.41 (q, J=6.4 Hz, 2H, Hc2), 3.06-2.97 (m, 4H, Hb4eq, HalO), 2.64 (t, J=7.2Hz, 2H, Hcl), 2.10-2.02 (m, 2H, Hb4ax), 1.84-1.77(m, 3H, Hb2 et Hb3eq), 1.38 (dq, J=2.6, 12.1 Hz, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 144.1 (Cal l), 142.9 (Cal4), 129.5(Cal2), 129.5 (Cel l), 129.1 (CclO), 126.1 (Cal3), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.3 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 46.1 (Ca9), 40.6(Cc2), 35.7 (Cb2), 34.7 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 612.2756 [M+H] + ; found: 612.2747.

4-(2-(4-nitrophenyl)ethylamino)-7-((l-(2-(quinolin-4-ylam ino)ethyl) piperidin-4- yl)methoxy)quinazoline (AE) (7 mg, 12 μηιοΐ, 33%) from 76 (37μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.41 (s, 1H, Hal), 8.40 (d, J= 5.3Hz, 1H, Hc5), 8.21 (brt, J=5.5 Hz, 1H, HNH), 8.18-8.14(m, 3H, Hal3 et Hc8), 8.09 (d, J=9.2 Hz, 1H, Ha4), 7.78 (dd, J=1.2, 8.4 Hz, 1H, Hel l), 7.61(m, 1H, HclO), 7.57-7.52 (m, 2H, Hal2), 7.43 (m, 1H, Hc9), 7.12 (dd, J=2.73, 9.25 Hz, 1H, Ha5), 7.06 (d, J=2.64, 1H, Ha7), 7.04 (brt, J=5.54 Hz, 1H, HNH), 6.48 (d, J=5.37 Hz, 1H, Hc4), 3.98 (d, J=5.9Hz, 2H, Hbl), 3.83- 3.77 (m, 2H, Ha9), 3.41 (q, J=8.44 Hz, 2H, Hc2), 3.12 (brt, J=6.9 Hz, 2H, HalO), 3.04-2.97 (m, 2H, Hb4eq), 2.66-2.61 (m, 2H, Hcl), 2.10-2.02 (m, 2H, Hb4ax), 1.84-1.76(m, 3H, Hb2 et Hb3eq), 1.44-1.31 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 146.5 (Cal l), 137.7 (Cal4), 130.5 (Cal2), 129.5 (Cel l), 129.1 (CclO), 123.8 (Cal3), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.4 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 41.9 (Ca9), 40.6(Cc2), 35.7 (Cb2), 34.8 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 578.2879 [M+H] + ; found: 578.2891.

4-(2-(4-isopropylphenyl)ethylamino)-7-((l-(2-(quinolin-4- ylamino)ethyl) piperidin-4-yl)methoxy)quinazoline (AF) (1.2 mg, 2.0 μηιοΐ, 6%) from 77 (37μηιο1): a16

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.40 (d, J= 5.2Hz, IH, Hc5), 8.22 (brt, J=5.9 Hz, IH, HNH), 8.16 (m, IH, Hc8), 8.10 (d, J=9.3 Hz, IH, Ha4), 7.78 (dd, J=1.2, 8.4 Hz, IH, Hel l), 7.61(m, IH, HclO), 7.45-7.40 (m, IH, Hc9), 7.20-7.09 (m, 4H, Hal2, Hal 3), 7.12 (dd, J=2.56, 9.12 Hz, IH, Ha5), 7.06 (d, J=2.2Hz, IH, Ha7), 7.04 (brt, J=5.3 Hz, IH, HNH), 6.48 (d, J=5.4 Hz, IH, Hc4), 3.99 (d, J=5.78Hz, 2H, Hbl), 3.73-3.68 (m, 2H, Ha9), 3.44-3.38 (m, 2H, Hc2), 3.04-2.98 (m, 2H, Hb4eq), 2.88-2.82 (m, 2H, HalO), 2.66-2.61 (m, 2H, Hcl), 2.10-2.02 (m, 2H, Hb4ax), 1.83-1.77(m, 3H, Hb2 et Hb3eq), 1.26- 1.16 (m, 8H, Hb3ax et Hal 6).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156.1 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 146.5 (Cal4), 137.4 (Cal l), 129.5 (Cal2), 129.3 (Cal3), 129.1 (Cel l), 129.1 (CclO), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.2 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 42.5 (Ca9), 40.6(Cc2), 35.7 (Cb2), 34.7 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 575.3498 [M+H] + ; found: 575.3496.

4-(2-(4-methoxyphenyl)ethylamino)-7-((l-(2-(quinolin-4-yl amino)ethyl) piperidin-4-yl)methoxy)quinazoline (AG) (15 mg, 27 μηιοΐ, 71%) from 78 (38μηιο1): a15

1H NMR (500 MHz ; DMSO) δ 8.41 (s, IH, Hal), 8.40 (d, J= 5.37Hz, IH, Hc5), 8.18 (brt, J=5.40 Hz, IH, HNH), 8.16 (m, IH, Hc8), 8.12 (d, J=9.18 Hz, IH, Ha4), 7.78 (dd, J=0.9, 8.3 Hz, IH, Hel l), 7.61(m, IH, HclO), 7.43 (m, IH, Hc9), 7.20-7.15 (m, 2H, Hal2), 7.12 (dd, J=2.6, 9.1 Hz, IH, Ha5), 7.06 (d, J=2.62, IH, Ha7), 7.04 (brt, J=5.44 Hz, IH, HNH), 6.88-6.82 (m, 2H, Hal3), 6.48 (d, J=5.4 Hz, IH, Hc4), 3.98 (d, J=5.9Hz, 2H, Hbl), 3.72 (s, 3H, Hal5), 3.73-3.64 (m, 2H, Ha9), 3.44-3.38 (m, 2H, Hc2), 3.04-2.96 (m, 2H, Hb4eq), 2.89 (brt, J=7.1 Hz 2H, HalO), 2.63 (t, J=6.8 Hz, 2H, Hcl), 2.10-2.01 (m, 2H, Hb4ax), 1.84-1.74(m, 3H, Hb2 et Hb3eq), 1.44-1.31 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 158.1 (Cal4), 156.1

(Cal), 151.8 (Ca8), 151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 131.8 (Cal l), 130.1 (Cal2), 129.5 (Cel l), 129.1 (CclO), 124.6 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.2 (Ca5), 114.2 (Cal3), 109.6 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 55.4 (Cal5), 53.5 (Cb4), 42.7 (Ca9), 40.6(Cc2), 35.7 (Cb2), 34.2 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 563.3134 [M+H] + ; found: 563.3145.

4-(2-(4-aminophenyl)ethylamino)-7-((l-(2-(quinolin-4-ylam ino)ethyl) piperidin- 4-yl)methoxy)quinazoline (AH) (7 mg, 13 μηιοΐ, 33%) from 79 (40μηιο1): a15

1H NMR (500 MHz ; DMSO) δ 8.42-8.36 (m, 2H, Hal et Hc5), 8.19-8.13 (m, 2H, HNH et Hc8), 8.12 (d, J=9.08 Hz, 1H, Ha4), 7.78 (dd, J=0.95, 8.45 Hz, 1H, Hel l), 7.61(m, 1H, HclO), 7.43 (m, 1H, Hc9), 7.11 (dd, J=2.5, 8.9 Hz, 1H, Ha5), 7.05 (d, J=2.46, 1H, Ha7), 7.04 (brt, J=5.28 Hz, 1H, HNH), 6.93-6.87 (m, 2H, Hal2), 6.52-6.48 (m, 2H, Hal3), 6.48 (d, J=5.4 Hz, 1H, Hc4), 4.86 (s, 2H, Hal5), 3.98 (d, J=5.9 Hz, 2H, Hbl), 3.67-3.58 (m, 2H, Ha9), 3.45-3.38 (m, 2H, Hc2), 3.05-2.97 (m, 2H, Hb4eq), 2.76 (brt, J=7.28 Hz 2H, HalO), 2.63 (t, J=6.76 Hz, 2H, Hcl), 2.11-2.01 (m, 2H, Hb4ax), 1.84-1.75(m, 3H, Hb2 et Hb3eq), 1.45-1.31 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.4 (Ca2), 156.2 (Cal), 151.8 (Ca8),

151.2 (Cc5), 150.2 (Cc7), 148.7 (Cc6), 147.2 (Cal4), 129.5 (Cal2), 129.5 (Cal l), 129.5 (Cel l), 128.4 (CclO), 124.7 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 117.2 (Ca5), 114.4 (Cal3), 109.6 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 42.9 (Ca9), 40.6(Cc2), 35.7 (Cb2), 34.4 (CalO), 29 (Cb3).

HRMS-ESI (m/z) calculated: 548.3137 [M+H] + ; found: 548.3142.

4-(2-(quinolin-4-yl)ethylamino)-7-((l-(2-(quinolin-4-ylam ino)ethyl) piperidin-4- yl)methoxy)quinazoline (AI) (3.2 mg, 5.3 μηιοΐ, 23%) from 80 (23.3μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.49 (s, IH, Hal), 8.43 (brt, J=5.7 Hz, IH, HNH), 8.42 (d, J= 5.3Hz, IH, Hal3), 8.40 (d, J= 5.3Hz, IH, Hc5), 8.17 (d, J=6.2 Hz, IH, Hal8), 8.16 (d, J=5.6Hz, IH, Hc8), 8.12 (d, J=9.2Hz, IH, Ha4), 7.78 (d, J=8.4 Hz, 2H, Hel l and Hal5), 7.61(t, J=7.2Hz, 2H, HclO and Hal6), 7.47 (t, J=5.5Hz, IH, HNH), 7.43 (t, J=7.6Hz, 2H, Hal7 and Hc9), 7.14 (dd, J=2.5, 9.0Hz, IH, Ha5), 7.09 (d, J=2.5Hz, IH, Ha7), 7.06 (brt,J=5.1Hz, IH, HNH), 6.70(d, J=5.4Hz, IH, Hal2), 6.48(d, J=5.4Hz, IH, Hc4), 3.99 (d, J=5.7Hz, 2H, Hbl), 3.82 (q, J=6.1Hz, 2H, Ha9), 3.58 (q, J=6.2Hz, 2H, HalO), 3.41 (m, 2H, Hc2), 3.00 (brd, 2H, Hb4eq), 2.63 (t, J=7.0Hz, 2H, Hcl), 2.06 (t, J=10.5Hz, 2H, Hb4ax), 1.865-1.74 (m, 3H, Hb3eq and Hb2), 1.443-1.32 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.3 (Ca6), 159.8 (Ca2), 156.0 (Cal), 151.8 (Ca8), 151.2 (Cc5 and Cal3), 150.4 (Cc3), 150.3 (Cal l), 148.7 (Cal4), 148.6 (Cc6), 129.5 (Cel l and Cal5), 129.2 (CclO and Cal6), 124.7 (Ca4), 124.4 (Cal7),127.3 (Cal3), 122.0 (Cc9 and Cal8), 121.8 (Ca4), 119.3 (Cal9), 119.2 (Cc7), 117.5 (Ca5), 109.5 (Ca3), 108.0 (Ca7), 98.7 (Cc4), 98.6 (Cal2), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 42.1 (CalO), 40.6 (Hc2), 39.3 (Ca9), 35.7 (Cb2), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 599.3242 [M+H] + ; found: 599.3241.

4-(2-( naphthalen-l-yl)ethylamino)-7-((l-(2-(quinolin-4-ylamino)eth yl) piperidin-4-yl)methoxy)quinazoline (AJ) (13 mg, 22 μηιοΐ, 63%) from 81 (35μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.50 (s, IH, Hal), 8.40 (d, J= 5.3Hz, IH, Hc5), 8.38 (brt, J=5.6 Hz, IH, HNH), 8.16 (d, J=7.9Hz, IH, Hc8), 8.14 (d, J=9.4 Hz, IH, Hal9), 8.11 (d, J=8.0Hz, IH, Ha4), 7.79 (d, J=8.4 Hz, IH, Hel l), 7.75 (dd, J=1.8, 7.5Hz, IH, Hal5), 7.61(ddd, J=0.8, 6.5, 7.7Hz, IH, Hcl0),7.45-7.38 (m, 3H, Hal7, Hc9 and Hal8), 7.31 (t, J=7.9Hz, IH, Hal3), 7.15 (dd, J=2.7, 9.3 Hz, IH, Ha5), 7.11 (d, J=7.8Hz, IH, Hal4), 7.09 (d, J=2.6Hz, IH, Ha7), 7.04 (brt,J=5.2Hz, IH, HNH), 6.70(d, J=7.8Hz, IH, Hal2), 6.48-45 (m, 2H, Hc4HNH), 3.99 (d, J=5.9Hz, 2H, Hbl), 3.86 (q, J=6.0Hz, 2H, Ha9), 3.51 (q, J=6.0Hz, 2H, HalO), 3.41 (q, J=6.3 Hz, 2H, Hc2), 3.05-2.97 (m, 2H, Hb4eq), 2.66- 2.60 (m, 2H, Hcl), 2.10-2.02 (t, J=6.9 Hz, 2H, Hb4ax), 1.85-1.75 (m, 3H, Hb3eq and Hb2), 1.43-1.31 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.3 (Ca6), 159.8 (Ca2), 156.0 (Cal), 151.8 (Ca8), 151.2 (Cc5), 150.2 (Cc3), 148.7 (Cc6), 144.4 (Cal l), 134.5 (Ca21), 129.5 (Cel l), 129.1 (CclO), 128.4 (Cal5), 127.3 (Cal3), 126.0 (Cal7), 124.7 (Cal9), 124.4 (Ca4), 124.3 (Cal 8), 123.3 (Ca20), 121.9 (Ca4), 121.8 (Cc9), 119.2 (Cc7), 117.4 (Ca5), 115.8 (Cal4), 109.5 (Ca3), 108.0 (Ca7), 103.2 (Cal2), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 43.2 (CalO), 40.6(Hc2), 39.7 (Ca9), 35.7 (Cb2), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 598.32893 [M+H] + ; found: 598.3295. 4-(2-(phenylamino)ethylamino)-7-((l-(2-(quinolin-4-ylamino)e thyl) piperidin-4- yl)methoxy)quinazoline (AK) (11 mg, 20 μηιοΐ, 50%) from 82 (40μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.42 (s, IH, Hal), 8.40 (d, J= 5.2Hz, IH, Hc5), 8.26 (brt, J=5.5 Hz, IH, HNH), 8.16 (d, J=7.9Hz, IH, Hc8), 8.13 (d, J=9.0Hz, IH, Ha4), 7.78 (dd, J=1.0, 8.3 Hz, IH, Hel l), 7.61(ddd, J=1.0, 6.8, 8.1Hz, IH, Hcl0),7.42 (ddd, j=1.0, 6.9, 7.9Hz, IH, Hc9), 7.13 (dd, J=2.5, 9.0 Hz, IH, Ha5), 7.1 1-7.02 (m, 3H, Ha7 and Hal3), 6.64 (d, J=7.8Hz, 2H, Hal2), 6.52 (t, J=7.3Hz, 2H, Hal4), 6.48 (d, J=7.3Hz, IH, Hc4), 5.81 (brt, J=5.8Hz, IH, HNH), 3.99 (d, J=5.8Hz, 2H, Hbl), 3.69 (q, J=6.4Hz, 2H, Ha9), 3.41 (q, J=6.8 Hz, 2H, Hc2), 3.30 (q, J=6.40Hz, 2H, HalO), 3.05-2.97 (m, 2H, Hb4eq), 2.63 (t, J=6.8Hz, 2H, Hcl), 2.05 (t, J=6.9 Hz, 2H, Hb4ax), 1.86-1.74 (m, 3H, Hb3eq and Hb2), 1.45-1.32 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.6 (Ca2), 156.0 (Cal), 151.8 (Ca8), 151.1 (Cc5), 150.2 (Cc3), 149.1(Cal l), 148.7 (Cc6), 129.5 (Cel l), 129.4 (Cal3), 129.1 (CclO), 124.4 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 119.2 (Cc7), 117.3 (Ca5), 116.0 (Cal4), 112.4 (Cal2), 109.6 (Ca3), 107.9 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 42.4 (CalO), 40.5(Hc2), 40.2 (Ca9), 35.7 (Cb2), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 548.3133 [M+H] + ; found: 548.3140.

4-((N-(l-benzylpiperidin-4-yl))amino)-7-((l-(2-(6,7-dimet hoxyquinolin-4- ylamino)ethyl)piperidin-4-yl)methoxy)quinazoline (AL) (10 mg, 17 μηιοΐ, 48%) from 83 (34.7μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.40 (d, J= 5.3Hz, IH, Hc5), 8.37 (s, IH, Hal), 8.22 (d, J=9.3Hz, IH, Ha4), 8.16 (dd, J=0.9, 7.8Hz, IH, Hc8), 7.78 (dd, J=1.0, 8.4Hz, IH, Hel l), 7.73 (d, J=7.6 Hz, IH, Ha7), 7.60 (ddd, J=1.3, 7.3, 8.4Hz, IH, HclO), 7.42 (ddd, j=1.3, 7.3, 8.4Hz, IH, Hc9), 7.36-7.29 (m, 4H, Hal4 and Hal5), 7.27-7.23 (m, IH, Hal6), 7.13-7.05 (m, 3H, Ha5, Hc4 and Ha7), 7.10 (dd, J=2.6, 9.1Hz, IH, Ha5), 7.04 (d, J=2.6Hz, IH, Ha7), 7.02 (brt, J=5.3Hz, IH, HNH), 6.47 (d, J=5.4Hz, IH), 4.20-4.12 (m, IH, Ha9), 3.97 (d, J=5.9Hz, 3H, Hbl), 3.49 (s, 2H, Hal2), 3.41 (q, J=6.0,13.0Hz, 2H, Hc2), 3.01 (brd, J=11.2Hz, 2H, Hal leq), 2.87 (brd, J=11.7Hz, 2H, Hb4eq), 2.64 (m, 2H, Hcl,), 2.06 (m, 4H, Hal lax and Hb4ax), 1.94-1.86 (m, 3H, HalOeq), 1.86-1.74 (m, 2H, Hb2 and Hb3eq), 1.65 (ddd, J=3.3, 11.9Hz, 2H, HalOax), 1.45-1.31 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 158.8 (Ca2), 156.0 (Cal), 151.9 (Ca8), 151.1 (Cc5), 150.2 (Cc3), 148.7 (Cc6), 139,2 (Cal3), 129.5 (Cel l), 129.1 (Cal4), 129.1 (CclO), 128.6 (Cal5), 127.3 (Cal5), 124.9 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 119.2 (Cc7), 117.0 (Ca5), 109.5 (Ca3), 107.8 (Ca7), 98.7 (Cc4), 72.7 (Cbl), 62.6 (Cal2), 56.6 (Ccl), 53.6 (Cal l), 52.8 (Cb4), 48.2(Ca9), 40.5 (Cc2), 35.7 (Cb2), 31.7 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 602.3602 [M+H] + ; found: 602.3603.

Compound 84 was synthesized following the same procedure as for Compound 44 from compound 42.

4-((3-phenylpropyl)amino)-7-(0-(( V-Boc)pyrrolidin-4-ylmethoxy)) quinazoline

(84)

1H NMR (500 MHz ; DMSO) δ 8.39 (s, IH, Hal), 8.17 (d, J=9.1Hz, IH, Ha4), 8.13 (brt, J=5.1 Hz, IH, HNH), 7.32-7.22 (m, 4H, Hal3 and Hal4), 7.20-7.16 (m, IH, Hal5), 7.11 (dd, J=2.4, 8.7Hz, IH, Ha5) , 7.08 (d, J=2.5Hz, IH, Ha7), 4.16- 4.05 (m, 2H, Hbl), 3.57-3.48 (m, 3H, Ha9 and Hb5), 3.44-3.24 (m, 2H, Hb4), 3.17-3.09 (m, IH, Hb5), 2.75- 2.60 (m, 3H, Hal l and Hb2), 2.11-2.01 (m, IH, Hb3), 1.96 (quint, J=7.3Hz, 2H, HalO), 1.80-1.69 (m, IH, Hb3), 1.41 (s, 9H, HBoc).

13 C NMR (125MHz; DMSO) δ 162.0 (Ca6), 159.5 (Ca2), 156.0 (Cal), 154.0 (CBoc), 151.5 (Ca8), 142.2(Cal2), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.8 (Ca4), 117.1 (Ca5), 109.6 (Ca3), 107.9 (Ca7), 78.6 and 78.4 (Cbl), 62.9 (CBoc), 49.0 and 48.7 (Cb5), 45.6 and 45.4 (Cb4), 40.5 (Ca9), 38.3 and 37.4 (Cb2), 33.1 (Cal l), 30.8 (CalO), 28.6 (CBoc), 28.4and 27.4 (Cb3).

HRMS-ESI (m/z) calculated: 463.2704 [M+H] + ; found: 463.2659. 4-((3-phenylpropyl)amino)-7-0-(pyrrolidin-3-ylmethoxy)quinaz oline (85)

A mixture of 84 (lOOmg; 216μηιο1) in TFA was stirred for lh at room temperature. TFA was removed. The residue was diluted with dichloromethane and the organic phase was washed with saturated Na 2 C0 3 . The solvent was removed and 85 was obtained as pale blue foam (60mg; 166μιηο1, 76%).

1H NMR (500 MHz ; DMSO) δ 8.38 (s, IH, Hal), 8.15 (d, J=9.5Hz, IH, Ha4), 8.09 (brt, J=5.7 Hz, IH, HNH), 7.32-7.22 (m, 4H, Hal3 and Hal4), 7.18 (m, IH, Hal5), 7.11 (dd, J=2.9,9.5Hz, IH, Ha5) 7.07 (d, J=2.1Hz, IH, Ha7), 4.03- 3.97 (m, 2H, Hbl), 3,56-3.49 (m, 2H, Ha9), 2.93-2.79 (m, 2H, Hb5 and Hb4), 2.76-2.60 (m, 4H, Hb5, Hb4 and Hal l), 2.49-2.41 (m, 1H, Hb2), 1.94 (quint, J=7.0Hz, 2H, HalO), 1.90-1.80 (m, 1H, Hb3), 1.50- 1.39 (m, lH, Hb3).

13 C NMR (125MHz; DMSO) δ 162.2 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.7 (Ca8), 142.2(Cal2), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 117.2 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 71.2 (Cbl), 50.4 (Cb5), 46.7 (Cb4), 40.5 (Ca9), 38.7 (Cb2), 33.2 (Cal l), 31.7 (CalO), 29.4(Cb3).

HRMS-ESI (m/z) calculated: 363.2180 [M+H] + ; found: 363.2195.

4-((3-phenylpropyl)amino)-7-((l-(2-(quinolin-4-ylamino)et hyl)pyrrolidin-3-yl) methoxy)quinazoline (AM)

To a solution of 85 (15mg; 41μηιο1), K 2 C0 3 (l lmg; 80μηιο1) and a catalytic amount of KI in DMF (0.5mL) was added 26 (16mg; 80μmol). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0 to 10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0 to 80% CH 3 CN) to afford Compound AM as a white powder (7mg; 13μιηο1; 31 >).

1H NMR (500 MHz ; DMSO) δ 8.38 (s, 1H, Hal), 8.37 (d, J= 5.3Hz, 1H, Hc5),

8.16 (d, J=7.8Hz, 1H, Hc8), 8.15 (d, J=9.2Hz, 1H, Ha4), 8.11 (brt, J=5.9 Hz, 1H, HNH), 7.77 (dd, J=1.0, 8.2 Hz, 1H, Hel l), 7.59 (ddd, J=1.0, 6.8, 8.2Hz, 1H, HclO), 7.41 (ddd, j=1.0, 6.8, 8.1Hz, 1H, Hc9), 7.32-7.22 (m, 4H, Hal3 and Hal4), 7.18 (m, 1H, Hal5), 7.13- 7.05 (m, 3H, Ha5, Hc4 and Ha7), 6.47 (d, J=5.4Hz, 1H), 4.06- 3.97 (m, 2H, Hbl), 3.46-3.39 (m, 2H, Hc2), 2.93 (q, J=6.1Hz, 2H, Ha9), 2.82-2.72 (m, 3H, Hcl+Hb5), 2.72-2.57 (m, 6H, Hb4, HalO, Hb2 and Hal l), 2.53-2.43 (m, 1H, Hb5), 1.99-1.86 (m, 1H, Hb3), 1.31-1.24 (m, 1H, Hb3). 1J C NMR (125MHz; DMSO) δ 162.0 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.7 (Ca8), 151.1 (Cc5), 150.2 (Cc3), 148.7 (Cc6), 142.2(Cal2), 129.5 (Cel l), 129.1 (Ccl0),128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 124.3 (Cc9), 121.9 (Cc8), 119.2 (Cc7), 117.1 (Ca5), 109.6 (Ca3), 108.0 (Ca7), 98.67 (Cc4), 71.6 (Cbl), 57.7 (Ccl), 53.8 (Cb4), 41.9 (Cc2), 40.4 (Ca9), 35.9 (Cb2), 33.7 (Cal l), 31.7 (CalO), 30.8 (Cb3).

HRMS-ESI (m/z) calculated: 533.3024 [M+H] + ; found: 533.3025.

Compound AN was synthesized following the general procedure above as for Compound G from 45.

To a solution of 45 (80mg; 212μιηο1), K 2 C0 3 (59mg; 424μιηο1) and a catalytic amount of KI in DMF (lmL) was added 26 (115mg; 424μιηο1). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed 15 with water and brine and dried over sodium sulfate. The solvent was removed. The crude product was used without further purification and was solubilized in TFA. The reaction mixture was stirred at room temperature for lh. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0→10% MeOH/NH 3 ) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compound AN as a white powder 23mg; 45μιηο1; 21%).

1H NMR (500 MHz ; DMSO) δ 8.38 (s, 1H, Hal), 8.15 (d, J=9.1Hz, 1H, Ha4), 8.09 (brt, J=5.5 Hz, 1H, HNH), 7.32-7.21 (m, 4H, Hal3 and Hal4), 7.17 (t, J=7.0Hz, 1H, Hal5), 7.11 (dd, J=2.9, 9.2Hz, 1H, Ha5), 7.05 (d, J=2.4Hz, 1H, Ha7), 6.45-6.36 (m, 4H, Hc4 and Hc5), 3.97 (d, J=5.9Hz, 2H, Hbl), 3.53 (q, J=6.2Hz, 2H, Ha9), 3.00 (t, J=6.6 Hz, 2H, Hc2), 2.97-2.85 (m, 2H, Hb4eq), 2.68 (t, J=8.0Hz, 2H, Hal l), 2.47 (t, J=7.1Hz, 2H, Hcl), 2.01- 1.90 (m, 2H, HalO and Hb4ax), 1.83-1.73 (m, 3H, Hb3eq and Hb2), 1.42-1.29 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 156.0 (Cal), 151.7 (Ca8), 142.2(Cal2), 140.7 (Cc3), 139.6 (Cc6), 128.8 (Ccal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 117.2 (Ca5), 115.9 (Cc4), 114.2 (Cc5), 109.5 (Ca3), 107.9 (Ca7), 72.7 (Cbl), 57.8 (Ccl), 53.5 (Cb4), 42.1 (Cc2), 40.5 (Ca9), 35.8 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 511.3180 [M+H] + ; found: 511.3180.

Compounds AO to AS were synthesized following the same procedure Compound G from 45.

To a solution of 0.1 M of 45, K 2 CO 3 (2eq) and a catalytic amount of KI in DMF was added the desired chloro-derivative (2eq). The mixture was stirred at 65°C overnight then was diluted with ethyl acetate. The organic phase was washed with water and brine and dried over sodium sulfate. The solvent was removed and the residue was purified by silica gel flash chromatography using a linear gradient of ammonia IN in methanol (0→10% MeOH/NHs) in dichloromethane or by reversed phase HPLC using a linear acetonitrile gradient with 0.01% of TEA (0→80% CH 3 CN) to afford Compounds AO to AS.

4-((3-phenylpropyl)amino)-7-((l-(2-(7-chloroquinolin-4-yl amino)ethyl) piperidin-4-yl)methoxy)quinazoline (AO) (15 mg, 26μηιο1, 97%) from 45 (27μmol):

1H NMR (500 MHz ; DMSO) δ 8.41 (d, J= 5.3Hz, IH, Hc5), 8.38 (s, IH, Hal), 8.23 (d, J=9.1Hz, IH, Hc8), 8.15 (d, J=9.2Hz, IH, Ha4), 8.09 (brt, J=5.5Hz, IH, HNH), 7.79 (d, J=2.3Hz, IH, Hel l), 7.46 (dd, j=2.2, 7.3, 8.9Hz, IH, Hc9), 7.32-7.15 (m, 5H, Hal3, Hal4 and Hal 5), 7.11 (dd, J=2.6, 9.1Hz, IH, Ha5), 7.05 (d, J=2.6Hz, IH, Ha7), 6.51 (d, J=5.4Hz, IH, Hc4), 3.98 (d, J=5.9Hz, 2H, Hbl), 3.53 (q, J=6.1Hz, 2H, Ha9), 3.41 (q, J=6.5Hz, 2H, Hc2), 3.01 (brd, J=11.2Hz, 2H, Ha4eq), 2.68 (t, J=7.5Hz ; 2H, Hal l,) 2.62 (t, J=7.1Hz, 2H, Hcl,), 2.06 (t, J= 11.1Hz, 2H, Hb4ax), 1.95 (q, J= 7.5Hz, HalO), 1.83-1.73 (m, 3H, Hb3eq and Hb2), 1.43-1.31 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 156.1 (Cal), 152.4 (Cc5), 151.8 (Ca8), 150.4 (Cc3), 149.5 (Cc6), 142.2 (Cal2), 133.8 (CclO), 128.8 (Cal4), 128.7 (Cal3), 128.0 (Cel l), 126.2 (Cal5), 124.7 (Ca4), 124.5 (Cc9), 124.4 (Cc8), 117.9 (Cc7), 117.2 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 99.2 (Cc4), 72.7 (Cbl), 56.6 (Ccl), 53.5 (Cb4), 40.7 (Ca9), 40.5 (Cc2), 35.7 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 581.2790 [M+H] + ; found: 581.2791.

4-((3-phenylpropyl)amino)-7-((l-(2-(7-methoxyquinolin-4-y lamino)ethyl) piperidin-4- l)methoxy)quinazoline (AP) (43 mg, 75μηιο1, 71%) from 45 (106μηιο1):

1H NMR (500 MHz ; DMSO) δ 8.38 (s, IH, Hal), 8.32 (d, J= 5.3Hz, IH, Hc5), 8.15 (d, J=9.2Hz, IH, Ha4), 8.10 (brt, J=5.4Hz, IH, HNH), 8.06 (d, J=9.1Hz, IH, Hc8), 7.32-7.22 (m, 4H, Hal3 and Hal4) 7.21-7.15 (m, 2H, Hal5 and Hel l), 7.11 (dd, J=2.4, 9.1Hz, IH, Ha5), 7.08-7.03 (m, 2H, Ha7 and Hc9), 6.96 (brt, J=5.2Hz, IH, HNH), 6.36 (d, J=5.5Hz, IH, Hc4), 3.97 (d, J=5.7Hz, 2H, Hbl), 3.87 (s, 3H, Hcl2), 3.53 (q, J=6.4Hz, 2H, Ha9), 3.41-3.36 (m, 2H, Hc2), 3.00 (brd, J=10.9Hz, 2H, Ha4eq), 2.68 (t, J=7.7Hz ; 2H, Hal l) 2.61 (t, J=6.9Hz, 2H, Hcl), 2.04 (t, J= 11.1Hz, 2H, Hb4ax), 1.95 (quint, J= 7.4Hz, HalO), 1.85-1.73 (m, 3H, Hb3eq and Hb2), 1.44-1.31 (m, 2H, Hb3ax). 1J C NMR (125MHz; DMSO) δ 162.1 (Ca6), 160.0 (Cc3), 159.5(Ca2), 156.1 (Cal), 151.8 (Ca8), 151.5(Cc5), 150.6 (CclO), 150.3 (Cc6), 142.2(Cal2), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 123.3 (Cc8), 117.2 (Ca5), 116.0 (Cc9), 113.7 (Cc7), 109.5 (Ca3), 108.3 (Cel l), 107.9 (Ca7), 97.6 (Cc4), 72.7 (Cbl), 56.7 (Ccl), 55.6 (Ccl2), 53.5 (Cb4), 40.6 (Cc2), 40.5 (Ca9), 35.7 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 577.3286 [M+H] + ; found: 577.3296.

4-((3-phenylpropyl)amino)-7-((l-(2-(6,7-dimethoxyquinolin -4- ylamino)ethyl)piperidin-4-yl)methoxy)quinazoline (AQ) (9 mg, 15 μηιοΐ, 56%) from 45

1H NMR (500 MHz ; DMSO) δ 8.37 (s, 1H, Hal), 8.22 (d, J= 5.3Hz, 1H, Hc5), 8.15 (d, J=9.3Hz, 1H, Ha4), 8.11 (brt, J=5.4Hz, 1H, HNH), 7.45 (s, 1H, Hc8), 7.32-7.21 (m, 4H, Hal3 and Hal4), 7.21-7.15 (m, 2H, Hal5 and Hel l), 7.11 (dd, J=2.6, 9.1Hz, 1H, Ha5), 7.05 (d, J=2.6Hz, 2H, Ha7), 6.84 (brt, J=5.2Hz, 1H, HNH), 6.37 (d, J=5.5Hz, 1H, Hc4), 3.98 (d, J=5.9Hz, 2H, Hbl), 3.89 (s, 3H, Hcl3), 3.87 (s, 3H, Hcl2), 3.56-3.49 (m, 2H, Ha9), 3.41-3.36 (m, 2H, Hc2), 3.00 (brd, J=10.8Hz, 2H, Ha4eq), 2.68 (t, J=7.7Hz ; 2H, Hal l) 2.63 (t, J=6.9Hz, 2H, Hcl), 2.06 (t, J= 10.8Hz, 2H, Hb4ax), 1.95 (quint, J= 7.4Hz, HalO), 1.86-1.75 (m, 3H, Hb3eq and Hb2), 1.47-1.32 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5(Ca2), 156.1 (Cal), 151.7 (Cc3),

151.5(Ca8), 149.3 (Cc9), 148.9 (Cc5), 148.0 (CclO), 145.4 (Cc6), 142.2(Cal2), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 117.2 (Ca5), 113.1 (Cc7), 109.5 (Ca3), 108.7 (Cel l), 107.9 (Ca7), 101.1 (Cc8), 97.9 (Cc4), 72.7 (Cbl), 56.9 (Ccl), 56.3 (Ccl3), 55.8 (Ccl2), 53.6 (Cb4), 40.7 (Cc2), 40.3 (Ca9), 35.7 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 607.3391 [M+H] + ; found: 607.3391. 4-((3-phenylpropyl)amino)-7-((l-(2-(methyl(quinolin-4-yl)ami no)ethyl) piperidin-4-yl)methoxy)quinazoline (AR) (13 mg, 23μηιο1, 85%) from 45 (27μηιο1)::

1H NMR (500 MHz ; DMSO) δ 8.61 (d, J= 5.1Hz, IH, Hc5), 8.38 (s, IH, Hal), 8.21 (d, J=8.5Hz, IH, Hc8), 8.15 (d, J=9.2Hz, IH, Ha4), 8.09 (brt, J=5.4 Hz, IH, HNH), 7.91 (dd, J=0.7,8.3Hz, IH, Hel l), 7.66 (t, J=7.3Hz, IH, Hc9), 7.51(t, J=7.3Hz, IH, HclO), 7.32-7.22 (m, 4H, Hal3 and Hal4), 7.18 (t, J=7.3Hz, IH, Hal5), 7.10 (dd, J=2.5, 9.1Hz, IH, Ha5), 7.04 (d, J=2.6Hz, IH, Ha7), 6.93 (d, J=5.4Hz, HI, Hc4), 3.93 (d, J=5.9Hz, 2H, Hbl), 3.53 (q, J=5.8Hz, 2H, Ha9), 3.40 (t, J=6.3Hz, 2H, Hc2), 2.97 (s, 3H, Hcl2), 2.85 (brd, J=10.7Hz, 2H, Ha4eq), 2.68 (t, J=7.7Hz, 2H, Hal l,) 2.64 (t, J=6.2Hz, 2H, Hcl,), 2.02-1.88 (m, 4H, Hb4ax and HalO), 1.79-1.63 (m, 3H, Hb3eq and Hb2), 1.29-1.15 (m, 2H, Hb3ax).

13 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 157.1 (Cc3), 156.1 (Cal), 151.8 (Ca8), 150.8 (Cc5), 149.8 (Cc6), 142.2(Cal2), 129.9 (Cel l), 129.1 (CclO), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.9 (Cc9), 124.7 (Ca4), 124.6 (Cc8), 123.1 (Cc7), 117.1 (Ca5), 109.5 (Ca3), 108.8 (Cc4), 107.9 (Ca7), 72.6 (Cbl), 55.9 (Ccl), 54.2 (cc2), 53.6 (Cb4), 40.5 (Ca9), 40.2 (Ccl2), 35.6 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 561.3337 [M+H] + ; found: 561.3339.

4-((3-phenylpropyl)amino)-7-((l-(2-(quinolin-6-ylamino)et hyl)piperidin-4- yl)methoxy)quinazoline (AS) (14 mg, 26μιηο1, 96%) from 45 (27μmol):

1H NMR (500 MHz ; DMSO) δ 8.48 (dd, J= 1.63, 4.18Hz, IH, Hel l), 8.38 (s, IH, Hal), 8.15 (d, J=9.2Hz, IH, Ha4), 8.09 (brt, J=5.5Hz, IH, HNH), 7.99 (dd, J=0.9, 7.4Hz, IH, Hc9), 7.70 (d, J=9.1Hz, IH, Hc4), 7.32-7.22 (m, 6H, Hal3, Hal4, Hc5 and HclO), 7.18 (dt, J=1.4, 7.3Hz, IH, Hal5), 7.12 (dd, J=2.5, 9.1Hz, IH, Ha5), 7.06 (d, J=2.6Hz, IH, Ha7), 6.72 (d, J=5.4Hz, HI, Hc8), 5.99 (t, J=5.3Hz, IH, HNH), 3.98 (d, J=5.9Hz, 2H, Hbl), 3.53 (q, J=6.0Hz, 2H, Ha9), 3.24 (q, J=5.9Hz, 2H, Hc2), 2.99 (brd, J=11.2Hz, 2H, Ha4eq), 2.68 (t, J=7.7Hz, 2H, Hal l), 2.59 (t, J=6.7Hz, 2H, Hcl,), 2.03 (brt, j=10.8Hz, 2H, Hb4ax), 1.94 (quint, J=7.4Hz, 2H, HalO), 1.85-1.75 (m, 3H, Hb3eq and Hb2), 1.45-1.35 (m, 2H, Hb3ax).

1 3 C NMR (125MHz; DMSO) δ 162.1 (Ca6), 159.5 (Ca2), 156.1 (Cal), 151.8 (Ca8),

174.3 (Cc3), 145.4 (Cel l), 142.7 (Cc6), 142.2(Cal2), 133.6 (Cc9), 130.5 (Cc7), 129.8 (Cc4), 128.8 (Cal4), 128.7 (Cal3), 126.2 (Cal5), 124.7 (Ca4), 122.1 (Cc5), 121.7 (CclO), 117.2 (Ca5), 109.5 (Ca3), 107.9 (Ca7), 101.6 (Cc8), 72.7 (Cbl), 57.3 (Ccl), 53.6 (Cb4), 41.1 (cc2), 40.5 (Ca9), 35.7 (Cb2), 33.1 (Cal l), 30.8 (CalO), 29.0 (Cb3).

HRMS-ESI (m/z) calculated: 547.3180 [M+H] + ; found: 547.3182.

II. Biological tests of the compounds according to the invention

DNMT3A Assay.

DNMT3A enzyme inhibition was adapted from the restriction-based fluorescence assay protocol described in Ceccaldi et al. (ChemBioChem 2011, 12, 1337-45). Briefly, a 5'- labelled biotin oligonucleotide is hybridized to its complementary strand labelled with 6- carboxyfluorescein at the 3'-end into a 384 well microplate (black Optiplates; Perkin Elmer) pre-coated with avidin. The duplex contains a unique CpG site overlapping with a restriction site of a methylation sensitive restriction enzyme. The human C-terminal DNMT3A (a.a. 623-908), produced as described in Gros et al. (Nucleic Acids Research 2013 August 25), was added in each well (200 ng/well) and mixed with the chemical compounds at desired concentrations and freshly prepared AdoMet (20 μΜ final concentration) to start the reaction in a total volume of 50 μί. After 1 hour incubation at 37°C each well were washed three times with PBS, Tween-20 0.05%, NaCl (500 mM) and three more times with PBST. Specific fluorescent signals were detected with the methylation-sensitive restriction enzyme HpyCH4IV (NEB) as described and measured on a Perkin Elmer Envision detector. The percentage of inhibition is reported. The formula used to calculate the percentage of inhibition is [(X-Y)/ X] x 100, where X is the signal determined in the absence of the inhibitor and Y is the signal obtained in the presence of the inhibitor. The concentration at which 50 % of efficacy of inhibition is observed (EC50) was determined by analysis of a concentration range of the tested compound in triplicates. The non- linear regression fittings with sigmoidal dose-response (variable slope) were performed with GraphPad Prism 4.03 (GraphPad Software).

DNMT1 Assay.

His-DNMTl (182kDa, human) was cloned, expressed and purified as described in Halby et al. (ChemBioChem 2012, 13, 157-65). The reaction was performed in a ΙΟμΙ ^ total reaction volume in low volume NBS™ 384-well microplates (Corning), containing the tested compound (up to 1% DMSO), ΙμΜ of a SAM/[methyl- 3 H] SAM (3TBq/mmol, PerkinElmer) mix in a ratio of 3-to-l (isotopic dilution 1 *:3), 0.3μΜ of biotinylated hemimethylated DNA duplex (5 ' -G ATm CGCm CG ATGm CGm CG AATm CGm CG AT mCGATGmCGAT-3 ' and BIOT-5'-ATCGCATCGATCGCGATTCGCGCATCGGCG ATC-3'), and 90nM of DNMT1 in methylation buffer (20mM HEPES pH 7.2, lmM EDTA, 50mM KC1, 25μg/mL BSA). The reaction was incubated at 37°C for 2 hours. 8μί are then transferred into a streptavidin 96-well scintillant coated Flashplate™ (PerkinElmer) containing 190μΕ of 20μΜ SAH in 50mM Tris-HCl pH 7.4. The Flashplate™ was agitated at room temperature for 1 hour, washed three times with 200μί of 0.05% Tween®-20 in 50mM Tris-HCl pH 7.4, and read in 200μΕ of 50mM Tris-HCl pH 7.4 on TopCount NXT (PerkinElmer). The results of these tests obtained with the compounds of the invention are indicated below:

DNMT1 (% of inhibition) DNMT3A (% of inhibition)

Compound

100μΜ 32μΜ 10μΜ 32μΜ 20μΜ 10μΜ ECso μΜ

A 97 - - 85 76 60 10

B 86 - - - 67 - -

E - - - 51 27 - -

F 90 - - 87 76 59 10

G 91.2 24.5 - - 97 81 2.4

H 94.9 - - 69 44 28 -

1 99.3 - - 68 32 16 -

J 33.7 - - 72 65 47 -

K - - - 44 40 10 -

L 45 - - 26 - 10 -

M 100 100 - 91 - 89 1.1

N 69 24 - 83 - 86 1.1

0 98.4 38 - 99 - 80 4.9

P 22 26 - 81 - 43 13.1

Q 99.6 74 - 68 - 61 5.7

R 98.4 45 - 99 - 65

S 90 - - 95 - 52

AA 97 - - 91 - 82 2,7

AB 97 42 - 100 - - -

AC 97 75 - 100 - 88 1 ,9

AD 100 59 - 93 - - -

AE 74 54 - 72 - 61 1 , 1

AF - - - 83 - 53

AG 59 77 - 100 - 82 3,4

AH 97 59 - 96 - 68 -

Al 99 99 - 99 - 86 1

AJ 94 94 - 90 - 91 -

AK 93 93 - 5 - 76 -

AL 81 81 - 93 - 72 8

AM 100 - - 81 - - -

AN - - - 65 - - -

AO 79 - - 62 - 70 -

AP 90 - - 80 - 70 2,6 DNMT1 (% of inhibition) DNMT3A (% of inhibition)

Compound

100μΜ 32μΜ 10μΜ 32μΜ 20μΜ 10μΜ ECso μ

AQ 100 61 - 95 - 95 1 ,4

AR - - - 66 - 66 4,7

AS - - - 60 - 60 -

Anti-proliferative activity.

- On KG-1 and Karpas 299 cells:

KG-1 and Karpas299 human leukemia cells were obtained from the ATCC (USA) and cultivated in RPMI 1640 medium (with HEPES and Glutamine, BE12-115F, Lonza, France) supplemented with, respectively, 20% and 15% foetal calf serum (Lonza, France), at 37°C and under 5% C0 2 . To measure the anti-proliferative properties of tested molecules, 2x10 4 cells are seeded at day 0 in a 96 wells plate. The compounds to be tested, stored at - 20°C as 10 "2 M stock solution in 100% DMSO, are freshly diluted on day 1 in RPMI 1640 medium, before adding a dose range of 3.2 nM to 10 μΜ to the cells. This treatment is repeated on day 2 and 3, and on day 4 cell viability is assessed using the ATPLite kit from Perkin (ATPlite 1 Step Luminescence Assay System, ref 3016739), following the provider instructions. The raw data are analyzed with GraphPad Prism software (v4.03) to generate EC50 values corresponding to the compound concentrations giving 50% reduction in cell viability. The values presented are the mean results of at least two independent experiments. The 95%) confidence intervals for these EC50 values are also indicated.

- On WM266.4, U87MG and PANC1 cells:

The antiproliferative activity of compounds was measured in vitro using the ATP quantification method "ATPlite one step assay" (Perkin Elmer ref 6016739) according to the manufacturer conditions.

Briefly, WM266.4 cells (human melanoma cells) (5 x 104 cells per ml in RPMI 1640 medium, 10% FBS, 2 mM glutamine, 50 U/mL penicillin/streptomycin and 1.25 μg/mL fungizone), PANC1 cells (human pancreatic carcinoma cells) (2 x 104 cells per ml in DMEM medium, 10% FBS, 2 mM glutamine, 50 U/mL penicillin/streptomycin and 1.25 μg/mL fungizone), and U87MG cells (human neuronal gliablastoma - astrocytoma - cells) (3 x 104 cells per ml in MEM medium, 10% FBS, 2 mM glutamine, 50 U/mL penicillin/streptomycin and 1.25 μg/mL fungizone) were seeded in 96-well plates, incubated for 24 h and treated with 8 increasing concentrations of test compounds diluted in cells respective mediums, or vehicle in triplicate.

Cells were then incubated for 72 h at 37 °C in humidified 5% C0 2 atmosphere.

At the end of the experiment, cell viability was evaluated by determining the level of ATP released by viable cells.

EC50 values were determined with curve fitting analysis method (non linear regression model with a sigmoid dose response, variable Hill slope coefficient) provided by the Prism Software (GraphPad). Results were expressed as average EC50 values (concentration of tested compound that inhibits 50% of the maximum effect for the considered compound).

The results of these tests obtained with the compounds of the invention are indicated below: