OA18891A - New bicyclic derivatives, a process for their preparation and pharmaceutical compositions containing them. - Google Patents

New bicyclic derivatives, a process for their preparation and pharmaceutical compositions containing them. Download PDF

Info

Publication number
OA18891A
OA18891A OA1201700513 OA18891A OA 18891 A OA18891 A OA 18891A OA 1201700513 OA1201700513 OA 1201700513 OA 18891 A OA18891 A OA 18891A
Authority
OA
OAPI
Prior art keywords
group
branched
linear
phenyl
mmol
Prior art date
Application number
OA1201700513
Inventor
Olivier Geneste
Maïa CHANRION
James Edward Paul Davidson
James Brooke MURRAY
l-Jen CHEN
Andras Kotschy
Szabolcs SIPOS
Balazs Balint
Marton Csékei
Levente Ondi
Agnes Proszenyak
Zoltan Szlavik
Zoltan Szabo
Original Assignee
Les Laboratoires Servier
Vernalis (R&D) Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Les Laboratoires Servier, Vernalis (R&D) Limited filed Critical Les Laboratoires Servier
Publication of OA18891A publication Critical patent/OA18891A/en

Links

Abstract

Compounds of formula (I);

Description

The present invention relates to new bicyciic dérivatives, to a process tor their préparation and to pharmaceutical compositions containing them.
The compounds of the present invention are new and hâve very valuable pharmacological characteristics in the field of apoptosis and cancerology.
Apoptosis, or programmed cell death, is a physiological process that is crucial for embryonic development and maintenance of tissue homeostasîs.
Apoptotic-type cell death involves morphological changes such as condensation of the nucléus, DNA fragmentation and also biochemical phenomena such as the activation of caspases which cause damage to key structural components of the cell, so inducing its disassembly and death. Régulation ofthe process of apoptosis is complex and involves the activation or repression of several intracellular signalling pathways (Cory S. et al.. Nature Review Cancer 2002, 2, 647-656).
Deregulation of apoptosis is involved in certain pathologies. Increased apoptosis is associated with neurodegenerative diseases such as Parkinson s disease, Alzheimer s disease and ischaemia. Conversely, déficits in the implémentation of apoptosis play a significant rôle in the development of cancers and their chemoresistance, in auto-immune diseases. inflammatory diseases and viral infections. Accordingly, absence of apoptosis is one ofthe phenotypic signatures of cancer (Hanahan D. et al., Cell 2000,100, 57-70).
The anti-apoptotic proteins of the Bcl-2 family are associated with numerous pathologies. The involvement of proteins ofthe Bcl-2 family is described in numerous types of cancer, such as colon cancer, breast cancer, small-cell lung cancer, non-small-cell lung cancer, bladder cancer, ovarian cancer, prostate cancer, chronic lymphoid leukaemia, lymphoma, myeloma. acute myeloid leukemia, pancreatic cancer, etc. Overexpression of the antiapoptotic proteins of the Bcl-2 family is involved in tumorigenesis, in résistance to chemotherapy and in the clinical prognosis of patients affected by cancer. Notably, Mcl-1, an anti-apoptotic Bcl-2 family member, is overexpressed in various types of cancer (Beroukhim R. et al., Nature 2010, 899-905). There is, therefore, a therapeutic need for compounds that inhibit the anti-apoptotic activity of the proteins of the Bcl-2 family.
ORIGINAL
- 2In addition to being new, the compounds of the présent invention hâve pro-apoptotic properties making it possible to use them in pathologies involving a defect in apoptosis, such as, for example, in the treatment of cancer and of immune and auto-immune diseases.
The présent invention relates more especially to compounds of formula (I):
wherein:
♦ A represents the group
in which 1 is linked to the W group and 2 is linked to the phenyl ring, wherein:
- E represents a furyl, thienyl or pyrrolyl ring,
- X], X3, X.I and X5 independently of one another represent a carbon atom or a nitrogen atom,
- X? represents a C-R2i group or a nitrogen atom, and
- ( ) means that the ring is aromatic,
R] represents a halogen atom, a linear or branched (Cj-Côjalkyl group, a linear or branched (C2-C6)alkenyi group, a linear or branched (C2-C6)alkynyl group, a linear or branched (C|-C6)polyhaloalkyl group, a hydroxy group, a hydroxy(C|-C6)alkyl group, a linear or branched (Cj-C&)alkoxy group, -S-(C|-Cé)alkyl, a cyano gioup, a nitro group, -alkylfCo-Côf-NRitRii’, -O-alkylfCi-Cej-NRnRn , -O-alkyl(Ci-C6)-Ri2, -C(0)-ORu, -O-C(O)-Rlb -C(O)-NRnRii’, -NRn-C(O)-Rii’,
ORIGINAL
-NRii-C(O)-ORn’, -alkyl(Ci-C6)-NRii-C(O)-Rii\ -SCb-NRuRn’, -SO2-alkyl(Cj-Cé), ♦ R2, R3, Ri and R5 independently of one another represent a hydrogen atom, a halogen atom. a linear or branched (Ci-Cé)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, a linear or branched (C|-C6)polyhaloalkyl, a hydroxy group, a hydroxy(Ci-C6)alkyl group, a linear or branched (C,-C6)alkoxy group, a -S-(C,-C6)alkyl group, a cyano group, a nitro group, -alkyl(Co-Cé)-NR||Rn’, -0-alkyl(C|-C6)-NRnRn , -O-alkyl(C!-C6)-Rl2, -C(O)-ORu, -O-C(O)-Rn, -C(O)-NRiiRh’, -NRh-C(O)-Ru’, |0 -NRu-C(O)-ORn’, -alkyl(Ci-C6)-NRu-C(O)-Rir, -SCb-NRuRn’, or
-SO2-alkyl(Ci-C6), or the substituents of the pair (Ri, R2) form together with the carbon atoms carrying them an aromatic or non-aromatic ring composed of from 5 to 7 ring members, which may contain from l to 3 heteroatoms selected from oxygen, sulphur and ] 5 nitrogen. it being understood that resulting ring may be substituted by from l to 2 groups selected from halogen, linear or branched (Ci-Cé)alkyl, -alkyl(C0-C6)-NRliRii’, -NR13R13’, -alkyl(C0-C6)-Cyi or oxo, ♦ Ré and R? independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (CrC6)alkyl group, a linear or branched (C2-C6)alkenyl 20 group, a linear or branched (C2-C6)alkynyl group, a linear or branched (Ci-C6)polyhaloalkyl, a hydroxy group, a linear or branched (Ci-C6)alkoxy group, a -S-(Ci-C6)alkyl group, a cyano group, a nitro group, -alkyl(Co-C6)-NR|]Rn , -O-Cyi, -alkyl(C0-C6)-Cyi, -alkenyl(C2-C6)-Cyl, -alkynyl(C2-C6)-Cy17 -O-alkyl(C|-C6)-Ri2, -C(O)-ORn, -O-C(O)-Rn, -C(O)-NRuR|i’, -NRu-C(O)-Rii’, 25 -NRh-C(O)-0Rii’, -alkyl(Ci-C6)-NRn-C(O)-Rn’, -SO2-NRhRh ,
-SO2-alkyl(CrC6), or the substituents of the pair (Ré, R7), when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them an aromatic or nonaromatic ring composed of from 5 to 7 ring members, which may contain from 1 to 30 3 heteroatoms selected from oxygen, sulphur and nitrogen. it being understood that resulting ring may be substituted by a group selected from a linear or branched (Ci-Cé)alkyl group, -NRi3Rb’, -alkyl(Co-Cé)-Cyi or an oxo,
ORIGINAL ♦ W represents a -CH2- group, a -NH- group or an oxygen atom, ♦ R8 represents a hydrogen atom, a linear or branched (C]-C8)alkyl group, a -CHRaRb group, an aryl group, a heteroaryl group, an arylalkyl(C]-C6) group, or a heteroarylalkyl(Ci-C6) group, ♦ R9 represents a hydrogen atom, a linear or branched (Cj -C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, -Cy2, -alkyl(CrC6)-Cy2, -alkenyl(C2-C6)-Cy2, -alkynyl(C2-C6)-Cy2, -Cy2-Cy3, -alkynyl(C2-C6)-O-Cy2, -Cy2-alkyl(C0-C6)-0-alkyl(Co-C6)-Cy3, a halogen atom, a cyano group, -C(O)-R[j, or -C(O)-NR|3R|5’, ♦Rio represents a hydrogen atom, a linear or branched (CpCôjalkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, an arylalkylfCi-Ce) group, a cycloalkylalkyl(Ci-C&) group, a linear or branched (C i-C6)polyhaloalkyl, -alkyl(C i-C6)-O-Cy4, or the substituents of the pair (R9, Rio), when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them an aromatic or nonaromatic ring composed of from 5 to 7 ring members, which may contain from 1 to J heteroatoms selected from oxygen, sulphur and nitrogen, ♦ Ru and Ru’ independently of one another represent a hydrogen atom, a linear or branched (C |-Cô)alkyl group, or the substituents of the pair (Ru, Ru’) torm together with the nitrogen atom carrying them an aromatic or non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom trom 1 to j heteroatoms selected from oxygen, sulphur and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a hydrogen atom, or a linear or branched (Ci-Côjalkyl group, r12 represents -Cys, -Cys-alkyl(Co-C6)-0-alkyl(Co-C6)-Cy6, -Cy5-alkyl(C0-C6)-Cy6, -Cy5-alkyl(Co-C6)-NR|i-alkyl(Co-C6)-Cy6.
-Cy5-Cy6-0-alkyl(Co-C6)-Cy7, -C(O)-NRhRii’, -NRnRn’, -OR(i, -NRn-C(O)-Rn’, -O-alkyl(Ci-C6)-ORii, -SO2-Rn, -C(O)-ORn, or
-NH-C(O)-NH-Rn, ♦ Ri3, Rî3’, R15 and Ri>’ independently of one another represent a hydrogen atom, or an optionally substituted linear or branched (C j-Côjalkyl group.
ORIGINAL ♦ R.|4 represents a hydrogen atom, a hydroxy group or a hydroxy(C]-C6)alkyl group, ♦ R2j represents a hydrogen atom, a halogen atom, a linear or branched (Ci-C6)alkyl group, or a cyano group, ♦ Ra represents a hydrogen atom or a linear or branched (C i-Cr,)alkyl group, ♦ Rh represents a -O-C(O)-O-Rc group, a -O-C(O)-NRcRc’ group, or a -O-P(O)(ORC)2 group, ♦ Rc and IV independently of one another represent a hydrogen atom, a linear or branched (Ci-C$)alkyl group, a cycloalkyl group, a (Ci-C6)alkoxy(C|-C6)alkyl group, a(Ci-C6)alkoxycarbonyl(Ci-C6)alkyl group, or the substituents of the pair (Rc, R/) form together with the nitrogen atom carrying them a non-aromatic ring composed oi from 5 to 7 ring members, which may contain in addition to the nitrogen atom from 1 to 3 heteroatoms selected from oxygen and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a linear or branched (Ci-Cé)alkyl group, ♦ Cyi, Cy2, Cy3, Cy4, Cy5, Cy6 and Cy7 independently of one another, represent a cycloalkyl group, a heterocycloalkyl group, an aryl group, or a heteroaryl group, ♦ n is an integer equal to 0 or 1, it being understood that:
- “aryl” means a phenyl, naphthyl, biphenyl, indanyl or indenyl group,
- “heteroaryl” means any mono- or bi-cyclic group composed of from 5 to 10 ring members, having at least one aromatic moiety and containing from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen,
- “cycloalkyl” means any mono- or bi-cyclic non-aromatic carbocyclic group containing from 3 to 10 ring members,
- “heterocycloalkyl” means any mono- or bi-cyclic non-aromatic carbocyclic group containing from 3 to 10 ring members, and containing from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen, which may include fused, bridged or spiro ring Systems, it being possible for the aryl, heteroaryl, cycloalkyl and heterocycloalkyl groups so defined and the alkyl, alkenyl, alkynyl, alkoxy groups, to be substituted by from 1 to 4 groups
ORIGINAL selected from optionally substituted linear or branched (Ci-C6)alkyl, optionally substituted linear or branched (C2-C6)alkenyl, optionally substituted linear or branched (C2-C6)alkynyl, optionally substituted linear or branched (Ci-C6)alkoxy, optionally substituted (C|-C6)alkyl-S-, hydroxy, oxo (or A-oxide where appropriate), nitro, cyano, C(O)-OR’, -O-C(O)-R’, -C(O)-NR’R”, -O-C(O)-NR’R”, -NR’R”, -(C=NR’)-OR”, -OP(O)(OR’)2, -O-P(O)(O'M+)2, linear or branched (Ci-Côjpolyhaloalkyl, trifluoromethoxy, halogen, or an aldohexose of formula:
in which each R’ is independent;
it being understood that R’ and R” independently of one another represent a hydrogen atom or an optionally substituted linear or branched (C|-C6)alkyl group, and M represents a pharmaceutically acceptable monovalent cation.
with the proviso that
their enantiomers, diastereoisomers and atropisomers, and addition salts thereof with a 15 pharmaceutical ly acceptable acid or base.
Advantageously, the présent invention relates to compounds of formula (I) wherein:
♦ R| and R2 independently of one another represent a halogen atom, a linear or branched (C|-C6)alkyl group, a hydroxy group, a linear or branched (CrC6)alkoxy group, or the substituents of the pair (Ri, R2) form together with the carbon atoms carrying them an aromatic ring composed of from 5 to 7 ring members, which may contain
ORIGINAL
-7from l to 3 nitrogen atoms, ♦ R3 represents a hydrogen atom, a halogen atom, a linear or branched (Ci-C6)alkyi group, a hydroxy group, a linear or branched (Ci-Céjalkoxy group, or -O-alkyl(C rCej-NRi i Ri i ♦ R4 and R5 independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (Ci-Cèjalkyl group, a hydroxy group, a linear or branched (CrC6)alkoxy group, ♦ R6 and R7 independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (CpCJalkyl group, a linear or branched (Ci-C6)polyhaloalkyl group, a hydroxy group, a linear or branched (Ci-C6)alkoxy group, a cyano group, a nitro group, -alkyl(Cû-C6)-NRuRu’, -alkyl(Co-C6)-Cyi, -O-alkyl(CrC6)-Ri2, or -C(O)-NRnRn’, ♦ Rs represents a hydrogen atom, a linear or branched (Ci-C8)alkyl group, or a -CHRaRb group, ♦ R9 represents a hydrogen atom, a linear or branched (Ci-C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, -Cy2, or a halogen atom, ♦ Rio represents a hydrogen atom, a linear or branched (Ci-C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, an arylalkyl(Ci-C6) group, a cycloalkylalkyl(Ci-C6) group, a linear or branched (C|-C6)polyhaloalkyl, or -alkyl(Ci-C6)-O-Cy4, or the substituents of the pair (R9, Rio) when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen, ♦ Ru and Ru’ independently of one another represent a hydrogen atom, a linear or branched (CrC6)alkyl group, or the substituents of the pair (Ru, R11’) torm together with the nitrogen atom carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from 1 to 3 heteroatoms selected from oxygen and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a linear or branched (Ci-C^alkyl group,
ORIGINAL ♦ R12 represents -Cy, or -Cy5-alkyl(C0-C6)-Cy6, ♦ W represents a -N H- group or an oxygen atom.
it being possible for the aryl, heteroaryl, cycloalkyl and heterocycloalkyl groups so defined and the alkyl, alkenyl, alkynyl, alkoxy groups, to be substituted by from l to 4 groups selected from optionally substituted linear or branched (Cj-Céfalkyl, optionally substituted linear or branched (Ci-Ce)alkoxy, hydroxy, oxo (or /V-oxide where appropriate), -C(O)-OR’, -C(O)-NR’R”, -O-C(O)-NR’R”, -NR’R”, -O-P(O)(OR’h, -O-P(O)(O’M+)2, linear or branched (Ci-Cèjpolyhaloalkyl, halogen, or an aldohexose of formula.
in which each R’ is independent;
it being understood that R’ and R” independently of one another represent a hydrogen atom or an optionally substituted linear or branched (C, -Cô)alkyl group and M represents a pharmaceutically acceptable monovalent cation.
More especially, compounds of formula (I) to which preference is given are compounds 15 wherein n is an integer equal to l.
In another embodiment oi the invention, an advantageous possibility consists oi compounds of formula (l-a):
ORIGINAL
wherein Rb R2, R3, R4, Rs, Rr„ R?. R«, R9, R«4, Χι, X2, X3 and W are as defined for formula (I). More especially, compounds of formula (l-a) to which preference is given are
More particularly, compounds of formula (I-a) to which preference is given are compounds wherein
IO
ORIGINAL
- ΙΟ-
In another embodiment of the compounds of formula (I-b):
invention, an advantageous possibility consists of
(Lb) wherein Rb R2, R3, Ri, R5, Rô, R7, Rs, Ro, Ru, Xi, X2, X3 and W are as defined for formula (I). More especially, compounds of formula (I-b) to which preference is given are compounds wherein
ORIGINAL
In another embodiment of the invention, an advantageous possibility consists of compounds of formula (l-c):
(l-c) wherein Ri, R?, R3, R-ι, R5, Re- R7, R«, Ro, Rio, R14, Χι, X2, X3 and W are as defined for formula (I). More especially, compounds of formula (l-c) to which preference is given are compounds wherein
More particularly, compounds of formula (l-c) to which preference is given are compounds wherein
ORIGINAL
in another embodiment of the invention, an advantageous possibility consists of compounds of formula (I-d):
wherein Rh R3, Rj, R4, R5, % R7, Rs, R9, Rio, Ru, Xi, X2, X3 and W are as defined for formula (I). More especially, compounds of formula (I-d) to which preterence is given are compounds wherein
In another embodiment of the invention, an advantageous possibility consists of 10 compounds of formula (l-e):
ORIGINAL
wherein Rh R2, R3, R+, Rs, R*, R7, Rs, Rg. Ri4, Xi, X2, X3 and W are as defined for formula (I). More especially, compounds of formula (I-e) to which preterence is given are compounds wherein
Advantageously,
Compounds of formulae (I-a). (l-b), (ï-c) and (I-e) are particularly preferred. Compounds of formulae (I-a) and (I-b) are even more preferred.
IO ln another embodiment ol the invention, an advantageous possibility consists of compounds of formula (I-f):
ORIGINAL
wherein E, Rj, R2, R3, R-i- R5, Ro, R7, Rs, R9- Rio, Ri4, Xi, X2, Xî, X-h X? and W are as defined for formula (I).
Atropisomers are stereoisomers arising because of hindered rotation about a single bond, where energy différences due to steric strain or other contributors create a barrier to rotation that is hîgh enough to allow for isolation of individual conformers. For example, for compounds of formula (I-b) (the same can be donc for compounds of formula (I-a), (I-c), (I-d) and (l-e)), atropisomers are as follows:
IO Preferred atropisomer is (Sa) for compounds of formula (I-a), (I-b), (I-c) and (I-d).
Preferred atropisomer is (Ra) for compounds of formula (l-e).
Advantageously, at least one of the groups selected from R2, R3, R4 and R5 does not represent a hydrogen atom.
ORIGINAL
Preferably, R|4 represents a hydrogen atom.
R2i represents preferably a hydrogen atom, a fluorine atom, a methyl group or a cyano group. More preferably, R2) represents a hydrogen atom or a fluorine atom. Even more preferably, R2j represents a hydrogen atom.
In the preferred compounds of the invention, Ri represents a linear or branched (C|-C6)alkyl group or a halogen atom. More preferably, Ri represents a methyl group, an ethyl group. a bromine atom or a chlorine atom. Even more preferably, R| represents a methyl group or an ethyl group.
Advantageously, R2 represents a halogen atom, a hydroxy group, a linear or branched (Ci-Cô)alkoxy group. More preferably, R2 represents a methoxy group, a hydroxy group, a fluorine atom, a bromine atom or a chlorine atom. Even more preferably, R2 represents a chlorine atom.
In some preferred embodiment of the invention, when the substituents oi the pair (Rj, R2) form together with the carbon atoms carrying them an aromatic ring,
represents
R3 advantageously represents a hydrogen atom, a hydroxy group, a linear or branched (Ci-C6)alkoxy group or -O-alkyltCi-Cô/NRiiRn'. Advantageously, R3 represents -O-alkyKCi-Côj-NRuRn’.
R, and R> preferably represent a hydrogen atom.
In an advantageous embodiment, the substituents of the pair (R|, R5) are identical and the
ORIGINAL
- 16substituents of the pair (R2, R4) are identical. In the preferred compounds of the invention, the substituents of the pair (Ri, R5) are identical and represent a (Cj-C^jalkyl group, preferably a methyl group, whereas the substituents of the pair (R?, R-ι) are identical and represent a halogen atom, preferably a chlorine atom, or a hydrogen atom.
In the preferred compounds of the invention,
wherein Ri i and Ru’ are as defined for formula (l).
In another embodiment of the invention, R& represents a hydrogen atom, an optionally substituted linear or branched (Ci-Cô)alkoxy group or a -O-alkyl(C|-C6)-Ri2 group.
IO Advantageously, Rf) represents a 2,2,2-trifluoroethoxy group, a methoxy group, or a -O-alkyl(Ci-C6)-Rt2 group.
R7 preferably represents a hydrogen atom.
In the preferred compounds of the invention,
represents
wherein R12 is as defined for formula (I).
In another embodiment of the invention, an advantageous possibility consists of
ORIGINAL
- 17compounds of formula (I-g):
wherein Rj, R&, R7, Rg, R9, Rio> Rii, Rii'> R14· Xi, X2» X3, Χ4> X5, W and R are as defined for formula (I).
Preferably, Rg represents a hydrogen atom, a -CHRaRb group, an optionally substituted linear or branched (Ci-Cg)alkyl group, or a heteroarylalkyl(Ci-C6) group. Preferably, Rg represents a -CHRaRb group in which Ra represents a hydrogen atom or a methyl group and Rb represents a -O-C(O)-O-(C,-C8)alkyl group; a -O-C(O)-O-cycloalkyl group; a -O-C(O)-NRcRJ group, in which Rc and RJ independently of one another represent a IO hydrogen atom, a linear or branched (Ci-Cg)alkyl group, a (Ci-CJalkoxylCi-Côjalkyl group, a (CrC6)alkoxycarbonyl(Ci-C6)alkyl group, or the substituants of the pair (Rc, RJ) form together with the nitrogen atom carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from l to 3 heteroatoms selected from oxygen and nitrogen; or a -O-P(O)(OH)2 group. Preferred Rg groups are as follows: hydrogen; methyl; ethyl; (5-methyl-2-oxo-l,3-dioxol-4-yl)methyl; a -CHRaRb group in which Ra represents a methyl group and Rb represents a -O-C(O)-O-CH2CH3 group or a -O-C(O)-N(CH3)2 group. Even more preferably, Rg represents hydrogen.
In the preferred compounds of the invention, R9 represents a hydrogen atom, a halogen atom, a linear or branched (CrC6)alkyl group, a linear or branched (C2-C6)alkenyl group, a
ORIGINAL
- 18linear or branched (C2-C6)alkynyl group, an aryl group or a heteroaryl group. More preferably, R9 represents a prop-l -yn-l-yl group, a phenyl group or a furan-2-yl group. In a more preferred embodiment, R9 represents a prop-l-yn-l-yl group, a 4-fluorophenyl group or a 5-fluorofuran-2-yl group. Even more preferentially, R9 represents a 4-fluorophenyl group.
In the advantageous possibility consisting in compounds of formula (l-c), preferred Rio sroups are as follows: hydrogen; methyl; isopropyl; 2,2,2-triiluoroethyl; benzyl, 4-methoxybenzyI; phenethyl; 3-phenyl-propyl; cyclopropylmethyl; cyclopentylethyl; naphthalen-l-ylmethyl; 2-(naphthalen-l-yloxy)ethyl; but-2-yn-l-yl; prop-2-en-lyl; but-3-en-l-yl. In another embodiment, the substituents of the pair (R9, Rio) when grafted onto two adjacent atoms, form together with the carbon and nitrogen atoms carrying them a non-aromatic ring composed of from 5 to 6 ring members.
In the advantageous possibility consisting in compounds of formula (I-d), Rio preferably represents a hydrogen atom or a halogen atom.
In the preferred compounds of the invention. Ru and Ru independently of one another represent a linear or branched (Cj-Cojalkyl group, or the substituents of the pair (Ri i, Ru ) form together with the nitrogen atom carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a hydrogen atom, a linear or branched (CrUjalkyl group. More preferably, Ru and Ru’ represent a methyl group, or the substituents of the pair (Rlt, Ru’) form together a 4-methyl-piperazinyl group or a 4-ethyl-piperazinyl group. In a more preferred embodiment, the substituents of the pair (Ru, Rii’) form together a 4-methyl-piperazinyl group. In another preferred embodiment, Ru and Ru’ represent a methyl group.
Advantageously, Ri? represents -Cy> or -Cy5-alkyl(Co-Câ)-Cy6. Preferably, Ri? represents -Cy5 or -Cy5-Cy6.
ORIGINAL
- 19Cy5 preferably represents a heteroaryl group, particularly, a pyrimidinyl group, a pyrazolyl group, a triazolyl group, a pyrazinyl group or a pyridinyl group. More preferably, Cy5 represents a pyrimidin-4-yl group, a pyrazol-5-yl group, or a pyrazin-2-yl group. In the preferred compounds ofthe invention, Cys represents a pyrimidin-4-yl group.
In another embodiment of the invention, Cy5 represents a heteroaryl group which is substituted by an optionally substituted linear or branched (Ci-Côjalkyl group, an optionally substituted linear or branched (C]-Cô)alkoxy group, a -NR’R group, or a linear or branched (CrCôjpolyhaloalkyl group, it being understood that R’ and R” independently of one another represent a hydrogen atom or an optionally substituted linear or branched (Ci-C6)alkyl group.
Cy6 preferably represents a phenyl group.
Other compounds ofthe invention to which preference is given are those wherein.
Rl2 represents
in which p is an integer equal to 0 or l and Ri6 represents a hydrogen atom, a hydroxy group, an optionally substituted linear or branched (C|-C6)alkyl group, a linear or branched (C|-C6)alkoxy group, a -O-(CHR|7-CHR|8-O)q-R* group, a -O-P(O)(OR )2 group, a -O-P(O)(O’M+)2 group, a -O-C(O)-NR]9R2() group, a di(C|-C6)alkylamino(C|-C6)alkoxy group. a halogen atom, or an aldohexose of formula:
in which each R’ is independent;
ORIGINAL
-20it being understood that:
♦ R’ represents a hydrogen atom or a linear or branched (Cj-Côjalkyl group, ♦ R|7 represents a hydrogen atom or a (C]-C6)alkoxy(Ci-C6)alkyl group, ♦ Rie represents a hydrogen atom or a hydroxy(C|-Cé)alkyl group, ♦ Rio represents a hydrogen atom or a (Ci-C6)alkoxy(C]-C6)alkyl group, ♦ R2o represents a (Ci-Cô)alkoxy(C]-C6)alkyl group, a -(CHaX-NRnRn’ group or a -(CHaJrO-ÎCHRn-CHRjg-Ojq-R’ group, ♦ q is an integer equal to 1, 2 or 3 and r is an integer equal to 0 or 1, ♦ M+ represents a pharmaceuticaily acceptable monovalent cation.
The aldohexose according to the invention is preferably D-mannose. Preferably, the group -(CIDp-Riô is located at ortho position of the phenyl group.
Among the preferred compounds of the invention there may be mentioned:
j - (2/î)-2-{[5-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy]phenyl}-6-(4fluorophenyI)furo[2,3-r/]pyrimidin-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
- (27?)-2-{ [5- {3-chloro-2-ethyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl} -6-(4fluorophenyl)furo[2,3-(7|pyrimidin-4-yl]oxy} -3-(2- {[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy} phenyl)propanoic acid;
- A-[(5S„)-5-i 3-chloro-2-nicthyl-4-[2-(4-methylpiperazin-l -yl)ethoxv]phcnyl}-6-(4- fluorophenyl)furo[2,3-J]pyrirnidin-4-yl]-2- {[2-(2-methoxyphenyl)pyrirnidin-4-yl] methoxy} -Ώ-pheny lalanine;
(27î)-2-{ [(35^)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)-l-benzothiophen-4-yl]oxy}-3-(2-{[2-(2methoxyphenyl)pyrimidin-4-yl]methoxy} phenyl)propanoic acid;
- (2/?)-2-{[(3Sa)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl jpyrimidin4-yl]methoxy}phenyl)propanoic acid;
ORIGINAL
- (2/?)-2-{[(3Sfl)-3-{3-chloro-2-methy1-4-[2-(4-niethylpiperazin-l-yl)ethoxy]phenyl}6-fluoro-2-(4-fluorophenyl)-1 -benzofuran-4-yl]oxy} -3-(2-{ [2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy} phenyl)propanoic acid;
- (2/?)-2-{[3-{(3SÎI)-3-chloro-2-methyI-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-
2-(4-fluorophenyl)-1 -methyl-1 H-indol-4-yl]oxy} -3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy} phenyl)propanoic acid;
- (2Æ)-2-{[(3SJ-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl)2-(4-fluorophenyl)thieno[2,3-à]pyridin-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
- (27Q-2-[5-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]-6-(4fluorophenyl)-7-methyl-pyrrolo[2,3-7]pyrimidin-4-yl]oxy-3-[2-[[2-(2-methoxy phenyl)pyrimidin-4-yl]methoxy]phenyl]propanoic acid;
l-[(dimethylcarbamoyl)oxy]ethyl (2Æ)-2-{[(3Se)-3-{3-chloro-2-methyl-4-[2-(4methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fluoiOphenyl)thieno[2,3-Z>]pyridin-415 yl]oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy}phenyl)propanoate;
l-[(ethoxycarbonyl)oxy]ethyl (2/?)-2-{[(3S’a)-3-{3-chloro-2-methyl-4-[2-(4methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fluorophenyl)thieno[2,3-6]pyridin-4yl]oxy}-3-(2-{[2-(2-methoxyphenyI)pyrimidin-4-yl]methoxy}phenyl)propanoate;
- ;V-[3- {3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl} -2-(4- fluorophenyl)thieno[2,3-6]pyridin-4-yl]-2- {[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy}-D-phenylalanine;
- Af-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2-(4fluorophenyl)thieno[3,2-c]pyridin-4-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy} phenylalanine;
- 2- {[(3 Ra)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl} -2-(4fluorophenyl)imidazo[ 1,2-c]pyrimidin-5-y l]oxy} -3-(2- {[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy} phenyl)propanoic acid.
The invention relates also to a process for the préparation of compounds of formula (1), which process is characterized in that there is used as starting material the compound of 30 formula (Il-a):
ORIGINAL
-22A.
/f Zi (Il-a) wherein Z| represents bromine or iodine, Z? represents chlorine, bromine or hydroxy, and A is as defined for formula (I) in which 1 is linked to the Z2 group and 2 is linked to the Z| group, which compound of formula (Il-a) is subjected to coupling with a compound of formula (III):
Alk
(III) wherein R6, R?, Ru, W and n are as defined for formula (I), and Alk represents a linear or branched (C j-C6)alky1 group, to yield the compound of formula (IV):
Alk
(IV) wherein R6, R7, Rij. A, W and n are as defined for formula (1), and Zi and Alk are as defined before, compound of formula (IV) which is further subjected to coupling with compound of formula (V):
ORIGINAL
(V)
B rb2o orb1 wherein Ri, R2, R3, Ri and R5 are as defined for formula (I), and RB1 and RB2 represent a hydrogen atom, a linear or branched (Ci-Cô) alkyl group, or Rm and RB2 form ^ith the oxygen carrying them an optionally methylated ring, to yîeld the compound of formula (VI):
(VI) wherein R,, R2, R3 Rt, Rs, Rô, Rî, Ru, A, W and n are as defined for formula (I) and Alk is as defined before.
the Alk-O-C(O)- ester function of which compound of formula (VI) is hydrolyzed to yield the carboxylic acid, which may optionally be reacted with an alcohol of formula Rs’-OH or a chlorinated compound of formula Rg’-Cl wherein Rs represents a linear or branched (Ci-C8)alkyl group, a -CHRaRb group, an aryl group, a heteroaryl group, an arylalkyl(Ci-C6) group, or a heteroarylalkyl(Ci-C6) group, Ra and Rb are as defined for formula (I), to yield the compound of formula (I), which may be purified according to a conventional séparation technique, which is converted, if desired, into its addition salts with a
ORIGINAL
-24pharmaceutically acceptable acid or base and which is optionally separated into its isomers according to a conventional séparation technique, it being understood that at any moment considered appropriate during the course of the process described above, some groups (hydroxy, amino...) of the starting reagents or ot the synthesis intermediates can be protected, subsequently deprotected and functionalized, as required by the synthesis.
In another embodiment of the invention, compounds of formula (I) may be obtained using an alternative process, which process is characterised in that there is used as starting materiai the compound of formula (Il-b):
ζ/Λ\ν <H-b) wherein Z3 represents iodine, Z4 represents chlorine, hydroxy, and A is as defined for formula (I) in which 1 is linked to the Z4 group and 2 is linked to the Z3 group, which compound of formula (ΙΙ-b) is subjected to coupling with a compound of formula (V):
B ΒΒ2θ θ^ΒΙ wherein Ri, R?, R3, R-i and R5 are as defined for formula (I), and RBi and RB2 represent a hydrogen atom, a linear or branched (CTCô) alkyl group, or RBi and RB2 form with the oxygen carrying them an optionally methylated ring, to yield the compound of formula (VII):
ORIGINAL
R2 (VII) wherein R(, R2, R3 R4, R5 and A are as defined for formula (I), and Z4 is as defined betore, compound of formula (Vil) which is further subjected to coupling with compound of formula (III):
(III) wherein R6, R7, Ru, W and n are as defined for formula (I), and Alk represents a linear or branched (Ci-C6)alkyl group, to yield the compound of formula (VI):
(VI)
IO wherein Rh R2, R3 R4, Ri, R(>, R?, Ri4, A, W and n are as defined for formula (I) and Alk is as defined before,
ORIGINAL
-26the Alk-O-C(O)- ester function of which compound of formula (VI) is hydrolyzed to yield the carboxylic acid, which may optionally be reacted with an alcohol of formula Rg -OH or a chlorinated compound of formula Rg’-Cl wherein Rg’ represents a linear or branched (Ci-C8)alkyl group, a -CHRaRb group, an aryl group, a heteroaryl group, an arylalkyl(Ci-C6) group, or a heteroarylaikyl(Ci-C6) group, Ra and Rb are as defined for formula (I), to yield the compound of formula (I), which may be purified according to a conventional séparation technique, which is converted, if desired. into its addition salts with a pharmaceutically acceptable acid or base and which is optionally separated into its isomers according to a conventional séparation technique, it being understood that at any moment considered appropriate during the course of the process described above, some groups (hydroxy, amino...) of the starting reagents or of the synthesis intennediates can be protected, subsequently deprotected and functionalized, as required by the synthesis.
The compounds of formulae (Π-a), (ΙΙ-b), (III), (V), Rg’-OH and Rg -Cl are either commercially available or can be obtained by the person skilled in the art using conventional Chemical reactions described in the literature.
Pharmacological study of the compounds of the invention has shown that they hâve proapoptotic properties. The ability to reactivate the apoptotic process in cancerous cells is of 20 major therapeutic interest in the treatment of cancers and of immune and auto-immune diseases.
More especially, the compounds according to the invention will be useful in the treatment of chemo- or radio-resistant cancers.
Among the cancer treatments envisaged there may be mentioned, without implying any 25 limitation, treatment of cancers of the bladder, brain, breast and utérus, chronic lymphoid leukemia, cancer of the colon, esophagus and liver, lymphoblastic leukemia, acute myeloid
ORIGINAL
-27leukemia, lymphomas, melanomas, malignant haemopathies, myelomas, ovarian cancer, non-small-cell lung cancer, prostate cancer, pancreatic cancer and small-cell lung cancer.
The present invention relates also to pharmaceutical compositions comprising at least one compound of formula (I) in combination with one or more pharmaceutically acceptable excipients.
Among the pharmaceutical compositions according to the invention there may be mentioned more especially those that are suitable for oral, parentéral, nasal, per- or trans-cutaneous, rectal, perlingual, ocular or respiratory administration, especially tablets or dragées, sublingual tablets, sachets, paquets, capsules, glossettes, lozenges, suppositories, creams, ointments, dermal gels, and drinkable or injectable ampoules.
The dosage varies according to the sex, âge and weight of the patient, the administration route, the nature of the therapeutic indication, or ot any associated treatments, and ranges from 0.01 mg to l g per 24 hours in one or more administrations.
Furthermore, the present invention relates also to the combination ot a compound of formula (I) with an anticancer agent selected from genotoxic agents, mitotic poisons, antimetabolites, protéasome inhibitors, kinase inhibitors and antibodies, and also to pharmaceutical compositions comprising that type of combination and their use in the manufacture of médicaments for use in the treatment ot cancer.
Advantageously, the présent invention relates to the combination ot a compound oi formula (I) with an EGFR, inhibitor, and also to pharmaceutical compositions comprising that type of combination.
In another embodiment, the present invention relates to the combination of a compound of formula (I) with a mTOR/PI3K inhibitor, and also to pharmaceutical compositions comprising that type of combination.
In a preferred embodiment, the present invention relates to the combination ot a compound of formula (I) with a MEK inhibitor, and also to pharmaceutical compositions comprising that type of combination.
ORIGINAL
-28Preferably, the présent invention relates to the combination of a compound of formula (I) with a HER2 inhibitor, and also to pharmaceutical compositions comprising that type of combination.
Advantageously, the présent invention relates to the combination of a compound of formula (I) with a RAF inhibitor, and also to pharmaceutical compositions comprising that type of combination.
In another embodiment, the présent invention relates to the combination of a compound of formula (I) with a EGFR/HER2 inhibitor, and also to pharmaceutical compositions comprising that type of combination.
In a preferred embodiment. the présent invention relates to the combination of a compound of formula (I) with a taxane, and also to pharmaceutical compositions comprising that type of combination.
In another embodiment, the présent invention relates to the combination of a compound oi formula (I) with a protéasome inhibitor, an immunomodulator or an alkylating agent, and also to pharmaceutical compositions comprising that type of combination.
The combination of a compound of formula (I) with an anticancer agent may be administered simultaneously or sequentially. The administration route is preferably the oral route, and the corresponding pharmaceutical compositions may allow the instantaneous or delayed release of the active ingrédients. The compounds of the combination may moreover be administered in the form of two separate pharmaceutical compositions, each containing one of the active ingrédients, or in the lorm ot a single pharmaceutical composition, in which the active ingrédients are in admixture.
The compounds of the invention may also be used in combination with radiotherapy în the treatment of cancer.
Finally, the compounds of the invention may be linked to monoclonal antibodies or fragments thereof or linked to scaffold proteins that can be related or not to monoclonal antibodies.
Antibody fragments must be understood as fragments of Fv, scFv, Fab, F(ab')2, F(ab’), scFv-Fc type or diabodies, which generally hâve the same specificity of binding as the
ORIGINAL
-29antibody from which they are descended. According to the présent invention, antibody fragments of the invention can be obtained starting from antibodies by methods such as digestion by enzymes, such as pepsin or papain. and/or by cleavage of the disulfide bridges by Chemical réduction. In another manner, the antibody fragments comprised in the présent invention can be obtained by techniques of genetic recombination likewise well known to the person skilled în the art or else by peptide synthesis by means of, for example, automatic peptide synthesîzers such as those supplîed by the company Applied Biosystems, etc.
Scaffold proteins that can be related or not to monoclonal antibodies are understood to mean a protein that contains or not an immunoglobulin fold and that yields a binding capacity similar to a monoclonal antibody. The man skilled in the art knows how to select the protein scaffold. More particularly, it is known that, to be selected, such a scaffold should display several features as follow (Skerra A., J. Mol. Recogn. 2000,13, 167-187): phylogenetically good conservation, robust architecture with a well-known threedimensional molecular organization (such as, for example, crystallography or NMR), small size, no or only a low degree of post-translational modifications, easy to produce, express and purify. Such a protein scaffold can be, but without limitation, a structure selected from the group consisting in fibronectin and preterentially the tenth fibronectin type III domain (FNfnlO), lipocalin, anticalin (Skerra A., J. Biotechnol. 2001, 74(4):257-75), the protein Z derivative from the domain B of staphylococcal protein A, thioredoxin A or any protein with a repeated domain such as an ankyrin repeat (Kohl et al., PNAS 2003, 100(4), 1700-1705), armadillo repeat, leucine-rich repeat or tetratricopeptide repeat. There could also be mentioned a scaffold derivative from toxins (such as, for example, scorpion, insect, plant or mollusc toxins) or protein inhibitors of neuronal nitric oxide synthase (PIN).
The following Préparations and Examples illustrate the invention but do not limit it in any way.
General Procedures
ORIGINAL
- 30All reagents obtained from commercial sources were used without further purification. Anhydrous solvents were obtained from commercial sources and used without further drying.
Flash chromatography was performed on ISCO CombiFlash Rf 200i with pre-packed silica-gel cartridges (RediSep®.^ Gold High Performance).
Thin layer chromatography was conducted with 5 x 10 cm plates coated with Merck Type 60 F254 silica-gel.
Micro wave heating was performed in an Anton Parr Mono Wave or CEM Discover® instrument.
Préparative HPLC purifications were performed on an Armen Spot Liquid Chromatography System with a Gemini-NX* 10 μΜ Cl8, 250 mm χ 50 mm i.d. colunm running at a flow rate of 118 mL min'1 with UV diode array détection (210 - 400 nm) using 25 mM aqueous NH4HCO3 solution and MeCN as eluents unless specified otherwise.
Analytical LC-MS: The compounds of the présent invention were characterized by high performance liquid chromatography-mass spectroscopy (HPLC-MS) on Agitent HP 1200 with Agilent 6140 quadrupole LC/MS, operating in positive or négative ion electrospray ionisation mode. Molecular weight scan range is 100 to 1350. Parallel UV détection was done at 210 nm and 254 nm. Samples were supplied as a 1 mM solution in ACN, or in THF/H2O (1:1) with 5 pL loop injection. LCMS analyses were performed on two instruments, one of which was operated with basic, and the other with acidic eluents.
Basic LCMS: Gemini-NX, 3 pm, Cl8, 50 mm x 3.00 mm i.d. column at 23 °C, at a flow rate of 1 mL min'1 using 5 mM ammonium bicarbonate (Solvent A) and acetonitrile (Solvent B) with a gradient starting from 100% Solvent A and finishing at 100% Solvent B over various/certain duration of time.
Acidic LCMS: ZORBAX Eclipse XDB-C18, 1.8 pm, 50 mm x 4.6 mm i.d. column at 40 °C, at a flow rate of 1 mL min'1 using 0.02% v/v aqueous formic acid (Solvent A) and
ORIGINAL
-3I0.02% v/v formic acid in acetonitrile (Solvent B) with a gradient starting from 100% Solvent A and finishing at 100% Solvent B over various/certain duration of time.
’H-NMR measurements were performed on Bruker Avance III 500 MHz spectrometer and Bruker Avance III 400 MHz spectrometer, using DMSO-d& or CDCI3 as solvent. lH NMR data is in the form of delta values, given in part per million (ppm), using the residual peak of the solvent (2.50 ppm for DMSO-dô and 7.26 ppm for CDCI3) as internai standard. Splitting patterns are designated as: s (singlet), d (doublet), t (triplet), q (quartet), quint (quintet), m (multiplet), br s (broad singlet), dd (doublet of doublets), td (triplet of doublets), dt (doublet of triplets), ddd (doublet of doublet of doublets).
Combination gas chromatography and low resolution mass spectrometry were performed on Agilent 6850 gas chromatograph and Agilent 5975C mass spectrometer using 15 m* 0.25 mm column with 0.25 pm HP-5MS coating and hélium as carrier gas. Ion source: ΕΓ, 70 eV, 230°C, quadrupole: 150°C, interface: 300°C.
HRMS were determined on a Shimadzu IT-TOF, ion source température 200°C, ESI +/-, ionization voltage: (+-)4.5 kV. Mass resolution min. 10000.
Elementary analyses were performed on a Thermo Flash EA 1112 Elemental Analyzer.
List of abbreviations
Abbreviation N a me
2-Me-THF 2-methyl-tetrahydrofurane
abs. absolute
Ac acetyl
AIBN 2- [( 1 -cyano-1 -methyl -ethy 1 )azo] -2-methyl-propaneni t ri le
AtaPhos bis(di-/É77-butyl(4-dimethylaminophenyl)phosphine) dichloropalladium(Il)
B1NAP (2,2'-bis(diphenylphosphino)-1,1 ’-binaphthyl)
ORIGINAL
-32-
cc. concentrated
dba dibenzylideneacetone
DCM methylene chloride
DEAD diethyl azodicarboxylate
DEE diethyl ether
DIPA diisopropylamine
DIPEA diisopropylethylamine
DMA dimethylacetamide
DME l ,2-dimethoxyethane
DMF d i methy l form am i de
DMSO dimethyl sulfoxide
dppf l, r-bis(diphenylphosphino)ferrocene
DTAD di-ie/7-butyl azodicarboxylate
EDC.HCl V-(3-dimethylaminopropyl)-7V'-ethylcarbodiimide hydrochloride
eq. équivalent
Et ethyl
H1L1C hydrophilic interaction liquid chromatography
HMDS hexamethyldisilazane
’Pr isopropyl
LDA lithium diisopropylamide
MCPBA //ze/a-chloroperoxybenzoic acid
Me methyl
MeCN acetonitrile
MTBE methyl ze/7-butyl ether
MW microwave
NBS A7-bromosuccinimide
nBu n-butyl
NCS V-chlorosuccinimide
Ph phenyl
PPA polyphospholic acid
rac. racemic
ORIGINAL
- jj-
r.t. room température
S2Me2 dimethyl disulfide
SPhos 2-dicyclohexylphosphino-2',6'-dimethoxybiphenyl
TBAF tetrabutyl ammonium fluoride
TBAOH tetrabutyl ammonium hydroxyde
lBu rerCbutyl
TEA triethylamine
TFA trifluoroacetic acid
THF tetrahydrofurane
TIPSCl triisopropyIsilyl chloride
TLC thin layer chromatography
Ts tosyl
X-Phos 2-dicyclohexylphosphino-2’,4',6’-triisopropylbiphenyl
General Procedure la l eq. Préparation la, 2 eq. from the appropriate lactic ester derivative, 10 mL/mmol ‘BuOH and 5 eq. CS2CO3 were placed in a flask and stirred at 55 °C until no further conversion was observed. Then the mixture was concentrated under reduced pressure, neutralized with IM aqueous HCl solution, diluted with brine and extracted with EtOAc. The combined organic phases were dried over Na^SO^, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents unless otherwise stated.
General Procedure 1b l eq. Préparation la, 2 eq. from the appropriate amino acid derivative, IO mL/mmol DMSO and 3 eq. K2CO3 were placed in a flask and stirred at 45 °C until no further conversion was observed. Then the mixture was neutralized with l M aqueous HCl solution, diluted with brine and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via HILIC chromatography unless otherwise stated.
ORIGINAL
- 34General Procedure II
Step A l eq. from the appropriate 5-bromo-furo[2,3-d]pyrimidyl-lactic ester dérivative, 1.25 eq. from the appropriate boronic acid dérivative, 10 mol% AtaPhos and 3 eq. CS2CO3 were dissolved in a l:l mixture of dioxane and water (10 mL/mmoI 5-bromo-furo[2,3-iZ] pyrimidyl-lactic ester dérivative) and stirred at 105 °C in a MW reactor until no further conversion was observed. Then the mixture was neutralized with IM aqueous HCl solution, diluted with brine and extracted with THF. The combined organic phases were dried over MgSÛ4, fîltered and the filtrate was concentrated under reduced pressure. The 10 crude product was purified using preparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents.
Step B
The obtained intermediate was dissolved in a l:l mixture of dioxane and water (25 mL/mmol) and 10 eq. LiOHxH2O was added. The mixture was stirred at r.t. until no 15 further conversion was observed. Then it was diluted with brine, neutralized with 2M aqueous HCl, extracted with DCM. The combined organic phases were dried over Na2SC>4, fîltered and the filtrate was concentrated under reduced pressure. The diastereoisomers were purified and separated by preparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents.
General Procedure III l eq. from the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative, 3 eq. from the appropriate amino acid dérivative, 10 mL/mmol DMSO and 4 eq. K2CO3 were stirred at 15O°C until no further conversion was observed. The mixture was acidified with IM aqueous HCl solution, the precipitate was fîltered and purified via preparative reversed 25 phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents.
General Procedure IVa l eq. from the appropriate 5-bromo-pyrrolo[2,3-i/]pyrimidine dérivative, 3 eq. from the appropriate boronic acid dérivative, 3 eq. TBAOH, 0.2 eq. palladium acetate, 0.4 eq.
ORIGINAL
- 35tricyclohexylphosphonium tetrafluoroborate and 3.5 mL/mmol DME were stirred under N2 atmosphère at 120 °C in a MW reactor until no further conversion was observed. Then the mixture was filtered through Celite and washed with MTBE and water. The layers were separated, the aqueous layer was washed with MTBE. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 40 mM aqueous NH4()Ac (pH = 4) solution and MeCN as eluents.
General Procedure IVb l eq. from the appropriate 5-iodo-pyrrolo[2,3-/7] pyrimidine dérivative, 3 eq. from the appropriate boronic acid dérivative, 3 eq. TBAOH, 0.2 eq. palladium acetate, 0.4 eq. butyidi-l-adamantylphosphine and 7 mL/mmol DME were stirred under N2 atmosphère at reflux until no further conversion was observed. Then the mixture was filtered through Celite and concentrated under reduced pressure. The residue was purified via flash chromatography using DCM and MeOH as eluents.
General Procedure V l eq. from the appropriate benzofuran-4-ol dérivative, 2.5 eq. from the appropriate lactic ester dérivative, 2.5 eq. DTAD and 2.5 eq. PPh3 were dissolved in dry toluene (20 mL/mmol) and stirred at 55 °C until no further conversion was observed. Then the mixture was concentrated and the residue was purified via flash chromatography using heptane and EtOAc as eluents.
General Procedure VI eq. from the appropriate 3-bromo-benzofuran dérivative, 2 eq. from the appropriate boronic acid dérivative, 2 eq. Cs2CO3, 10 mol% Ataphos, 1.5 eq. tri-/e/7butylphosphonium tetrafluoroborate and THF (10 mL/mmol) and water (4 mL/mmol) were stirred under N2 atmosphère at 110°C in a MW reactor until no further conversion was observed. Then the mixture was acidified with IM aqueous HCl solution and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 25 mM aqueous NH4HCO3
ORIGINAL
-36solution and MeCN as eluents. The obtained intermediate was dissolved in dioxane:water l:l (10 mL / mmol), 10 eq. LiOHxH2O was added and the mixture was stirred at r.t. until no further conversion was observed. Then the mixture was diiuted with water, acidified with IM aqueous HCl solution and extracted with DCM. The combined organic phases 5 were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure.
The crude product was purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents.
Préparation la: 5-bromo-4-chloro-6-(4-fluorophenyl)furo[2,3-rf]pyrimidine
Step À ; 2~(4-fluorobenzoyl)propanedinitrile
IO 8l mL IM NaOEt solution in EtOH (8l mmol) was cooled to 0 °C and 6.14 g malononitrile (93 mmol) was added. The mixture was stirred at 0 °C for l hour, then 16.8 g 2-bromo-l-(4-fluorophenyl)ethanone (77.4 mmol) was added. The mixture was stirred at 0 °C for l hour, then at r.t. until no further conversion was observed. The volatiles were removed under reduced pressure, and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 2-(4-fluorobenzoyl)propanedinitrile. ’H NMR (400 MHz. CDCI3): 8.1 (m, 2H), 7.24 (m, 2H), 4.41 (t, IH), 3.75 (d, 2H)
Step B: 2-amino-5-(4-fluorophenyl)furan-3-carbonitrile
6.56 g 2-(4-fluorobenzoyl)propanedinitrile (28.5 mmol) was dissolved in 140 mL AcOH and 6 g Amberlite 15H+ was added. The mixture was stirred at 90 °C until no further 20 conversion was observed. Then the mixture was filtered, the filtrate was concentrated under reduced pressure. The residue was recrystallized from DCM to obtain 2-amino-5-(4fluorophenyl)furan-3-carbonitrile. lH NMR (400 MHz, DMSO-dè): 7.69 (m, 2H), 7.24 (m, 211),6.96 (s, IH)
Step C: 6-(4-fluorophenyl)-3}3-furo[2,3-à]pyrimidin-4-one
1290 mg 2-amino-5-(4-lluorophenyl)furan-3-carbonitrile (6.38 mmol) and 25.5 mL acetic formic anhydride were placed in a flask and stirred at r.t. for 30 minutes. Then, the volatiles were evaporated under reduced pressure. The residue was dissolved in 51 mL
ORIGINAL
-37AcOH and heated in a MW reactor at l60°C for 30 minutes, then at 18O°C for 15 minutes. Then the mixture was cooled to r.t., and the precipitate was filtered to obtain 6-(4-fluorophenyl)-37/-furo[2,3-i/]pyrimidin-4-one. 'H NMR (500 MHz, DMSO-dû): 12.66 (br s, IH), 8.15 (s, IH), 7.99 (m, 2H), 7.47 (s, IH), 7.33 (m, 2H)
Step D: 5-bromo-6-(4-fluorophenyl)-3}3-furo[2,3-à]pyrimidin-4-one
1704 mg 6-(4-fluorophenyl)-3//-furo[2,3-i/]pyrimidin-4-one (7.4 mmol) was dissolved in 74 mL AcOH, then 1182 mg bromine (7.4 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was then filtered, the filtrate was concentrated under reduced pressure. The residue was digerated with 15 mL MeOH, filtered and dried on air to obtain 5-bromo-6-(4-fluorophenyl)-3L/-furo[2,3-</]pyrimidin-4one. MS: (M-H)+ = 309.0
Step E: Préparation la
1680 mg 5-bromo-6-(4-fluorophenyl)-3/f-turo[2,3-i7]pyriniidin-4-one (5.44 mmol) was dissolved in 12.7 mL POCl3 (136 mmol) and 690 pi. DMA (5.44 mmol) was added. The mixture was stirred at 110 °C until no further con\rersion was observed. The mixture was then cooled to 0 °C and poured into ice-water. The crude product was isolated by filtration and purified via flash chromatography using heptane and EtOAc as eluents to obtain Préparation la. '14 NMR (400 MHz. DMSO-db): 8.87 (s, 114), 8.16 (m, 214), 7.47 (m, 2H)
Préparation lb: 5-bromo-4-chloro-6-ethyl-7H-pyrrolo|2,3-i/|pyrimidine
Step A: 6-amino-5-[(2-elhyl-l,3-dioxolan-2-yl)rneihylJpyriniidin-4-oI
257 mg 6-amino-5-[(2-ethyl-l ,3-dioxolan-2-yl)methyl]-2-sulfanyl-pyrimidin-4-ol (0.1 mmol), 0.77 mL aqueous cc. NH3 solution, 768 mg Raney-Ni and 11 mL water were placed in a flask under N2 atmosphère and heated to reflux until no further conversion was observed. The warm reaction mixture was then filtered through Celite and washed with warm water. The filtrate was concentrated under reduced pressure. The crude product (6-amino-5-[(2-ethyl-l,3-dioxolan-2-yl)niethyl]pyrimidin-4-oI) was used without further purification.
ORIGINAL
-38*H NMR (400 MHz. DMSO-d6) δ: 11.44 (br s, IH), 7.70 (s, 1 H), 6.07 (s, 2H), 3.89 (m, 4H), 2.62 (s, 2H), 1.53 (m, 2H), 0.81 (t. 3H)
MS (M+H): 226.2
Step B: 6-ethyl- 7W-pyrrolo[2,3-d]pyrimidin-4-ol
4.193 g 6-amino-5-[(2-ethyl-l,3-dioxolan-2-yl)methyl]pyrimidin-4-ol (18.6 mmol) was dissolved in 280 mL 0.2M aqueous HCl solution. The mixture was stirred at r.t. until no further conversion was observed. The precipitate was fîitered, washed with water and dried to obtain 6-ethyl-7//-pyrrolo[2,3-i/]pyrimidin-4-ol.
'H NMR (400 MHz, DMSO-d6) δ: 11.67 (s, IH), 7.75 (s, IH), 6.12 (t. IH), 2.56 (m. 2H),
1.21 (t,3H)
MS (M+H): 164.2
Step C: 5-bromo-6-ethyl- 7Y{-pyrrolo[2,3-d]pyrimidin-4-ol
1.63 g 6-ethyl-7/7-pyrrolo[2,3-i/]pyrimidin-4-ol (10 mmol) was dissolved in 20 mL DMF and cooled to 0 °C. 1 mL bromine (20 mmol) was added and the mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with water and aqueous NaiSiCh solution and extracted with DCM. The combined organic layers were washed with brine, dried over MgSC>4, fîitered and the filtrate was concentrated under reduced pressure to obtain 5-bromo-6-ethyl-7F/-pyrrolo[2,3-iZ]pyrimidin-4-ol.
'H NMR (400 MHz, DMSO-d6) δ: 12.08 (s, IH), 11.83 (s, IH), 7.80 (d. IH), 2.60 (q, 2H),
1.16 (t,3H)
MS (M+H): 243.8
Step D: Préparation 1b
1936 mg 5-bromo-6-ethyl-777-pyrrolo[2,3-i/]pyrimidin-4-ol (8 mmol), 4.5 mL POCI3 and 969 mg ÀGV-dirnethylaniline (8 mmol) were placed in a fîask and stirred at 100 °C until no further conversion was observed. The mixture was then poured into ice-water and extracted with DCM. The combined organic layers were washed with brine, dried over MgSÛ4, fîitered and the filtrate was concentrated under reduced pressure to obtain Préparation 1b.
'H NMR (400 MHz, CDC13) δ: 9.79 (s, 1 H), 8.59 (s, 1 H), 2.91 (q, 2H), 1.37 (t, 3H)
ORIGINAL
-39MS (M+H): 260.0
Préparation le: 3-bromo-2-(4-fluorophenyl)benzofuran-4-oi
Step A: 2-(4-fluorophenyl)benzofuran-4-ol
2.37 g 2-bromoresorcinol (12.5 mmol) was dissolved in 30 mL dry THF under N2 atmosphère and 4.17 mL TEA (30 mmol) and 1.92 mL AcCI (27 mmol) were added respectively. After stirring the mixture for 5 minutes, 2.4 g l-ethynyl-4-fiuorobenzene (20 mmol), 561 mg Pd(OAc)2 (2.5 mmol), 1.45 g tri-rm-butylphosphonium tetrafluoro borate (5 mmol), 476 mg Cul (2.5 mmol) and 10 mL dry DIP A were added and the mixture was stirred at 80 °C until no further conversion was observed. Then 2 g LiOHxH2O was added and the mixture was stirred at 80 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain 2-(4-fluorophenyl)benzofuran-4-ol. lH NMR (400 MHz, DMSO-dô) δ: 10.00 (s, IH), 7.91 (m, 2H), 7.38 (s, IH), 7.31 (t, 2H), 7.10 (t, IH), 7.04 (d, lH), 6.63 (dd, IH)
Step B: [2-(4-fluorophenyl)benzofuran-4-yl] acetate
456 mg 2-(4-fluorophenyl)benzofuran-4-ol (2 mmol) was dissolved in 10 mL dry THF then 156 pL AcCI (2.2 mmol) and then 306 pL TEA (2.2 mmol) were added carefully. The mixture was stirred under N2 atmosphère until no further conversion was observed. The solvent was then removed under reduced pressure, and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain [2-(4-fluorophenyl) benzofuran-4-yl] acetate. 1H NMR (400 MHz, CDCI3) δ: 7.84 (m, 2H), 7.42 (d, IH), 7.28 (t, IH), 7.15 (t,2H), 7.02 (d, IH), 6.86 (s, 1 H), 2.42 (s, 3H)
Step C: [3-bromo-2-(4-fluorophenyl)benzofuran-4-yl] acetate
688 mg [2-(4-fluorophenyl)benzofuran-4-yl] acetate (2.54 mmol) and 589 mg NBS (3.31 mmol) were dissolved in 20 mL MeCN and stirred at 70 °C until no further conversion was observed. The solvent was then removed under reduced pressure, and the residue was purified via flash chromatography using heptane and EtOAc as eluents to
ORIGINAL
-40obtain [3-bromo-2-(4-fluorophenyl)benzofuran-4-yl] acetate. ’H NMR (400 MHz, CDCl3) δ: 8.11 (m, 2H), 7.44 (dd, IH), 7.34 (t, IH), 7.19 (m, 2H), 7.00 (dd, IH), 2.45 (s, 3H)
Slep D: Préparation le
175 mg [3-bromo-2-(4-iluorophenyl)benzofuran-4-yl] acetate (0.5 mmol) and 150 pL IM NaOEt in EtOH solution and 5 mL EtOH were stirred at r.t. under N2 atmosphère until no further conversion was observed. The mixture was diluted with 50 mL aqueous cc. NH4CI solution and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated to give Préparation le. 'H NMR (400 MHz, DMSO-df,) δ: 10.16 (br s, IH), 8.08 (m, 2H), 7.38 (m, 2H), 7.17 (t, IH), 7.08 (d, IH), 6.70 (d,lH)
Préparation Id: 3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-ol
Step A: 5-fluoro-2-iodo-benzene-l,3-diol
3.81 g (29.7 mmol) 5-fluorobenzene-l,3-diol was dissolved in 600 mL water and 8.08 g (31.8 mmol) iodine was added at 0 °C and the mixture was stirred for 30 minutes. Then pH was adjusted to 3 with NaHCO3 solution and the mixture was stirred until no further conversion was observed. Then pH was adjusted to 8 (with NaHCO3 solution), 20 g Na2S2O3 was added and the mixture was extracted with EtOAc. Combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated and purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-fluoro-2-iodo-benzene-
1,3-diol. 'H NMR (400 MHz, DMSO-d6): 10.54 (s, 2H), 6.19 (d, 2H)
Step B: (3-acetoxy-5-fhioro-2-iodo-phenyl) acetate
4.78 g 3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-ol (18.8 mmol) was dissolved in 150 mL THF and 5.70 g TEA (56.5 mmol) was added, then 4.267 g Ac2O (41.4 mmol) was added dropwise at r.t. The mixture was stirred until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain (3-acetoxy-5-fluoro-2-iodophenyl) acetate. 'H NMR (400 MHz, DMSO-d6): 7.24 (d, 2H), 2.34 (s, 6H)
ORIGINAL
-41Step C: 6-fluor0-2-(4-fliiorophenyl)benzofuran-4-ol
5.9 g (3-acetoxy-5-fluoro-2-iodo-phenyl) acetate ( 17.45 mmol) was dissolved in 70 mL dry THF and 70 mL dry DIPA under N2 atmosphère, then 3.77 g l-ethynyl-4-fluorobenzene (31.4 mmol), 587 mg Pd(OAc)2 (2.62 mmol), 1.52 g tri-/er/-butylphosphonîum tetrafluoroborate (5.24 mmol), and 500 mg Cul (2.62 mmol) were added and the mixture was stirred at 60 °C until no further conversion was observed. Then 2.93 g LiOHxH2O was added and the mixture was stirred at 60 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain 6-fluoro-2-(4-fluorophenyl)benzofuran-4-ol. ’H NMR (400 MHz, DMSOd6): 10.60 (s, IH), 7.89 (m, 2H), 7.38 (s, IH), 7.32 (m, 2H), 6.99 (m, IH), 6.48 (dd, IH)
Step D: [6-jhtoro-2-(4-fluoraphenyl)benzofuran-4-yl] acetate
2.49 mg 6-fluoro-2-(4-fluorophenyl)benzofuran-4-ol (10.1 mmol) was dissolved in 50 mL dry THF then 791 pL AcCl (11.1 mmol) and then 1.55 mL TEA (11.1 mmol) were added carefully. The mixture was stirred under N2 atmosphère until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain [6-fluoro-2-(4fluorophenyl)benzofuran-4-yl] acetate. ’H NMR (400 MHz, DMSO-d6): 7.95 (m, 2H), 7.57 (m, IH), 7.46 (s, IH), 7.37 (m, 2H), 7.09 (dd, IH), 2.40 (s, 3H)
Step E: [3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl] acetate
2.96 g [6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl] acetate (10.27 mmol) and 2.28 g NBS (12.84 mmol) were dissolved in 120 mL MeCN and stirred at 60 °C until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain [3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl] acetate. [H NMR (400 MHz, DMSO-d6): 8.07 (m, 2H), 7.69 (dd, IH), 7.44 (m, IH), 7.19 (m, 2H), 7.09 (dd, IH), 2.41 (s, 3H)
Step F: Préparation Id
ORIGINAL
3,35 g [3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl] acetate (9.12 mmol) and
8.67 mL IM NaOEt in EtOH solution and 90 mL EtOH were stirred at r.t. under N2 atmosphère until no further conversion was observed. The mixture was diluted with 50 mL aqueous cc, NH4CI solution and extracted with DCM. The combined organic phases were dried over Na2SÛ4, filtered and the filtrate was concentrated to give Préparation Id. 'H NMR (400 MHz. DMSO-dti): 10.78 (s, IH), 8.06 (m, 2H), 7.40 (m, 2H), 7.06 (dd, IH), 6.54 (dd, IH)
Préparation 2a: Ethyl (2JÎ)-2-acetoxy-3-(2-hydroxyphenyI)propanoate and
Préparation 2b: Ethyl (2S)-2-acetoxy-3-(2-hydroxyphenyl)propanoatc
Step A: [2-(Bromomethyl)phenyl]acetate
60.07 g 2-methylphenyl acetate (400 mmol) and 106.8 g NBS (600 mmol) were placed in a l L flask. 500 mL cyclohexane was added. and then with intensive stirring 3.284 g AIBN (20 mmol) was added over 30 minutes. The mixture was stirred at 80 °C until no further conversion was observed, then cooled to r.t. The precipitate was filtered off and washed with cyclohexane. The mother liquor was concentrated under reduced pressure, and the crude product was used in Step B without further purification.
Step B: Préparations 2a and 2b
23.10 g anhydrous LiCl (545 mmol) and 65.36 g anhydrous ZnCl2 (479,6 mmol) were placed in a 2 L flask, then dried at 160 °C under O.l mmHg for l hour. After cooling to r.t. under argon atmosphère, 26.49 g magnésium tumings ( 1090 mmol) and l L dry pre-cooled (0 °C) THF were added, The resuiting mixture was immersed into an ice-bath, and then stirred for 30 minutes. 100 g [2-(bromomethyl)phenyl] acetate (crude product from Step A, -436 mmol) was dissolved in 120 mL dry THF and was added to the precooled inorganics over 15 minutes. After addition of the reagent the resuiting mixture was stirred for 45 minutes while keeping the température between 0-5 °C. Then 64.82 mL ethyl 2-oxoacetate (654 mmol, 50 % in toluene) was added over 5 minutes and the resuiting mixture was stirred for another 15 minutes. The remaining inorganics were removed by filtration, and the filtrate was diluted with 500 mL MeOH. It was stirred until the
ORIGINAL
-43intramolecular acetyl group migration from the phenolic oxygen to the alkyl oxygen was complété. Then 30 mL acetic acid was added the volatiles were evaporated under reduced pressure. 350 mL water was added to the residue and it was extracted with EtOAc. The combined organic layers were washed with saturated aqueous NaHCOj and with brine, and then dried over MgSO4, filtered and the fîltrate was concentrated under reduced pressure. Then 100 mL hexane was added and it was stirred for 30 minutes at 0 °C. The formed whîte crystals were collected by filtration and washed with hexane. ’H NMR (500 MHz. DMSO-dô) δ: 9.53 (s, IH), 7.06 (t, IH), 7.04 (d, IH), 6.79 (d, IH), 6.71 (t, IH), 5.10 (dd, IH), 4.05 (q, 2H), 3.06 (dd, IH), 2.94 (dd, IH), 2.00 (s, 3H), l.09 (t, 3H)
The enantiomers were separated via chiral chromatography. Column: OD; Eluents: heptane / EtOH; the enantiomer eluting earlier was collected as Préparation 2b with
99.8 % ee and the enantiomer eluting later was collected as Préparation 2a with 99.9 % ee.
Préparation 2c: Ethyl (2Æ)-2-hydroxy-3-[2-|]2-(2-methoxyphenyl)pyriniidin-4-yl] methoxy] phenyl] propanoate
Step A: (2R)-2-hydroxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]niethoxy]phenyl] propanoic acid
30.3 g Préparation 2a (120 mmol), 38.9 g Préparation 5b (180 mmol) and 47.2 g triphenyl phosphine (180 mmol) were dissolved in 120 mL dry toluene, then 82 mL DEAD (180 mmol, 40 % in toluene) was added. The mixture was stirred at 50 °C under nitrogen atmosphère until no further conversion was observed. The volatiles were evaporated under reduced pressure. Then 300 mL DEE was added, the mixture was sonicated and filtered, washed with DEE. The fîltrate was concentrated under reduced pressure. The residue was dissolved in 125 mL THF, then 24 g NaOH (0.6 mol) dissolved in 125 mL water was added. The mixture was stirred at 50 °C until no further conversion was observed. The pl i was set to 5 with cc. HCl, and the volatiles were removed under reduced pressure. 100 mL water and 350 mL DCM were added, the mixture was stirred at 0 °C and the precipitate was filtered, washed with cold water and DCM and dried under reduced pressure to obtain (2Æ)-2-hydroxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoic acid. 'H-NMR (400 MHz, DMSO-df,) δ: 8.88 (d, IH), 7.80 (d, IH), 7.55 (dd, IH), 7.49-
ORIGINAL
-447.44 (m, IH), 7.26 (dd, IH), 7.17-7.H (m, 2H), 7.06 (t, IH), 6.98 (d, IH), 6.88 (t, IH),
5.22 (s, 2H), 4.50 (d, IH), 3.81 (dd. IH), 3.77 (s, 3H), 3.73 (dd, IH), 2.44 (dd, IH)
Step B: Préparation 2c
51.7 g (27?)-2-hydroxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoic acid (136 mmol) was dissolved in 520 mL EtOH, then 20 mL cc. H7SO4 was added. The mixture was stirred at 60 °C until no further conversion was observed. Then it was diluted with water, neutralized with aqueous saturated NaHCCfe solution and extracted with dichloromethane. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure and purified via flash chromatography using EtOAc and MeOH as eluents to obtain Préparation 2c. HRMS calculated for C23H24N2O5: 408.1685, found: 409.1757 (M+H)
Préparation 2d: Ethyl (2A)-2-hy<lrox)-3-|2-||2-(2-inethoxyphenyl)pyrinii<iin-4-vl| methoxy]phenyl]propanoate
Préparation 2d was synthesized the way as Préparation 2c, but starting from Préparation 2b instead of Préparation 2a.
Préparation 2e: Ethyl (27ï)-2-hydroxy-3-(2-methoxyphenyl)propanoate and
Préparation 2f: Ethyl (2S)-2-hydroxy-3-(2-methoxyphcnyl)propanoate
The enantiomers of ethyl 2-hydroxy-3-(2-methoxyphenyl)propanoate were separated via chiral chromatography; Column: AD, Eluent: 2-PrOH; the enantiomer eluting earlier was collected as Préparation 2e with 99.8 % ee. The enantiomer eluting later was collected as Préparation 2f with 97.8 % ee.
Préparation 2g: Ethyl (27?)-2-hydroxy-3-[2-(pyrazin-2-ylinethoxy)phenyI|propanoate
Step A: Ethyl (2R)-2-acetoxy-3-[2-(pyrazin-2-ylmelhoxy)phenyl]propanoate eq. Préparation 2a, 2 eq. of pyrazin-2-ylmethanol and 2 eq. triphenylphosphine were dissolved in dry toluene (0.2M for the phénol), then 2 eq. DT AD was added. The mixture was stirred at 50 °C under nitrogen atmosphère. After reaching an appropriate conversion
ORIGINAL
-45the volatiles were removed under reduced pressure. The crude intermediate was purified via flash chromatography using heptane and EtOAc as eluents to obtain ethyl (2R)-2acetoxy-3-[2-(pyrazin-2-ylmethoxy)phenyl]propanoate.
Step B: Préparation 2g
Ethyl (2Æ)-2-acetoxy-3-[2-(pyrazin-2-ylmethoxy)phenyl]propanoate was dissolved in éthanol (0.5M) then 2 mol% NaOEt solution (l.OM in éthanol) was added. The resulting mixture was stirred at r.t. Additional NaOEt solution was added if conversion was not complété. The mixture was concentrated to half of its volume, then water and brine was added, and it was extracted with EtOAc. The combined organics were dried over Na2SÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using DCM and methanol as eluents to obtain Préparation 2g. 'H NMR (400 MHz, DMSO-d6) δ: 8.88 (s, IH), 8.64 (dd, 2H), 7.22-7.16 (m, 2H), 7.06 (d, IH), 6.89 (t, IH), 5.46 (d, IH), 5.27 (dd, 2H), 4.29 (dq, lH),4.00 (q, 2H), 3.09 (dd, IH), 2.79 (dd, IH), 1.08 (t, 3H)
Préparation 2h: Ethyl (2S)-2-hydroxy-3-[2-(2,2,2-trifluoroethoxy)phenyl|propanoate
Slep A: Ethyl (2S)-2-hydroxy-3-(2-hydroxyphenyl)propanoate
13,633 g Préparation 2b (54 mmol) was dissolved in 200 mL dry EtOH, then 30 mL NaOEt solution (IM in EtOH) was added and the mixture was stirred at r.t. If needed, the addition of the NaOEt solution was repeated until the cleavage of the acetyl group was complété. The mixture was diluted with 600 mL water and it was extracted with EtOAc. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The obtained ethyl (2S)-2-hydroxy-3-(2hydroxyphenyl)propanoate was used in the next step without further purification.
Slep B: Préparation 2h
9.18 g ethyl (2Sr)-2-hydroxy-3-(2-hydroxyphenyl)propanoate (43.7 mmol) was dissolved in 130 mL dry DMF, then 6.040 g K2CO3 (43.7 mmol) was added. After 5 minutes stirring
7.7 mL 2,2,2-trifluoroethyl trifluoromethanesulfonate (48 mmol) was added over 5 minutes. The resulting mixture was stirred until no further conversion was observed. The
ORIGINAL
-46reaction mixture was diluted with brine, then extracted with EtOAc. The combined organic layers were dried over Na3SO4, fîltered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents. 'H NMR (500 MHz, DMSO-d6) δ: 7.23 (t, IH), 7.18 (d, IH), 7.06 (d, IH), 6.95 (t, IH), 5.50 (d, IH), 4.75 (q, 2H), 4.22 (m, IH), 4.02 (q, 2H), 3.00 (dd, IH),
2.76 (dd, IH), l.09 (t, 3H)
Préparation 2i: (27?)-2-amino-3-|2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy| phenyl]propanoic acid
Step A: ethyl (2R)-2-amino-3-(2-hydroxyphenyl)propanoate hydrochloride
653 mg (27?)-2-amino-3-(2-hydroxyphenyl)propanoic acid hydrochloride (3.0 mmol) was dissolved in 6 mL HCl (I.25 M in EtOH) and stirred at 60 °C until no further conversion was observed. Then the reaction mixture was carefully diluted with 10% aqueous NaHCO3 solution and extracted with DCM. The combined organic phase was dried over Na2SO4, fîltered and the filtrate was concentrated under reduced pressure. The product should be stored in freezer.
’H NMR (500 MHz, DMSO-d6) Ô: 7.05-6.95 (m, 2H), 6.72 (dm, IH), 6.69-6.63 (m, IH), 4.02 (q, 2H), 3.65 (dd. IH), 2.84 (dd, IH), 2.78 (dd, IH), l.12 (t, 3H)
HRMS calculated for C,, H j 5NO3: 209.1052; found: 210.1128 (M+H)
Step B: ethyl (2R)-2-amino-3-[2-[[2-(2-meihoxyphenyl)pyriniidin-4-yl]methoxy]phenyl] propanoate
3.96 g ethyl (2Æ)-2-amino-3-(2-hydroxyphenyl)propanoate hydrochloride (18.9 mmol) was dissolved in 200 mL dry toluene, then 5.69 g PPh3 (21.7 mmol), 4.69 g Préparation 5b (21.7 mmol) were added and the mixture was heated to 35 °C, then 5.0 g DTAD (21.7 mmol) was added and the mixture was stirred at 45 °C until no further conversion was observed. Then the mixture was concentrated under reduced pressure and purified via flash chromatography using EtOAc and MeOH as eluents.
'H NMR (500 MHz, DMSO-d6) δ: 8.92 (d, IH), 7.61 (d, IH), 7.55 (dd, IH), 7.46 (td, IH),
7.20 (td, IH), 7.17 (dd, IH), 7.15 (dd, IH), 7.06 (td, IH), 7.04 (dd, IH), 6.91 (td, IH),
ORIGINAL
-475.27/5.23 (d. 2H), 4.01 (q, 2H), 3.76 (s, 3H), 3.68 (dd, IH), 3.08 (br, 2H), 3.03/2.83 (dd, 2H), 1.07 (t, 3H)
HRMS calculated for C23H25N3O4: 407.1845; found: 408.1928 (M+H)
Step C: Préparation 2i
3.20 g ethyl (2/?)-2-ammo-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyI] propanoate (7.85 mmol) was dissolved in 10 mL THF, then 10 mL water and 420 mg LiOHxHjO (10 mmol) were added and the mixture was stirred at r.t. until the hydrolysis was complété. Then it was diluted with water and neutralized with 2 M aqueous HCl solution. The formed precipitate was filtered, washed with water and dried to obtain Préparation 2i.
’H NMR (500 MHz, DMSO-d6) δ: 8.88 (d, IH), 7.82 (d, IH), 7.54 (dd, IH), 7.47 (m, IH), 7.27 (dd. IH), 7.23 (t, IH), 7.16 (d. IH), 7.06 (t. IH), 7.05 (d, IH), 6.93 (t, IH), 5.26 (s, 2H), 3.76 (s, 3H), 3.59 (dd, IH), 3.49/2.83 (dd, 2H)
HRMS calculated for C21H21N3O4: 379.1532; found: 380.1610 (M+H)
Préparation 3a: 2-Chloro-3-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl) phénol
Step A: (4-Bromo-2-chloro-phenoxy)-tnmelhyl-silane
20.8 g 4-bromo-2-chloro-phenol (100 mmol) was dissolved in 150 mL dry THF then
24.2 g HMDS (150 mmol) was added. The reaction mixture was stirred at 85 °C under argon atmosphère for 1.5 hours then concentrated under reduced pressure. The resulted crude product was used without further purification. 'H NMR (200 MHz, CDCI3): 7.49 (d, IH), 7.23 (dd, IH), 6.75 (d, 1 H), 0.26 (s, 9H)
Step B: 4-Bromo-2-chloro-3~methyl-phenol mL nBuLi solution in hexanes (120 mmol, 2.5M in hexanes) was added dropwise to a solution of 12.1 g dry DIPA (120 mmol) in 250 mL dry THF at -78 °C under argon atmosphère. The mixture was stirred for 30 minutes at the same température then 28.0 g (4-biOino-2-chloro-phenoxy)-trimethyl-silane (100 mmol) was added dropwise. After
2.5 hours, 21.3 g Mel (150 mmol) was added dropwise then the cooling bath was removed
ORIGINAL
-48and the mixture was stirred ovemight. The reaction was quenched with 100 mL aqueous NH3 solution and 200 mL saturated aqueous NH4CI solution and extracted with EtOAc. The organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The resulting dark mass was refluxed with pure hexane several times (150-150 mL aliquots) and decanted leaving a black tar behind. The combined organic phases were concentrated under reduced pressure affording 19.0 g crude product, which was used without further purification. NMR (200 MHz, CDCl3) δ: 7.32 (d, IH), 6.76 (d, l H), 5.62 (s, l H), 2.49 (s, 3H)
Step C: (4-Bromo-2-chloro-3-methyl-phenoxy)-trimethyl-silane
20.8 g HMDS (129 mmol) was added to the solution of 19.0 g 4-bromo-2-chloro-3-methylphenol (86.0 mmol) in 150 mL dry THF. The mixture was stirred at 85 °C under argon balloon for 1.5 hours and then concentrated under reduced pressure. The obtained product was used without further purification. ’H NMR (200 MHz, CDCl3) δ: 7.30 (d, IH), 6.63 (d. IH), 2.50 (s,3H), 0.28 (s, 9H)
Step D: Préparation 3a
A solution of 25.2 g (4-bromo-2-chloro-3-methyl-phenoxy)-trimethyl-silane (86.0 mmol) in 250 mL dry THF was cooled to -78 °C under argon and then 38 mL nBuLi solution (94.6 mmol, 2.5M in hexanes) was added dropwise. After 5 minutes, 19.2 g 2-isopropoxy-
4,4,5,5-tetramethyl-l ,3,2-dioxaborolane (103 mmol) was added dropwise. The cooling bath was removed and the mixture was slowly allowed to warm up to r.t. Then the mixture was added to 200 mL saturated aqueous NH4CI solution and extracted with EtOAc. The combined organic layers were concentrated under reduced pressure and passed through a pad of silica gel using hexane and EtOAc as eluents. The crude product was recrystallized from a mixture of EtOAc and hexane to obtain Préparation 3a. 1 FI NMR (500 MHz, DMSO-dû) δ: 10.40 (s, 1 H), 7.42 (d, IH), 6.80 (d, IH), 2.49 (s, 3H), 1.27 (s, 12H)
Préparation 3b: l-|2-[2-Chloro-3-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2yI)phenoxy]ethyl]-4-methyl-piperazine
10.0 g Préparation 3a (37.2 mmol,), 8.7 g 2-(4-methylpiperazin-l-yl)ethanol (60.3 mmol) and 15.8 g PPh3 (60.3 mmol) were dissolved in 100 mL dry toluene and then 27 mL
ORIGINAL
-49DEAD (60.3 mmol, 40 % solution in toluene) was added dropwise. The mixture was stirred at 50 °C under argon atmosphère until no further conversion was observed. The volatiles were evaporated under reduced pressure and 100 mL Et2O was added. The precipitated white crystals were filtered off and washed with Et2O. The filtrate was concentrated under reduced pressure and purified via flash chromatography using CHCl3 and MeOH as eluents. The resulting light brown oil was crystallized from hexane to give Préparation 3b as an off-white solid. 'H NMR (500 MHz, DMSO-de) δ: 7.56 (d, IH),
6.99 (d, IH), 4.15 (t, 2H), 2.72 (t, 2H), 2.51 (s, 3H), 2.50 (br s, 4H), 2.29 (br s, 4H), 2.13 (s, 3H), 1.29 (s, 12H)
Préparation 3c: 2-(3-chloro-2-methy)-phenyl)-5,5-dimethyl-l,3,2-dioxaborinane
4.94 g (3-chloro-2-methylphenyl)boronic acid (29 mmol) and 3.021 g neopentyl-glycol (29 mmol) were stirred at r.t. in the presence of Amberlite 15IΓ (dried with toluene) until no further conversion was observed. The mixture was then filtered through Celite and washed with 2-Me-THF. The filtrate was concentrated under reduced pressure to obtain Préparation 3c. 'H NMR (400 MHz, CDCI3): 7.59 (dd, IH), 7.38 (dd, IH), 7.10 (t, IH),
3.79 (s, 4H), 2.57 (s, 3H), 1.05 (s, 6H)
Préparation 4: Ethyl (27î)-2-I5-bromo-6-(4-fluorophenyl)furo[2,3-rf]pyrimidin-4-yl] oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl|methoxy|phenyl]propanoate
Using General procedure la and Préparation 2c as the appropriate lactic ester dérivative, Préparation 4 was obtained. MS: (M+H)+ =700.4
Préparation 5a: (E)-4-(Dimethylaniino)-l,l-dimethoxy-but-3-en-2-one
502.1 g i,l-dimethoxypropan-2-one (4.25 mol) and 506.4 g l.l-dimethoxy-AUV-dimethylmethanamine (4,25 mol) were mixed in a 2 L flask and stirred at 105 °C for 3 hours. The formed MeOH was removed continuously via distillation. When MeOH formation stopped (at 65 °C head température) the reaction mixture was vacuum distilled (decreasing the pressure slowly to 30 mbar) to remove side products and unreacted starting materials. The crude product was distilled at 0.1 mbar. Fractions were collected between 107-118 °C head température (bath température 160-165 °C) to give a yellow oil. NMR (500 MHz,
ORIGINAL
-50DMSO-d6) δ: 7.59 (d, IH), 5.17 (d. IH), 4.42 (s, IH), 3.25 (s, 6H), 3.09 (s, 3H), 2.78 (s, 3H)
Préparation 5b: [2-(2-Methoxyphenyl)pyrimidin-4-yl]methanol
Step A: 4-(dimethoxymethyl)-2-(2-methoxyphenyl)pyrimidine
To the mixture of 1.2 eq. 2-methoxybenzamidine acetic acid sait and 1 eq. Préparation 5a in dry methanol (0.5 mL / mmol), 1.2 eq. NaOEt was added portionwise and the mixture was stirred at 75 °C until no further conversion was observed. Then the reaction mixture was cooled and concentrated under reduced pressure. Water was added to the residue and it was extracted with DCM. The combined organic layers were dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give 4-(dimethoxymethyl)-2-(2-methoxyphenyl)pyrimidine. 'H NMR (400 MHz. DMSO-d<,) δ:
8.93 (d, IH), 7.55-7.44 (m. 3H), 7.16 (d, IH), 7.06 (m, 1 H), 5.31 (s, 1 H), 3.76 (s, 3H), 3.37 (s, 6H)
Step B: Préparation 5b
261 mg 4-(dimethoxymethyl)-2-(2-metho.xyphenyl)pyrimidine (1.0 mmol) was dissolved in 2 mL HCI in dioxane (4M solution), then 2 mL water was added and this mixture was stirred at 50 °C for 16 hours. The reaction mixture was cooled to 0 °C. then 320 mg NaOH (8.0 mmol) was added portionwise. The pH was adjusted to 8 using 10 % aqueous K2COj solution, then 76 mg sodium borohydride (2.0 mmol) was added and the mixture was stirred for 30 minutes at 0 °C. The reaction mixture was diluted with 5 mL water and extracted with EtOAc. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give Préparation 5b. ‘H NMR (400 MHz, DMSO-d6) Ô: 8.84 (d, IH), 7.50-7.42 (m, 3H), 7.14 (d, IH), 7.03 (m, IH), 5.66 (t, IH), 4.58 (d, 2H), 3.75 (s, 3H)
Préparation 6: (27î)-2-|(7-benzyl-5-bromo-6-ethyl-pyriOlo[2,3-r/]pyrimidin-4-yl) amino]-3-phenyl-propanoic acid
ORIGINAL
-5IStep A: 7-benzy!-5-bromo-4-chloro-6-elhyl-pyrrolo[2,3-d]pyrimidine
255 mg NaH (6.38 mmol) and 50 mL dry THF were charged into a 50 mL Schienk tube under N2 atmosphère and the slurry was cooled to 0 °C. Then L792 g Préparation 1b (5.8 mmol) was added. After stirring the mixture for 30 minutes at 0 °C, 773 pL benzyl bromide (6.38 mmol) was added and the mixture was aliowed to warm up to r.t., and stirred until no further conversion was observed. The mixture was then diluted with saturated aqueous NH4Cl solution, and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 7-benzyl-5-bromo-4-chloro~6-ethyl-pyrrolo[2,3-if[ pyrimidine.
'H NMR (400 MHz, CDCI3) δ: 8.60 (s, IH), 7.33-7.26 (m, 3H), 7.06-7.04 (m, 2H), 5.54 (s, 2H), 2.79 (q,2H), l .07 (t, 3H)
MS (M+H): 351.8
Step B: Préparation 6
Using General Procedure III and 7-benzyl-5-bromo-4-chloro-6-ethyl-pyrrolo[2,3-i/] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, Préparation 6 was obtained. MS (M+H): 279.2
Préparation 7a: /V-|2-bcnzyloxy*6-(2,2-dibromovinyl)phenyl|-3-chloro-2-niethyI-4triisopropylsilyloxy-aniline
Step A: (4-Bromo-2-chloro-phenoxy)-triisopropyl-silane
200 g 4-bromo-2-chloro-phenol (0.97 mol) and 126 mL TIPSCI (Ί.18 mol) were dissolved in l .6 L DCM. 167 g îmidazole (2.45 mol) was added and the mixture was stirred at r.t. for 2 hours. Then the volatiles were evaporated under reduced pressure and the residue was dissolved in 1.5 L EtOAc. The mixture was washed with brine, dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The triisopropylsilyl hydroxide impurity was removed by distillation (12O°C at 0.01 mmHg). The residue was filtered
ORIGINAL
-52through a short pad of silica with hexane and concentrated under reduced pressure. The product (colourless oil) was used in the next step without further purification.
'H NMR. (400 MHz. CDCI3) ô: 7.49 (d. IH), 7.21 (dd, IH), 6.78 (d, IH), l.3l (septet, 3H),
I.l4 (d, 18H)
MS (El, 70 eV) m/z (% relative intensity, [ion]): 63 (30), 79 (24), 93 (41), 170 (17), 235 (19), 251 (16), 265 (24), 293 (23), 319 (77), 321 (100), 323 (28), 362 (l, [M+])
Step B: (4-Bromo-2-chloro-3-inethyl-phenoxy)-triisopropyl-silane
76.0 mL dry DIPA (0.54 mol) was dissolved in 1.2 L dry THF under argon atmosphère and
51.2 mL nBuLi solution (0.512 mol, 10M in hexanes) was added dropwise at -78 °C. The mixture was stirred for 45 minutes at the same température. Then 178 g (4-bromo-2chloro-phenoxy)-triisopropyl-silane (0.488 mol) was added dropwise at -78 °C and the white suspension was stirred until no further conversion was observed. Then 36.5 mL Me! (0.586 mmol) was added at this température and the reaction mixture was stirred overnight without further cooling. The volatiles were evaporated under reduced pressure. The residue was dissolved in 1.5 L EtOAc, washed with brine. The organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was filtered through a short pad of silica using hexane as eluent and concentrated under reduced pressure to obtain the product as pale yellow oil. ’H NMR (400 MHz, CDCI3) ô: 7.30 (d, IH), 6.68 (d, IH), 2,53 (s, 3H), 1.32 (septet, 3H), 1.14 (d, 18H)
Step C: N-benzyl-3-chloro-2-niethyl-4-triisopropylsilyloxy-aniline
7.56 g (4-bromo-2-chloro-3-methyl-phenoxy)-triisopropyi-silane (20 mmol) and 4,29 g benzylamine (40 mmol) were dissolved in 16 mL dry toluene, then 450 mg Pd2dba3 (0.5 mmol), 450 mg X-Phos (l mmol) and 9.77 g Cs2CO3 (30 mmol) were added and the mixture was stirred at l00°C until no further conversion was observed. Then it was filtered through Celite, and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using hexane and EtOAc as eluents to obtain A-benzyl-3-chloro-2-methyl-4-triisopropylsilyloxy-aniline.
Step D: 3-chloro-2-methyl-4-tnisopropyIsilyloxy-aniline
ORIGINAL
3.50 g jV-benzyl-3-chloro-2-methyl-4-triisopropylsilyloxy-aniline (8.66 mmol) was dissolved in 100 mL MeOH and 20 mL EtOAc, then 80 mg 10 % Pd/C was added and the mixture was stirred under 1 bar H2 atmosphère until no further conversion was observed. Then it was fîitered through Celite, and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using hexane and EtOAc as eluents to obtain 3-chloro-2-methyl-4-triisopropyIsilyloxy-aniline.
’H NMR (400 MHz, DMSO-d6) δ: 6.58 (d, IH), 6.50 (d, IH), 4.68 (s, 2H), 2.11 (s, 3H),
1.24 (m,3H), 1.06 (d, 18H)
MS: (M+H)+ = 314.2
Step Et 3-benzyloxy-2-brotno-benzaldehyde
4.554 g 2-bromo-3-hydroxybenzaldehyde (22.65 mmol), 4.262 g benzyl bromide (24.92 mmol) and 4.696 g K2CO3 (33.98 mmol) were dissolved in 20 mL DMSO and stirred at 50 °C until no further conversion was observed. The mixture was then poured into water. The precipitate was fîitered to give 3-benzyloxy-2-bromo-benzaldehyde. MS (El, 70 eV) m/z (% relative intensity, [ion]): 65 (10), 91 (100), 290 (5, [M+]), 292 (5, [M+])
Step F; 3-benzyloxy-2-(3-chloro-2-tiieihyl-4-triisopropylsilyloxy-anilino)benzaldehyde
5.0 g 3-benzyloxy-2-bromo-benzaldehyde (17.17 mmol), 5.391 g 3-chloro-2-methyl-4triisopropylsilyloxy-aniline (17.17 mmol), 16.782 g Cs2CO3 (51.51 mmol), 393 mg Pd2dba3 (0.43 mmol) and 535 mg rac. BINAP (0.86 mmol) were mixed in 85 mL toluene and stirred at 120 °C until no further conversion was observed. The volatiles were removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 3-benzyloxy-2-(3-chloro-2-methyl-4-triisopropylsilyloxyanilino) benzaldehyde. MS: (M+H/ = 524.2
Step G: Préparation 7a
7.7 g 3-benzyloxy-2-(3-chloro-2-methyl-4-triisopropylsilyloxy-anilino)benzaldehyde (14.69 mmol) and 7.308 g carbon tetrabromide (22.03 mmol) were dissolved in 160 mL DCM at 0 °C, then 11.56 g PPh3 (44.07 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. Then the solvent was removed under reduced pressure, the residue was dissolved in Et2O. Then heptane was added and the formed
ORIGINAL
-54precipitate was filtered, the filtrate was concentrated under reduced pressure. Then heptane was added, and the mixture was stirred for 10 minutes and filtered again. The filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to give Préparation 7a.
’H NMR (400 MHz, DMSO-d6) δ: 7.28-7.23 (m, 5H), 7.19 (s, IH), 7.11 (dd, 2H), 7.05 (d, IH), 6.60 (d, IH), 6.41 (s, IH), 6.22 (d, IH), 5.08 (s, 2H), 2.30 (s, 3H), 1.25 (m, 3H), l.05 (d, 18H)
MS: (M+H)+ = 680.0
Préparation 7b: Ethyl (2Æ)-2-[l-[3-chIoro-2-methyl-4-[2-(4-methylpiperazin-l-yl) ethoxy]phenyl|-2-(4-fluorophenyl)indol-7-yl|oxy-3-(2-methoxyphenyl)propanoate
Step A: [4-[7-benzyloxy-2-(4-fliiorophenyl)indol-l-yl]-2-chloro-3-methyl-phenoxy]tri isopropyl -silane
2720 mg Préparation 7a (4 mmol), Hl9 mg 4-fluorophenylboronic acid (8 mmol), 4245 mg K.3PO4 (20 mmol), 90 mg Pd(OAc)2 (0.4 mmol) and 328 mg SPhos (0.8 mmol) were mixed in 60 mL dry toluene under N2 atmosphère and stirred at l00°C until no further conversion was observed. Then the solvent was removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to give [4-[7-benzyloxy-2-(4-fluorophenyl)indol-1 -yl]-2-chloro-3-methyl-phenoxy]triisopropyl-silane. *H NMR (400 MHz, CDCI3) Ô: 7.33 (d, 2H), 7.29-1.22 (m, 2H), 7.18 (d, IH), 7.16 (d, IH), 7.10 (t. 2H), 6.94 (d, IH), 6.92-6.84 (m, 4H), 6.73 (s, IH), 6.61 (d, IH),
4.94 (d, 1 H), 4.89 (d, IH), 1.97 (s, 3H), 1.31 (m, 3H), 1.13 (t, 18H)
Step B: 4-[7-benzyloxy-2-(4-fluorophenyl)indol-l-yl]-2-chloro-3-methyl-phenol
2600 mg [4-[7-benzyloxy-2-(4-fluorophenyl)indol-1 -yl]-2-chloro-3-methy 1-phenoxy]triisopropyl-silane (2.96 mmol), 2.96 mL TBAF solution (2.96 mmol, IM in THF) and 50 mL THF were stirred at r.t. until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to give 4-[7-benzyloxy-2-(4-fluorophenyl)indol-l-yl]2-chloro-3-methyl-phenol.
ORIGINAL
-55’HNMR (400 MHz. DMSO-dû) δ: 10.27 (br s, IH), 7.28-7.18 (m, 6H), 7.10 (t, 2H), 7.07-
6.99 (m, 2H), 6.85-6.77 (m,3H),6.75 (s, IH), 6.72 (d, lH), 4.95 (d, lH), 4.90 (d, IH), 1.75 (s, 3H)
MS: (M+H)+ = 458.0.
Step C: 7-benzyloxy-l -[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2(4-fl uorophenyl) indol e
I.2 g 4-[7-benzyloxy-2-(4-fluorophenyl)indol-l-yl]-2-chIoro-3-methyl-phenol (2.1 mmol), 606 mg l-(2-hydroxyethyl)-4-methylpiperazine (4.2 mmol) and 2.1 g PPh2 (6.3 mmol) were dissolved in 50 mL dry toluene under N2 atmosphère and the mixture was cooled to 0 °C. Then I45l mg DTAD (6.3 mmol) was added and the mixture was heated to 45 °C and stirred until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc and MeOH as eluents to give 7-benzyioxy-l-[3-chloro-2-methyl-4-[2-(4methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluorophenyl)indole. MS: (M+H)T - 584.2
Step D: l-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluoro phenylflndol- 7-ol
1280 mg 7-benzyloxy-l-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy]phenyl]2-(4-fluorophenyl)indole (2.19 mmol) was dissolved in 100 mL EtOH, then 100 mg 10 % Pd/C was added. The mixture was stirred under l bar H2 atmosphère at r.t. until no further conversion was observed. Then the mixture was filtered through Celite and the filtrate was concentrated to give l -[3-chloro-2-methyl-4-[2-(4-methylpiperazîn-l-yl)ethoxy]phenyl]-2(4-fluorophenyl)indol-7-ol.
‘H NMR (400 MHz, DMSO-d6) δ: 9.04 (br s, IH), 7.25 (dd, 2H), 7.17-7.03 (m, 4H), 6.94 (d. IH), 6.86 (t, IH), 6.70 (s, IH), 6.47 (d, IH), 4.13 (m, 2H), 2.72 (t, 2H), 2.58-2.42 (br s, 4H), 2.40-2.17 (br s, 4H), 2.14 (s, 3H), l .86 (s, 3H)
MS: (M+H)+= 494.2
Step E: Préparation 7b
494 mg l -[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy]phenyl]-2-(4-fluoro phenyl)indoI-7-ol (l mmol), 449 mg Préparation 2f (2 mmol) and 786 mg PPh2 (3 mmol)
ORIGINAL
-56were dissolved in 10 mL dry toluene under N2 atmosphère and the mixture was cooled to 0 °C. Then 691 mg DTAD (3 mmol) was added and the mixture was heated to 45 °C and stirred until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and
EtOAc and MeOH as eluents to give Préparation 7b as a mixture of diastereoisomers.
‘H NMR (500 MHz, DMSO-d6) δ: 7.43/6.98 (d, IH), 7.28 (m, 2H), 7.23/7.24 (d, IH), 7.17/7.18 (t, IH), 7.14 (m, 2H), 7.12/6.88 (d, IH), 6.95/6.94 (t, IH), 6.91/6.91 (d, 1 H), 6.79/6.78 (s, IH), 6.73/6.75 (t, IH), 6.52/6.60 (d, IH), 6.46/6.40 (d. IH), 4.85/4.76 (dd, IH), 4.25-4.01 (m, 2H), 4.01-3.89 (m, 2H), 3.77/3.76 (s, 3H), 2.70-2.60 (m, 3H), 2.54-2.30 (m, 5H), 2.21 (brs,4H), 2.13/2.09 (s, 3H), 1.59/2.08 (s, 3H), 0.99/0.98 (t, 3H)
MS: (M+H)+ = 700.0
Example 1 : (2Æ)-2-{[5-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yI)ethoxy] phenyl}-6-(4-nuorophenyI)furo[2,3-rf]pyriniidin-4-yi]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid
Using General Procedure II and Préparation 4 as the appropriate 5-bromo-furo[2,3-i/| pyrimidine dérivative and Préparation 3b as the appropriate boronic acid dérivative. Example 1 was obtained as a mixture of diastereoisomers. HRMS calculated for C47H44CIFN6O7: 858.2944, found: 430.1547 and 430.1555 (M+2H)
Example 2: (2Æ)-2-{|5-{3-chloro-2-ethvI-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}20 6-(4-fluoroplienyl)furo|2,3-r/]pyrimidin-4-yI]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl|methoxy}phenyl)propanoic acid
Siep A : l-[2-(4-bromo-2-chloro-phenoxy)ethyl]-4-methyl-piperazine
10.373 g 4-bromo-2-chlorophenol (50 mmol), 14.442 g 2-(4-methylpiperazin-l-yl)ethanol (100 mmol) and 26.229 g PPI13 (100 mmol) were dissolved in 250 mL dry toluene under 25 N2 atmosphère, then 23.027 g DTAD (100 mmol) was added. The mixture was stirred at °C until no further conversion was observed. The volatiles were evaporated under
ORIGINAL
-57reduced pressure and the residue was purified via flash chromatography using EtOAc and MeOH as eluents. MS (M+H): 333.0
Step B'. l-[2-(4-bromo-2-chloro-3-ethyl-phenoxy)ethyl]-4-methyl-piperazine
2.0 g l-[2-(4-bromo-2-chloro-phenoxy)ethyl]-4-methyl-piperazine (6 mmol) was dissolved in 50 mL dry THF under N? atmosphère and was cooled to -78 °C. 6 mL LDA solution (12 mmol in 2M THF) was added and the mixture was stirred for 3 hours, then 982 mg iodoethane (6.3 mmol) was added and the mixture was allowed to warm up to r.t. It was quenched with saturated aqueous NH4CI solution, extracted with EtOAc. The combined organic layer was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. MS (M+H): 360.8
Step C: l-[2-[2-chloro-3-ethyl-4-(4,4,5,5-letramethyl-i,3,2-dioxaborolan-2-yl)phenoxy] ethyI]-4-inethyl-pîperazme
2099 mg l-[2-(4-bromo-2-chloro-3-ethyl-phenoxy)ethyl]-4-methyl-piperazine (5.8 mmol) was dissolved in 30 mL dry THF under N2 atmosphère and was cooled to -78 °C. 4.65 mL nBuLi solution (l L61 mmol in 2.5M THF) was added dropwise. It was stirred for 5 hours, then 2.6 mL 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaboroIane (12.77 mmol) was added and the mixture was stirred for 30 minutes. Then it was allowed to warm up to r.t. and it was concentrated under reduced pressure. The crude product was purified via flash chromatography using EtOAc and MeOH as eluents. MS (M+H): 409.2
Step D: Example 2
Using General Procedure II and Préparation 4 as the appropriate 5-bromo-furo[2,3-cZ] pyrimidine derivative and l-[2-[2-chloro-3-ethyl-4-(4,4,5,5-tetramethyl-1,3,2dioxaboro!an-2-yl)phenoxy]ethyl]-4-methyl-piperazine as the appropriate boronic acid derivative, Example 2 was obtained as a mixture of diastereoisomers. HRMS calculated for C48H46C1FN6O7: 872.3101, found: 437.1620 and 437.1620 (M+2H)
Example 3: (27?)-2-{|5-{3-chloro-2-mcthyl-4-[2-(4-mcthylpiperazin-l-yl)ethoxy| phenyl}-6-(4-fluorophenyl)furo[2,3-f/]pyrimidin-4-yl]oxy}-3-(2-rnethoxyphenyl) propanoic acid
ORIGINAL
-58Step A: Ethyl (2R)-2-[5-btx)mo-6-(4-fluorophenyl)fiiro[2,3-à]pyrimidm-4-yl]oxy-3-(2methoxyphenyl)propanoate
Using General procedure la and Préparation 2e as the appropriate lactic ester dérivative, ethyl (2Æ)-2-[5-bromo-6-(4-fluorophenyl)furo[2,3-i/]pyrimidin-4-yl]oxy-3-(2-methoxy phenyl)propanoate was obtained. NMR (400 MHz, DMSO-dô): 8.53 (s, IH), 8.10 (m, 2H), 7.47-7.36 (m, 3H), 7.23 (m, IH), 6.96 (m, IH), 6.89 (t, IH), 5.58 (m, IH), 4.12 (q, 2H), 3.79 (s, 3H), 3.36 (m, IH), 3.21 (m, 1 H), 1.11 (t, 3H)
Step B: Example 3
Using General Procedure II and ethyl (2Æ)-2-[5-bromo-6-(4-fluorophenyl)furo[2,3-c/] pyrimidin-4-yl]oxy-3-(2-methoxyphenyl)propanoate as the appropriate 5-bromo-furo [2,3-rf]pyrimidine dérivative and Préparation 3b as the appropriate boronic acid dérivative, Example 3 was obtained as a mixture of diastereoisomers. HRMS calculated for C36H36CIFN4O6: 674.2307, found: 675.2367 and 675.2364 (M+H)
Example 4: (2/f)-2-{[5-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy] phenyl}-6-(4-fluorophenyl)furo|2,3-i/|pyrimidin-4-yl|oxy}-3-|2-(pyrazin-2-ylmethoxy) phenyl]propanoic acid
Step A: Ethyl (2R)-2-[5-bromo-6-(4-flitorophenyl)furo[2,3-<3]pyrimidin-4-yl]oxy-3-[2(pyrazin-2-ylmethoxy)phenyl]propanoate
Using General procedure la and Préparation 2g as the appropriate lactic ester dérivative, ethyl (2/?)-2-[5-bromo-6-(4-fluorophenyl)furo[2,3-iZ]pyrimidin-4-yl]oxy-3-[2-(pyrazin-2ylmethoxy)phenyl]propanoate was obtained. MS: (M+H)+ = 595.0
Step B: Example 4
Using General Procedure II and ethyl (27?)-2-[5-bromo-6-(4-lluorophenyl)furo[2,3-if| pyrimidin-4-yl]oxy-3-[2-(pyrazin-2-ylmethoxy)phenyI]propanoate as the appropriate
5-bromo-furo[2,3-i/]pyrimidine dérivative and Préparation 3b as the appropriate boronic acid dérivative, Example 4 was obtained as a mixture of diastereoisomers. HRMS calculated for C4oH38C1FN606: 752.2525, found: 753.2645 and 753.2606 (M+H)
ORIGINAL
-59Example 5: (2/f)-2-{[6-(5-chlorofuran“2-yl)-5-{3-chloro-2-methyl-4-|2-(4-methyl piperazin-l-yl)ethoxy|phenyl}furo[2,3-rf]pjTÏmidin-4-yl]oxy}-3-[2-(pyrazin-2-yl niethoxy)phenyl]propanoic acid
Step A: 2-[2-(2-furyl)-2-oxo-ethyl]propanedinitrile
46.2 mL IM NaOEt solution in EtOH (46.2 mmol) and 400 mL EtOH were cooled to 0 °C and 3.2 g malononitrile (48.4 mmol) was added. The mixture was stirred at 0 °C for l hour, then 8.35 g 2-bromo-l-(2-furyl)ethanone (44 mmol) was added. The mixture was stirred at 0 °C for l hour, then at r.t. until no further conversion was observed. The volatiles were removed under reduced pressure, the residue was digerated in Et2O, fîltered, then purified 10 via flash chromatography using DCM and EtOAc as eluents to obtain 2-[2-(2-furyl)-2-oxoethyl]propanedînitrile. MS: (M+H)+ = 175.2
Step B: 2-amino-5-(2-furyl)furan-3-carbonitrile
4.587 g 2-[2-(2-furyl)-2-oxo-ethyl]propanedinitrile (26.34 mmol) was dissolved in 150 mL EtOH and 4.6 g Amberlite 15H was added. The mixture was stirred at 90 °C until no 15 further conversion was observed. The mixture was then fîltered, washed with DCM and EtOAc. The filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain 2-amino-5-(2-furyl)furan-3carbonitrile. MS: (M+H)+ = 175.4
Step C: 6-(2-furyl)-3\d-furo[2,3-d]pyrûnidin-4-one
1310 mg 2-amino-5-(2-furyl)furan-3-carbonitrile (7.52 mmol) and 30 mL acetic formic anhydride were placed in a flask and stirred at r.t. for 30 minutes. Then the volatiles were evaporated under reduced pressure and the residue was dissolved in 60 mL AcOH, and irradiated at 180 °C for 50 minutes. The mixture was cooled to r.t., and the crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain
6-(2-furyl)-3/7-furo[2,3-i/]pyrimidin-4-one. ’H NMR (400 MHz, DMSO-d6): 12.68 (br s, l H), 8.14 (s, l H), 7.84 (m, l H), 7.08 (s, l H), 6.94 (d, l H), 6.67 (m, l H)
Step D: 6-(5-chloro-2-furyl)-3H-furo[2,3-d]pyrimidin-4-one
ORIGINAL
-601.183 g 6-(2-furyl)-3i/-furo[2,3-J]pynmidin-4-one (5.85 mmol) was dissolved in 55 mL THF and 860 mg NCS (6.44 mmol) was added. The mixture was stirred at 40 °C until no further conversion was observed. The mixture was cooled to 0 °C, and the precipitate was filtered. and dried to obtain 6-(5-chloro-2-furyl)-3/7-furo[2,3-i7]pyrimidin-4-one. MS: (M+H)+ = 237.0
Step E: 5-bromo-6-(5-chloro-2-furyl)-3}3-furo[2,3-<3]pyrimidin-4-one)
1000 mg 6-(5-chloro-2-furyl)-3/7-furo[2,3-i/]pyrimidin-4-one (4.23 mmol) was dissolved in 40 mL AcOH, then 776 mg bromine (4.86 mmol) was added. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure. The residue was digerated with DCM then filtered to obtain 5-bromo-6(5-chloro-2-furyl)-3//-furo[2,3-i/]pyriniidin-4-onc. MS: (M-H)+ = 314.8
Step F: 5-bromo-4-chloro-6-(5-chloro-2-fitryl)furo[2,3-à]pyrimidine
1110 mg 5-bromo-6-(5-chloro-2-furyl)-3i/-furo[2,3-i/|pyrimidin-4-one (3.52 mmol) was dissolved in 8.21 mL POC13 (88.1 mmol) then 447 pL DMA (3.52 mmol) was added. The mixture was stirred at 110 °C until no further conversion was observed. The mixture was then cooled to -78 °C and ice was added. It was sonicated then the precipitate was filtered. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-(5-chloro-2-furyl)furo[2,3-i/]pyrimidine. MS: (M+H)+ = 335.0
Step G: Ethyl (2R)-2-[5-bromo-6-(5-chloro-2-furyl)furo[2,3-à]pyrimidin-4-yl]oxy-3-[2(pyrazin-2-ylmelhoxy)phenyl]propanoate eq. 5-bromo-4-chloro-6-(5-chloro-2-furyl)furo[2,3-i7]pyrimidine, 2 eq. Préparation 2g.
mL/mmol 'BuOH and 5 eq. CS2CO3 were placed in a flask and stirred at 55 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure, diluted with brine, neutralized with IM aqueous HCl solution, and extracted with EtOAc. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give ethyl (2/?)-2-[5-bromo-6-(5ORIGINAL
-61chloro-2-furyl)furo[2,3-rf]pyrîmidin-4-yl]oxy-3-[2-(pyrazin-2-ylmethoxy)phenyl] propanoate. MS: (M+H)+ = 601.0
Step H: Example 5
Using General Procedure II and ethyl (2Æ)-2-[5-bromo-6-(5-chloro-2-furyl)furo[2,3-i/J pyrimidin-4-yl]oxy-3-[2-(pyrazin-2-ylmethoxy)phenyl]propanoate as the appropriate
5-bromo-furo[2,3-<7]pyrimidine derivative and Préparation 3b as the appropriate boronic acid derivative, Example 5 was obtained as a mixture of diastereoisomers. HRMS calculated for C38H36Cl2N6O7: 758.2023, found: 759.2119 and 759.2156 (M+H)
Example 6: (2Æ)-3-{2-[(l-/ert-buty l-lH-pyrazol-5-yl)methoxy]phenyl}-2-{|5-{3-chloro4-[2-(dimethylamino)ethoxy]-2-methylphenyl}-6-(4-fluorophenyl)furo[2,3-rf] pyrimidin-4-yl]oxy}propanoic acid
Step A: 1 -\ert-biityl-5-(dimethoxymethyl)-liï-pyrazole
1.2 eq. /erz-butylhydrazine hydrochloride and 1 eq. Préparation 5a was dissolved in dry methanol (0.5 mL/mmol), then 1.2 eq NaOEt was added portionwise and the mixture was stirred at 75 °C for 2 hours. The reaction mixture was cooled and concentrated under reduced pressure. The residue was diluted with water and it was extracted with DCM. The combined organic phases were dried over MgSCC, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give l-/<?r/-butyl-5-(dimethoxymethyl)-l/Z-pyrazole. ‘H NMR (400 MHz, DMSO-d6) δ: 7.34 (d, IH), 6.34 (d. IH), 5.74 (s, IH), 3.24 (s, 6H),
1.57 (s, 9H). We also obtained l-/e/7-butyl-3-(dimethoxymethyl)-l/7-pyrazole. 'H NMR (400 MHz, DMSO-dô) δ: 7.75 (d, IH), 6.18 (d, IH), 5.34 (s, IH), 3.24 (s, 6H), 1.50 (s, 9H)
Step B: (1 Aert-Butyl-lW-pyrazol-5-yl)methanol eq. l-/e/7-butyl-5-(dimethoxymethyl)-l/f-pyrazole was stirred with IM aqueous HCl solution (3 mL/mmol) at 50 °C until no further conversion was observed. The reaction mixture was cooled to 0 °C, then 2.85 eq. solid NaOH was added portionwise. The pH was adjusted to 8 using 10% aqueous K2CO3 solution, then 2 eq. sodium borohydride was added portionwise, keeping the température below 5 °C and stirred at 0 °C until no further
ORIGINAL
-62conversion was observed. The mixture was extracted with EtOAc, the combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc to obtain (l-/er/-butyl-lH-pyrazol-5-yl)methanol. ’H NMR (400 MHz, DMSO-d6) δ: 7.27 (d, l H), 6.19 (d, l H), 5.31 (t, l H), 4.61 (d, 2H), l .56 (s, 9H)
Step C: (2R)-3-[2-[(2Aer(-butylpyrazol-3-yl)methoxy]phenyl]-2-hydroxy-propanoic acid
2.51 g Préparation 2a (9.96 mmol), 2.0 g (l-/e/7-butyl-lA-pyrazol-5-yl)methanol (l3 mmol) and 3.39 g triphenyl phosphine (13 mmol) were dissolved in 12 mL dry toluene, then 5.9 mL DEAD (13 mmol) was added. The mixture was stirred at 50 °C under nitrogen atmosphère until no further conversion was observed. The volatiles were evaporated under reduced pressure, Then 30 mL Et2O was added, the mixture was sonicated and filtered (to remove PPh3 and PPh3O). The filtrate was concentrated under reduced pressure. The residue was dissolved in THF, and then 2 g NaOH dissolved in 8 mL water wras added. The mixture was stirred at 50 °C until no further conversion was observed. Then it was acidified with 2M aqueous HCl solution, and THF was removed under reduced pressure. The residue was extracted with DCM, dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to obtain (2Æ)-3-[2-[(2-ie/7-butylpyrazol-3yl)methoxy]phenyl]-2-hydroxy-propanoic acid. MS (M+H): 319.0
Step D: Ethyl (2R)-3-[2-[(2-tecPbutylpyrazol-3-yl)methoxy]phenyl]-2-hydroxy-propanoate
7.2 g (2Æ)-3-[2-[(2-/e/7-butylpyrazol-3-yl)methoxy]phenyl]-2-hydroxy-propanoic acid was dissolved in 75 mL EtOH, then 2 mL cc. H2SO4 was added. The mixture was stirred at 60 °C until no further conversion was observed. Then it was diluted with water, neutralized with saturated aqueous NaHCO3 solution and extracted with dichloromethane. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure and purified via flash chromatography using EtOAc and MeOH as eluents to obtain ethyl (2Æ)-3-[2-[(2-ie/7-butylpyrazol-3-yl)methoxy] phenyl]-2-hydroxy-propanoate. MS (M+H): 347.0
Step E: Ethyl (2ty-2-[5-bromo-6-(4-fluorophenyl)furo[2,3-à]pyrimidin-4-yl]oxy-3-[2-[(2\ext-butylpyrazol-3-yl)niethoxy]phenyl]propanoate
ORIGINAL
-63Using General procedure la and ethyl (2/?)-3-[2-[(2-Zc/7-butylpyrazol-3-yl)methoxy] phcnylJ-2-hydroxy-propanoate as the appropriate lactic ester dérivative, ethyl (2Æ)-2-[5bromo-6-(4-fluorophenyl)furo[2,3-iT]pyrimidin-4-yl]oxy-3-[2-[(2-/e/7-butyl-pyrazol-3-yl) methoxy]phenyl]propanoate was obtained. MS (M+H): 636.6-638.6
Step F: 2-[2-chloro-3-methyl-4-(4,4,5,54etramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]N. N -dimelhyl-ethanamine
10.0 g Préparation 3a (37.2 mmol), 5.366 g N,N-dimethylethanolamine (60.3 mmol) and
15.8 g PPh3 (60.3 mmol) were dissolved in 100 mL dry toluene and then 27 mL DEAD (60.3 mmol, 40 % solution in toluene) was added dropwise. The mixture was stirred at 50 °C under argon atmosphère until no further conversion was observed. The volatiles were evaporated under reduced pressure and 100 mL Et2O was added. The precipitated white crystals were filtered off and washed with Et2O. The filtrate was concentrated under reduced pressure and purified via flash chromatography using CHCl3 and MeOH as eluents. The resulting light brown oil was crystallized from hexane to give 2-[2-chloro-3methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]-/V,V-dimethylethanamine.
’H NMR (200 MHz. CDCl3) δ: 7.63 (d, IH), 6.75 (d, IH), 4.15 (t, 2H), 2.81 (t, 2H), 2.60 (s, 3H), 2.38 (s, 6H), 1.33 (s. 12H) MS (M+H): 340.1
Step G: Example 6
Using General Procedure II and ethyl (2/?)-2-[5-bromo-6-(4-fluorophenyl)furo[2,3-</] pyrimidin-4-yl]oxy-3-[2-[(2-/ei7-butylpyrazol-3-yl)methoxy]phenyl]propanoate as the appropriate 5-bromo-furo[2,3-d]pyrimidine dérivative and 2-[2-chloro-3-methyl-4(4,4,5,5-tetramethyl-l ,3.2-dioxaborolan-2-yl)phenoxy]-AOV-dimethyl-ethananiinc as the appropriate boronic acid dérivative, Example 6 was obtained. HRMS calculated for C4oH4iClFN506: 741.2729, found: 742.2813 and 742.2808 (M+H) for the two diastereomers
Example 7: Ar-[(5S'fl)-5-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-lyl)ethoxy|phenyI}-6-(4-fluorophenyl)furo[2,3-rfJpyrimidin-4-yl]-2-methoxy-DORIGINAL
-64phenylalanine
Step A: (2K)-2-[[5-bromo-6-(4-fluorophenyl)furo[2,3-à]pyrimidin-4-yl]amino]-3-(2melhoxyphenyljpropanoic acid
Using General Procedure Ib and (27i)-2-amino-3-(2-methoxyphenyl)propanoic acid as the appropriate amino acid dérivative, (2/?)-2-[[5-bromo-6-(4-fluorophenyl)furo[2p-i/] pyrimidin-4-yI]amino]-3-(2-methoxyphenyl)propanoic acid was obtained. MS: (M+H)+ =
487.8
Step B: Example 1 eq. (27?)-2-[[5-bromo-6-(4-fluorophenyl)furo[2,3-i/]pyrimidin-4-yl]amino]-3-(2-methoxy phenyl)propanoic acid, 1.5 eq. Préparation 3b, 5 mol% AtaPhos and 2 eq. CS2CO3 were stirred in a 1:1 mixture of THF and water (10 mL/mmol 5-bromo-furo[2,3-i/]pyrimidine dérivative) and heated to 110°C in a MW reactor until no further conversion was observed. Then the mixture was diluted with brine, the pH was set to 4 with 1M aqueous HCl solution, and was extracted with DCM. The combined organic phases were dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The obtained mixture of diastereoisomers were purified and separated via HILIC chromatography. Example 7 was obtained as the later eluting diastereoisomer. HRMS calculated for C36H37CIFN5O5: 673.2467, found: 337.6286 (M+2H)
Exaniple 8: /V-[(55a)-5-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)etlioxy]phenyl}-6-(4-fluorophenyl)furo[2,3-if]pyrimidin-4-yl|-2-{[2-(2methoxyphenyl)pyrimidin-4-yl]methoxy}-Z)-phenylalanine and
Examplc 9: /V-[(5Ra)-5-{3-chloro-2-niethyl-4-|2-(4-methylpiperazin-lyl)ethoxy]phenyl}-6-(4-f1uorophenyl)furo|2,3-i/|pyrimidin-4-yI]-2-{|2-(2methoxyphenyl)pyrimidin-4-yl|methoxy}-£>-phenyIalanine
Step A: (2ty-2-[[5d>romo-6-(4-fluorophenyl)furo[2,3-à]pyrimidin-4-yl]amino]-3-(2hydroxyphenyl)propani) ic acici
ORIGINAL
-65Using General Procedure Ib and D-(Æ)-2-amino-3-(2-hydroxy-phenyl)-propionic acid as the appropriate amino acid dérivative, (27î)-2-[[5-bromo-6-(4-fluorophenyl)furo[2,3-iZ] pyrimidin-4-yl]amino]-3-(2-hydroxyphenyl)propanoic acid was obtained, MS: (M+H) = 473,6
Step B: Ethyl (2V.)-2-[[5-bromo-6-(4-Jluorophenyl)furo[2,3-d]pyrimidin-4-yl] amino]-3-[2[[2-(2-methoxyphenyl)pyrimidin-5-yl]methoxy]phenyl]propanoate
163 mg (2Æ)-2-[[5-bromo-6-(4-fluorophenyl)furo[2,3-</]pyrimidin-4-yI]amino]-3-(2hydroxyphenyl)propanoic acid was dissolved in 3 mL HCl solution (1.25M in EtOH) and stirred at 60 °C until no further conversion was bserved. The mixture was concentrated under reduced pressure, diluted with water. The precipitate was filtered and purified via flash chromatography using heptane and EtOAc as eluents to obtain ethyl (27?)-2-[[5bromo-6-(4-fluorophenyl)furo[2,3-t/]pyrimidin-4-yl]amino]-3-(2-hydroxyphenyl) propanoate. MS: (M+H)+ = 501.6
Step C: Ethyl (2^)-2-[[5-bromo-6-(4-flttorophenyl)furo[2,3-d]pyrhnidin-4-yl]amino]-3-[2[[2 - (2-methoxyphenyl) pyrim idin-5 -y!] methoxy]phenyl]propa> loate
500 mg ethyl (2Æ)-2-[[5-bromo-6-(4-fluorophenyl)furo[2,3-</|pyrimidin-4-yl]amino]-3-(2hydroxyphenyl)propanoate (1 mmol), 540 mg Préparation 5b (2.5 mmol) and 656 mg PPh3 (2.5 mmol) were dissolved in 20 mL dry toluene under N2 atmosphère, then 576 mg DTAD (2.5 mmol) was added. The mixture was stirred at 60 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to give ethyl (2Æ)-2-[[5-bromo-
6-(4-fluorophenyl)furo[2,3-i/]pyrimidin-4-yl]amino]-3-[2-[[2-(2-methoxyphenyl) pyrimidin-5-yl]methoxy]phenyl]propanoate. HRMS (M+H)+: 698.1402
Step D: Examples 8 and 9 eq. ethyl (27?)-2-[[5-bromo-6-(4-fluoiOphenyl)furo[2,3-i/]pyrimidin-4-yl]amino]-3-[2[[2-(2-methoxyphenyl)pyrimidin-5-yl]methoxy]phenyI]propanoate, 1.5 eq.
Préparation 3b, 5 mol% AtaPhos and 2 eq. Cs2CO3 were stirred in a 1:1 mixture of THF and water (10 mL/mmol 5-bromo-furo[2,3-</]pyrimidine dérivative) and heated to 70 °C and stirred until no further conversion was observed. Then the mixture was diluted with
ORIGINAL
- 66brine, the pH was set to 4 with IM aqueous HCl solution, and was extracted with DCM. The combined organic phases were dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude intermediate was purified via flash chromatography using DCM and MeOH as eluents. Then it was dissolved in dioxaneiwater l:l (20 mL/mmol) and 10 eq. LiOH*H2O was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with brine, neutralized with 2M aqueous HCl solution, extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to obtain a mixture of diastereoisomers. They were separated and purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents. Example 8 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C47H45CIFN7O6: 857.3104, found: 429.6637 (M+2H). Example 9 was obtained as the later eluting diastereoisomer. HRMS calculated for C47H45C1FN7O6: 857.3104, found: 429.6648 (M+2H)
Example 10: /V-|7-methyl-5-(naplithalen-l-yl)-7//-pyrrolo|2,3-rf|pyrimidin-4-yl]-Z>phcnylalaninc
Step A : 4-chloro-5-iodo-7-methyl-pyrrolo[2,3-à]pyrimidine
Into a 50 mL Schlenk tube under N2 atmosphère 220 mg NaH (5.5 mmol) and 40 mL dry THF were charged and the slurry was cooled to 0 °C. Then 1471 mg 4-chloro-5-iodo-7f/pyrrolo[2,3-i/]pyrimidine (5 mmol) was added. After 30 minutes stirring, 346 pL Mel (5.5 mmol) was added and the mixture was allowed to warrn up to r.t., and stirred until no further conversion was observed. The mixture was then diluted with saturated aqueous NH4CI solution, and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure to obtain 4-chloro-5-iodo-7-methyl-pyrrolo[2,3-t/]pyrimîdme.
’H NMR (400 MHz, DMSO-d6) δ: 8.65 (s, IH), 7.98 (s, IH), 3.83 (s, 3H) MS: (M+H)+ = 294.0
Step B: 4-chloro- 7-methyl-5-(/-naphthyl)pyrrolo[2,3-dJpyrimidine
ORIGINAL
-67l eq, 4-chloro-5-iodo-7-methyl-pyrrolo[2,3-c/]pyrimidine, l.l eq. l-naphthaleneboronic acid neopentyl glycol ester, l.l eq. silver carbonate, O.l5 eq, Pd(PPh3)4 and 2-Me-THF (l5mL/mmol 5-iodo-pyrrolo[2,3-fr]pyrimidine dérivative) were stirred under N2 atmosphère at 110 °C until no further conversion was observed. The mixture was diiuted with brine, neutralized with IM aqueous HCl solution, and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give 4-chloro-7-methyl-5-(lnaphthyl)pyrrolo[2,3-<7]pyrimidine. MS: (M+H)+ = 294.2
Step C: Example 10
Using General Procedure III and 4-chloro-7-methyl-5-(l-naphthyl)pyrrolo[2,3-i/] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, Example 10 was obtained. HRMS calculated for C26H22N4O2: 422.1743, found: 423.1804 (M+H)
Example 11: A'-|5-(naphthalen-l-yl)-7/7-pyrrolo|2,3-<'/|pyrimidin-4-yl|-Dphenylalanine
Step A: 7-(benzenesïtlfonyl)-4-chloro-5-iodo-pyrrolo[2.3-d]pyrimidine
Into a 50 mL Schlenk tube under N2 atmosphère 220 mg NaH (5.5 mmol) and 40 mL dry THF were charged and the slurry was cooled to 0 °C. Then 1471 mg 4-chloro-5-iodo-7//pyrrolo[2,3-i/]pyrîmidine (5 mmol) was added. After 30 minutes stirring, 1.4 mL benzenesulfonyl chloride (5.25 mmol) was added and the mixture was allowed to warm up to r.t., and stirred until no further conversion was observed. The mixture was then diiuted with saturated aqueous NH4CI solution, and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. Then it was digerated with MTBE, then filtered to obtain 7-(benzenesulfonyl)-4-chloro-5-iodo-pyrrolo[2,3-i/]pyrimidine, ’H NMR (400 MHz, CDC13) Ô: 8.75 (s, IH), 8.22 (m, 2H), 7.95 (s, IH), 7.67 (m, IH), 7.56 (m, 2H)
MS: (M+H)+ = 419.8
ORIGINAL
-68Step B: 7-(benzenesulfonyl)-4-chloro-5-(l~naphthyl)pyrrolo[2,3-à]pyrimidine l eq. 7-(benzenesulfonyl)-4-chloro-5-iodo-pyrrolo[2,3-i/]pyrimidine, l.l eq.
I-naphthaleneboronic acid neopentyl glycol ester, l.l eq. silver carbonate, 0.15 eq. Pd(PPh3)4 and 2-Me-THF (15 mL/mmol 5-iodo-pyirolo[2,3-i7]pyrimidine derivative) were stirred under N2 atmosphère at 110 °C until no further conversion was observed. The mixture was diluted with brine, neutralized with IM aqueous HCl solution, and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the fîltrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give 7-(benzenesulfonyl)-4-chloro-
5-( l -naphthyl)pyrrolo[2,3-i/]pyrimidine.
!H NMR (400 MHz, CDCl3) δ: 8.82 (s, IH), 8.31 (m, 2H), 7.94 (m, 2H), 7.84 (s, IH), 7.71 (m, IH), 7.60 (m, 2H), 7.56-7.48 (m, 3 H), 7.48-7.38 (m, 2H)
MS: (M+H)+= 420.0
Step C: Example 11
Using General Procedure III and 7-(benzenesulfonyl)-4-chloro-5-(l-naphthyl)pyrrolo [2,3-c/]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-^pyrimidine derivative and D-phenylalanine as the appropriate amino acid derivative, Example 11 was obtained. HRMS calculated for C25H20N4O2: 408.1586, found: 409.1670 (M+H)
Example 12: /V-I7-benzyl-6-ethyl-5-(naphthalen-l-yl)-7/7-pyrrolo[2,3-i/]pyrimidin-4yl]-Z)-phenylalanine, diastereoisomer 1 and
Exaniple 13: /V-|7-benzyl-6-ethyl-5-(naphthalen-l-yI)-7/f-pyrrolo[2,3-i/|pyrimidin-4yI]-D-phenylalanine, diastereoisomer 2
Using General Procedure IVa and Préparation 6 as the appropriate 5-bromo-pyrrolo [2,3-t7|pyrimidine derivative and 1-naphthaleneboronic acid neopentyl glycol ester as the appropriate boronic acid derivative, Example 12 was obtained as the earlier eluting diastereoisomer. HRMS calculated for CvjHkiN^L: 526.2369, found: 527.2431 (M+H).
ORIGINAL
-69Example 13 was obtained as the later eluting diastereoisomer. HRMS calculated for C34H30N4O2: 526.2369, found: 527.2423 (M+H)
Example 14: Ai-{6-ethyl-5-(naphthalen-l-yl)-7-[2-(naphthalen-l-yIoxy)ethyl]-7/7pyrrolo[2,3-rf]pyrimidin-4-yI}-D-phenylalanine, diastereoisomer 1 and
Example 15: jV-{6-ethyl-5-(naphthalen-l-yI)-7-|2-(naphthalen-l-yloxy)ethyI]-7Hpyrrolo|2,3if]pyrimidin-4-yl)-£>-phenylalanine, diastereoisomer 2
Step A: 5-bronio-4-chloro-6-ethyl-7-[2-(l-naphthyloxy)ethyl]pyrrolo[2,3-d]pyrimidine mg 2-(l-naphthyloxy)ethanol (0.5 mmol), 131 mg PPh3 (0.5 mmol) and 66 mg Préparation 1b (0.25 mmol) were dissolved in 2.5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 230 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyl-
7-[2-( 1 -naphthyloxy)ethyl]pyrrolo[2,3-i/]pyrimidine.
’H NMR (400 MHz, DMSO-d6) δ: 8.69 (s, IH), 7.80 (dd, 2H), 7.51-7.31 (m, 4H), 6.94 (d, IH), 4.90 (t, 2H), 4.52 (t, 2H), 3.08 (q, 2H), 1.26 (t, 3H)
MS: (M+H)+ = 430.0
Step B: (2R)-2-[[5-bromo-6-ethyl-7-[2-(l-naphthyloxy)eihyl]pyrrolo[2,3-d]pyrimidin-4yl]amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-[2-(l-naphthyloxy) ethyl]pyrrolo[2,3-c/|pyriniidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative (2Æ)-2-[[5-bromo-
6-ethyl-7-[2-(l-naphthyloxy)ethyl]pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenylpropanoic acid was obtained.
'H NMR (400 MHz, DMSO-dfi) δ: 12.96 (br s, 1 H), 8.24 (s, IH), 7.88 (d, IH), 7.82 (d, IH), 7.52-7.32 (m, 4H), 7.29-7.15 (m, 5H), 6.94 (d, IH), 6.38 (d, IH), 4.94 (q, IH), 4.72 (t, 2H), 4.45 (t, 2H), 3.28 (m, IH), 3.18 (dd, 1 H), 2.92 (q, 2H), 1.19 (t,3H)
MS: (M+H)+ = 559.2
ORIGINAL
-70Step C: Examples 14 and 15
Using General Procedure IVa and (27î)-2-[[5-bromo-6-ethyl-7-[2-(l-naphthyloxy) ethyl]pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate
5-bromo-pyrrolo[2,3-c/]pyrimidine derivative and 1 -naphthaleneboronic acid neopentyl glycol ester as the appropriate boronic acid derivative, Example 14 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C39H34N4O3: 606,2631, found. 607.2711 (M+H). Example 15 was obtained as the later eluting diastereoisomer. HRMS calculated for C39H34N4O3: 606.2631, found: 607.2705 (M+H)
Example 16: MI6-ethyl-5-(naphtha)en-l-yl)-7-(2-phenylethyl)-7H-pyrrolo[2,3-rf| pyrimidin-4-yl]-D-phenylalanine, diastereoisomer 1 and
Example 17: jV-[6-ethyl-5-(naphthalen-l-yl)-7-(2-phcnylethyl)-7/7-pyrrolo[2,3-rf] pyrimidin-4-yl]-D-phenylalanine, diastereoisomer 2
Step A: 5-bromo-4-chloro-6-ethyl-7-phenethyl-pyrrolo[2.3-d]pyrimidine
3.1 mL 2-phenylethanol (25.9 mmol), 3.397 g PPh3 (12.95 mmol) and 3.40 g Préparation 1b (12.95 mmol) were dissolved in 110 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 11.87 mL DEAD (40% in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyl-
7-phenethyl-pyrrolo[2,3-J]pyrimidine.
'H NMR (400 MHz, DMSO-d6) δ: 8.61 (s, 111), 7.32-7.16 (m, 3H), 7.11 (m, 2H), 4.51 (t, 2H), 3.06 (t, 2H), 2.70 (q, 2H), 1.10 (t, 3H)
MS: (M+H/ = 364.0
Step B: (2R)-2-[(5-bromo-6-ethyl-7-phenelhyl-pyrrolo[2,3-à]pyrimidin-4-yl)amino]-3phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-phenethyl-pyrrolo[2,3-ri] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine derivative and
ORIGINAL
-71D-phenylalanine as the appropriate amino acid dérivative (2/?)-2-[(5-bromo-6-cthyl-7phenethyl-pyrrolo[2,3-i/]pyrimidin-4-yl)amino]-3-phenyl-propanoic acid was obtained.
’H NMR (400 MHz, DMSO-d6) δ: 12.80 (br s, IH), 8.20 (s, IH), 7.34-7.17 (m, 8H), 7.13 (m, 2H), 6.45 (d, lH),4.9l (q, IH), 4.33 (t, 2H), 3.31 (dd, lH),3.!8(dd, IH), 3.00 (t. 2H), 2.55 (q, 2H), l .04 (t, 3H)
MS: (M+H)+= 493.2
Step C: Examples 16 and 17
Using General Procedure IVa and (2/î)-2-[(5-bromo-6-ethyl-7-phenethyl-pyrrolo[2,3-i/] pyrimidin-4-yl)amino]-3-phenyl-propanoic acid as the appropriate 5-bromo-pyrrolo[2,3-tZ] pyrimidine dérivative and 1-naphthaleneboronic acid neopentyl glycol ester as the appropriate boronic acid dérivative, Example 16 was obtained as the earlier eluting diastereoisomer. HRMS calculated tor C35H32N4O2: 540.2525, found: 541.2592 (M+H). Example 17 was obtained as the later eluting diastereoisomer. HRMS calculated for C35H32N4O2: 540.2525, found: 541.2619 (M+H)
Example 18: /V-[6-ethyl-5-(naphthalen-l-yl)-7-(3-phenylpropyl)-7H-pyrrolo[2,3-i/] pyrimidin-4-yl]-Z>-phenylalanine, diastereoisomer 1 and
Example 19: /V-|6-ethyl-5-(naphthalen-l-yl)-7-(3-phenylpropyl)-7//-pyrrolo[2,3-rf] pyrimidin-4-yl|-Z>-phenylalanine, diastereoisomer 2
Step A: 5-bromo-4-chloro-6-ethyl-7-(3-phenylpropyl)pyrrolo[2,3-à]pyrimidine
3.52 mL 3-phenyl-propanol (25.9 mmol), 3.397 g PPI13 (12.95 mmol) and 3.4 g Préparation 1b (12.95 mmol) were dissolved in 110 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 11.87 mL DEAD (40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyl-
7-(3-phenylpropyl)pyrrolo[2,3-ô/]pyrimidine.
'H NMR (400 MHz, DMSO-d6) δ: 8.60 (s, 1 H), 7.31-7.22 (m, 2H), 7.21-7.13 (m, 3H),
4.32 (t, 21-1), 2.85 (q, 2H), 2.65 (t, 2H), 2.05 (m, 2H), 1.16 (t, 3H)
ORIGINAL
-72MS: (M+H)+ = 378.0
Step B: (2R.)-2-[[5-bromo-6-ethyl-7-(3-phenylpropyl)pyrrolo[2,3-dJpyrimidin-4-yl] amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-(3-phenylpropyl)pyrrolo [2,3-t7]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, (2/?)-2-[[5-bromo-6-ethyl-7-(3phenylpropyl)pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained. 'H NMR (400 MHz, DMSO-d6) δ: 12.95 (br s, IH), 8.15 (s, IH), 7.33-7.12 (m, 10H), 6.35 (d, IH), 4.94 (q, IH), 4J6 (t, 2H), 3.28 (dd, IH), 3.16 (dd, IH), 2.68 (q, 2H), 2.61 (t, 2H),
1.97 (m.2H), l .09 (t, 3H)
MS: (M+H)+ = 507.2
Step C: Examples /8 and 19
Using General Procedure IVa and (27?)-2-[[5-bromo-6-ethyl-7-(3-phenylpropyl) pyrrolo[2,3-t/] pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromopyrrolo[2,3-c/] pyrimidine dérivative and l-naphthaleneboronic acid neopentyl glycol ester as the appropriate boronic acid dérivative, Example 18 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C36H34N4O2: 554.2682, found: 555.2742 (M+H). Example 19 was obtained as the later eluting diastereoisomer. HRMS calculated for C36H34N4O2: 554.2682, found: 555.2756 (M+H)
Example 20: Ar-[(57fn)-5-(3-chloro-2-methyIphenyl)-6-ethyl-7-methyl-7/7-pyrrolo[2,3i/]pyrimidin-4-yl]-£>-plicnykilanine and
Example 21:7V-[(55'<f)-5-(3-chloro-2-metliylphenyI)-6-ethyl-7-mcthyl-7/7-pyrroloj2,3rf]pyrïmidin-4-yl]-Z>-phenyIalanine
Step A: 5-bromo-4-chloro-6-ethyl-7-methyl-pyrrolo[2,3-d]pyrimidine mg Préparation 1b (0.25 mmol) was dissolved in I mL dry THF, then 20.3 pL dry MeOH (0.5 mmol) and 0.5 mL cyanomethylenetributylphosphorane solution (0.5 mmol, 1M in toluene) was added. The mixture was stirred at r.t. until no further conversion was
ORIGINAL
-73observed. The volatiles were removed under reduced pressure. The residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-
6-ethyl-7-methyl-pyrrolo[2,3-i/]pyrimidine.
H NMR (400 MHz, CDCl3) Ô: 8.56 (s, IH), 3.84, (s, 3H), 2.91 (q, 2H), 1.26 (t, 3H)
MS: (M+H)+ = 274.0
Step B: (2R)-2-[[5-bromo-6-ethyl-7-methyl-pyrrolo[2,3-à]pyrimidin-4-yl]amino]-3phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-methyl-pyrrolo[2,3-t7] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-r(|pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, (2/?)-2-[[5-bromo-6-ethyl-7methyl-pyrrolo[2,3-cf] pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (500 MHz, DMSO-d6) δ: 13.05 (br s, IH), 8.17 (s, IH), 7.32-7.25 (m. 2H), 7.25-
7.18 (m, 3H), 6.32 (d, IH), 4.97 (m, IH), 3.68, (s, 3H), 3.29 (dd, IH), 3.18 (dd, IH), 2.75 (q,2H), 1.13 (t, 3H)
MS: (M+H)+ = 403.0
Step C: Examples 20 and 21
Using General Procedure IVa and (2A)-2-[[5-bromo-6-ethyl-7-methyl-pyrrolo[2,3-i/] pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromo-pyrrolo[2,3-i/] pyrimidine derivative and Préparation 3c as the appropriate boronic acid dérivative, Example 20 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C25H2îC1N4O3: 448.1666, found: 449.1753 (M+H). Example 21 was obtained as the later eluting diastereoisomer. HRMS calculated for C25H25CIN4O2: 448.1666, found: 449.1752 (M+H)
Example 22: jV-|(5/?a)-5-(3-chloro-2-mcthylphenyl)-7-(cyclopropylmethyl)-6-ethyl-7//pyrrolo|2,3-rf]pyrimidin-4-yl]-Z)-plienylalanine and
Example 23: A/-[(5Sfl)-5-(3-chloro-2-methylphenyl)-7-(cyclopropylmethyl)-6-ethyl-7Hpyrrolo|2,3-i/]pyrimidin-4-yl]-£>-phenylaIanine
ORIGINAL
- 74Step A: 5-bromo-4-chloro-7-(cyclopropylmethyl)-6-ethyl-pyrrolo[2,3-d]pyrimidine mg Préparation 1b (0.25 mmol) was dissolved in l mL dry THF, then 40 pL cyclopropanemethanol (0.5 mmol) and 0.5 mL cyanoinethyienetributylphosphorane solution (0.5 mmol, IM in toluene) was added. The mixture was stirred at r.t. until no further conversion was observed. The volatiles were removed under reduced pressure. The residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-7-(cyclopropylmethyl)-6-ethyl-pyrrolo[2,3-t/]pyrimidine.
'H NMR (400 MHz, CDClj) Ô: 8.54 (s, IH), 4.18 (d, 2H), 2.94 (q, 2H), 1.29 (t, 3H), l.24l.l4(m, lH), 0.60-0.51 (m, 2H), 0.51-0.43 (m,2H)
MS: (M+H)+= 314.0
Step B: (2R)-2-[[5-bromo-7-(cyctopropylmethyl)-6-ethyl-pyrrolo[2,3-d]pyrimidin-4-yl] amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-7-(cyclopropylmethyl)-6-ethylpyrrolo[2,3-iZ]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-c/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, (2/?)-2-[[5-bromo-7(cyclopropylmethyl)-6-ethyl-pyrrolo[2,3-i7]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (500 MHz, DMSO-d6) δ: 13.05 (br s, IH), 8.15 (s, IH), 7.32-7.26 (m, 2H), 7.26-
7.20 (m, 3H), 6.34 (d, IH), 4.94 (m. IH), 4.05 (d, 2H) 3.29 (dd, IH), 3.18 (dd, IH), 2.78 (q, 2H), 1.28-1.20 (m, 1 H), 1.16 (t, 3H), 0.47-0.42 (m, 2H), 0.42-0.37 (m, 2H)
MS: (M+H)+ = 443.0
Step C: Examples 22 and 23
Using General Procedure IVa and (27?)-2-[[5-bromo-7-(cyclopropylmethyl)-6-ethylpyrroio[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromopyrrolo[2,3-t/]pyrimidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Example 22 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C28H29CIN4O2: 488.1979, found: 489.2064 (M+H). Example 23 was obtained as the later eluting diastereoisomer. HRMS calculated for C28H29CIN4O2: 488.1979, found: 489.2048 (M+H)
ORIGINAL
-75Example 24: /V-[(57?fl)-5-(3-chloro-2-methylphenyl)-6-ethyl-7-(prop-2-en-l-yl)-7//pvrrolo[2,3-rf]pyrimidin-4-yl]-D-phenylalanine and
Example 25: ?V-|(5Sn)-5-(3-chloro-2-methylphenyI)-6-ethyl-7-(prop-2-en-l-yl)-7//pyrrolo|2,3-r/|pyrimidin-4-yl]-£>-phenylalanine
Step A: 7~allyl-5-bromo-4-chloro-6-ethyl-pyrrolo[2,3-d]pyrimidine mg Préparation 1b (0.25 mmol) was dissolved in l mL dry THF, then 34 pL allylalcohol (0.5 mmol) and 0.5 mL cyanomethylenetributylphosphorane solution (0.5 mmol, l M in toluene) was added. The mixture was stirred at r.t. until no further conversion was observed. The volatiles were removed under reduced pressure. The residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 7-allyl-5-bromo-4chloro-6-ethyl-pyrrolo[2,3-i/]pyrimidine.
‘H NMR (400 MHz, CDCl3) δ: 8.57 (s, IH), 6.02-5.90 (m, IH), 5.25-5.16 (m, IH), 5.00-
4.85 (m, 3H), 2.87 (q, 2H), l .26 (t, 3H) MS: (M+H)+ = 300.0
Step B: (2R)-2-[[7-allyl-5-bromo-6-ethyl-pyrrolo[2,3-d]pyriniidin-4~yl]amino]-3-phenylpropanoic acid
Using General Procedure [Il and 7-allyl-5-bromo-4-chloro-6-ethyl-pyrrolo[2,3-<7] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-r/]pyrimidine derivative and D-phenylalanine as the appropriate amino acid derivative, (2Æ)-2-[[7-allyl-5-bromo-6ethyl-pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (500 MHz, DMSO-d6) δ: 13.06 (br s, IH), 8.16 (s, IH), 7.34-7.26 (m, 2H), 7.267.l9(m, 3H), 6.35 (d. I H), 6.01-5.89 (m, IH), 5.l0(dd, IH), 5.01-4.93 (m, IH), 4.87-4.73 (m, 3H), 3.29 (dd. IH), 3.18 (dd, IH), 2.70 (q, 2H), 1.12(1, 3H)
MS: (M+H)+= 429.0
Step C: Examples 24 and 25
Using General Procedure IVa and (2À)-2-[[7-allyl-5-bromo-6-ethyl-pyrrolo[2,3-r/] pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromo-pyrroIo[2,3-c/j pyrimidine derivative and Préparation 3c as the appropriate boronic acid derivative,
ORIGINAL
-76Example 24 was obtained as the earlier eluting dîastereoisomer. HRMS calculated for C27H27CIN4O2: 474.1823, found: 475.1908. Example 25 was obtained as the later eluting dîastereoisomer. HRMS calculated for C27H27CIN4O2: 474,1823, found: 475.1909
Example 26:7V-|7-(but-2-yn-l-yl)-(5/fa)-5-(3-chloro-2-methylphenyl)-6-ethyl-7£fpyrrolo[2,3-rf]pyrimidin-4-yl]-Z)-phenylalanine and
Example 27: ZV-|7-(but-2-yn-l-yl)-(55'fl)-5-(3-chloro-2-methylphenyl)-6-ethyl-7Hpyrrolo|2,3-rf]pyrimidin-4-yl]-Z>-phenyIalanine
Step A: 5-bromo-7-but-2-ynyl-4-chloro-6-ethyl-pyrrolo[2,3-d]pyrimidine pL 2-butyn-l-ol (0.5 mmol), 131 mg PPha (0.5 mmol) and 66 mg Préparation 1b (0.25 mmol) were dissolved in 2.5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 230 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-7-but-2-ynyI-4-chIoro-6-ethylpyrrolo[2,3-i/] pyrimidine.
'H NMR (400 MHz. CDCI3) Ô: 8.59 (s, IH), 5.03 (q, 2H), 2.99 (q, 2H), 1.77 (t, 3H), 1.33 (t, 3H)
MS: (M+H)+ = 312.0
Step B: (2R)-2-[(5-bromo- 7-but-2-ynyl-6-ethyl-pyrrolo[2,3-à]pyrimidin-4-yl)amino]-3phenyi-propanoic acid
Using General Procedure III and 5-broino-7-but-2-ynyl-4-chloro-6-ethyl-pyrrolo[2,3-tZ] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative (2Æ)-2-[(5-bromo-7-but-2-ynyl-
6-ethyl-pyrrolo[2,3-t/]pyrimidin-4-yl)amino]-3-phenyl-propanoic acid was obtained.
fH NMR (500 MHz, DMSO-d6) δ: 13.25 (br s, IH), 8.19 (s, 1 H), 7.30-7.24 (m, 2H), 7.24-
7.16 (m, 3H), 6.45 (d, IH), 5.02-4.96 (m, 2H), 4.93 (q, IH), 3.30 (dd, IH), 3.19 (dd, 1 H),
2.80 (q, 2H), 1.74 (t, 3H), 1.19 (t,3H)
MS: (M+H)+=441.0
ORIGINAL
-77Step C: Examples 26 and 2 7
Using General Procedure IVa and (2Æ)-2-[(5-bromo-7-but-2-ynyl-6-ethyi-pyrrolo[2,3-J] pyrimidin-4-yl)amino]-3-phenyl-propanoic acid as the appropriate 5-bromo-pyrrolo[2,3-c/] pyrimidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Example 26 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C28H27CIN4O2: 486.1823, found: 487.1893 (M+H). Example 27 was obtained as the later eluting diastereoisomer. HRMS calculated for C28H27CIN4O2: 486.1823, found: 487.1893 (M+H)
Example 28: JV-|(5/f„)-5-(3-chloro-2-methylphenyl)-6-ethyl-7-(2,2,2-trifluoroethyl)7H-pyrrolo[2,3-rf|pyrinndin-4-yl]-Z>-phenylalanine and
Example 29:7V-[(55, ü)-5-(3-chIoro-2-methylphenyl)-6-ethyl-7-(2,2,2-trifluoroethyI)-777pyrrolo[2,3-i/lpyrimidin-4-yl|-£>-phenylalanine
Step J; 5-bromo-4-chloro-6-ethyl-7-(2,2,2-trifluoroethyl)pyrrolo[2,3-à]pyrimidine pL trifluoroethanol (1 mmol), 262 mg PPhj (1 mmol) and 130 mg Préparation 1b (0.5 mmol) were dissolved in 5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 460 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethy 1-7-(2,2,2-trifluoro ethyl)pyrrolo[2,3-i/J pyrimidine.
‘1-1 NMR (400 MHz, CDCI3) δ: 8.62 (s, IH), 4.90 (q, 2H), 2.94 (q, 2H), 1.28 (t, 3H)
MS: (M+H)+ = 342.0
Step B: (2R)-2-[[5-bromo-6-ethyl-7-(2,2,2-trifluoroethyl)pyrrolo[2,3-à]pyrimidin-4-yl] amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-(2,2,2-trifluoroethyl)pyrrolo [2,3-i/]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimîdine dérivative and D-phenylalanine as the appropriate amino acid dérivative (27?)-2-[[5-bromo-6-ethyl-7ORIGINAL
-78(2,2,2-trifluoroethyl)pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
‘H NMR (500 MHz, DMSO-d6) Ô: I3.l l (br s, IH), 8.23 (s, IH), 7.33-7.26 (m, 2H), 7.26-
7.19 (m, 3H), 6.44 (d, IH), 5.12 (q, 2H), 5.00-4.93 (m, IH), 3.30 (dd, IH), 3.20 (dd, IH), 2.78 (q, 2H), l.l4(t, 3H)
MS: (M+H/= 471.0
Step C: Examples 28 and 29
Using General Procedure IVa and (2À)-2-[[5-bromo-6-ethyl-7-(2,2,2-trifluoroethyl) pyrrolo[2,3-iZ|pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromopyrrolo[2,3-i/]pyrimidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Example 28 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C26H24CIF3N4O2: 516.1540, found: 517.1624 (M+H). Example 29 was obtained as the later eluting diastereoisomer. HRMS calculated for C26H24CIF3N4O2: 516.1540, found: 517.1606 (M+H)
Example 30:2V-[(5/ta)-5-(3-chloro-2-methylphenyl)-7-(2-cyclopentv’lethyl)-6-ethyl-7Wpyrrolo[2,3-i/]pyrimidin-4-yl]-Z)-phenylalaninc and
Example 31 : j'V-|(55a)-5-(3-chioiO-2-methylphenyl)-7-(2-cyclopentylethyl)-6-ethyl-7//pyrrolo[2,3-rf|pyrimïdin-4-yl]-Z>-phenylalanine
Step A : 5-bromo-4-chloro-7-(2-cyclopentylethyl)-6-ethyl-pyrrolo[2.3-à]pyrimidine
124 pL 2-cyclopentylethanol (1 mmol), 262 mg PPh3 (1 mmol) and 130 mg Préparation lb (0.5 mmol) were dissolved in 5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 460 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-7-(2cyclopentylethyl)-6-ethyl-pyrrolo[2,3-c/]pyrimidine.
1H NMR (400 MHz, CDCI3) δ: 8.55 (s, 1 H), 4.31 -4.20 (m, 2H), 2.89 (q, 2H), 1.91 -1.72 (m, 5H), 1.69-1.57 (m, 2H), 1.57-1.46 (m, 2H), 1.28 (t, 3H), 1.23-1.05 (m, 2H)
ORIGINAL
-79MS: (M+H)+ = 356.0
Step B: (2R)-2-[[5-bromo-7-(2-cyclopentylethyl)-6-ethyl-pyrrolo[2,3-à]pyrimidin-4-yl] am ino]-3~phenyl-propano ic acid
Using General Procedure III and 5-bromo-4-chloro-7-(2-cyclopentylethyl)-6-ethylpyrrolo[2,3-(/]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-</]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative (2J?)-2-[[5-bromo-7-(2cyclopentylethyl)-6-ethyl-pyrrolo[2,3-i7]pyrimidin-4-yI]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (500 MHz, DMSO-d6) S: 13.04 (br s, IH), 8.17 (s, IH), 7.32-7.26 (m, 2H), 7.25-
7.19 (m, 3H), 6.32 (d, IH), 5.00-4.92 (m, IH), 4.17-4.09 (m. 2H), 3.29 (dd, 1 H), 3.18 (dd, IH), 2.74 (q, 2H), 1.79-1.70 (m, 3H), 1.70-1.62 (m, 2H), 1.60-1.50 (m, 2H), 1.50-1.42 (m, 2H), 1.15 (t,3H), 1.12-1.01 (m, 2H)
MS: (M+H)+ = 485.2
Step C: Examples 30 and 31
Using General Procedure IVa and (2Æ)-2-[[5-bromo-7-(2-cyclopentylethyl)-6-ethylpyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromopyrrolo[2,3-i/]pyrimidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Example 30 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C31H35CIN4O2: 530.2449, found: 531.2528 (M+H). Example 31 was obtained as the later eluting diastereoisomer. HRMS calculated for C31H35CIN4O2: 530.2449, found: 531.2547 (M+H)
Example 32:7V-|(57?„)-5-(3-chloro-2-methylphenyl)-6-ethyl-7-(naphthalen-lylmethyl)-7H-pyrrolo [2,3-(/] pyrimidin-4-yl|-Z>-plienylalanme and
Example 33:7V-|(5Sa)-5-(3-chloro-2-methylphenyl)-6-ethyI-7-(naphthalen-l-ylmethyI)7//-pyrroIo|2,3-i/|pyrimidin-4-yl|-/)-phenylalanine
Step A: 5-bromo-4-chloro-6-ethyl-7-(l-naphthylmethyl)pyrrolo[2,3-à]pyrimidine
ORIGINAL
-80158 mg l-naphthalenemethanol (1 mmol), 262 mg PPhj (1 mmol) and 130 mg Préparation 1b (0.5 mmol) were dissolved in 5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 460 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40°C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyI-
7-( 1 -naphthylmethyl)pyrrolo[2,3-f/]pyrimidine.
‘H NMR (400 MHz, CDC13) δ: 8.58 (s, IH), 8.09 (d, IH), 7.95-7.89 (m, IH), 7.79 (d, IH), 7.66-7.54 (m, 2H), 7.25 (t. IH), 6.45 (dd, IH), 6.03 (s, 2H), 2.76 (q, 2H), 1.08 (t, 3H) MS: (M+H)+ = 400.0
Step B: (2R.)-2-[[5-bromo-6-ethyl-7-(l-naphlhylmethyl)pyrrolo[2,3-d]pyrinüdin-4-yl] amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-(l-naphthylmethyl)pyrrolo [2,3-i/]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative (2Æ)-2-[[5-bromo-6-ethyl-7-(lnaphthylmethyl)pyrrolo[2,3-c/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (500 MHz, DMSO-d6) δ: 13.14 (br s, IH), 8.27 (d, IH), 8,15 (s, IH), 7.98 (d, IH), 7.83 (d, IH), 7.66-7.56 (m, 2H), 7.37-7.20 (m, 6H), 6.48 (d, IH), 6.40 (d, IH), 5.94 (s, 2H), 4.99 (q, IH), 3.33 (dd. IH), 3.22 (dd, IH), 2.62 (q, 2H), 0.89 (t, 3H) MS: (M+Hf = 529.0
Step C: Examples 32 and 33
Using General Procedure IVa and (2Æ)-2-[(5-bromo-6-ethyl-7-(l-naphthylmethyl)pyrrolo [2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate 5-bromopyrrolo[2,3-i/]pyrimidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Examplc 32 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C35H31CIN4O2: 574.2136, found: 575.221 1 (M+H). Example 33 was obtained as the later eluting diastereoisomer. HRMS calculated for C35H31CIN4O2: 574.2136, found: 575.2203 (M+H)
ORIGINAL
-81Examplc 34: /V-[(5Æa)-5-(3-chloro-2-methyIphenyl)-6-ethyl“7-(4-methoxybeiizyl)-7//pyrroIo[2,3-i/]pyrimidin-4-yl]-Z>-phenylalanine and
Examplc 35: Ar-|(5Sa)-5-(3-chloro-2-mcthylphenyl)-6-ethyl-7-(4-methoxybenzy 1)-7//pyrrolo(2,3-rf]pyrimidin-4-yl]-Z)-phenylaIanine
Step A: 5-bromo-4-cldoro-6-ethyl-7-[(4-methoxyphenyl)methyl]pyrrolo[2,3-à]pyrimidîne 138 mg 4-methoxybenzyl alcohol (1 mmol), 262 mg PPhj (1 mmol) and 130 mg Préparation 1b (0.5 mmol) were dissolved in 5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 460 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyl-
7-[(4-methoxyphenyl)methyl]pyrrolo[2,3-7]pyrimidine. MS: (M+H)+ = 380.0
Step B: (2Rj-2-[[5-bromo-6-ethyl-7-[(4-methoxyphenyl)methyl]pyrrolo[2,3-à]pyrimidin-4yl]amino]-3-phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyi-7-[(4-methoxyphenyl) methyl]pyrrolo[2,3-i/]pyrÎmidine as the appropriate 4-chloro-pyrrolo[2,3-7]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, (2Æ)-2-[[5bromo-6-ethyl-7-[(4-methoxyphenyl)methyl]pynolo[2,3-i7]pyrimidin-4-yl]aniino]-3phenyl-propanoic acid was obtained.
'H NMR (500 MHz. DMSO-d6) δ: 13.07 (br s, IH), 8.20 (s, IH), 7.33-7.17 (m, 5H), 7.03 (d, 2H), 6.85 (d, 2H), 6.37 (d, IH), 5.37 (s, 2H), 4.99 (q, IH), 3.69 (s, 3H), 3.31 (dd, IH),
3.20 (dd, IH), 2.65 (q, 2H), 0.91 (t. 3H)
MS: (M+H)+ = 508.8
Step C: Examples 34 and 35
Using General Procedure IVa and (2/?)-2-[[5-bromo-6-ethyI-7-[(4-methoxyphenyl) methyl]pyrrolo[2,3-7]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid as the appropriate
5-bromo-pyrrolo[2,3-7]pyriniidine dérivative and Préparation 3c as the appropriate boronic acid dérivative, Example 34 was obtained as the earlier eluting diastereoisomer.
ORIGINAL
- 82HRMS calculated for C32H3]CIN4O3: 554.2085, found: 555.2176 (M+H). Example 35 was obtained as the later eluting diastereoisomer. HRMS calculated for C32H31CIN4O3: 554.2085, found: 555.2140 (M+H)
Example 36: jV-[7-benzyl-(5iï(,)-5-(3-chloro-2-methylphenyl)-6-ethyl-7/7-pyrrolo[2,3rf]pyrimidin-4-yI]-Z>-phenylalanine and
Example 37: Ai-[7-benzyl-(5Xfl)-5-(3-chloro-2-methylphenyI)“6-ethyl-7/f-pyrrolo[2,3-i/| pyrimidin-4-yl]-Z)-phenylalanine
Using General Procedure IVa and Préparation 6 as the appropriate 5-bromo-pyrrolo [2,3-i/|pyrimidine derivative and Préparation 3c as the appropriate boronic acid derivative, Example 36 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C31H29CIN4O2: 524.1979, found: 525.2048 (M+H). Example 37 was obtained as the later eluting diastereoisomer. HRMS calculated for C31H29CIN4O2: 524.1979, found: 525.2064 (M+H)
Example 38: jV-|(5/fa)-5-(3-chloro-2-methylphenyl)-6-etliyl-7-(propan-2-yl)-7/7pyrrolo[2,3-rf|pyrÎmidin-4-yl]-£)-phenylalanine and
Example 39: /V-|(55„)-5-(3-chloro-2-methylphenyl)-6-ethyl-7-(propan-2-yl)-7Hpyrrolo[2,3-rf|pyrimidin-4-yl]-£>-plienylalanine
Slep A : 5-bromo-4-chloro-6-ethyl-7-isopropyl-pyrrolo[2,3-d]pyrimidine pL 2-propanol (1 mmol), 262 mg PPI13 (1 mmol) and 130 mg Préparation 1b (0.5 mmol) were dissolved in 5 mL dry THF under N2 atmosphère and cooled to 0 °C. Then 460 pL DEAD (0.5 mmol, 40 % in toluene) was added dropwise. The mixture was stirred at 40 °C until no further conversion was observed. Then the volatiles were removed under reduced pressure and the residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain 5-bromo-4-chloro-6-ethyl-7-isopropyl-pyrrolo [2,3-i/]pyrimidine.
ORIGINAL
-83*H NMR (400 MHz. CDCl3) 6: 8.53 (s, IH), 4.71 (sp, IH), 2.92 (q, 2H), 1.72 (d, 6H), 1.25 (t, 3H)
MS: (M+H)' =302.0
Step B: (2B.)-2-[(5-bromo-6-ethyl-7-isopropyl-pyrrolo[2,3-d]pyrimidin-4-yl)amino]-3phenyl-propanoic acid
Using General Procedure III and 5-bromo-4-chloro-6-ethyl-7-isopropyl-pyrrolo[2,3-i7] pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-iZ]pyrimidine derivative and D-phenylalanîne as the appropriate amino acid derivative (27?)-2-[(5-bromo-6-ethyl-7isopropyl-pyrrolo[2,3-i7]pyrimidin-4-yl)amino]-3-phenyl-propanoic acid was obtained.
‘H NMR (500 MHz. DMSO-d6) δ: 13.04 (br s, IH), 8.14 (s, IH), 7.35-7.17 (m. 5H), 6.33 (d, IH), 4.95 (q, IH), 4.64 (sp, IH), 3.28 (dd, IH), 3.17 (dd, IH), 2.76 (q, 2H), 1.59 (d, 6H), 1.11 (t, 3H)
MS: (M+H)+ = 431.2
Step C: Examples 38 and 39
Using General Procedure IVa and (27?)-2-[(5-bromo-6-ethyl-7-isopropyl-pyrrolo[2,3-i/] pyrimidin-4-yl)amino]-3-phenyl-propanoic acid as the appropriate 5-bromo-pyrrolo[2,3-r/] pyrimidine derivative and Préparation 3c as the appropriate boronic acid derivative, Example 38 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C27H29CIN4O2: 476.1979, found: 477.2057 (M+H). Example 39 was obtained as the later eluting diastereoisomer. HRMS calculated for C27H29CIN4O2: 476.1979, found: 477.2063 (M+H)
Example 40: (2/ï)-2-[(7-benzyl-(55'„)-5-{3-chloro-2-methyl-4-[2-(4-methylpipcrazin-lyI)ethoxy]phenyl}-6-ethyI-7H'-pyrrolo[2,3-rf| pyrimidm-4-yl)oxy]-3-phenylpropanoic acid
Step A: 7-benzyl-5-bronio-4-chloro-6-ethyl-pyirolo[2,3-àJpyrimidine
255 mg NaH (6.38 mmol) and 50 mL dry THF were charged into a 50 mL Schlenk tube under N2 atmosphère and the slurry was cooled to 0 °C. Then 1.792 g Préparation 1b (5.8 mmol) was added. After stirring the mixture for 30 minutes at 0 °C, 773 pL benzyl
ORIGINAL
-84bromide (6.38 mmol) was added and the mixture was allowed to warm up to r.t., and stirred until no further conversion was observed. The mixture was then diluted with saturated aqueous NH4CI solution, and extracted with DCM. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 7-benzyl-5-bromo-4-chloro-6-ethyl-pyrrolo[2,3-i/] pyrimidine. 'H NMR (400 MHz, CDClj) δ: 8.58 (s, IH), 7.35-7.20 (m, 3H), 7.10-6.96 (m, 2H), 5.52 (s, 2H), 2.78 (q, 2H), 1.05 (t, 3H)
Step B: Methyl (2R)-2-(7-benzyl-5-bromo-6-ethyl-pyrrolo[2,3-8]pyrimidin-4-yl)oxy-3phenyl-propanoate
1.639 g 7-benzyl-5-bromo-4-chloro-6-ethyl-pyrrolo[2,3-i7]pyrimidine (4.67 mmol) was dissolved in 47 mL dry DMSO, then 2.948 g methyl (2Æ)-2-hydroxy-3-phenyl-propanoate (16.4 mmol) and 7.234 g Cs2CO3 (22.2 mmol) were added and the mixture was stirred at 100 °C under N2 atmosphère until no further conversion was observed. Then it was diluted with water and brine, extracted with DCM. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and 'Pr2O as eluents to obtain methyl (2Æ)-2-(7-benzyl-5-bromo-6-ethyl-pyrrolo[2,3-i/]pyrimidin-4-yl)oxy-3-phenyl-propanoate. ‘H NMR (400 MHz, CDC13) δ: 8.29 (s, IH), 7.47 (d, 2H), 7.36-7.19 (m, 6H), 7.06-6.96 (m, 2H), 5.60 (dd, IH), 5.47 (s, 2H), 3.73 (s, 3H), 3.41-3.28 (m, 2H), 2.72 (q, 2H), 1.03 (t, 3H) MS: (M - H)' = 494.2
Step C: Methyl (2R.)-2-[7-benzyl-(5S3)-5-[3-chloro-2-methyl-4-hydroxphenyl]-6-ethylpyrrolo [2,3-8]pyrim idin-4 -yl] oxy-3 -phenyl-propanoate
A mixture of 1.20 g methyl (2Æ)-2-(7-benzyl-5-bromo-6-ethyl-pyrrolo[2,3-i(]pyrimidin-4yl)oxy-3-phenyl-propanoate (2.43 mmol), 1.98 g Préparation 3a (7.21 mmol), 110 mg Pd(OAc)2 (0.49 mmol), 350 mg butyl-diadamantylphosphine (0.98 mmol), and 7.35 mL IM aqueous TBAOH in 18 mL DME was heated under MW irradiation at 100 °C until no further conversion was observed. The reaction mixture was filtered through Celite. Water was added to the filtrate, it was acidified to pH = 4 and extracted with MTBE. The
ORIGINAL
-85combined organic phases were dried over Na^SCL, filtered and the filtrate was concentrated under reduced pressure.
The residue was heated in a mixture of 10 mL MeOH and 40 pL cc. H2SO4 until no further conversion was observed. The volatiles were removed under reduced pressure, the residue 5 was diluted with water, the pH was set to 5, and it was extracted with DCM. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl (2Æ)-2-[7-benzyl-(5S’ÎJ)-5-[3-chloro-2-methyl-4hydroxphenyl]-6-ethyl-pyrroIo[2,3-i7]pyrimidin-4-yl]oxy-3-phenyl-propanoate as the later I0 eiuting diastereoisomer. lH NMR (500 MHz, DMSO-d&) δ: 10.14 (s, IH), 8.27 (s, IH), 7.34-7.27 (m, 2H), 7.27-7.22 (m, IH), 7.17-7.07 (m, 4H), 7.05 (d, 2H), 6.98 (dd, IH), 6.64 (d, 2H), 5.60 (d, IH), 5.51 (d, IH), 5.43 (dd, IH), 3.56 (s, 3H), 3.00 (dd, IH), 2.85 (dd, IH), 2.60-2.51 (m, IH), 2.48-2.38 (m, IH), 2.04 (s, 3H), 0.84 (t. 3H)
Step D: Example 40
139 mg methyl (2/î)-2-[7-benzyl-(5S, a)-5-[3-chloro-2-methyl-4-hydroxphenyl]-6-ethylpyrrolo[2,3-i/]pyrimidin-4-yl]oxy-3-phenyl-propanoate (0.25 mmol), 72 mg l-(2-hydroxyethyl)-4-methylpiperazine (0.50 mmol) and 166 mg resin bound PPh3 (0.5 mmol) were dissolved in 3 mL dry toluene under N2 atmosphère, then 115 mg DTAD (0.5 mmol) was added. The mixture was stirred at 50 °C until no further conversion was 20 observed. The mixture was then diluted with DCM, filtered and the filtrate concentrated under reduced pressure, and purified via flash chromatography using heptane, EtOAc and MeOH as eluents. The obtained intermediate was dissolved in 10 mL MeOH, then 500 mg LiOHxH2O was added, and the mixture was stirred at 50 °C until no further conversion was observed. The mixture was diluted with brine, neutralized with IM aqueous HCl 25 solution and extracted with DCM. The organic layer was dried over Na2SÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 40 mM aqueous NH4OAc solution (pH = 4, adjusted with AcOH) and MeCN as eluents to obtain Example 40. HRMS calculated for C3sH42ClN5O4: 667.2925, found: 668.2992 (M+H)
Example 41: iV-[6-bromo-7-(but-3-en-l-yl)-(5/ffl)-5-(3-chloro-2-methylphenyl)-7/7ORIGINAL
-86pyrrolo[2,3-if]pyrimidin-4-yl]-Z)-phenylalanine and
Example 42: 7V-[6-bromo-7-(but-3-en-l-yl)-(55ir)-5-(3-chloro-2-methylpiienyl)-7Hpyrrolo[2,3-i/]pyrimidin-4-yl]-Z)-phenylalanine
Step A: 7-but-3-enyl-4-chloro-5-iodo-pyrrolo[2,3-à]pyrimidine
5.0 g 4-chloro-5-iodo-7Z/-pyrrolo[2,3-i/]pyrimÎdine (17 mmol), 2.842 g K2CO3 (20.57 mmol), 2.15 mL 4-bromo-l-butene (20.57 mmol) and 26 mL dry DMF were stirred at r.t. under N2 atmosphère until no further conversion was observed. Then the mixture was poured into water and extracted with EtOAc. The combined organic layers were washed with water, dried over MgSOj, fîitered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc to obtain 7-but-3-enyl-4-chloro-5-iodo-pyrrolo[2,3-i/]pyrimidine.
'H NMR (400 MHz, CDCI3) Ô: 8.62 (s, IH), 7.38 (s, IH), 5.82-5.69 (m, IH), 5.08 (s, IH), 5.04 (dd, IH), 4.33 (t, 2H), 2.60 (q, 2H)
MS; (M+H)+ = 334.0
Step B: (2\A)-2-[(7-but-3-enyl-5-iodo-pyrrolo[2.3-<5]pyrimidin-4-yl)amino]-3-phenylpropanoic acid
Using General Procedure III and 7-but-3-enyl-4-chloro-5-iodo-pyrrolo[2,3-i7]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine derivative and D-phenylalanine as the appropriate amino acid derivative, (2Â)-2-[(7-but-3-enyl-5-iodo-pyrrolo[2,3-i/]pyrimidin4-yl)amino]-3-phenyl-propanoic acid was obtained. ’H NMR (400 MHz, CDCI3) δ: 8.32 (s, IH), 7.38 (s, IH), 7.35-7.28 (m, 31-1), 7.28-7.22 (m, 2H), 7.02 (s, IH), 6.28 (d, IH), 5.80-
5.67 (m, IH), 5.09-5.04 (m, IH), 5.04-5.00 (s, IH), 4.94-4.85 (m, IH), 4.22 (t, 2H), 3.51 (dd, IH), 3.30 (dd, IH), 2.54 (q, 2H)
Step C: (2\k)-2-[[7-but-3-enyl-5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-d]pyrimidin-4-yl] ami) 10]-3 -phenyl-pi -opanoic acid
Using General Procedure IVb and (2Æ)-2-[(7-but-3-enyl-5-iodo-pyrrolo[2,3-i/]pyrimidin-4yl)amino]-3-phenyl-propanoic acid as the appropriate 5-iodo-pyrrolo[2,3-<f]pyrimidme derivative and Préparation 3c as the appropriate boronic acid derivative, (2Æ)-2-[[7-but-3ORIGINAL
- 87enyl-5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenylpropanoic acid was obtained. 'H NMR (500 MHz, DMSO-dô) Ô: 12.86 (br s, IH), 8.24 (s, IH), 7.55-7.43 (m, IH), 7,33-6.95 (m, 6H), 6.89-6.80 (m, 2H), 5.84-5.40 (m. IH), 5.08-
4.93 (m, 3H), 4.84 (br s, IH), 4.37-4.15 (m, 2H), 3,l6(d, IH), 2.85 (dd, IH), 2.56 (q, 2H), 2.22-2.04 (s, 3H).
Step D: Examples 41 and 42
512 mg (2Æ)-2-[[7-but-3-enyl-5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-i/]pyrimidin-4-yl] amino]-3-phenyl-propanoic acid (1 mmol) was dissolved in 4.5 mL dry DMF and 187 mg NBS (i mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was then poured into water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 0.1 % aqueous TFA solution and MeCN as eluents to obtain Example 41 as the earlier eluting diastereoisomer. HRMS calculated for C26H24BrClN4O2: 538.0771, found: 541.0831 (M+H). Example 42 was obtained as the later eluting diastereoisomer. HRMS calculated for C26H24BrClN4O2: 538.0771, found: 541.0835 (M+H)
Example 43: jV-[6-bronio-(5Æa)-5-(3-chloro-2-methylphenyl)-7-(prop-2-en-l-yl)-7//pyrrolo[2,3-//jpyiiniidin-4-vl|-£>-phenylalanine and
Example 44: 7V-|6-bromo-(55„)-5-(3-chloro-2-methylphenyl)-7-(prop-2-en-I-yl)-7/7pyrrolo|2,3-rf]pyriniidin-4-yl]-£)-phenylalanine
Step A: 7-allyl-4-chloro-5-iodo-pyirolo[2,3-d]pyrimidine
176.5 mg 4-chloro-5-iodo-7/7-pyiTolo[2,3-if|pyrimidine (0.6 mmol), 100.7 mg K2CO3 (0.73 mmol), 63 pL allyl bromide (0.73 mmol) and 1 mL dry DMF were stirred at r.t. under N2 atmosphère until no further conversion was observed. Then the mixture was poured into water and extracted with EtOAc. The combined organic layers were washed with water, dried over MgSO4. filtered and the filtrate was concentrated under reduced
ORIGINAL
-88pressure. The crude product was purified via flash chromatography using heptane and EtOAc to obtain 7-allyl-4-chloro-5-iodo-pyrrolo[2,3-</|pyrimidine. MS: (M+H)+ = 320.0
Step B: (2I3)-2-[(7-allyl-5-iodo-pyrrolo[2,3-à]pyrinûdin-4-yl)amino]-3-phenyl-propanoic acid
Using General Procedure III and 7-allyl-4-chloro-5-iodo-pyrrolo[2,3-i/]pyrimidine as the appropriate 4-chloro-pyrrolo[2,3-i/]pyrimidine dérivative and D-phenylalanine as the appropriate amino acid dérivative, (27î)-2-[(7-allyl-5-iodo-pyrrolo[2,3-i/]pyrimidin-4-yl) amino]-3-phenyl-propanoic acid was obtained.
‘H NMR (400 MHz, DMSO-d6) δ: 13.09 (br s, IH), 8.20 (s, IH), 7.43 (s, IH), 7.34-7.18 (m, 5H), 6.52 (bd, IH), 6.05-5.90 (m, IH), 5.15 (dd. IH), 5.07-4.94 (m, 2H), 4.74 (d, 2H),
3.38 (dd, IH), 3.l5(dd, lH)
MS: (M+H)+ - 449.0
Step C: (2R)-2-[[7-allyl-5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-à]pyrimidin-4-yl] amino]-3-phenyl-propanoic acid
Using General Procedure IVb and (2Æ)-2-[(7-aliyl-5-iodo-pyrrolo[2,3-</]pyrimidin-4-yl) amino]-3-phenyl-propanoic acid as the appropriate 5-iodo-pyrrolo[2,3-t/]pyrimÎdine dérivative and Préparation 3c as the appropriate boronic acid dérivative, (2Æ)-2-[[7-allyl-
5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-i/]pyrimidin-4-yl]amino]-3-phenyl-propanoic acid was obtained.
'H NMR (400 MHz, DMSO-d6) δ: 12.89 (br s, IH), 8.23 (s, IH), 7.59-7.42 (br, IH), 7.31-
7.10 (m, 6H), 6.91-6.81 (br, 2H), 6.12-5.98 (m, IH), 5.16 (dd, IH), 5.09-4.96 (m, 2H), 4.90-4.76 (br, 3H), 3.17 (dd, l H), 2.86 (dd. I H), 2.23-2.04 (br s, 3H)
MS: (M+H)+ = 447.0
Step D: Examples 43 and 44
447 mg (2/?)-2-[[7-allyl-5-(3-chloro-2-methyl-phenyl)pyrrolo[2,3-cf]pyrimidin-4-yl] amino]-3-phenyl-propanoic acid (l mmol) was dissolved in 4.5 mL dry DMF and 187 mg NBS (l mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was then poured into water, extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and the fîltrate was
ORIGINAL
-89concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using O.l % aqueous TFA solution and MeCN as eluents to obtain Example 43 as the earlier eluting diastereoisomer. HRMS calculated for C25H22BrClN4O2: 524.0615, found: 525.0675 (M+H). Example 44 was obtained as the later eluting diastereoisomer. HRMS calculated for C25H22BrClN4O2: 524.0615, found; 525.0674 (M+H)
Example 45; (2«)-2-[(7-benzyl-5-{3-chloro-2-mcthyl-4-[2-(4-methylpipcrazin-l-yl) ethoxy]phenyl}-7W-pyrrolo|2,3-rf]pyrimidin-4-yl)oxy]-3-phenylpropanoic acid
Step A: 7-benzyl-4-chloro-5-iodo-pyrrolo[2,3-d]pyrimidine
1.68 g 4-chloro-5-iodo-7/7-pyrrolo[2,3-i/]pyrimidine (6 mmol), 1.24 mL benzyl alcohol (12 mmol), 3.144 g PPh3 (12 mmol) and 60 mL dry THF were cooled to 0 °C under N2 atmosphère, then 5.5 mL DEAD solution (12 mmol, 40 % in toluene) was added and the mixture was stirred at 40 °C until no further conversion was observed. Then the mixture was poured into water and extracted with Et2O. The combined organic layers were washed with water, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc to obtain 7-benzyl-4-chloro-5-iodo-pyrrolo[2,3-J]pyrimidine.
‘H NMR (500 MHz, DMSO-d6) δ: 8.67 (s, IH), 8.12 (s, IH), 7.32 (t, 2H), 7.28 (t. IH),
7.28 (d, 2H), 5.47 (s, 2H)
MS (M+H): 369.9
Step B: Methyl (2^)-2d7-benzy4'5âodo-pyrrolo[2,3-à]pyiiimdin-4-yl)oxy-3-phenylpropanoate eq. 7-benzyl-4-chloro-5-iodo-pyrrolo[2,3-i7]pyrimidine, 3 eq. methyl (2/?)-2-hydroxy-3phenyl-propanoate, 3 eq. Cs2CO3 and dry DMSO (6 mL/mmol) were stirred at 100 C until no further conversion was observed. The mixture was acidified with IM aqueous HCl solution, and extracted with DCM. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give methyl (2/?)-2-(7benzyl-5-iodo-pyrrolo[2,3-i/]pyrimidin-4-yl)oxy-3-phenyl-propanoate. MS (M+H): 514.1
ORIGINAL
-90Step C: Example 45
Using General Procedure IVb and methyl (27?)-2-(7-benzyl-5-iodo-pyrroIo[2,3-i7] pyrimidin-4-yl)oxy-3-phenyl-propanoate as the appropriate 5-iodo-pyrrolo[2,3-</] pyrimidine dérivative and Préparation 3b as the appropriate boronic acid dérivative, methyl (2tf)-2-[7-benzyl-5-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy] phenyl]pyrrolo[2,3-i/]pyrimidin-4-yH oxy-3-phenyl-propanoate was obtained. It was dissolved in dioxane:water l:l (20 mL/mmol) and 10 eq. LiOH^HiO was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diiuted with brine, neutralized with 2M aqueous HCl solution, extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 0.1 % aqueous TFA solution and MeCN as eluents to obtain Example 45. HRMS calculated for C36H38CIN5O4: 639.2612, found: 640.2654 (M+H)
Example 46: /V-[5-(3-chIoro-2-mcthylphenyl)-7,8-(lihydro-6ff-pyrimido[5,4-i>] pyrrolizin-4-yll-i)-phenylalanine
210 mg 1:1 mixture of Examples 43 and 44 (mixture of the two diastereoisomers, 0.4 mmol) was dissolved in 3 mL MeOH and 70 pL cc. H2SO4 (1.2 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was poured into icy water, neutralized with saturated aqueous NaHCO3 solution and extracted with EtOAc. The combined organic phases were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. Then it was dissolved in dry THF (6 mL/mmol), and was cooled to 0 °C. 5 eq. 9-borabicyclo[3.3.1]nonane solution (0.5M in THF) was added and the mixture was stirred at r.t. until no further conversion was observed. Then 20 eq. 2M aqueous NaOH solution and 20 mol% PdC12xdppf was added. The mixture was stirred at 80 °C until no further conversion was observed. Then it was filtered through Celite, washed with EtOAc. The layers of the filtrate were separated, the aqueous layer was acidified to pH 3 with 2M aqueous HCl solution, then extracted with EtOAc. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative
ORIGINAL
-91reversed phase chromatography using 40 mM aqueous NH4OAC solution (pH = 4, adjusted with AcOH) and MeCN as eluents to obtain Example 46 as a mixture of diastereoisomers. HRMS calculated for C25H23CIN4O2: 446.1510, found: 447.159 and 447.1591 (M+H)
Example 47:7V-[(57îa)-5-(3-chloro-2-methylphenyl)-6,7,8,9-tetrahydropyrimido|5,4-i»| indolizin-4-yl]-Z)-phenylalanine and
Example 48: A-[(5S„)-5-(3-chloro-2-niethylphenyl)-6,7,8,9-tetrahydropyriniido|5,4-/)| indolizin-4-yl]-Z>-phenylalanine
1.29 g 1:1 mixture of Examples 41 and 42 (mixture of the two diastereoisomers,
2.3 mmol) was dissolved in 10 mL MeOH and 0.4 mL cc. H2SO4 (6.9 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was poured into icy water, neutralized with saturated aqueous NaHCO3 solution and extracted with EtOAc. The combined organic phases were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. Then it was dissolved in dry THF (6 mL/mmol), and was cooled to 0 °C. 5 eq. 9-borabicyclo[3.3.1]nonane solution (0.5M in THF) was added and the mixture was stirred at r.t. until no further conversion was observed. Then 20 eq. 2M aqueous NaOH solution and 20 mol% PdCl2 xdppf was added. The mixture was stirred at 80 °C until no further conversion was observed. Then it was filtered through Celite, washed with EtOAc. The layers of the filtrate were separated, the aqueous layer was acidified to pH 3 with 2M aqueous HCl solution, then extracted with EtOAc. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 0.1 % aqueous TFA solution and MeCN as eluents. Example 47 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C26H25C1N4O2: 460.1666, found: 461.1747 (M+H). Example 48 was obtained as the later eluting diastereoisomer. HRMS calculated for C26H25C!N4O2: 460.1666, found: 461.1752 (M+H)
Example 49: (2Æ)-2-{|(3Sa)-3-(3-chloro-4-hydroxy-2-methylphenyl)-2-ethyl-lbenzothiophen-4-yl|oxy}-3-phenylpropanoic acid
ORIGINAL
-92and
Example 50: (2/?)-2-{|(37îi,)-3-(3-chloro-4-hydroxy-2-methylphenyl)-2-ethyl-lbenzothiophen-4-yl]oxy}-3-phenylpropanoic acid
Step A: (2R)-2-(2-ethylbemothiophen-4-yl)oxy-3-phenyl-propanoic acid
270 mg (2Æ)-2-hydroxy-3-phenyl-propanoic acid (1.63 mmol), 40 mg Cul (0.21 mmol) and 325 mg CS2CO3 (l mmol) were measured into a 7 mL vial equipped with screw cap and rubber septum. The vial was purged with argon and 5 mL dry DMF and 288 mg 2-ethyl-4-iodo-benzo[6]thiophene (l mmol) were added by syringe. The mixture was stirred at HO °C in dark for 20 hours. Ail further steps were carried out in dark or at red 10 light. 10 mL water was added and the pH was set to 3 with 2M aqueous HCl solution. Then it was extracted with EtOAc. The combined organic layer was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified on a preparative TLC plate (siiica layer, toluene:AcOH 9:l eluent) to obtain (2/?)-2-(2ethylbenzothiophen-4-yl)oxy-3-phenyl-propanoic acid. ’H NMR. (500 MHz, DMSO-dô) δ: 15 12.53 (br s, IH), 7.42-7.36 (m, 3H), 7.30 (t, 2H), 7.25-7.18 (m, IH), 7.13 (t, IH), 7.07 (br,
IH), 6.65 (d, lH), 4.98 (dd, lH), 3.29 (dd, lH), 3.22 (dd, IH), 2.89 (q, 2H), I.30 (t,3H)
Step B: Methyl (2R)-2-(2-ethylbenzothiophen-4-yl)oxy-3-phenyl-propanoate
1.434 g (2/Î)-2-(2-ethylbenzothiophen-4-yl)oxy-3-phenyl-propanoic acid (4.39 mmol) was dissolved in 20 mL MeOH and 20 pL cc. H2SO4 was added. The mixture was stirred at 20 80 °C until no further conversion was observed. The mixture was concentrated under reduced pressure, then diluted with water, neutralized with saturated aqueous NaHCO3 solution and extracted with DCM. The combined organic phases were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure to obtain methyl (27î)-2-(2-ethylbenzothiophen-4-yl)oxy-3-phenyl-propanoate. *H NMR (400 25 MHz. CDCI3) Ô: 7.46-7.33 (m, 5H), 7.33-7.26 (m, IH), 7.16 (bd, IH), 7.13 (t, IH), 6.65 (d, IH), 4.99 (dd, IH), 3.75 (s, 3H), 3.46-3.32 (m, 2H), 3.01-2.9I (m, 2H), 1.42 (t, 3H)
Step C: Methyl (2R.)-2-(2-ethyl-3-iodo-benzothiophen-4-yl)oxy-3-phenyl-propanoate
1.278 g methyl-(2R)-2-(2-ethylbenzothiophen-4-yl)oxy-3-phenyl-propanoate (3.75 mmol), 2.284 g I2 (9 mmol), and 2.5 g Ag2SO4 (8 mmol) were dissolved in 10 mL EtOH and
ORIGINAL
-93stirred at r.t. until no further conversion was observed. The mixture was then filtered, the filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain 860 mg methyl (2/?)-2-(2-ethyl-3,7-diiodobenzothiophen-4-yl)oxy-3-phenyI-propanoate that was dissolved in 20 mL THF, 150 mg 10 % Pd/C was added and the mixture was stirred at r.t. under 4 bar H2 atmosphère until no further conversion was observed. Then it was filtered through Celite, the filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl (2/?)-2-(2-ethyl-3-iodo-benzothiophen-4-yl)oxy-3phenyl-propanoate. ’H NMR (500 MFIz, DMSO-d6) δ: 7.53 (d, IH), 7.49-7.41 (m, 2H), 7.34-7.27 (m, 2H), 7.26-7.18 (m, 2H), 6.77 (d, IH), 5.33 (dd, IH), 3.61 (s, 3H), 3.43 (dd, IH), 3.32 (dd, 1 H), 2.94-2.85 (m, 2H), 1.25 (t, 3H)
Step D: Examples 49 and 50
320 mg methyl (2/?)-2-(2-ethyl-3-iodo-benzothiophen-4-yl)oxy-3-phenyl-propanoate (0.686 mmol) and 368 mg Préparation 3a (1.37 mmol) were dissolved in 4 mL 2-Me-THF under N2 atmosphère, then 1.37 mL TBAOH solution (1.37 mmol, IM in THF) and 49 mg AtaPhos (0.069 mmol) were added and the mixture was stirred at 90 °C in a closed vial until no further conversion was observed. Then it was diluted with 30 mL DCM, washed with 10 mL IM aqueous HCl solution. The organic layer was concentrated under reduced pressure, then dissolved in 5 mL MeOH. 100 mg LiOHxHiO was added, and the mixture was stirred at 50 °C until no further conversion was observed. Then it was diluted with brine, neutralized with IM aqueous HCl solution and extracted with DCM. The organic layer was dried over Na^SOj, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 0.1 % aqueous TFA solution and MeCN as eluents. Example 49 was obtained as the later eluting diastereoisomer. HRMS calculated for C26H23CIO4S: 466.1006, found: 465.0956 (M-H). Example 50 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C26H23CIO4S: 466.1006, found: 465.0971 (M-H)
Example 51 : (2Æ)-2-|((3tSTJ-3-{3-cliloiO-2-rnethyI-4-|2-(4-iiiethylpipei;izin-l-vl)ethoxy | phenyl}-2-ethyI-l-benzothiophen-4-yl)oxy]-3-phenylpropanoic acid
ORIGINAL
-94Step A: Methyl (2R)-2-[3-(3-chloro~4-hydroxy-2-methyl-phenyl)-2-ethyl-benzothiophen-4yl] oxy-3 -phenyl -propanoate
140 mg Example 49 (0.3 mmol) was dissolved in 3 mL MeOH and 50 pL cc. H2SO4 was added. The mixture was stirred at 80 °C until no further conversion was observed. The mixture was concentrated under reduced pressure and the residue was diluted with water, neutralized with saturated aqueous NaHCO3 solution and extracted with DCM. The combined organic phases were washed with brine, dried over MgSÛ4, fîltered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl (2Æ)-2-[3-(3-chloro4-hydroxy-2-methyl-phenyl)-2-ethyl-benzothiophen-4-yl]oxy-3-phenyl-propanoate.
‘H NMR (500 MHz, DMSO-d6) δ: 10.02 (s, IH), 7.49 (d, IH), 7.23-7.12 (m, 4H), 7.02 (d, IH), 6.92 (d, IH), 6.89-6.86 (m, 2H), 6.62 (d, IH), 5.01 (dd, IH), 3.50 (s, 3H), 2.72 (dd, lH), 2.60-2.51 (m, 2H), 2.38 (dd, lH), l .96 (s, 3H), l. 12 (t, 3H)
Step B: Example 51 mg methyl (2Æ)-2-[3-(3-chloro-4-hydroxy-2-methyl-phenyl)-2-ethyl-benzothiophen-4yl]oxy-3-phenyl-propanoate (0.13 mmol), 23 mg l-(2-hydroxyethy!)-4-methylpiperazine (0.156 mmol) and 41 mg PPh3 (0.156 mmol) were dissolved in 2 mL dry THF under N2 atmosphère, then 36 mg DTAD (0.156 mmol) was added. The mixture was stirred at 50 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure, and purified via flash chromatography using heptane, EtOAc and MeOH as eluents. The obtained intermediate was dissolved in 5 mL MeOH, then 100 mg LiOHxH2O was added, and the mixture was stirred at 50 °C until no further conversion was observed. Then it was diluted with brine, neutralized with 1 M aqueous HCl solution and extracted with DCM. The organic layer was dried over Na2SO4, fîltered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 40 mM aqueous NH4OAC solution (pH = 4, adjusted with AcOH) and MeCN as eluents to obtain Example 51. HRMS calculated for C33H37C1N2O4S: 592.2163, found: 593.2238 (M+H)
Example 52: (2Æ)-2-{|(3/ï(,)-3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-lyI)ethoxy]phenyl}-2-(4-fluorophenyl)-l-benzotliiophen-4-yl|oxy}-3-(2-{[2-(2ORIGINAL
-95methoxyphenyl)pyrimidin-4-yl] methoxy}phenyl)propanoic acid and
Example 53: (27î)-2-{[(35(J)-3-{3-chloro-2-methyl-4-|2-(4-methyIpiperazin-l-yl)ethoxy] phenyI}-2-(4-fluorophenyl)-l-benzothiophen-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid
Step A: (3-bromophenyl) N,N-diethylcarbamate
5.0 g 3-bromophenol (28.9 mmol) and 4.31 g diethylcarbamoyl chloride (31.8 mmol) were dissolved in 50 mL pyridine and stirred at l00°C until no further conversion was observed. Then the mixture was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain (3-bromophenyl) Λζ/V-diethylcarbamate. MS (El, 70 eV) m/z (% relative intensity, [ion]): 56 (9), 72 (42), 100 (100), 174 (4), 176 (4), 271 (4, [M+]), 273 (4, [M+])
Step B: (3-bromo-2-iodo-phenyl) NN-diethylcarbamate
2.72 g (3-bromophenyl) AÇ/V-diethylcarbamate (10 mmol) was dissolved in 50 mL dry THF under N2 atmosphère and cooled to -78 °C. 6 mL LDA solution (12 mmol, 2M in THF, heptane, ethyl benzene) was added and the mixture was stirred al -78 °C for 30 minutes. Then 3.18 g I2 (12.5 mmol) was added and the mixture was stirred at -78 °C for 30 minutes then it was allowed to warm up to r.t. Then the mixture was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain (3-bromo-2-iodo-phenyl) ZV,/V-diethylcarbamate. 'H NMR (400 MHz, DMSO-d6) 5:
7.60 (dd, IH), 7.35 (t, IH), 7.17 (dd, IH), 3.47 (q, 2H), 3.31 (q, 2H), 1.27 (t. 3H), 1.14 (t, 3H)
Step C: [3-bromo-2-[2-(4-fluorophenyl)ethynyl]phenyl] N,N-dîelhylcarbamate
2.60 g (3-bromo-2-iodo-phenyl) MA-diethylcarbamate (6.53 mmol), 863 mg 1 -ethynyl-4fluorobenzene (7.19 mmol), 229 mg Pd(PPh3)2Cl2 (0.33 mmol), 130 mg copper(l) iodide (0.65 mmol) and 1.43 g diethylamine (19.6 mmol) were dissolved in 25 mL dry DMF and stirred at 50 °C until no further conversion was observed. The mixture was diluted with water and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The
ORIGINAL
-96crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain [3-bromo-2-[2-(4-fluorophenyl)ethynyl]phenyl] AÇ/V-diethylcarbamate. MS (El, 70 eV) m/z (% relative intensity, [ion]): 56 (2), 72 (35), 100 (100), 261 (2), 263 (2), 389 (2, [M+]), 391 (2, [M+])
Step D: [2-[2-(4-fluorophenyl)ethynyl]-3-methylsulfanyl-phenyl] N,N-diethylcarbamate
2.5 g [3-bromo-2-[2-(4-fluorophenyl)ethynyl]phenyl] jV,/V-diethylcarbamate (6.56 mmol) was dissolved in 65 mL dry THF and cooled to -78 °C, then 4.3 mL nBuLi solution (6.88 mmol, 1.6M in hexanes) was added. The mixture was stirred at -78 °C for 30 minutes. Then 742 mg S2Me2 (7.87 mmol) was added and the mixture was stirred at -78 °C for 30 minutes, then it was allowed to warm up to r.t. The mixture was then concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain [2-[2-(4-fiuorophenyl)ethynyl]-3-methylsulfanyl-phenyl] A’.jV-dicthylcarbamate. MS (El, 70 eV) m/z (% relative intensity, [ion]): 56 (2), 72 (46), 100 (100), 342 (40), 357 (1, [M+])
Step E: [2-(4-fluorophenyl)-3-iodo-benzothiophen-4-ylJ N.N-diethylcarbamate
1100 mg 2-[2-(4-fluorophenyl)ethynyl]-3-methylsulfanyl-phenyl] A’jV-dicthyicarbamate (3.08 mmol) and 937 mg 12 (3.7 mmol) were dissolved in 20 mL DCM and stirred at r.t. until no further conversion was observed. The mixture was then diluted with 10 % aqueous Na2S2O3 solution and extracted with DCM. The combined organic layers were washed with brine, to give [2-(4-fluorophenyl)-3-iodo-benzothiophen-4-yl] /V./V-diethylcarbamate.
’H NMR (400 MHz, CDC13) δ: 7.74 (dd, IH), 7.56 (m. 2H), 7.40 (t, IH), 7.18 (m, 2H),
7.12 (dd, 1 H), 3.60 (q,2H), 3.46 (q, 2H), 1.36 (t, 3H), 1.26 (t.3H)
MS (El, 70 eV) m/z (% relative intensity, [ion]): 72 (42), 100 (100), 170 (16), 342 (37), 369(5), 469 (1, [M+])
Step F: [3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluoro phenyl)benzothiophen-4-yl] N.N-diethyÎcarbainate eq. [2-(4-fluorophenyl)-3-iodo-benzothiophen-4-yl] ?V,N-diethylcarbamate, 2 eq. Préparation 3b, 2 eq. Cs2CO3, 0.1 eq. Ataphos and THF:water 3:1 (10 mL/mmol benzothiophene dérivative) were stirred under N2 atmosphère at 70 °C until no further
ORIGINAL
-97conversion was observed. The mixture was diluted with water and extracted with DCM. The organic phase was dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give [3-[3-chloro-2-methyl-4-[2-(4-methyipiperazin-lyl)ethoxy]phenyl]-2-(4-fluorophenyl)benzothiophen-4-yl] A,A/-diethylcarbamate. MS: (M+H)+= 610.2
Slep G: 3-[3-chloro-2-methylM-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluoro phenyl)benzothiophen-4-ol
1.8 g [3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluoro phenyl)benzothiophen-4-yl] /V,A-diethylcarbamate (3 mmol) was dissolved in 80 mL EtOH and 1.2 g NaOH (30 mmol) was added. The mixture was stirred at 80 °C until no further conversion was observed. The mixture was concentrated under reduced pressure and purified via flash chromatography using DCM and MeOH as eluents to obtain 3-[3-chloro2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluorophenyl)benzothiophen4-ol. MS:(M+H)+ = 511.2
Step H: Examples 52 and 53
470 mg 3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluoro phenyl)benzothiophen-4-ol (0.92 mmol), 1.12 g Préparation 2d (2.76 mmol) and 726 mg PPh3 (2.76 mmol) were dissolved in 10 mL dry toluene, then 635 mg DTAD (2.76 mmol) was added. The mixture was stirred at 50 °C until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents. The formed intermediate was dissolved in 10 mL dioxane:water 1:1,400 mg LiOHxH2O was added, and the mixture was stirred at r.t. until no further conversion was observed. It was neutralized with 2M aqueous HCl solution and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents. Example 52 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C49H46C1FN4O6S: 872.2811, found: 437.1457
ORIGINAL
-98(M+2H). Example 53 was obtained as the later eluting diastereoisomer. HRMS calculated for C49H46ClFN4O6S: 872.2811, found: 437.1491 (M+2H)
Example 54: 2-benzyl-3-[3-(3-chloro-4-hydroxy-2-methylphenyl)-2-ethyl-lbenzothiophen-4-yl]propanoic acid
Step A: Methyl (Z)-2-benzyl-3-(2-ethylbenzothiophen-4-y!)prop-2-enoate
576 mg 2-ethyl-4-iodo-benzo[ô]thiophene (2 mmol), 717 mg methyl 2-benzylacrylate (4 mmol), 556 pL TE A (4 mmol) and 24 mg PdCl2 (0.1 mmol) were dissolved in 10 mL DMF and stirred at 130 °C in a MW reactor until no further conversion was observed. The mixture was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl (Z)-2-benzyl-3-(2ethylbenzothiophen-4-yl)prop-2-enoate. '14 NMR (400 MHz, CDCI3) ratio of diastereoisomers 1.00 ! Ο.ΊΊ — major / minor, δ: 8.06-8.28 (s, IH), 7.68-7.76 (d, 111), 7.44-
6.98 (m, 8H), 4.25-3.93 (s, 2H), 3.78-3.82 (s, 3H), 2.97-2.99 (q, 214), 1.41-1.43 (t, 3H)
Step B: Methyl 2-benzyl-3-(2-eihylbenzothiophen-4-yl)propanoate
432 mg methyl (Z)-2-benzyl-3-(2-ethylbenzothiophen-4-yl)prop-2-enoate (1.28 mmol), 137mg 10% Pd/C, 5 mL AcOH and 20 mL MeOH were stirred under 4 bar FL atmosphère at r.t. until no further conversion was observed. The mixture was filtered through Celite, the filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl 2-benzyl-3-(2ethylbenzothiophen-4-yl)propanoate. *H NMR (400 MHz, CDCI3) Ô: 7.61 (d, IH), 7.j87.05 (m, 7H), 6.80 (s, 114), 3.50 (s, 3H), 3.28-3.18 (m, IH), 3.11-3.00 (m, 314), 2.90 (q, 214), 2.86-2.77 (m, IH), 1.35 (t, 3H)
Step C: Methyl 2-benzyl-3-(2-ethyl-3-iodo-benzothiophen-4-yl)propanoate
346 mg methyl 2-benzyl-3-(2-ethyibenzothiophen-4-yl)propanoate (1.02 mmol), 305 mg I2 (1.2 mmol) and 468 mg Ag2SO4 (1-5 mmol) were dissolved in 5 mL EtOH and stirred at r.t. until no further conversion was observed. The mixture was filtered, the filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain methyl 2-benzyl-3-(2-ethyl-3-iodo-benzothiophen-4OR1GINAL
-99yl)propanoate. lH NMR (400 MHz, CDCI3) δ: 7.67 (dd, IH), 7.28-7.06 (ni, 7H), 4.29-4.17 (m. IH), 3.80-3.71 (m, IH), 3.32 (s, 3H), 3.28-3.21 (m. IH), 3.08-3.00 (m, 2H), 2.97 (q, 2H), l.35(t,3H)
Step D: Example 54 l eq. methyl 2-benzyl-3-(2-ethyl-3-iodo-benzothiophen-4-yl)propanoate, 2 eq. Préparation 3a, 2 eq. TBAOH solution (IM in water), O.l eq. Ataphos and 2-Me-THF (5 mL/mmol benzothiophene dérivative) were stirred under N2 atmosphère at 100 °C until no further conversion was observed. The mixture was diluted with water and extracted with DCM. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The formed intermediate was dissolved in MeOH (5 mL/mmol benzothiophene dérivative), 10 eq. LiOHxH2O was added, and the mixture was stirred at r.t. until no further conversion was observed. It was neutralized with 2M aqueous HCl solution and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified using préparative reversed phase chromatography using 0.1 % aqueous TFA solution and MeCN as eluents to give Example 54. HRMS calculated for C27H25C1O3S: 464.1213, found: 463.1158 (M-H)
Example 55: (2/Î)-2-{((lÆfl)-l-{3-chloro-2-methyl-4-12-(4-methylpiperazin-lyl)ethoxy]phenyl)-2-(4-fluorophenyl)-l//-indol-7-yl]oxy}-3-(2methoxyphenyl)propanoic acid and
Example 56: (2Æ)-2-{[(15(/)-l-{3-chloro-2-niethyl-4-I2-(4-methylpiperazin-l-yl)ethoxy| phenyl)-2-(4-fluorophenyl)-l//-indol-7-yl]oxy}-3-(2-methoxyphenyl)propanoic acid
600 mg Préparation 7b (0.86 mmol) was dissolved in 20 mL dioxane:water 1:1 and 600 mg LiOHxFbO was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with water, acidified with IM aqueous HCl solution and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure and purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents.
ORIGINAL
- 100Example 55 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C38H39CIFN3O5: 671.2562, found: 672.2618 (M+H). Example 56 was obtained as the later eluting diastereoisomer. HRMS calculated for C38H39CÎFN3O5: 671.2562, found: 672.2652 (M+H)
Example 57: (2«)-2-{[3-chloro-(lSa)-l-{3-chloro-2-methyI-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl}-2-(4-fluorophenyl)-l H-indol-7-yl|oxy}-3-(2methoxyphenyl)propanoic acid
240 mg Préparation 7b (0.34 mmol) was dissolved in 3 mL DCM and 46 mg NCS (0.34 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with water and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. Then it was dissolved in 5 mL dioxane:water 1:1 and 140 mg LiOHxH2O was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with water, acidified with IM aqueous HCl solution and extracted with DCM. The combined organic phases were dried over NaiSCfr, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents. Example 57 was obtained as the later eluting diastereoisomer. HRMS calculated for C3sH38C12FN3O5: 705.2173, found: 706.2227 (M+H)
Example 58: (2Æ)-2-{[(l/ffl)-l-{3-chloro-2-mcthyl-4-|2-(4-inethylpiperazin-lyI)ethoxy]phenyl}-2-(furan-2-yl)-lH-indol-7-yl|oxy}-3-(2-methoxyphenyl)propanoic acid and
Example 59: (27f)-2-{[(lSfl)-l-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy] phenyl}-2-(furan-2-yl)-l//-indol-7-yl]oxy}-3-(2-methoxyphenyl)propanoic acid and
Example 60: (27î)-2-{[(15u)-l-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yl)ethoxy] phenyl}-2-(5-fluorofuran-2-yl)-lF7-indol-7-yl]oxy}-3-(2-methoxyphenyl)propanoic acid
ORIGINAL
- ΙΟΙStep A: 4-[7-benzyloxy-2-(5-fluoro-2-furyl)indol-1 -yl]-2-chloro-3-methyl-phenol
1360 mg Préparation 7a (2 mmol), 848 mg 2-(5-fluoro-2-furyl)-4,4,5,5-tetramethyl-1,3,2dioxaborolane (4 mmol), 2123 mg K3PO4 (10 mmol), 45 mg Pd(OAc)2 (0.2 mmol) and 164 mg SPhos (0.4 mmol) were dissolved in 30 mL dry toluene and stirred at 75 °C until no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents. Then 2 mL TBAF solution (2 mmol, IM in THF) and 25 mL THF were added and the mixture was stirred at r.t. until no further conversion was observed. Then the mixture was concentrated under reduced pressure, and the residue was purified via flash chromatography using heptane and EtOAc as eluents to give 4-[7-benzyloxy-2-(5-fiuoro-2furyl)indol-l-yl]-2-chloro-3-methyl-phenol. MS: (M+H)+ = 448.0
Step B: 7-benzyloxy-1 -[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]-2(5-fluoro-2-fitryl)indole
650 mg 4-[7-benzyloxy-2-(5-fluoro-2-furyI)indol-l -yl]-2-chloro-3-methyl-phenol (1.01 mmol), 288 mg l-(2-hydroxyethyl)-4-methylpiperazine (2 mmol) and 786 mg PPI13 (3 mmol) were dissolved in 20 mL dry toluene. Then 690 mg DTAD (3 mmol) was added and the mixture was stirred at 45 °C until no further conversion was observed. Then it was concentrated under reduced pressure, and was purified via flash chromatography using DCM and MeOH as eluents to give 7-benzyloxy-l-[3-chloro-2-methyl-4-[2-(4-methyl piperazin-l-yl)ethoxy]phenyl]-2-(5-fluoro-2-furyl)indole. MS: (M+H)+ = 574.2
Step C: The mixture of 1 -[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l -yl)ethoxy]phenyl]2-(2-furyl)indol-7-ol and /-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy] phenyl]-2-(5-fluoro-2-furyT)indol-7-ol
1300 mg 7-benzyloxy-1 -[3-chloiO-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]2-(5-fluoro-2-furyl)indole (2.26 mmol) was dissolved in 100 mL MeOH and 100 mg 10 % Pd/C was added. The mixture was stirred under 1 bar H2 atmosphère at r.t. overnight. The mixture was filtered through Celite and the filtrate was concentrated under reduced pressure to give a 7:3 mixture of l-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl]-2-(2-furyl)indoI-7-ol (MS: (M+H)+ = 466.2) and l-[3-chloro-2-methylORIGINAL
- 1024-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(5-fluoro-2-furyl)indol-7-ol (MS: (M+H)T = 484.2).
Step D: Examples 58, 59 and 60
465 mg of the 7:3 mixture of l-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl]-2-(2-furyl)indol-7-ol and l -[3-chloro-2-methyl-4-[2-(4-methylpiperazinl-yl)ethoxy]phenyl]-2-(5-fluoro-2-furyl)indol-7-ol (l mmol), 449 mg ethyl (25)-2hydroxy-3-phenyl-propanoate (2 mmol) and 786 mg PPhj (3 mmol) were dissolved in 10 mL dry toluene. Then 691 mg DTAD (3 mmol) was added and the mixture was stirred at 45 °C until no further conversion was observed. Then it was concentrated under reduced pressure, and the residue was purified via flash chromatography using DCM and MeOH as eluents. Then it was dissolved in 5 mL dioxanerwater l:l and 140 mg LiOH^HiO was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with water, acidified with l M aqueous HCl solution and extracted with DCM. The combined organic phases were dried over Na2SÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents. Example 58 was obtained as the earlier eluting dîastereoisomer. HRMS calculated for C36H38ClN3O6: 643.2449, found: 644.2512 (M+H). Example 59 was obtained as the later eluting dîastereoisomer. HRMS calculated for C36H38CIN3O6: 643.2449, found: 644.2521 (M+H). Example 60 was obtained as the later eluting dîastereoisomer. HRMS calculated for C36H37CIFN3O6: 661.2355, found: 662.2411 (M+H)
Example 61: (2Æ)-2-{|(3/fa)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl}-2-(4-fliiorophenyl)-l-benzofiiran-4-yl]oxy}-3-(2methoxyphenyl)propanoic acid and
Example 62: (2Æ)-2-{[(35'„)-3-{3-chloro-2-methyI-4-[2-(4-methylpipcrazin-l-yl)ethoxy| phenyI}-2-(4-fluorophenyl)-l-benzofuran-4-yl|oxy}-3-(2-niethoxyphenyl)propanoic acid
Step A: Ethyl (2S>)-3-(2-methoxyphenyl)-2-(p-tolylsulfonyloxy)propanoate
ORIGINAL
- 1033000 mg Préparation 2f (13.38 mmol) was dissolved in 10 mL pyridine and 2933 mg TsCl (15.38 mmol) was added at 0 °C. The mixture was stirred at r.t. until no further conversion was observed. Then the mixture was diluted with water and extracted with EtOAc. The combined organic phases were washed with IM aqueous citric acid solution, dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give ethyl (2S)-3-(2-methoxyphenyl)-2-(p-tolylsulfonyloxy)propanoate. MS (El, 70 eV) m/z (% relative intensity, [ion]): 65 (7), 77 (14), 91 (49), 123 (33), 133 (33), 165 (100), 207 (65), 307(13),512(7, [M+])
Step B: Ethyl (2B]-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-(2-methoxy phenyl)propanoate eq. Préparation le, 1.5 eq. ethyl (2S)-3-(2-rnethoxyphenyl)-2-(/?-tolylsulfonyloxy) propanoate, 2 eq. K2CO3 and DMSO (10 mL/mmol benzofurane dérivative) were stirred at 60 °C under N2 atmosphère until no further conversion was observed. Then it was diluted with brine, neutralized with IM aqueous HCl solution and extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to give ethyl (2Æ)-2-[3-bromo-2-(4fluoropheny[)benzofuran-4-yl]oxy-3-(2-methoxyphenyl)propanoate. MS (El, 70 eV) m/z (% relative intensity, [ion]): 91 (56), 133 (41), 165 (100), 207 (93), 281 (26), 305 (9), 512 (3, [M+]), 514 (3, [M+])
Step C: Examples 61 and 62
Using General Procedure VI and ethyl (2/î)-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4yl]oxy-3-(2-methoxyphenyl)propanoate as the appropriate 3-bromo-benzofuran dérivative and Préparation 3b as the appropriate boronic acid dérivative, Examplc 61 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C38H38C1FN2O6: 672.2402, found: 673.2465 (M+H). Example 62 was obtained as the later eluting diastereoisomer, HRMS calculated for C38H38C1FN2O6: 672.2402, found: 673.2486 (M+H)
Exaniple 63: (27i)-2-{[(3/fa)-3-{3-chloro-2-niethyl-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl}-2-(4-fluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{[2-(2OR1GINAL
- 104methoxyphenyl)pyrimidin-4-y[]methoxy}phenyl)propanoic acid and
Example 64: (2ft)-2-{[(35a)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy] phenyl)-2-(4-fluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{[2-(2-niethoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid
Step A: Ethyl (2R)-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-[2-(2-methoxy phenyl)pyrimidin-4-yl]oxyphenyl]propanoate
Using General Procedure V and Préparation le as the appropriate benzofuran-4-ol dérivative and Préparation 2d as the appropriate lactic ester dérivative, ethyl (2Æ)-2-[310 bromo-2-(4-fIuorophenyl)benzofuran-4-yl]oxy-3-[2-[2-(2-methoxyphenyl)pyrimidin-4yl]oxyphenyl] propanoate was obtained. MS: (M+H)+ = 699.2
Step B: Examples 63 and 64
Using General Procedure VI and ethyl (2/?)-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4yl]oxy-3-[2-[2-(2-methoxyphenyl)pyrimidin-4-yl]oxyphenyl]propanoate as the appropriate 15 3-bromo-benzofuran dérivative and Préparation 3b as the appropriate boronic acid dérivative, Example 63 was obtained as the earlier eluting diastereoisomer. HRMS calculated for C49H46CIFN4O7: 856.3039, found: 429.1582 (M+2H). Examplc 64 was obtained as the later eluting diastereoisomer. HRMS calculated for C49H46CIFN4O7: 856.3039, found: 429.1604 (M+2H)
Example 65: (2Æ)“2-{[(3Sa)-3-{3-chIoro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy] phcnyl}-2-(4-fiuorophenyl)-l-benzofuran-4-yl]oxy}-3-[2-(2,2,2-trifluoroethoxy) phenyijpropanoic acid
Step A: Ethyl (2R)-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-(2.2,2trifluoroethoxy)phenyl]propanoate
Using General Procedure V and Préparation le as the appropriate benzofuran-4-ol dérivative and Préparation 2h as the appropriate lactic ester dérivative, ethyl (27?)-2-[3bromo-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-(2,2,2-trifluoroethoxy)phenyl] propanoate was obtained. MS: (M+Na)+ = 604.4
ORIGINAL
- 105Slep B: Example 65
Using General Procedure VI and ethyl (27?)-2-[3-bromo-2-(4-fluorophenyl)benzofuran-4yl]oxy-3-[2-(2,2,2-triiluoroethoxy)phenyl]propanoate as the appropriate 3-bromobenzofuran derivative and Préparation 3b as the appropriate boronic acid derivative, 5 Example 65 was obtained as the later eluting diastereoisomer. HRMS calculated for C39H37CiF4N2O6: 740.2276, found: 74I.2372 (M+H)
Example 66: (2Æ)-2-{|(3S0)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy| phenyI}-6-fluoro-2-(4-fluorophenyl)-l-benzofuran-4-yl|oxy)-3-|2-(2,2,2-trifluoro ethoxy)phenyl| propanoic acid
Slep A: Ethyl (2R)-2-[3-bromo-6-fluoro-2-(4-fluorophenyl)benzofiiran-4-y}]oxy-3-[2(2,2,2-trifluoroethoxy)phenyl]propanoate
Using General Procedure V and Préparation Id as the appropriate benzofuran-4-ol derivative and Préparation 2h as the appropriate lactic ester derivative, ethyl (27?)-2-[3bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-(2,2,2-trifluoroethoxy) phenyl]propanoate was obtained. ’H NMR (400 MHz. DMSO-d6): 8.07 (m, 2H), 7.43 (m, 3H), 7.27 (m, 2H), 7.11 (m, IH), 6.98 (m, IH), 6.55 (dd, IH), 5.23 (m, IH), 4.82 (q, 2H),
4.12 (q, 2H), 3.37 (m, IH), 3.25 (m, IH), l. 10 (t, 3H)
Step B: Example 66
Using General Procedure VI and ethyl (2/?)-2-[3-biOmo-6-fluoro-2-(4-lluorophenyl) 20 benzofuran-4-yl]oxy-3-[2-(2,2,2-trifluoroethoxy)phenyl]propanoate as the appropriate
3-bromo-benzofuran derivative and Préparation 3b as the appropriate boronic acid derivative, Example 66 was obtained as the later eluting diastereoisomer. HRMS calculated for C39H36CIF5N2O6: 758.2182, found: 759.2244 (M+H)
Example 67: (2/ï)-2-{[(35'„)-3-{3-chloro-2-mcthyI-4-[2-(4-methylpiperazin-l-yl)ethoxy] 25 phenyl}-6-fluoro-2-(4-iluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{|2-(2-methoxy phenyl)pyrimidin-4-yI]methoxy)phenyl)propanoic acid
ORIGINAL
- 106Sien A: Ethyl (2^)-2-[3-bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-[[2(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate
Using General Procedure V and Préparation Id as the appropriate benzofuran-4-ol dérivative and Préparation 2d as the appropriate lactic ester dérivative, ethyl (2Λ)-2-[3bromo-6-fluoro-2-(4-fluorophenyl)benzofuran-4-yl]oxy-3-[2-[[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy]phenyl]propanoate was obtained. ’H NMR (400 MHz, DMSOd6): 8.86 (d, IH), 8.05 (m, 2H), 7.61 (d, IH), 7.52 (dd. IH), 7.48-7.38 (m, 4H), 7.25 (m, IH), 7.21 (m. IH), 7.12 (m, 2H), 7.03 (td, IH), 6.94 (td, IH), 6.67 (dd, IH), 5.40 (m, IH), 5.26 (s, 2H), 4.15 (q, 2H), 3.75 (s , 3H), 3.56 (m, IH), 3.30 (m, 1 H), 1.12 (t, 3H)
Step B: Example 67
Using General Procedure VI and ethyl (2Â)-2-[3-bromo-6-fluoro-2-(4-fluorophenyl) benzofuran-4-yl]oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate as the appropriate 3-bromo-benzofuran dérivative and Préparation 3b as the appropriate boronic acid dérivative, Example 67 was obtained as the later eluting diastereoisomer. HRMS calculated for C49H45CIF2N4O7: 874.2945, found: 438.1543 (M+2H)
Example 68: (27î)-2-{|(3/?0)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)ethoxy]plienyl)-2-(4-fiuorophenyl)-l-methyl-l//-indol-4-yl]oxy}-3-(2-{|2-(2mcthoxyphenyl)pyrimidin-4-yl]methoxy}phenyl)propanoic acid and
Example 69: (2ff)-2-{|(35(f)-3-{3-chloro-2-methyi-4-|2-(4-methylpiperazin-l-yI)etlioxy| phenyl}-2-(4-fluoroplienyl)-l-metliyl-l/f-indol-4“yl]oxy}-3-(2-{|2-(2-methoxyphenyl) pyrimidin-4-yl|methoxy}phenyl)propanoic acid
Step A : l-(benzenesidfonyl)-4-benzyloxy-indole
7.0 g 4-benzyloxy-l/7-indole (31.35 mmol) was dissolved in 60 mL dry DMF and 1.317 g NaH (32.92 mmol, 60 % on minerai oil) was added at 0 °C. The mixture was stirred for 1 hour, then 6.09 g benzenesulfonyl chloride (34.48 mmol) was added dropwise and the mixture was stirred at 0 °C until no further conversion was observed. Then it was diluted with water and extracted with DCM. The combined organic phases were dried over
ORIGINAL
-107Na2SO4, filtered and the filtrate was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain l-(benzenesulfonyl)-
4-benzyloxy-indole.
'H NMR (400 MHz, DMSO-d6) δ: 7.97 (d, 2H), 7.72 (d. IH), 7.69 (t, IH), 7.59 (t, 2H), 7.54 (d, IH), 7.47 (d, 2H), 7.39 (t, 2H), 7.33 (d, IH), 7.27 (t,lH), 6.89 (d, IH), 6.85 (d, 1 H), 5.20 (s, 2H)
MS (El, 70 eV) m/z (% relative intensity, [ion]): 77 (32), 91 (100), 141 (18), 222 (6), 272 (11),363 (10, [M+])
Step B: 1 -(benzenesulfonyl)-4-benzyloxy-2-iodo-indole
5.08 g l-(benzenesulfonyl)-4-benzyloxy-indole (13.98 mmol) was dissolved in 140 mL dry THF. 8.54 mL LDA solution (15.38 mmol, 1.8M in THF-heptane-ethylbenzene) was added at -78 °C and the mixture was stirred for 1 hour. Then 4.26 g iodine (16.8 mmol) was added and the mixture was stirred for 1 hour at -78 °C. The mixture was quenched with saturated aqueous NH4C1 solution, extracted with EtOAc. The combined organic phases were washed with aqueous NajSjOj solution and water, then dried over NaiSO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain l-(benzenesulfonyl)-4-benzyloxy-2-iodo-indole. 'H NMR (400 MHz, DMSO-dô) δ: 7.86 (dd, 2H), 7.75 (d, 1 H), 7.70 (d, IH), 7.61 (t, 2H), 7.47 (dd, 2H), 7.39 (t, 2H), 7.33 (d, IH),
7.23 (t, IH), 7.18 (s, 1 H), 6.90 (d, IH), 5.20 (s, 2H)
Step C: l-(benzenesidfonyl)-4-benzyloxy-2-(4-fltiorophenyl)indole
5.8 g l-(benzenesulfonyl)-4-benzyloxy-2-iodo-indole (11.86 mmol) and 3.16 g 4-(4,4,5,5tetramethyl-l,3,2-dioxaboroIan-2-yl)fluorobenzene (14.22 mmol) were dissolved in 75 mL THF, then 7.73 g CS2CO3 (23.72 mmol), 420 mg Ataphos (0.59 mmol) and 25 mL water were added and the mixture was stirred at 70 °C under N2 atmosphère until no further conversion was observed. The mixture was then concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain l-(benzenesulfonyl)-4-benzyloxy-2-(4-fluorophenyl)indole. 'El NMR (400 MHz, DMSO-dû) δ: 7.79 (d, IH), 7.67 (m, IH), 7.60-7.48 (m, 6H), 7.43-7.25 (m, 8H), 7.00 (d, IH), 5.57 (s, 1 H), 5.22 (s, 2H)
ORIGINAL
- 108Step D: 1-(benzenesulfanyl)-4-benzyloxy-2-(4-fluorophenyl)-3-iodo-indole
4.92 g l-(benzenesulfonyI)-4-benzyloxy-2-(4-fluorophenyl)indole (I0.75 mmol), 3.69 g Ag2SO4 (l 1.83 mmol) and 3.0 g iodine (l 1.83 mmol) were stirred in 100 mL EtOH at r.t. until no further conversion was observed. Then the mixture was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents to obtain l-(benzenesulfonyl)-4-benzyloxy-2-(4-fluorophenyl)-3-iodo-indole. MS: (M+H)+ = 584.2
Step E: 1 -(benzenesidfonyl)-4d)enzyloxy-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 yl)ethoxy]phenyl]-2-(4-fluorophenyl)indole
5.5 g l-(benzenesulfonyl)-4-benzyloxy-2-(4-fluorophenyl)-3-iodo-indole (9.42 mmol).
4.46 g Préparation 3b (11,31 mmol), 6.14 g Cs2CO3 (18.84 mmol) and 354 mg Ataphos (0.5 mmol) were dissolved in 100 mL THFrwater 3:1 and stirred at 70 °C under N2 until no further conversion was observed. The mixture was concentrated under reduced pressure and purified via flash chromatography using heptane, EtOAc and MeOH as eluents to 15 obtain 1 -(benzenesulfonyl)-4-benzyloxy-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 yl)ethoxy] phenyl]-2-(4-fluorophenyl)indole.
’H NMR (400 MHz, DMSO-d6) δ: 7.85 (d, IH), 7.67 (t, IH), 7.61-6.90 (m, 2H), 7.53-7.47 (m, 4H), 7.4 (t. IH), 7.20-7.07 (m, 5H), 6.96 (d, IH), 6.77 (d, IH), 6.73 (d, IH), 6.66 (d, 2H), 4.96 (d, IH), 4.86 (d, IH), 4.09 (m, IH), 4.00 (m, IH), 3.34 (br s, 4H), 2.75 (t, 2H), 20 2.58 (br s, 4H), 2.30 (s, 3H), 1.81 (s, 3H)
MS: (M : H)' - 724.2
Step F: 4-benzyloxy-3-[3-chloro-2-meihyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2(4-fluorophenyl)-lW-indole
6.5 g 1 -(benzenesulfonyl)-4-benzyloxy-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 25 yl)ethoxy]phenyl]-2-(4-fluorophenyl)indole (8.97 mmol) was dissolved in 100 mL THF and 100 mL MeOH, then 28.3 g Ba(OH)2 x8 H2O (89.7 mmol) was added and the mixture was stirred at 70 °C until no further conversion was observed. The mixture was then filtered, the filtrate was concentrated under reduced pressure and purified via flash chromatography using DCM and MeOH as eluents to obtain 4-benzyloxy-3-[3-chloro-2-
ORIGINAL
- 109methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fluorophenyl)-lH-indole. MS: (M+H)+ = 584.2
Step G: 4-benzyloxy-3-[3-chloro-2-methyl-4-[2-(4-melhylpiperazin-l-yl)ethoxy]phenyl]-2(4-fh torophenyl)-!-methyl - indol e l .626 g 4-benzyloxy-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]2-(4-fluorophenyl)-l/f-indole (2.78 mmol) was dissolved in 25 mL dry DMF and cooled to 0 °C. Then 123 mg NaH (3.06 mmol, 60 % on minerai oil) was added and the mixture was stirred for l hour. Then 395 mg methyl iodide (2.78 mmol) was added and the mixture was stirred for l hour. The mixture was then poured into water and extracted with DCM. The combined organic phases were washed with brine, dried over MgSÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using DCM and MeOH as eluents to obtain 4-benzyloxy-3-[3-chloro-2methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]-2-(4-fluorophenyl )-1 -methyl-indole.
’H NMR (400 MHz, DMSO-d6) δ: 7.31 (dd, 2H), 7.24-7.10 (m, 7H), 6.97 (d, IH), 6.83-
6.76 (m, 3H), 6.68 (dd. IH), 5.01 (d, IH), 4.93 (d, IH), 4.14 (m, IH), 4.06 (m, IH), 3.63 (s, 3H), 3.10-2.60 (br s, 8H), 2.84 (br s, 2H), 2.58 (s, 3H), 2.04 (s, 3H)
Step H: 3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl]-2-(4-fhioro phenyl)-}-methyl-indol-4-ol
1.6 g 4-benzyloxy-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)eihoxy]phenyl]-2(4-fluorophenyl)-l-methyl-indole (2.68 mmol) was dissolved in 10 mL DCM and l eq. HBr (33 % solution in AcOH) was added. The mixture was stirred at r.t. until no further conversion was observed. The mixture was then diluted with 10% aqueous K2CO3 solution and extracted with DCM. The combined organic phases were washed with brine, dried over MgSÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using DCM and MeOH as eluents, then via préparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain 3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-lyl)ethoxy]phenyl]-2-(4-fluorophenyl)-l-methyl-indol-4-ol.
ORIGINAL
- IIO’H NMR (400 MHz, DMSO-d6) δ: 9.02 (s, IH), 7.29-7.15 (m, 4H), 7.06-6.92 (m, 2H),
6.86 (d, IH), 6.78 (d, IH), 6.38 (dd, IH), 4.07 (m, 2H), 3.58 (s, 3H), 2.70 (t, 2H), 2.58-2.40 (br s, 4H), 2.40-2.19 (br s, 4H), 2.19 (s, 3H), 2.09 (s, 3H)
MS: (M+H)+ = 508.2 5 Step I: Ethyl (2S)-3-[2-[[2-(2-methoxyphenyî)pyrimidin-4-yl]methoxy]phenyl]-2-(p-tolyl sulfonyloxy)propanoate
3.668 g Préparation 2d (8.97 mmol) was dissolved in 12 mL pyridine and L97 g TsCI (10.31 mmol) was added at 0 °C. The mixture was stirred at r.t. until no further conversion was observed. Then the mixture was diluted with water and extracted with EtOAc. The 10 combined organic phases were washed with IM aqueous citric acid solution, dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure to give ethyl (2S)-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]-2-(/7-tolylsulfonyloxy) propanoate.
'H NMR (400 MHz, DMSO-d6) δ: 8.93 (d, IH), 7.58 (dd. IH), 7.52-7.43 (m, 2H), 7.4315 7.34 (m, 2H), 7.26-7.15 (m, 4H), 7.13-7.04 (m, 2H), 6.93-6.83 (m, 2H), 5.12 (d, IH), 5.03-
4.92 (m, 2H), 4.01 (q, 2H), 3.79 (s. 3H), 3.26 (dd, IH), 3.01 (dd, IH), 2.36 (s, 3H), 1.12 (t, 3H)
MS: (M+H)+ = 563.2
Step J: Examples 68 and 69
60 mg 3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]-2-(4-fhioro phenyl)-l-methyl-indol-4-ol (0.12 mmol), 101 mg ethyl (2S)-3-[2-[[2-(2-methoxy phenyl)pyrimidin-4-yl]methoxy]phenyl]-2-(/>toly[sulfonyloxy)propanoate (0.18 mmol) and 80 mg Cs2CO3 (0.24 mmol) were dissolved in 2 mL dry DMF and stirred at 50 °C until no further conversion was observed. Then 2 eq. LiOH*H2O was added and mixture was 25 stirred at r.t. until no further conversion was observed. The mixture was concentrated and purified by preparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 68 as the earlier eluting diastereoisomer.
HRMS calculated for C5ûH49C1FN5O6: 869.3355, found: 435.6743 (M+2H). Example 69 was obtained as the later eluting diastereoisomer. HRMS calculated for C5oH49C1FN506: 30 869.3355, found: 435.6767 (M+2H)
ORIGINAL
- mExample 70: jV-[3-(3-chloro-2-methylphenyI)thieno|3,2-c]pyridin-4-yl|-Z)phenylalanine
Step A : 4-bromo-YE(dimethoxymethyl)thiophene-3-carboxamide
5.01 g 4-bromothiophene-3-carboxylic acid (24.2 mmol) was dissolved in 25 mL isopropyl acetate and 17.9 mL SOCl2 (242 mmol) was added and the mixture was stirred at 50 °C for 2 hours. Then the excess SOCl2 was distilled and the residue was dissolved in 25 mL isopropyl acetate and cooled to l0°C. 10.6 mL DIPEA (60.5 mmol) and 4.0 mL aminoacetaldehyde dimethyl acetal (36.3 mmol) were added. The mixture was allowed to warni up to r.t. and stirred under N2 atmosphère ovemight. The mixture was diluted with 10 % aqueous H3PO4 solution and extracted with isopropyl acetate. The combined organic phases were washed with 10% aqueous KH2PO4 solution and brine, then dried over Na2SC>4. fîitered and the filtrate was concentrated under reduced pressure to obtain 4-bromo-iV-(dimethoxymethyl)thiophene-3-carboxamide.
'H NMR (400 MHz, DMSO-d6) δ: 8.36 (t, IH), 7.93 (d, IH), 7.72 (d, IH), 4.48 (t, IH), 3.31-3.28 (m, 8H)
MS (M+H): 294.0
Step Bt 3-bromo-5Y\-thierio[3,2-e.]pyridin-4-orie mg 4-bromo-/V-(dimethoxymethyl)thiophene-3-carboxamide (O.l02 mmol) was dissolved in l mL PPA and stirred at i00°C under argon atmosphère until no further conversion observed. The mixture was then poured into ice, the formed precipitate was fîitered and washed with water to obtain 3-bromo-5/7-thieno[3,2-c]pyridin-4-one. MS (M+H): 229.9
Step C: 3-bromo-4-chloro-thieno[3,2-c]pyridine l .06 g 3-brorno-5//-thieno[3,2-c]pyridin-4-one (4.4 mmol), 560 pL NJV-dimethylaniline (4.4 mmol) and 8.37 mL POCI3 (88 mmol) were stirred at I00 °C until no further conversion observed. The reaction mixture was then poured into ice and extracted with DCM. The combined organic phases were washed with saturated aqueous NaHCO3 solution and brine, dried over Na2SC>4, fîitered and the filtrate was concentrated under
ORIGINAL
- H2reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 3-bromo-4-chloro-thieno[3,2-c]pyridine. MS (M+H): 247.9
Step D: 3,4-dibromothieno[3,2-cJpyridine
735 mg 3~bromo-4-chloro-thieno[3,2-c]pyridine (2.8 mmol) and 2.288 g bromotrimethylsilane (14.5 mmol) were dissolved in 15 mL propionitrile and stirred at 100 °C until no further conversion observed. The reaction mixture was then concentrated under reduced pressure and purified via preparative reversed phase chromatography using 40 mM aqueous NH4OAC solution (pH = 4, adjusted with AcOH) and MeCN as eluents to obtain 3,4-dibromothieno[3,2-c]pyridine. MS (M+H): 291.8
Step E: (2ty-2-[(3-bromothieno[3,2-c]pyridin-4-yl)ammo]-3-pheriyl-propanoic acid
340 mg 3,4-dibromothieno[3,2-c]pyridine (I.l6 mmoi) and 718 mg D-phenylalanine (4.35 mmol) were dissolved in 7.5 mL sulfolane, then 421 mg potassium fluoride (7.25 mmol) and 2.23 g 4,7,l3,l6,2l,24-hexaoxa-l,l0-diazabicyclo[8.8.8]hexacosane (5.8 mmol) were added and the mixture was stirred at 175 °C under argon atmosphère until no further conversion was observed. The reaction mixture was directly injected and purified via preparative reversed phase chromatography using 40 mM aqueous NH4OAC solution (pH = 4, adjusted with AcOH) solution and MeCN as eluents to obtain (27?)-2-|(3bromothieno[3,2-c]pyridin-4-yI)amino]-3-phenyl-propanoic acid.
Step F: Example 70
189 mg (2/?)-2-[(3-bromothieno[3,2-c]pyridin-4-yl)amino]-3-phenyl-propanoic acid (0.5 mmol), 341 mg (3-chloro-2-methylphenyl)boronic acid (2 mmol) were dissolved in 3.5 mL DME, then 72 mg butyldi-l-adamantylphosphine (0.2 mmol), 22 mg Pd(OAc)2 (O.l mmol) and 389 mg TBAOH (1.5 mmol) were added and the mixture was stirred at !00°C under argon atmosphère until no further conversion was observed. Then the mixture was poured into icy water, extracted with MTBE. The aqueous phase was acidified to pH 2 and extracted with DCM. The combined organic phases were dried over Na2SO4, fîltered and the filtrate was concentrated under reduced pressure. The crude product was purified via preparative reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 70. HRMS calculated for
ORIGINAL
- H3C23H19CIN2O2S: 422.0856, found: 423.0937 and 423.0919 for the two diastereomers (M+H)
Example 71: (27?)-2-{[(35(()-3-{3-cliloro-2-methyI-4-[2'(4-methylpiperazÎn-l-yI)ethoxy| phenyl}-2-(4-fluorophenyl)thieno[2,3-6]pyridm-4-ylJoxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yIjmethoxy}phenyl)propanoic acid
Step A: 2-chloro-3-[2-(4-fliiorophenyl)ethynyl]pyridine
In a dry flask 3.85 g 3-bromo-2-chloro-pyridine (20 mmol), 0.23 g Cul (1.2 mmol), 0.42 g PdCl2(PPlt3)2 (0.6 mmol) were added in 40 mL dry' TEA. After stirring for 10 minutes, 2.64 g l-ethynyl-4-fluoro-benzene (22 mmol) was added and the solution was heated to 100 °C and stirred ovemight. The reaction mixture was cooled down, diluted with water and then it was extracted with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 2-chloro-3-[2-(4fluorophenyl)ethynyl]pyridine. ’H NMR (500 MHz, DMSO-dô) δ 8.44 (dd, IH), 8.14 (dd, IH), 7.68 (t, 2H), 7.51 (dd, IH), 7.33 (t, 2H)
Step B: 2-(4-fluorophenyl)thieno[2,3-b]pyridine
2.95 g 2-chloro-3-[2-(4-fluorophenyl)ethynyl]pyridine (12.7 mmol) and 3.97 g Na2S (51 mmol) were placed in a 250 mL flask. 120 mL DMF was added and the mixture was stirred at 130 °C for 2 hours. Then the reaction mixture was cooled down, diluted with water and then it was extracted with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain 2-(4-fluorophenyl)thieno[2,3-Z>] pyridine. MS (M+H): 230.2
Step C: 2-(4-fluorophenyl)thieno[2,3-b]pyridine N-oxide
1.94 g 2-(4-fluorophenyl)thieno[2,3-ô]pyridine (8.4 mmol) was dissolved in DCM (50 mL) and cooled to 0 °C. 3.12 g MCPBA (12.6 mmol) was added portionwise and stirred at r.t. for 6 hours. Then it was concentrated under reduced pressure and the crude product was purified via flash chromatography using DCM and methanol as eluents. MS (M+H): 246.2
ORIGINAL
- 114Step D: 4-chloro-2-(4-fluorophenyl)thieno[2,3-\)]pyridine
1.56 g 2-(4-fluorophenyl)-7-oxido-thieno[2,3-ô]pyridin-7-ium (6.4 mmol) was dissolved in 50 mL CHCI3. 15.7 mL POCI3 (25.76 g, 168 mmol) was added and the reaction mixture was stirred at reflux température for 3 hours. Then it was cooled down, ice and saturated aqueous NaHCOs was added and it was extracted with CHCI3. The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was purified via flash chromatography using DCM and methanol as eluents to obtain 4-chloro-2-(4-fluorophenyl)thieno[2,3-ô]pyridine. MS (M+H): 264.0
Step E: 3-brotno-4-chloro-2-(4-fluorophenyl)thieno[2.3-b]pyridine
1.15 g Br? (7.2 mmol) was added dropwise to a mixture of 1.46 g 4-chloro-2-(4fluorophenyl)thieno[2,3-ô]pyridine (5.5 mmol), 0.52 g K2HPO4 (3.0 mmol), 0.46 g NaHCCfe (5.5 mmol) and 1.12 g MgSCL (9.2 mmol) in 20 mL CHCI3. The mixture was stirred overnight at reflux température. Then, the reaction was cooled down and filtered. The fîltrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using DCM and methanol as eluents to obtain 3-bromo-4-chloro-2(4-fluorophenyl)thieno[2,3-6]pyridine. ]H NMR (500 MHz, DMSO-d(,) δ 8.59 (d, IH),
7.76 (m, 2H), 7.71 (d, IH), 7.42 (m, 2H)
Step F: 3-bromo-2-(4-fluorophenyl)lhieno[2t3-b]pyridin-4-ol
The mixture of 0.206 g 3-bromo-4-chloro-2-(4-iluorophenyl)thieno[2,3-A]pyridine (0.6 mmol), 0.492 g sodium acetate (6 mmol), 12 mL AcOH and 0.18 mL H?O was heated at 150 °C via MW irradiation for 5 hours. Water was added and the product was collected by filtration. 'H NMR (500 MHz, DMSO-d6) δ 11.63 (br s, IH), 8.30 (br s, IH), 7.72 (m, 2H), 7.38 (m, 2H), 6.87 (br s, 1 H)
Step G: Ethyl (2R.)-2-[3-bromo-2-(4-jluorophenyl)thieno[2.3-b]pyridin-4-yl]oxy-3-[2-[[2(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate
0.324 g 3-bromo-2-(4-fluorophenyl)thieno[2,3-6]pyridin-4-ol (1 mmol), 0.613 g Préparation 2d (1.5 mmol), 0.691 g DTAD (3 mmol) and 0.787 g PPh3 (3 mmol) were dissolved in 10 mL dry THF under N2 atmosphère and the mixture was stirred at r.t. until
ORIGINAL
- Il5no further conversion was observed. The solvent was then removed under reduced pressure, the residue was purified via flash chromatography using heptane and EtOAc as eluents to give ethyl (27?)-2-[3-bromo-2-(4-fluorophenyl)thieno[2,3-ô]pyridin-4-yl]oxy-3[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate. 'H NMR (500 MHz, DMSO-dù) ô 8.86 (d, IH), 8.33 (d, IH), 7.72 (m, 2H), 7.61 (d, IH), 7.51 (dd, IH), 7.45 (td, IH), 7.44 (d. IH), 7.39 (m, 2H), 7.25 (td, IH), 7.14 (d, IH), 7.10 (d, IH), 7.03 (td, IH), 6.93 (t, IH), 6.88 (d, IH), 5.55 (dd. IH), 5.30 (d, IH), 5.26 (d, IH), 4.16 (m, 2H), 3.75 (s, 3H), 3.58 (dd, IH), 3.35 (dd, IH), 1.13 (t, 3H)
Step H: Example 71
0.288 g (2£)-2-[3-bromo-2-(4-fluorophenyI)thieno[2,3-6]pyridin-4-yl]oxy-3-|2-[[2-(2methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate (0.4 mmol), 0.472 g Préparation 3b (1.2 mmol), 0.028 g Ataphos (0.004 mmol) and 0.392 g Cs2CO3 (1.2 mmol) were dissolved in a mixture of dioxane (4 mL) and water (3 mL) and stirred under N2 at 70 °C until no further conversion was observed. Then the mixture was diluted with water and extracted with DCM. The combined organic phases were dried over Na2SO4 and concentrated under reduced pressure. The crude product was purified was purified via flash chromatography using DCM and methanol as eluents. The obtained intermediate was dissolved in a mixture of dioxane (7 mL) and water (7 mL) and 0.168 g LiOHxH2O (4 mmol) was added. The mixture was stirred at r.t. until no further conversion was observed. Then it was diluted with brine, neutralized with 2M aqueous HCl, extracted with DCM. The combined organic phases were dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The diastereoisomers were purified and separated by preparative reversed phase chromatography using 5 mM aqueous NH4HCO3 solution and MeCN as eluents. The diastereomer eluting later was collected as Example 71. HRMS calculated for C^^jCIFNjO^S: 873.2763; found 437.6441 (M+2H)
Example 72: (27?)-2-|5-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy| phenyl|-6-(4-fluoroplienyl)-7-methyl-pyrrolo[2,3-i/]pyrimidin-4-yl]oxy-3-|2-[[2-(2methoxyphenyI)pyrimidin-4-yl|methoxyjphenyl|propanoic acid
Step A: Ethyl 2-amino-5-(4-fluorophenyl)-l\A-pyn‘ole-3-carhoxylate
ORIGINAL
-neThe solution of 3330 mg ethyl 3-amino-3-imino-propanoate (20 mmol) and 4340 mg 2-bromo-l-(4-fluorophenyl)ethanone (20 mmol) in 40 mL éthanol was stirred at r.t. for 30 minutes, then 20 mL l M NaOEt solution in éthanol (20 mmol) was added at 0 °C, then it was stirred at 60 °C for 90 minutes. Additional 13 mL IM NaOEt solution in éthanol (13 mmol) was added at room température and it was stirred at 60 °C for further l hour. The reaction mixture was concentrated under reduced pressure, diluted with 40 mL water then it was extracted with ethyl acetate. The combined organic phase was dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified via flash chromatography using heptane and EtOAc as eluents to obtain ethyl 2-amino-5-(4-fluorophenyl)-l/7-pyrrole-3-carboxylate. NMR (400 MHz, DMSO-d6) δ: 10.75 (br s, IH), 7.52 (m, 2H), 7.14 (m, 2H), 6.44 (d, IH), 5.68 (br s, 2H), 4.14 (q, 2H),
1.25 (t, 3H)
Step B: 6-(4-fliiorophenyl)-3,7-dihydropyrrolo[2,3-d]pyrimidin-4-one
The solution of 6.83 g ethyl 2-amino-5-(4-fluorophenyl)-l//-pyrrole-3-carboxylate (27.5 mmol) and 12 mL formic acid in 50 mL formamide and 24 mL DMF was stirred at 160 °C for I6 hours in a sealed reaction vessel. The reaction mixture was cooled to room température; 150 mL 2-propanol was added. The precipitate was filtered, washed with heptane, then it was dried under reduced pressure to obtain 6-(4-fluorophenyl)-3,7dihydropyrrolo[2,3-J]pyrimidin-4-one. 'H NMR (400 MHz. DMSO-dé) δ: 12.36 (br s, IH), H.88 (br s, lH), 7.88 (m. 3H), 7.27 (t, 2H), 6.93 (s, lH)
Step C: 4-chloro-6-(4-fluorophenyl)-7Y{-pyrrolo[2,3-d]pyrimidine
The solution of 4.50 g 6-(4-fluorophenyl)-3,7-dihydropyrrolo[2,3-i/]pyrimidin-4-one (Ί 9.6 mmol) in 46 mL POCI3 (491 mmol) was stirred al 90 °C for 3 hours. It was concentrated under reduced pressure, the residue was poured onto ice. The pH was adjusted to 7 using solid K2CO3, then the mixture was extracted with ethyl acetate. The combined organic phase was washed with brine, then it was dried over MgSO4, filtered and the filtrate was concentrated under reduced pressure to give 4-chloro-6-(4-fIuorophenyl)7//-pyiTolo[2,3-i/]pyrimidine. 'H NMR (400 MHz, DMSO-d6) ô: 13.04 (br s, IH), 8.60 (s, IH), 8.08 (m, 2H), 7.37 (t, 2H), 7.10 (d, IH)
ORIGINAL
- 1I7Step D: 4-chloro-6-(4-fluorophenyl)-7-methyl-pyrrolo[2,3-d]pyrimidine
To the solution of l .87 g 4-chloro-6-(4-fluorophenyl)-7/7-pyrrolo[2,3-</]pyrimÎdine (7.55 mmol) in 38 mL DMF L286 g Mel (9.06 mmol) then Ll5 g K2CO3 (8.30 mmol) was added and it was stirred at r.t. for l hour. The reaction mixture was concentrated under reduced pressure. The residue was diluted with brine, and it was extracted with dichloromethane. The combined organic phase was dried over MgSCL, filtered and the filtrate was concentrated under reduced pressure, then the residue was purified via flash chromatography using heptane and ethyl acetate as eluents to obtain 4-chIoro-6-(4fluorophenyl)-7-methyl-pyrrolo[2,3-d]pyrimidine. 111 NMR (400 MHz, DMSO-d6) δ: 8.69 (s, l H), 7.79 (m, 2H), 7.42 (m, 2H), 6.80 (s, l H), 3.83 (s, 3H)
Step E: 5-bromo-4-chloro-6-(4-fliiorophenyl)-7-methyl-pyrrolo[2,3-d]pyrimidine
To the solution of 1.36 g 4-chloro-6-(4-fluorophenyl)-7-methyl-pyrrolo[2,3-i/]pyrimidine (5.20 mmol) in 16 mL acetic acid 5.46 mL IM Br2 solution in acetic acid (5.46 mmol) was added dropwise at 0 °C, then the reaction mixture was stirred at r.t. for 30 minutes. The reaction mixture was concentrated under reduced pressure, then the residue was diluted with saturated aqueous NaHCO3 solution and it was extracted with ethyl acetate. The combined organic phase was dried over MgSCL, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and ethyl acetate as eluents to obtain 5-bromo-4-chloro-6-(4-iluorophenyl)-7methyl-pyrrolo[2,3-<7]pyrimidine. ’H NMR (400 MHz, DMSO-dô) δ: 8.73 (s, IH), 7.70 (m, 2H), 7.47 (m, 2H), 3.69 (s, 3H)
Step F: Ethyl (2R)-2-[5-bromo-6-(4-jhtorophenyl)-7-methyl-pyrrolo[2,3-d]pyrimidin-4yl] oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy]phenyl]propanoate
845 mg 5-bromo-4-chloro-6-(4-lluorophenyl)-7-inethyl-pyrrolo[2,3-i.7]pyrimidine (2.48 mmol), 1.27 g Préparation 2c (3. H mmol) was dissolved in 10 mL DMF, then 2.43 g Cs2CO3 (7.44 mmol) was added and the mixture was stirred at 60 °C for 6 hours. The reaction mixture was concentrated under reduced pressure, it was diluted with brine, and then the mixture was extracted with ethyl acetate. The combined organic phase was dried over MgSCU filtered and the filtrate was concentrated under reduced pressure, then the residue was purified via flash chromatography using heptane and ethyl acetate
ORIGINAL
- usas eluents to obtain ethyl (2Æ)-2-[5-bromo-6-(4-fluorophenyl)-7-methyl-pyrrolo [2,3-J]pyrimidin-4-yl]oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate. MS (M+H): 712.0
Step G: Example 72
Using General Procedure II and ethyl (2Æ)-2-[5-bromo-6-(4-fluorophenyl)-7-methylpyrrolo[2,3-i/]pyrimidm-4-yl]oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy] phenyl]propanoate instead of 5-bromo-furo[2,3-r/]pyrimidyl-lactic ester, and using Préparation 3b as the appropriate boronîc acid dérivative, Example 72 was obtained as a mixture of diastereoisomers. HRMS calculated for C48H47CIFN7O6: 871.3260; found 436.6703 and 436.6710 (M+2H)
Example 73: 2-{|3-{3,5-dichloro-2,6-dimethyl-4-[2-(4-methylpiperazin-l-yl)etlioxy| phenyl}-2-(4-fluorophenyl)thïeno|2,3-ô|pyridin-4-yl]oxy}-3-(2-{|2-(2-methoxyphenyl) pyrimidin-4-yl] methoxy)phenyl)propanoic acid
Example 74: 2-{[3-{2,6-dimethyl-4-|2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2-(4fluorophenyl)thieno I2,3-6| pyridin-4-yl]oxy )-3-(2-{| 2-(2-methoxyphenyl)pyrimidin-4yl] methoxy)phenyl)propanoïc acid
Example 75: l-j(dimethylcarbamoyl)oxy]ethyl (2/î)-2-{|(3Srt)-3-{3-chloro-2-methyl-4[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fluorophenyl)thieno|2,3-h]pyridin-4yl]oxy)-3-(2-{[2-(2-niethoxyphenyl)pyrimidin-4-yl]methoxy}phcnyl)propanoate
591 mg dimethylamine hydrochloride (7.25 mmol) and 1.20 mL pyridine (14.9 mmol) were dissolved in 18 mL dry DCM under nitrogen atmosphère, then the mixture was cooled to -78 °C and 990 mg 1-chloroethyl chloroformate (6.9 mmol) was added. The reaction mixture was stirred at -78 °C until no further conversion was observed. The cold mixture was filtered and the filtrate was concentrated under reduced pressure (30 mbar) using a 30°C bath. Then it was dissolved in 2 mL dry DMF under nitrogen atmosphère, 60 mg Example 71 (0.069 mmol) and 223 mg Cs2CO3 (0.55 mmol) were added and the reaction mixture was stirred at r.t. until no further conversion was observed. Then the
ORIGINAL
- 119mixture was diluted with brine, extracted with EtOAc, The combined organic layer was dried over Na2SÛ4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via reversed phase chromatography using 5 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 75 as a mixture of diastereoisomers. HRMS calculated for C53H54ClFN6O8S: 988.3397; found: 495.1782 and 495.1772 (M+2H)
Example 76: l-[(ethoxycarbonyl)oxy|ethyl (27f)-2-{[(3S, a)-3-(3-chloro-2-methyl-4-[2(4-methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fIuorophenyl)thieno[2,3-ô]pyridin-4yl|oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4-yl|methoxy}phenyl)propanoate
668 mg EtOH (14.5 mmol) and 1.26 g pyridine (15.6 mmol) were dissolved in 18 mL dry DCM under nitrogen atmosphère, then the mixture was cooled to -78 °C and 1.98 g l-chloroethyl chloroformate (13.8 mmol) was added. The reaction mixture was stirred at -78 °C until no further conversion was observed. The cold mixture was filtered and the filtrate was concentrated under reduced pressure (30 mbar) using a 30 °C bath. Then it was dissolved in 2 mL dry DMF under nitrogen atmosphère, 60 mg Example 71 (0.069 mmol) and 223 mg Cs2CC>3 (0.55 mmol) were added and the reaction mixture was stirred at r.t. until no further conversion was observed. Then the mixture was filtered and the filtrate was purified via reversed phase chromatography using 5 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 76 as a mixture of diastereoisomers. HRMS calculated for C53H53C1FN5O9S: 989.3237; found: 990.3342 and 990.3314 (M+H).
Example 77: 2-{|3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yl)ethoxy|phenyl}-2(4-fluorophenyl)tliieno[2,3-Z»|pyridin-4-yi]oxy}-3-hydroxy-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yljmethoxy}phenyl)propanoic acid
Example 78: 2-{[3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yI)ethoxy]phenyl}-2(4-nuorophenyl)thieno[2,3-0|pyridin-4-yl]oxy}-4-hydroxy-3-(2-{|2-(2-mcthoxyphenyl) pyrimidin-4-yI] methoxy}phcnyl)butanoic acid
ORIGINAL
- 120Example 79: 2-0-[3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yI)ethoxy]phenyl}2-(4-fluorophenyl)thieno[2,3-/>]pyridin-4-yl]-3,4-dideoxy-3-(2-{)2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)pentonic acid
Exaniple 80: 2-{[3-{3-chloro-2-metliyl-4-l2-(4-rnethylpiperazin-l-yI)ethoxy]phenyl}-2(4-fluorophenyl)thieno|2,3-#|pyridin-4-yl]oxy}-3-[2-({2-[5-(hydroxymethyl)pyridin-3yl]pyrimidin-4-yl}methoxy)phenyl]propanoic acid
Example 81: 2-{|3-{3-chloro-2-methyl-4-I2-(4-methylpiperazin-l-yl)cthoxy]phenyl}-2(4-fIuorophenyl)thieno|2,3-ô]pyridin-4-yl]oxy}-3-{2-[(2-{2-[(2-hydroxycthoxy)mcthyl| phenyl}pyrimidin-4-yl)methoxy]phenyl}propanoic acid
Example 82: 2-{|3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yl)ethoxy|phcnyl}-2(4-fluorophenyl)thieno[2,3-i»]pyridin-4-yl]oxy}-3-{2-[(2-{4-[2-(dimethylamino)ethoxy| phenyl}pyrïmidin-4-yl)methoxy]phenyl}propanoic acid
Example 83: 2-{[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2(4-fluorophenyl)thieno[2,3-ô]pyridin-4-yl]oxy}-3-[2-({2-[3-(phosphonooxy)phenyl] pyrimidin-4-yl}methoxy)phenyl|propanoic acid
Example 84: ;V-[3-{3-chloro-2-mcthyl-4-|2-(4-niethylpiperazin-l-yl)cthoxy]phcnyl}-2(4-fluoroplienyl)thieno|2,3-/>|pyridin-4-yl|-2-{[2-(2-methoxyplienyl)pyrimidin-4-ylJ methoxy}-D-phenylalanine
Step A: ethyl (2B,)-2-[[3-bromo-2-(4-fluorophenyl)thieno[2.3-'o]pyridin-4-yl]ammo]-3-[2[[2-(2-methoxyphenyl)pyrhnidin-4-yl]methoxy]phenyl]propanoale
343 mg 3-bromo-4-chloro-2-(4-fluorophenyl)thieno[2,3-6]pyridine (Step E in Example 71, 1.0 mmol) and 455 mg Préparation 2i (1.20 mmol) were dissolved in 5 mL dry DMSO, then 978 mg Cs2CO3 (3.0 mmol) was added and the mixture was stirred under nitrogen atmosphère at 100 °C until no further conversion was observed. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was
ORIGINAL
- I2lconcentrated under reduced pressure. Then it was dissolved l .5 mL l .25 M HCl solution in EtOH, and the mixture was stirred at 60 °C until the ester formation was complété. Then it was carefully neutralized with saturated aqueous NaHCCfe solution, extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents.
'H NMR (500 MHz, DMSO-d6) δ: 8.77 (d, IH), 8.07 (d, IH), 7.6 (m, 2H), 7.52-6.88 (m, 8H), 7.37 (d, IH), 7.34 (m, 2H), 7.05 (d, IH), 6.57 (d, IH), 5.23/5.19 (d+d, 2H), 4.92 (m, IH), 4.12 (m, 2H), 3.73 (s, 3H), 3.44/3.25 (dd+dd. 2H), 1.14 (t, 3H)
HRMS calculated for CjôHjoBrFN^S: 712.1155; found: 357.0649 (M+2H)
Step B: ethyl (2R)-2-[[3-(3-chloro-4-hydroxy-2-methyl-phenyl)-2-(4-fuorophenyl)thieno [2,3-b]pyridin-4-yl]amino]-3-[2-[[2-(2-meihoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate
178 mg ethyl (2Æ)-2-[[3-bromo-2-(4-fluorophenyl)thieno[2,3-/j]pyridin-4-yl]amino]-3-[2[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate (0.249 mmol) and 107 mg Préparation 3a (0.4 mmol) were dissolved in l mL l,4-dioxane under nitrogen atmosphère, then 163 mg CS2CO3 (0.50 mmol), 0.5 mL water and 28 mg AtaPhos (0.04 mmol) were added and the mixture was stirred in a microwave reactor at 111 °C for 15 minutes. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain ethyl (27?)-2-[[3-(3chloro-4-hydroxy~2-methyl-phenyl)-2-(4-fluorophenyl)thieno[2,3-/>]pyridin-4-yl]amino]-
3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]piOpanoate as a mixture of atropoisomers. HRMS calculated for C^HjôClFNuOsS: 774.2079; found: 388. H 13 (M+2H)
Step C: Example 84 mg ethyl (2/î)-2-[[3-(3-chloro-4-hydroxy-2-methyl-phenyl)-2-(4-fluorophenyl)thieno [2,3-/>]pyridin-4-yI]amino]-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate (0.103 mmol), 43 mg 2-(4-methylpiperazin-l-yl)ethanol (0.30 mmol), and
ORIGINAL
- 12279 mg PPhj (0.30 mmol) were dissolved in l mL dry toluene, then 69 mg DTAD (0.30 mmol) was added and the mixture was stirred at 50 °C under nitrogen atmosphère until no further conversion was observed. Then the mixture was concentrated under reduced pressure and the residue was purified via flash chromatography using EtOAc and MeOH as eluents. The obtained ester dérivative was dissolved in l mL THF, then 80 mg LiOHxH2O and I mL water were added and the mixture was stirred at r.t. until the hydrolysis was complété. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 84 as a 7:3 mixture of diastereoisomers. HRMS calculated for C48H46ClFN6O5S: 872.2923; found: 437.1540 and 437.1538 (M+2H)
Example 85: 2-{[3-{3-chloro-2-methyL4-|2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2(4-fluorophenyl)thieno[3,2-c]pyndin-4-yI]oxy}-3-(2-{|2-(2-methoxyphenyl)pyrimidin4-yl]methoxy}phenyl)propanoic acid
Example 86:7V-[3-{3-chloro-2-methyl-4-|2-(4-mcthy[piperazin-l-yl)ethoxy|phenyl}-2(4-fluorophenyl)thieno|3,2-c|pyridin-4-yl|-2-{|2-(2-methoxyphenyl)pyrimidin-4-yI] metlioxyjphenylalanine
Siep A: 3-bromo-4-chloro-2-iodo-thieno[3.2-c]pyridine
4.97 g 3-bromo-4-chloro-thieno[3,2-c]pyridine (20.0 mmol) was dissolved in 50 mL dry THF under argon atmosphère and the mixture was cooled to -45 °C. Then 22 mL Mg(TMP)Cl*LiCl solution (22 mmol, 1 M in THF) was added dropwise and the mixture was stirred for 1 hour at -45 °C, then 1 hour at 0 °C, then it was cooled to -45 °C again. Then 5.58 g iodine (22 mmol, dissolved in 20 mL dry, cold THF) was added dropwise and the mixture was stirred at -45 °C for 2 hours. Then it was allowed to warm up to r.t. and concentrated under reduced pressure. The residue was poured onto 300 mL brine, and extracted with EtOAc. The combined organic phase was washed with saturated aqueous Na2S2O3 solution, saturated aqueous NH4CI solution, then with water and then dried over
ORIGINAL
- 123Na2SO4, filtered and the fîltrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using hexanes and EtOAc as eluents.
'H NMR (500 MHz, DMSO-dè) Ô: 8.27 (d, IH), 8.17 (d, IH)
HRMS calculated for C7H2BrClINS: 372.7824; found: 373.7916 (M+H)
Step B: 3-bromo-4-chloro-2-(4-fluorophenyl)thieno[3,2-c]pyridine
2.62 g 3-bromo-4-chloro-2-iodo-thieno[3,2-c]pyridine (7.0 mmol) and 2.33 g 2-(4fluorophenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (10.5 mmol) were dissolved in 18 mL THF under argon atmosphère, then 6.84 g Cs2CO3 (21 mmol), 18 mL water, 79 mg Pd(OAc)2 (0.35 mmol) and 297 mg ‘BuXPhos (0.70 mmol) were added and the mixture was stirred at 70 °C until no further conversion was observed. Then the volatiles were evaporated under reduced pressure. The residue was diluted with water and extracted with EtOAc. The combined organic phase was washed with saturated aqueous NH4C1 solution, then with brine and then dried over Na2SO4, filtered and the fîltrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using hexanes and EtOAc as eluents.
‘H NMR (500 MHz, DMSO-d6) δ: 8.35 (d, IH), 8.26 (d, IH), 7.74 (dd, 2H), 7.42 (t, 2H) HRMS calculated for Ci3H6BrClFNS: 340.9077; found: 341.9144 (M+H)
Step C: ethyl (2R)-2-[[3-bromo-2-(4-fliiorophenyl)thieno[3,2-c]pyridin-4-yl]amino]-3-[2[[2-(2-methoxy>phenyl)pyrhnidin-4-yl]methoxy]phenyl]propanoate
343 mg 3-bromo-4-chloro-2-(4-fluorophenyl)thieno[3,2-c]pyridine (1.0 mmol) and 455 mg
Préparation 2i (1,20 mmol) were dissolved in 5 mL dry DMSO, then 978 mg CS2CO3 (3.00 mmol) was added and the mixture was stirred under nitrogen atmosphère at 100 °C until no further conversion was observed. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the fîltrate was concentrated under reduced pressure. Then it was dissolved 1.5 mL 1.25 M HCl solution in EtOH, and the mixture was stirred at 60 °C until the ester formation was complété. Then it was carefully neutralized with saturated aqueous NaHCÛ3 solution, extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the fîltrate was concentrated under reduced pressure. The crude
ORIGINAL
- 124product was purified via flash chromatography using heptane and EtOAc as eluents. HRMS calculated for C^HaoBrEN^S: 712.1155; found: 713.1209 (M+H)
Step D: ethyl (2VL)-2-[[3-(3-chloro-4-hydroxy-2-methyl-phenyl)-2-(4-fluorophenyl)thieno [3,2-c]pyridin-4-yl]amino]-3-[2-[[2-(2-methoxyphenyl)pyrimldin-4-yl]methoxy]phenyl] propanoate mg ethyl (2/î)-2-[[3-bromo-2-(4-fluorophenyl)thieno[3,2-c]pyridtn-4-yl]amino]-3-[2[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate (0.043 mmol) and 24 mg Préparation 3a (0.09 mmol) were dissolved in 0.5 mL 1,4-dioxane under nitrogen atmosphère, then 33 mg Cs2CO3 (0.10 mmol), 0.5 mL water and 9.4 mg AtaPhos 10 (0.013 mmol) were added and the mixture was stirred in a microwave reactor at 111 °C for minutes. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, fîltered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain ethyl (2Æ)-2-[[j-(j15 chloro-4-hydroxy-2-methyl-phenyl)-2-(4-fluorophenyl)thieno[3,2-c]pyridin-4-yl]amino]-3[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate as a mixture ot atropoisomers. HRMS calculated for C43H36C1FN4O5S: 774.2079; found: 775.2134 (M+H)
Step E: Example 86 mg ethyl (2/?)-2-[[3-(3-chloro-4-hydroxy-2-methyl-pheny!)-2-(4-fluorophenyl)thieno 20 [3,2-c]pyridin-4-yl]amino]-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate (0.04 mmol), 15 mg 2-(4-methylpiperazin-l-yl)ethanol (0.10 mmol), and mg PPh3 (0.10 mmol) were dissolved in 1 mL dry toluene, then 23 mg DTAD (0.10 mmol) was added and the mixture was stirred at 50 °C under nitrogen atmosphère until no further conversion was observed. Then the mixture was concentrated under 25 reduced pressure and the residue was purified via flash chromatography using EtOAc and
MeOH as eluents. The obtained ester dérivative was dissolved in 1 mL THF, then 80 mg LÎOH*H2O and 1 mL water were added and the mixture was stirred at rt until the hydrolysis was complété. Then it was diluted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over 30 Na2SO4, fîltered and the filtrate was concentrated under reduced pressure. The crude
ORIGINAL
- 125product was purified via reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 86 as a 10:3 mixture of diastereoisomers. HRMS calculated for C48H46CIFN6O5S: 872.2923; found: 437.1549 and 437.1532 (M+2H)
Example 87: 2-{[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-25 (4-f|uorophenyl)thieno[2,3-clpyridin-4-ylloxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin4-yllmethoxy}phenyl)propanoic acid
Example 88: /V-|3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yl)ethoxylphenyl}-2(4-fluorophcnyl)tlneno[2,3-c|pyridin-4-yll-2-{[2-(2-niethoxyphenyl)pyrimidin-4-yl| methoxyjphenylalanine
Example 89: 2-{|3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy|phenyl}-2(4-fluorophenyl)thieno|2,3-i/]pyridazin-4-yl|oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimi(lin-4-yl|methoxy}phenyl)propanoic acid
Example 90: A-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxylphenyl}-2(4-fluorophenyl)thieno[2,3-rf|pyridazin-4-yl]-2-{[2-(2-methoxyplienyl)pyrimidin-4-yl] methoxyjphenylalanine
Example 91: 2-{|5-{3-chloro-2-methyl-4-I2-(4-methylpiperazin-l-yl)ethoxy|phenyl}-6(4-fluorophenyl)thieno|2,3-c]pyridazin-4-yl]oxy}-3-(2-{|2-(2-methoxyphenyl) pyrimidin-4-yl|methoxy}plienyl)propanoic acid
Example 92: A-I5-{3-chloro-2-mcthyI-4-[2-(4-methylpiperazin-l-yl)etlioxy]phenyI}-620 (4-fluorophenyl)thieno[2,3-c|pyridazin-4-yl]-2“{[2-(2-niethoxyphenyl)pyrimidin-4-yl| methoxyjphenylalanine
Example 93: 2-{[3-{3-chloro-2-methyl-4-I2-(4-methylpiperazin-l-yl)ethoxy|phenyl}-2(4-fluorophenyl)furo|2,3-Z>]pyridin-4-yl)oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4yl]methoxy}phenyl)propanoic acid
ORIGINAL
- 126Example 94: 7V-|3-{3-chloro-2-methyl-4-|2-(4-methyIpiperazin-l-yl)ethoxy]phenyl}-2(4-fluorophenyl)furo[2,3-i>]pyridin-4-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxyjphcnylalanine
Example 95: 2-{|3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-25 (4-fluorophenyl)furo[3,2-c]pyridin-4-yl|oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4yl]methoxy]phenyl)propanoic acid
Example 96:7V-[3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2(4-fluorophenyl)furo[3,2-c]pyridin-4-yl]-2-{|2-(2-methoxyphenyl)pyrimidin-4-yl| methoxy} phenylalanine
Example 97a: 2-{[(35„)-3-{3-chloro-2-methyl-4-|2-(4-methylpiperazin-lyl)ethoxy]phenyl}-2-(4-fluorophenyl)imidazo|l,2-c]pyrimidin-5-yl]oxy}-3-(2-{[2-(2.J» methoxyphenyl)pyrimidin-4-yI]methoxy}pheoyl)propanoic acid and j
Examplc 97b: 2-{[(3Æfl)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l15 yl)ethoxy|phenyl}-2-(4-fluorophenyI)imidazo|l,2-c]pyrimidin-5-yl]oxy}-3-(2-{|2-(2methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl)propanoic acid
Step A: 7-(2-bromo-l.1 -dimethoxy-ethyl)-4-fluoro-benzene
8.68 g 2-bromo-l-(4-fluorophenyl)ethanone (40.0 mmol) was dissolved in 80 mL MeOH, then 8.75 mL CH(OMe)j (80.0 mmol) and 380 mg TsOHxH2O (2.00 mmol) was added 20 and the mixture was stirred at reflux température until no further conversion was observed.
Then it was concentrated under reduced pressure and diluted with Et2O. It was washed with 10% aqueous K.2CO3 solution, dried over Na2SO4, fîitered and the filtrate was concentrated under reduced pressure. ’H NMR (250 MHz. CDCI3) δ: 7.53-7.44 (m, 2H), 7.11-7.01 (m, 2H), 3.60 (s, 2H), 3.22 (s, 6H)
Step B: 5-chloro-2-(4-fluorophenyl)imidazo[],2-c]pyrimidine
A high pressure reaction vessel made of Steel was charged with 648 mg 2-chloropyrimidin-
4-amine (5.0 mmol), 1.58 g l-(2-bromo-l,l-dimethoxy-ethyl)-4-fluoro-benzene
ORIGINAL
- 127(6.0 mmol), 123 mg Sc(OTf)3 (0.25 mmol) and 50 mL MeCN and the mixture was stirred at 120 °C for 24 hours. Then it was diluted with DCM and washed with saturated aqueous NaHCO3 solution. The aqueous layer was extracted with DCM. The combined organic layer was dried over MgSC>4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using hexanes and EtOAc as eluents. HRMS calculated for CjJ-LClFNj: 247.0312; found: 248.0397 (M+H)
Step C: 3-bromo-5-chloro-2-(4-fluorophenyl)imidazo[l, 2-c]pyrimidine
198 mg 5-chloro-2-(4-fluorophenyl)imidazo[l,2-c]pyrimidine (0.80 mmol) was dissolved in 4.8 mL DMF then 142 mg NBS (0.80 mmol) was added and the mixture was stirred at r.t. until the consomption of the starting material. Then the mixture was poured onto saturated aqueous NaHCO3 solution and the formed precipitate was filtered, washed with water. The crude product was purified via flash chromatography using hexanes and EtOAc as eluents.
'H NMR (500 MHz. DMSO-d6) Ô: 8.03 (m, 2H), 7.90 (d, IH), 7.73 (d, IH), 7.39 (m, 2H) HRMS calculated for C12H6BrClFN3: 324.9418; found: 325.9496 (M+H)
Step D: ethyl (2R)-2-[3-bromo-2-(4-fluorophenyl)imidazo[1.2-c]pyrimidin-5-yl]oxy-3-[2[[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy]phenyl]propanoate
102 mg 3-bromo-5-chloro-2-(4-fluorophenyl)imidazo[l,2-c]pyrimidine (0.312 mmol) and 140 mg Préparation 2c (0.344 mmol) were dissolved in 3 mL dry DMSO under nitrogen atmosphère, then 305 mg Cs2CO3 (0.936 mmol) was added and the mixture was stirred at r.t. until no further desired conversion was observed. Then it was diluted with brine and water, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents.
‘H NMR (500 MHz, DMSO-df,) δ: 8.20 (d, IH), 8.03 (m, 2H), 7.62 (d, IH), 7.56 (d, IH),
7.50 (dd, 111), 7.49 (dd, IH), 7.42 (ddd, IH), 7.35 (m, 2H), 7.27 (ddd, IH), 7.23 (d, IH),
7.13 (d, IH), 7.11 (d, IH), 7.01 (td, IH), 6.96 (td. IH), 5.80 (dd, IH), 5.31/5.27 (d+d, 2H), 4.18/4.15 (m+m, 2H), 3.75 (s, 3H), 3.62/3.36 (dd+dd, 2H), 1.12 (t, 3H)
HRMS calculated for CajHjgBrFNjO,: 697.1336; found: 698.1419 (M+H)
ORIGINAL
- 128Step E: ethyl (2R)-2-[3-(3-chloro-4-hydroxy-2-melhyl-phenyl)-2-(4-flnorophenyl)imidazo [l,2-c]pyrimidin-5-yl]oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl] propanoate
150 mg ethyl (2/î)-2-[3-bromo-2-(4-fiuorophenyI)imidazo[l,2-c]pyrimidin-5-yl]oxy-3-[25 [[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy]phenyl]propanoate (0.215 mmol) and
80.8 mg Préparation 3a (0.301 mmol) were dissolved in 1 mL THF under nitrogen atmosphère, then 140 mg Cs2CO3 (0.430 mmol), 0.2 mL water and 30.4 mg AtaPhos (0.043 mmol) were added and the mixture was stirred in a microwave reactor at 100 °C for 5 minutes. Then the mixture was diiuted with brine, neutralized with 2 M aqueous HCl solution and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using heptane and EtOAc as eluents to obtain a mixture of diastereoisomers. HRMS calculated for C42H35C1FN5O6: 759.2260; found: 760.2370 and 760.2344 (M+H)
Step F: Examples 97a and 97b
11.4 mg ethyl (2/?)-2-[3-(3-chloro-4-hydroxy-2-methyl-phenyl)-2-(4-fluorophenyl) imidazo[l,2-c]pyrimidin-5-yl]oxy-3-[2-[[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy] phenyl]propanoate (0.015 mmol), 7.2 mg 2-(4-methylpiperazin-Lyl)ethanoI (0.050 mmol), and 13.1 mg PPh3 (0.050 mmol) were dissolved in 1 mL dry toluene, then 11.5 mg DTAD (0.050 mmol) was added and the mixture was stirred at 50 °C under nitrogen atmosphère until no further conversion was observed. Then the mixture was concentrated under reduced pressure and the residue was purified via flash chromatography using EtOAc and MeOH as eluents. The obtained ester dérivative was dissolved in 1 mL THF, then 42 mg LiOH><H2O and 1 mL water were added and the mixture was stirred at r.t. until the hydrolysis was complété. Then it was diiuted with brine, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents. Example 97a was obtained as the earlier eluting
ORIGINAL
- 129diastereoisomer. HRMS calculated for C47H45CIFN7O6: 857.3104; found: 429.6626 (M+2H)
Example 97b was obtained as the later eluting diastereoisomer. HRMS calculated for C47H45C1FN7O6: 857.3104; found: 429.6638 (M+2H)
Example 98: Ar-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy]phenyl}-2(4-fluorophenyl)imidazo|l,2-c|pyrimidin-5-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4yljmethoxyjphenylalanine
Example 99: 2-{[3-{3-chi()r()-2-methvl-4-|2-(4-niethylpiperaziii-l-yl)ctlioxy|phenyli-2(4-fluorophenyl)imidazo[l,2-fl]pyrazin-5-yI]oxy}-3-(2-{|2-(2-methoxyphenyl) pyrimidin-4-y 1] methoxyjphenyl)propanoic acid
Example 100: 7V-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy|phcnyl}2-(4-fluorophenyl)imidazo[l,2-«]pyrazin-5-yl]-2-{|2-(2-methoxyphenyl)pyrimidin-4yl] methoxyjphenylalanine
Example 101: 2-{[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)imidazo[l,2-fl]pyrimidin-5-yl|oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-y!]methoxy}phenyl)propanoic acid
Example 102: Ar-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yI)ethoxy|phenyl}2-(4-iluorophenyI)imidazo[l,2-«]pyrimidin-5-yl]-2-{|2“(2-methoxyphenyI)pyrinndin4-yl]mcthoxy}-D-phenylalanine
Step A: 2-(4-fluorophenyl)-1 \A-imidazo[1,2-ά]pyrimidin-5-one
10.0 g 2-amino-7//-pyrimidin-4-one (90.0 mmol) and 9.77 g 2-bromo-l-(4-fluorophenyl) ethanone (45.0 mmol) were dissolved in 100 mL DMF and the mixture was stirred at 120 °C until no further conversion was observed. Then it was concentrated under reduced pressure and was diluted with EtOAc. Celite was added and the volatiles were evaporated under reduced pressure. The mixture was purified via flash chromatography using heptane and EtOAc as eluents. The regioisomer eluting earlier was collected as 2-(4-fluorophenyl)OR1GINAL
- 130l/7-imidazo[l,2-iï]pyrimidin-5-one. ’H NMR (500 MHz, DMSO-de) δ: I2.98 (br s, IH),
8.14 (s, IH), 7.97 (m, 2H), 7.90 (d, IH), 7.27 (m, 2H), 5.57 (d, IH)
Step B: 5-chloro-2-(4-fluorophenyl)imidazo[l,2-a]pyrimidine
1.36 g 2'(4-fluorophenyl)-l//-imidazo[l,2</]pyrimidin-5-one (5.9 mmol) and 16.6 mL POCI3 was stirred at 93 °C for 90 minutes, then the mixture was cooled to r.t. and concentrated under reduced pressure. The residue was poured onto icy-water. After the ice melted the formed precipitate was filtered, washed with water. *H NMR (500 MHz, DMSO-d6)ô: 8.65 (s, IH), 8.55 (d, IH), 8.l7(m, 2H), 7.45 (d, IH), 7.33 (m, 2H)
Step C: 3-bromo-5-chloro-2-(4yfluorophenyl)imidazo[l,2-a]pyrimidine
715 mg 5-chloro-2-(4-fluorophenyl)imidazo[l,2-iï]pyrimidÎne (2.89 mmol) was dissolved in 10 mL chloroform then 570 mg NBS (3.20 mmol) was added and the mixture was stirred at r.t. until the consumption of the starting material. Then the mixture was concentrated under reduced pressure and purified via flash chromatography using heptane and EtOAc as eluents.
'H NMR (500 MHz, DMSO-d6) δ: 8.52 (d, IH), 8.10 (m, 2H), 7.39 (m, 2H), 7.38 (d, IH) HRMS calculated for C|2H6BrClFN3: 324.9418; found: 325.9481 (M+H)
Step D: 5-chloro-3~[3-chloro-2-niethyl-4-[2-(4-melhylpiperazin-I-yl)elhoxy]phenyl]-2-(4fluorophenyl)imidazo[ 1,2-aJpyrimidine
620 mg 3-bromo-5-chloro-2-(4-fluorophenyl)imidazo[l,2-u|pyrimidine (1.93 mmol) and
2.37 g Préparation 3b (6.0 mmol) were dissolved in 10 mL THF under nitrogen atmosphère, then 1.30 g Cs2CO3 (4.00 mmol), 3 mL water and 273 mg AtaPhos (0.386 mmol) were added and the mixture was stirred in a microwave reactor at 110 °C for 10 minutes. Then the mixture was diluted with brine and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via flash chromatography using EtOAc and MeOH as eluents. LRMS calculated for C26H26C12FN5O: 513.15; found: 514.1 (M+H)
Step E: Example 102
ORIGINAL
- I3l341 mg 5-chloro-3-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-l -yl)ethoxy]phenyl]-2-(4fluorophenyl)imidazo[l,2-rt]pyrimidine (0.66 mmol) and 300 mg Préparation 2i (0.76 mmol) were dissolved in 3 mL dry DMSO under nitrogen atmosphère, then 652 mg Cs2CO3 (2.0 mmol) was added and the mixture was stirred in a microwave reactor at 160 °C for 10 minutes. Then it was diluted with brine and water, neutralized with 2 M aqueous HCl solution, and extracted with DCM. The combined organic phase was dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The crude product was purified via reversed phase chromatography using 25 mM aqueous NH4HCO3 solution and MeCN as eluents to obtain Example 102 as a mixture of diastereoisomers, HRMS calculated for C47H46C1FN8O5: 856.3264; found; 429.1687 and 429.1705 (M+2H)
Example 103: 2-{|3-{3-chloro-2-methyI-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)imidazo[l,2-fl]pyridin-5-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimîdin-4-yl]methoxy}phenyI)propanoic acid
Example 104: Ar-|3-{3-chloro-2-methyl-4-i2-(4-methylpiperazin-l-yl)ethoxy]phcnyl}2-(4-fluorophenyl)imidazo[l,2-«|pyridin-5-yll-2-{[2-(2-methoxyphenyl)pyrimidin-4yl]niethoxy}phenylalanine
ORIGINAL
- 132PHARMACOLOGICAL STUDY
EXAMPLE A: Inhibition of Mcl-l by the fluorescence polarisation technique
The relative binding potency of each compound was determined via Fluorescence Polarisation (FP). The method utilised a Fluorescein labelled ligand (Fluorescein-pAla5 Ahx-A-REIGAQLRRMADDLNAQY-OH; mw 2,765) which binds to the Mcl-l protein (such that Mcl-l corresponds to the UniProtKB® primary' accession number: Q07820) leading to an increased anisotropy measured in milli-polarisation (mP) units using a reader. The addition of a compound which binds competitively to the same site as the ligand will resuit in a greater proportion of unbound ligand in the System indicated by a decrease in 10 mP units.
Method 1: An 11 point serial dilution of each compound was prepared in DMSO and 2 μΐ transferred into fiat bottomed, low binding, 384-well plate (final DMSO concentration 5%). 38 μΙ of buffer (10 mM 4-(2-hydroxyethyl)-l-piperazineethanesulfonic acid [HEPES], 150 mM NaCl, 0.05 % Tween 20, pH 7.4), containing the Fluorescein labelled 15 ligand (final concentration 1 nM) and Mcl-l protein (final concentration 5 nM) was then added.
Assay plates were incubated ~2 hours at room température before FP was measured on a Biomek Synergy2 reader (Ex. 528 nm, Em. 640 nm, Cut off 510 nm) and mP units calculated. The binding of increasîng doses of test compound was expressed as a 20 percentage réduction in mP compared to a window established between ‘5 % DMSO only’ and ‘100% inhibition’ Controls. 11-point dose response curves were plotted with XL-Fit software using a 4-Parameter Logistic Model (Sigmoidal Dose-Response Model) and the inhibitory concentrations that gave a 50 % réduction in mP (IC50) were determined. Results obtained using Method 1 are presented in Table 1 below; 1C of Mcl-l inhibition obtained 25 using Method 1 are not underlined.
Method 2: An 11 point serial dilution of each compound was prepared in DMSO and 2 μΐ
ORIGINAL
- 133transferred into fiat bottomed, low binding, 384-well plate (final DMSO concentration
%). 38 μΐ of buffer (20 mM NaiHPO^, ImM EDTA, 50 mM NaCl, pH 7.4), containing the Fluorescein labelled ligand (final concentration lOnM) and Mcl-l protein (final concentration 10 nM) was then added.
Assay plates were incubated ~2 hours at room température before FP was measured on a Biomek Synergy2 reader (Ex. 528 nm. Em. 640 nm, Cut off 510 nm) and mP unîts calculated. The binding of increasing doses of test compound was expressed as a percentage réduction in mP compared to a window established between 5 % DMSO only and ‘100 % inhibition’ Controls (50 μΜ unlabelled ligand). 11-point dose response curves were plotted with XL-Fit software using a 4-Parameter Logistic Model (Sigmoidal DoseResponse Model) and the inhibitory concentrations that gave a 50 % réduction in mP (lC5o) were determined. Results obtained using Method 2 are presented in Table 1 below; IÇ5o of Mcl-l inhibition obtained using Method 2 are underlined.
The results show that the compounds of the invention inhibit interaction between the Mcl-1 protein and the fluorescent peptide described hereinbefore.
EXAMPLE B: In vitro cytotoxicity
The cytotoxicity studies were carried out on the H929 multiple myeloma tuniour line.
The cells are distributed onto microplates and exposed to the test compounds for 48 hours.
The cell viability is then quantified by a colorimétrie assay, the Microculture Tetiazolium Assay (Cancer Res., 1987, 47, 939-942).
The results are expressed in IC50 (the concentration of compound that inhibits cell viability by 50 %) and are presented in Table 1 below.
The results show that the compounds of the invention are cytotoxic.
ORIGINAL
- 134Table 1: IÇ50 of Mcl-1 inhibition (fluorescence polarisation test) and of cvtotoxicitv for H929 cells
Note: IC50 of Mcl-1 inhibition obtained using Method 2 are underlined.
IC* (M) Mcl-1 FP IC* (Μ) MTT H929 IC* (M) Mcl-1 FP IC* (Μ) MTT H929
Example 1 3.8E-09 2.41E-08 Example 30 6.4E-06 ND
Example 2 6.0E-09 I.45E-08 Example 31 7.9E-07 ND
Example 3 1.7E-08 3.64E-07 Example 32 3.5E-06 ND
Example 4 2.9E-08 3.29E-07 Example 33 2.6E-07 ND
Example 5 1.5E-08 6.I9E-07 Example 34 6.4E-06 ND
Example 6 8.9E-09 ND Example 35 2.9E-07 ND
Example 7 1.IE-07 7.57E-07 Example 36 6.5E-06 ND
Example 8 6.6E-09 I.78E-08 Example 37 5.3E-07 ND
Example 9 8.6E-08 6.89E-08 Example 38 67% @50 uM ND
Example 10 I.8E-O5 ND Example 39 77,75% @ 50 uM ND
Example 11 3.4E-05 ND Example 40 8.6E-07 / 3.3E-08 ND
Example 12 5.6E-07 ND Example 41 1.3E-05 ND
Example 13 6.6E-07 ND Example 42 4.5E-Q7 ND
Example 14 I.2E-05 ND Example 43 66.9% @ 50 uM ND
Example 15 7.3E-06 ND Example 44 2.5E-06 ND
Example 16 1.8E-06 ND Example 45 I.8E-06 ND
Example 17 3.8E-06 ND Example 46 71% @ 50 uM ND
Example 18 3.1E-06 ND Example 47 1.IE-05 ND
Example 19 3.3E-06 ND Example 48 5.9E-06 ND
Example 20 64.8% @ 50 iiM ND Example 49 3.9E-08 ND
Example 21 8.7E-06 ND Example 50 65.85% @ 10 uM ND
Example 22 74.2% @ 50 uM ND Example 51 3.6E-07 / 5.5E-09 I.I0E-05
Example 23 6.8E-06 ND Example 52 1.6E-06 ND
Example 24 I.8E-05 ND Example 53 2.2E-08 2.53E-08
Example 25 9.IE-06 ND Example 54 1.2E-07 ND
Example 26 5.9E-06 ND Example 55 55.35% @ 10 μΜ ND
Example 27 3.3E-07 ND Example 56 4.7E-08 ND
Example 28 63.25% @ 50 uM ND Example 57 1.7E-07 ND
Example 29 8.5E-06 ND Example 58 51.9% @ 10 μΜ ND
ORIGINAL
IC» (M) Mcl-1 FP ICW(M) MTT H929 ICm(M) Mcl-1 FP IC» (M) MTT H929
Example 59 3.6E-O8 I.24E-06 Example 83 ND ND
Example 60 1.9E-08 5.68E-07 Example 84 5.45E-09 I.09E-08
Example 61 52.8% @ 10 μΜ ND Examplc 85 ND ND
Example 62 8.2E-07 ND Example 86 3.05E-08 3.59E-08
Example 63 1.7E-07 ND Examplc 87 ND ND
Example 64 7.4E-09 4.7IE-08 Example 88 ND ND
Examplc 65 1.0E-06 ND Examplc 89 ND ND
Example 66 1.6E-06 ND Examplc 90 ND ND
Examplc 67 I.4E-08 8.36E-08 Examplc 91 ND ND
Example 68 I.2E-06 ND Example 92 ND ND
Examplc 69 2.4E-08 I.04E-07 Example 93 ND ND
Examplc 70 13.55% @ 10 uM ND Examplc 94 ND ND
Example 71 5.02E-09 9.08E-09 Examplc 95 ND ND
Example 72 I.55E-08 3.2E-08 Examplc 96 ND ND
Example 73 ND ND Example 97a 55% @ 10 μΜ I.16E-05
Example 74 ND ND Examplc 97b 4.I0E-08 4.59E-07
Example 75 5.6 IE-07 7.55E-08 Examplc 98 ND ND
Example 76 1.34E-07 1.01 E-08 Example 99 ND ND
Examplc 77 ND ND Examplc 100 ND ND
Example 78 ND ND Example 101 ND ND
Examplc 79 ND ND Examplc 102 no curve >3.00E-05
Example 80 ND ND Examplc 103 ND ND
Example 81 ND ND Examplc 104 ND ND
Examplc 82 ND ND
ND: not determined
For partial inhibitors, the percentage fluorescence polarization inhibition for a given concentration of the test compound is indicated. Accordingly, 45.1% @10 μΜ means that 45.1% fluorescence polarization inhibition is observed for a concentration of test compound equal to 10 μΜ.
ORIGINAL
- 136EXAMPLE C: Quantification of the cleaved form of PARP in vivo
The ability of the compounds of the invention to induce apoptosis, by measuring cleaved PARP levels, is evaluated in a xenograft model of AMO-l multiple myeloma cells.
I.IO7 AMO-l cells are grafted sub-cutaneously into immunosuppressed mice (SC1D strain). 12 to 14 days after the graft, the animais are treated by intraveinous or oral routes with the various compounds. After treatment, the tumour masses are recovered and lysed, and the cleaved form of PARP is quantified in the tumour lysâtes.
The quantification is carried out using the Meso Scale Discovery (MSD) ELISA platform test, which specifically assays the cleaved form of PARP. It is expressed in the form of an activation factor corresponding to the ratio between the quantity of cleaved PARP in the treated mice divided by the quantity of cleaved PARP in the control mice.
The results (presented in Table 2 below) show that the compounds of the invention are capable of inducing apoptosis in AMO-l tumour cells in vivo.
Table 2: Quantification of the cleaved form of PARP in vivo
PARP fold PARP fold PARP fold
Example l I57.5 Example 8 55.4 Example 67 29.3
Example 2 216.3 Example 53 40.2 Example 72 15.7
EXAMPLE D: Anti-tumour activity in vivo
The anti-tumour activity of the compounds of the invention is evaluated in a xenograft model of AMO-l multiple myeloma cells.
IxlO7 AMO-l cells are grafted sub-cutaneously into immunosuppressed mice (SCID strain).
to 8 days after the graft, when the tumour mass has reached about 150 mm3, the mice are treated with the various compounds in a daily schedule (5-day treatment). The tumour mass is measured twice weekly from the start of treatment.
ORIGINAL
- I37The compound of the invention has anti-tumour activity (tumour régression) in the AMO-l multiple myeloma model with ΔΤ/C (qualification parameter of the activity of a product. which is measured by subtracting the médian tumor volume on the day of last treatment from the médian tumor volume on the day of fîrst treatment / tumour volume of the 5 untreated control group on the day of last treatment) of -27 %. The results obtained show that the compounds of the invention induce significant tumour régression during the treatment period.
EXAMPLE E: Pharmaceutical composition: Tablcts
1000 tablets containing a dose of 5 mg ofa compound selected from Examples l to 104..........5g
Wheat starch g
Maize starch.........................................................................................20 g
Lactose..........................„ τη
.. D\J
Magnésium stéarate......................... ?
Silica...................................................................,....................... ]g
Hydroxypropylcellulose.................................................,.................................... 2g
ORIGINAL

Claims (4)

1. Compound of formula (I):
♦ A represents the group
ΙΟ in which 1 is linked to the W group and 2 is linked to the phenyl ring, wherein:
- E represents a furyl, thienyl or pyrrolyl ring,
- X[, X3, X4 and X5 independently of one another represent a carbon atom or a nitrogen atom.
- X2 represents a C-R21 group or a nitrogen atom, and
- I ) means that the ring is aromatic, ♦ R] represents a halogen atom, a linear or branched (Ci-C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, a linear or branched (C|-C6)polyhaloalkyl group, a hydroxy group, a hydroxyiCt-Côjalkyl group, a linear or branched (C|-C6)alkoxy group, -S-(Ci-C6)alkyl, a cyano group, a nitro group, -alkyl(Co-C6)-NR[|Rii', -O-alkyl(Ci-C6)-NRHRj|’, -O-alkyl(Ct-C6)-R12, -C(O)-ORn, -O-C(O)-Rn, -C(O)-NRnRlt’, -NRh-C(O)-Rii\ -NRn-CiOj-ORn’, -alkyl(C|-C6)-NR||-C(O)-R|i’, -SO^NRuRji’,
ORIGINAL
-139-SO2-alkyl(Ci-C6), ♦ R?, R3, R4 and R5 independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (Ci-C^jalkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, a linear or
5 branched (C|-C6)polyhaloalkyl, a hydroxy group, a hydroxytCi-C^Jalkyl group, a linear or branched (Ci-C&)alkoxy group, a -S-(C]-C6)alkyl group, a cyano group, a nitro group, -alkyl(Co-C6)-NRi]R||’, -O-alkyl(CrC6)-NR||Rn’, -O-alkyl(Ci-C6)-Ri2, -C(0)-ORn, -O-C(O)-RU, -CiOj-NRnRn’, -NRn-CtOj-Rn’, -NR]|-C(O)-OR]|’, -alkyl(C|-C6)-NRn-C(O)-Rir, -SO2-NRiiR,r, or
IO -SO2-alkyl(Ci-C6), or the substituents of the pair (Rj, R2) form together with the carbon atoms carrying them an aromatic or non-aromatic ring composed of from 5 to 7 ring members, which may contaîn from l to 3 heteroatoms selected from oxygen, sulphur and nitrogen, it being understood that resulting ring may be substituted by from l to 2 15 groups selected from halogen, linear or branched (Ci-CôJalkyl,
-alkyl(Co-C6)-NRi]R||’, -NRi3Ri3’, -aIkyl(Co-C6)-Cyi or oxo, J ♦ R6 and R7 independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (Ci-Cô)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, a linear or branched
20 (Ci-C6)polyhaloalkyl, a hydroxy group, a linear or branched (Ci-Céjalkoxy group, a -S-(C]-C6)alkyl group, a cyano group. a nitro group, -alkylfCo-Cftj-NRijRn’, -O-Cyi, -alkyl(Co-C6)-Cyi, -alkenyl(C2-C6)-Cyi, -alkynyl(C2-C6)-Cyi, -O-alkyl(Ci-C6)-R,2, -C(O)-ORi], -O-C(O)-Rn, -CÎOJ-NRhR,,’, -NRirC(O)-R|i\ -NRirC(O)-ORir, -alkyl(C|-C6)-NRn-C(O)-Rii·, -SO2-NR||RH’,
25 -SO2-alkyl(CrC6), or the substituents of the pair (R6, R7), when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them an aromatic or nonaromatic ring composed of from 5 to 7 ring members, which may contain from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen, it being understood thaï 30 resulting ring may be substituted by a group selected from a linear or branched (Ci-Côjalkyl group, -NRi3Ri3’, -alkyl(Co-C6)-Cyi or an oxo.
♦ W represents a -CH2- group, a -NH- group or an oxygen atom,
ORIGINAL
- MO- ♦ Rs represents a hydrogen atom, a linear or branched (Ci-C8)alkyl group, a -CHRaRh group, an aryl group, a heteroaryl group, an arylalkyKCj-Cô) group, or a heteroarylalkyl(Ci-C6) group, ♦ R9 represents a hydrogen atom, a linear or branched (CrC6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, -Cy2, -alkyl(Ci-C(,)-Cy2, -alkenyl(C2-Cé)-Cy2, -alkynyl(C2-C6)-Cy2, -Cy2-Cy3, -alkynyl(C2-C(,)-O-Cy2, -Cy2-alkyl(Co-C6)-0-alkyI(Co'C6)-Cy3, a halogen atom, a cyano group, -C(O)-Rl5, or -C(O)-NR|5R|5’, ♦ Rio represents a hydrogen atom, a linear or branched (C|-C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-C6)alkynyl group, an arylalkyl(C|-C6) group, a cycloalkylalkylfCi-Cù) group, a linear or branched (Ci -C6)polyhaloalkyl, -alkyl(C i -C6)-O-Cy4, or the substituents of the pair (R9, Rio), when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them an aromatic or nonaromatic ring composed of from 5 to 7 ring members, which may contain from l to 3 heteroatoms selected from oxygen, sulphur and nitrogen, ♦ Ru and Ru’ independently of one another represent a hydrogen atom, a linear or branched (C|-C6)alkyl group, or the substituents of the pair (Ru, Ru’) form together with the nitrogen atom carrying them an aromatic or non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from l to 3 heteroatoms selected from oxygen, sulphur and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a hydrogen atom, or a linear or branched (C|-C6)alkyl group, ♦ R]2 represents -Cy5, -Cy5-alkyl(C0-C6)-0-alkyl(Co-C6)-Cy6,
-Cy5-alkyi(C0-C6)-Cy6, -Cys-alkyKCo-Càj-NRn-alkyKCo-Côj-Cyf,,
-Cy5-Cy6-O-alkyl(CO-C6)-Cy7, -CfOj-NRnRn’, -NRuRh', -OR,., -NRii-C(O)-RH’, -O-alkyl(C|-C6)-ORn, -SO2-Rn, -C(O)-ORh, or -NH-C(O)-NH-Rn, ♦ Ris, Ri3\ Ris and R15’ independently of one another represent a hydrogen atom, or an optionally substituted linear or branched (Cj-Céjalkyl group, ♦ Rm represents a hydrogen atom, a hydroxy group, or a hydroxy(C]-C6)alkyl group.
ORIGINAL
- I4l- ♦ Rji represents a hydrogen atom, a halogen atom, a linear or a branched (C|-C6)alkyl group, or a cyano group, ♦ Ra represents a hydrogen atom or a linear or branched (C|-C6)alkyi group, ♦ Rb represents a -O-C(O)-O-Rc group, a -O-QOj-NRcRç’ group, or a -O-P(O)(ORe)2
5 group, ♦ Rc and Rf independently of one another represent a hydrogen atom, a linear or branched (CrC8)alkyI group, a cycloalkyl group, a (C|-C6)alkoxy(CrC6)alkyl group, a (C|-C6)alkoxycarbonyl(Ci-C6)alkyl group, or the substituents of the pair (Rc, Rc’) form together with the nitrogen atom 10 carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from 1 to 3 heteroatoms selected from oxygen and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a linear or branched (Ci-Cé)alkyl group, ♦ Cyi, Cy2, Cyj, Cy4, Cy5, Cy6 and Cy7 independently of one another, represent a
15 cycloalkyl group, a heterocycloalkyl group, an aryl group, or a heteroaryl group, ♦ n is an integer equal to 0 or 1, it being understood that:
- “aryl” means a phenyl, naphthyl, biphenyl, indanyl or indenyl group, heteroaryl” means any mono- or bi-cyclic group composed of from 5 to 10 ring 20 members, having at least one aromatic moiety and containing from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen, cycloalkyl means any mono- or bi-cyclic non-aromatic carbocyclic group containing from 3 to 10 ring members,
- heterocycloalkyl” means any mono- or bi-cyclic non-aromatic carbocyclic group
25 containing from 3 to 10 ring members, and containing from 1 to 3 heteroatoms selected from oxygen, sulphur and nitrogen. which may include fused, bridged or spiro ring Systems, it being possible for the aryl, heteroaryl, cycloalkyl and heterocycloalkyl groups so defined and the alkyl, alkenyl, alkynyl, alkoxy groups, to be substituted by from 1 to 4 30 groups selected from optionally substituted linear or branched (C|-C6)alkyl, optionally
ORIGINAL
- 142substituted linear or branched (C2-C6)alkenyl, optionally substituted linear or branched (C2-C6)alkynyl, optionally substituted linear or branched (C|-C6)alkoxy, optionally substituted (Ci-Cé)alkyl-S-, hydroxy, oxo (or jV-oxide where appropriate), nitro, cyano, -C(O)-OR’, -O-C(O)-R\ -C(O)-NR’R”, -O-C(O)-NR’R”, -NR’R”, -(C=NR’)-OR”, -O-P(O)(OR’)2, -O-P(O)(O*M+)2, linear or branched (Ci-Ce)polyhaloalkyl, trifluoromethoxy, halogen, or an aldohexose of formula:
in which each R' îs independent;
it being understood that R’ and R” independently of one another represent a hydrogen atom or an optionally substituted linear or branched (CrC6)alkyl group, and M+ represents a pharmaceutically acceptable monovalent cation.
their enantiomers, diastereoisomers and atropisomers, and addition salts thereof with a pharmaceutically acceptable acid or base.
2. Compound of formula (!) according to claim l, wherein:
♦ Rj and R2 independently of one another represent a halogen atom. a linear or branched (Ci-C6)alkyl group, a hydroxy group, a linear or branched (C|-C6)alkoxy group, or the substituents of the pair (Rb R2) form together with the carbon atoms carrying them an aromatic ring composed of from 5 to 7 ring members, which may contain from 1 to 3 nitrogen atoms,
ORIGINAL
- 143- ♦ Rj represents a hydrogen atom, a halogen atom, a linear or branched (C|-C&)alkyl group, a hydroxy group, a linear or branched (Ci-Cft)alkoxy group, or -O-alkyKC.-CôJ-NRnRn’, ♦ R4 and R5 independently of one another represent a hydrogen atom, a halogen
5 atom, a linear or branched (Ci-Côfalkyl group, a hydroxy group, a linear or branched (C|-C&)alkoxy group, ♦ R6 and R7 independently of one another represent a hydrogen atom, a halogen atom, a linear or branched (Ci-Cô)alkyl group, a linear or branched (Ci-C6)polyhaloalkyl group, a hydroxy group, a linear or branched (Ci-Cé)alkoxy
IO group, a cyano group, a nitro group, -alkyl(Co-C6)-NR||R]|’, -alkyl(Co-C6)-Cyi,
-O-alkyl(Cl-C6)-Ri2, or -C(O)-NR| [R| f, ♦ Rs represents a hydrogen atom, a linear or branched (Ci-Cg)alkyl group, or a -CHRaRb group, ♦ R9 represents a hydrogen atom, a linear or branched (Ci-C6)alkyl group, a linear or
15 branched (C2-Cô)alkenyl group, a linear or branched (C2-C6)alkynyl group, -Cy2, or a halogen atom, ♦ Rio represents a hydrogen atom, a linear or branched (C i -Côjalkyl group, a linear or branched (C2-Cô)alkenyl group, a linear or branched (C2-C6)alkynyl group, an arylalkyl(Ci-Cô) group, a cycloalkylalkyl(C|-C6) group, a linear or branched
20 (Ci-C6)polyhaioalkyl, or -alkyl(Ci-C6)-O-Cy4, or the substituents of the pair (R9, Rio) when grafted onto two adjacent carbon atoms, form together with the carbon atoms carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain from l to 3 heteroatoms selected from oxygen, sulphur and nitrogen,
25 ♦ R|| and Ru' independently of one another represent a hydrogen atom, a linear or branched (Ci-C&)alkyl group, or the substituents of the pair (Ru, Ru’) form together with lhe nitrogen atom carrying them a non-aromatic ring composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from l to 3 heteroatoms selected from 30 oxygen and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a linear or branched (C i -Côjalkyl group, ♦ Rj2 represents -Cy5 or -Cy5-alkyl(C(i-C6)-Cy6,
ORIGINAL
- I44♦ W represents a -NH- group or an oxygen atom, it being possible for the aryl, heteroaryl, cycloalkyl and heterocycloalkyl groups so defined and the alkyl, alkenyl, alkynyl, alkoxy groups, to be substituted by from i to 4 groups selected from optionally substituted linear or branched (CrC6)alkyl, optionally 5 substituted linear or branched (C]-C6)alkoxy, hydroxy, oxo (or N-oxide where appropriate), -C(O)-OR’, -C(O)-NR’R”, -O-C(O)-NR’R”, -NR’R”, -O-P(O)(OR’)2, -O-P(O)(OM )2, linear or branched (Ci-Côjpolyhaloalkyl, halogen, or an aldohexose of formula:
10 in which each R’ is independent;
it being understood that R’ and R” independently of one another represent a hydrogen atom or an optionally substituted linear or branched (Ci-C6)alkyl group and M’ represents a pharmaceutically acceptable monovalent cation.
3. Compound of formula (I) according to claim l, wherein n is an integer equal to l.
15 4. Compound of formula (!) according to claim l, which is compound of formula (l-a);
ORIGINAL
- 145-
(I-a) wherein Rj, R2, R3, R4, R5, R<„ R7, Rg, R9, Ri4, Xb X2, X3 and W are as defined in claim l.
5. Compound of formula (I) according to claim L which is compound of formula (I-b):
(I-b) wherein R], R2, R3, R4, R5, R*, R7, Rs, R9, Ri4, Xb X2, X3 and W are as defined in claim l.
6. Compound of formula (I) according to claim l, which is compound of formula (I-c):
ORIGINAL
- I46-
wherein Rt, R2, R3, R4, R5, Rr„ R7, Rs, R9, Rio, Ri4, Xi, X2, X3 and W are as defined in claim l.
7. Compound according to claim 6, wherein Rio represents hydrogen; methyl; isopropyl;
5 2,2,2-trifluoroethyl; benzyl; 4-methoxybenzyl; phenethyl; 3-phenyl-propyl;
cyciopropylmethyl; cyclopentylethyl; naphthalen-l-ylmethyl; 2-(naphthalen-lyloxy)ethyl; but-2-yn-l-yl; prop-2-en-lyl; or but-3-en-l-yl.
8. Compound of formula (!) according to claim l, which is compound of formula (I-d):
IO wherein Rb R2, R3, R4, R R/,, R7. Rs, R9, Rio, R14, X|, X2, X3 and W are as defined in
ORIGINAL
- 147claim I.
9. Compound according to claim 8, wherein Rl0 represents a hydrogen atom or a halogen atom.
10. Compound of formula (I) according to claim 1, which is compound of formula (I-e):
(i-e) wherein R|, R2, R3, R4, R5, R(,, R7, Rs, Rg, R14, Xb X2, X3 and W are as defined in claim 1.
11. Compound according to claim 1, wherein at least one of the groups selected from R2, R3, R4 and R5 does not represent a hydrogen atom.
10 12. Compound according to claim 1, wherein Rj4 represents a hydrogen atom.
13. Compound according to claim 1, wherein R2j represents a hydrogen atom, a fluorine atom, a methyl group or a cyano group.
14. Compound according to claim 1, wherein R| represents a linear or branched (C|-C&)alkyl group or a halogen atom.
15 15. Compound according to claim 1, wherein R2 represents a linear or branched (Cj-Côjalkoxy group, a hydroxy group or a halogen atom.
ORIGINAL
- I48-
16. Compound according to claim 1, wherein R3 represents a hydrogen atom, a hydroxy group, a linear or branched (C|-C6)alkoxy group or -O-alkyl(Ci-C6)-NR||Rn’.
17. Compound according to claim 1, wherein Rq and R5 represent a hydrogen atom.
18. Compound according to claim 1, wherein
represents
wherein Rj t and R] 1 ’ are as defined in claim 1.
19. Compound according to claim 1, wherein the substituents of the pair (Ri, R5) are identical and the substituents of the pair (R2, Rf) are identical.
20. Compound according to claim 1, wherein Rb represents a hydrogen atom, an optionally substituted linear or branched (Ci-C6)alkoxy group or a -O-alkyI(CrC6)-RI2 group.
21. Compound according to claim 1, wherein R7 represents a hydrogen atom.
22. Compound according to claim 1, wherein
ORIGINAL
- 149wherein Ru is as defined in claim l.
23. Compound according to claim l, which is compound of formula (I-g):
wherein R[, Rf>. R7, R8, R9, Rl0, Ru, Ru’, R|4, X|, X2, X3j X4, X5) W and E are as 5 defined in claim l.
24. Compound according to claim l, wherein R8 represents a hydrogen atom, a -CHRaRb group, an optionally substituted linear or branched (C|-C8)alkyl group, or a heteroarylalkyl(C|-C6) group.
25. Compound according to claim l, wherein R9 represents a hydrogen atom, a halogen
Ιθ atom, a linear or branched (Ci-C6)alkyl group, a linear or branched (C2-C6)alkenyl group, a linear or branched (C2-Cb)alkynyl group, an aryl group or a heteroaryl group.
26. Compound according to claim l, wherein Rj[ and Rn‘ independently of one another represent a linear or branched (Ci-Csjalkyl group, or the substituents of the pair (Ru, R|i’) torm together with the nitrogen atom carrying them a non-aromatic ring
15 composed of from 5 to 7 ring members, which may contain in addition to the nitrogen atom from l to 3 heteroatoms selected from oxygen, sulphur and nitrogen, it being understood that the nitrogen in question may be substituted by a group representing a hydrogen atom, a linear or branched (Cj-C6)alkyl group.
ORIGINAL
- ISO-
27. Compound according to daim l, wherein Rl2 represents -Cys or -Cy5-alkyl(Co-C6)-Cy6.
28. Compound according to claim 27, wherein Cys represents a heteroaryl group.
29. Compound according to claim 27, wherein Cyé represents a phenyl group.
5 30. Compounds according to claim 27 wherein
Ri2 represents
in which p is an integer equal to 0 or l and Rl6 represents a hydrogen atom, a hydroxy group, an optionally substituted linear or branched (C|-C6)alkyl group, a linear or branched (CrC6)alkoxy group, a -O-(CHR]7-CHRi8-O)q-R’ group, a -O-P(O)(OR’)2 group, a -O-P(O)(QM+)2 group, a -O-C(O)-NR|9R20 group, a di(C|-Cé)alkylamino(C|-C6)alkoxy group, a halogen atom, or an aldohexose of formula:
in which each R' is independent;
15 it being understood that:
♦ R’ represents a hydrogen atom or a linear or branched (Cj-C6)alkyl group, ♦ R|7 represents a hydrogen atom or a (C|-C6)aikoxy(Ci-C6)alkyl group.
ORIGINAL
- 151- ♦ Ri8 represents a hydrogen atom or a hydroxyiCj-CôJalkyl group, ♦ Ri9 represents a hydrogen atom or a (Ci-C6)alkoxy(CrC6)alkyl group, ♦ R20 represents a (Cl-C6)alkoxy(Cl-C6)alkyl group, a -(CH2)r-NRHR||’ group or a -(CH2)r-O-(CHRi7~CHR]8-O)q-R’ group,
5 ♦ q is an integer equal to l, 2 or 3 and r is an integer equal to 0 or i, ♦ M+ represents a pharmaceutically acceptable monovalent cation.
31. Compounds according to claim 30, wherein the aldexose is D-mannose,
32. Compounds according to claim l, which are:
(2Æ)-2-{[5-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl }-6-(4]0 fluorophenyl)furo[2,3-^pyrimidin-4-yI]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid; (2Æ)-2-{[5-{3-chloro-2-ethyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-6-(4fluorophenyl)furo[2,3-i/]pyrimidin-4-yl]oxy} -3-(2- {[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
15 - V-[(5SiJ)-5-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-6-(4fluorophenyl)furo[2,3-i/]pyrimidin-4-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy}-D-phenylalanine;
- (2Â)-2-{[(3xSj-3-{3-chloro-2-methyl-4-[2-(4-methyipiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl )-1 -benzothiophen-4-yl]oxy} -3-( 2-{[2-(2methoxyphenyl)pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
- (2/?)-2-{[(3S<I)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin4-yl]methoxy}phenyl)propanoic acid;
(2 J?)-2-{ [(35^)-3- {3-chloro-2-methyI-4-[2-(4-methyipiperazin-1 -yl )ethoxy]phenyl} 25 6-fluoro-2-(4-fluorophenyl)-l-benzofuran-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
(2 R)-2- {[3- {( jSa)-3-chloro-2-methy l-4-[2-(4-methylpiperazin-1 -y 1 )ethoxy]phenyl} 2-(4-fluorophenyl)-1 -methyl-1 /f-indol-4-yl]oxy}-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy}phenyl)propanoic acid;
ORIGINAL
- 152(2^)-2-{[(3S, a)-3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}2-(4-fluorophenyl)thieno[2,3-6]pyridin-4-yl]oxy)-3-(2-{[2-(2-methoxyphenyl) pyrimidin-4-yl]methoxy} phenyl)propanoic acid;
- (2Æ)-2-[5-[3-chloro-2-methyl-4-[2-(4-methylpiperazin-1 -yl)ethoxy]phenyl]-6-(4fluorophenyl)-7-methyl-pyrrolo[2,3-i/]pyrimidin-4-yl]oxy-3-[2-[[2-(2-methoxy phenyl)pyrimidin-4-yl]methoxy]phenyl]propanoic acid;
- l-[(dimethylcarbamoyl)oxy]ethyl (2Æ)-2-{[(3Sa)-3-{3-chloro-2-methyl-4-[2-(4methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fluorophenyl)thieno[2,3-6]pyridin-4yl]oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy}phenyl)propanoate;
- I -[(ethoxycarbonyl)oxy]ethyl (2R)-2-{[(35'a)-3-{3-chloro-2-methyl-4-[2-(4methylpiperazin-l-yl)ethoxy]phenyl}-2-(4-fiuorophenyl)thieno[2,3-ô]pyridin-4yl]oxy}-3-(2-{[2-(2-methoxyphenyl)pyrimidin-4-yl]methoxy}phenyl)propanoate;
- A43-{3-chloro-2-melhyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl}-2-(4fluorophenyl)thieno[2,3-6]pyridin-4-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy }-D-phenylalanine;
- jV-[3-{3-chloro-2-methyl-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyI}-2-(4fluorophenyl)thieno[3,2-c]pyridin-4-yl]-2-{[2-(2-methoxyphenyl)pyrimidin-4-yl] methoxy Jphenylalanine;
- 2-{[(3Æa)-3-{3-chloro-2-methyI-4-[2-(4-methylpiperazin-l-yl)ethoxy]phenyl )-2-(4fluorophenyl)iniidazo[l,2-c]pyrimidin-5-yl]oxy)-3-(2-{[2-(2-methoxyphenyI) pyrimidin-4-yl]methoxy)phenyl)propanoic acid.
33. Process for the préparation of a compound of formula (I) according to claim l, characterised in that there is used as starting material the compound of formula (ll-a):
7 ^A\7 (Il-a) wherein Zi represents bromine or iodine, Z2 represents chlorine, bromine or hydroxy, and A is as defined for formula (I) in which l is linked to the Z? group and 2 is linked to the Zi group, which compound of formula (II-a) is subjected to coupling with a compound of formula (III):
ORIGINAL
- 153-
(III) wherein R6, R7, Rl4, W and n are as defined for formula (l), and Alk represents a linear or branched (CrC6)aikyl group, to yield the compound of formula (IV):
(IV) wherein K,. R7, Ri4, A, W and n are as defined for formula (I), and Z] and Alk are as defined before, compound of formula (IV) which is further subjected to coupling with compound of formula (V):
(V) wherein Rt, R2, R3, R4 and R5 are as defined for formula (I), and RB1 and RB2 represent a hydrogen atom, a linear or branched (C,-C6) alkyl group, or RB, and RB2 form with the oxygen carrying them an optionally methylated ring,
ORIGINAL
- I54to yield the compound of formula (VI):
(VI) wherein Rls R2, R3 R-ι, Rs, R<„ R7, Ru, A, W and n are as defined for formula (I) and Alk îs as defined before, the Alk-O-C(O)- ester fonction of which compound of formula (VI) is hydrolyzed to yield the carboxylic acid, which may optionally be reacted with an alcohol of formula Rs’-OH or a chlorinated compound of formula Rs ?-Cl wherein Rs’ represents a linear or branched (Ci-Cs)alkyl group, a -CHRaRb group, an aryl group, a heteroaryl group, an arylalkyl(CrC6) group, or a heteroarylalkyl(Cj-C6) group, Ra and Rb are as defined for formula (I), to yield the compound of formula (l), which may be purified according to a conventional séparation technique, which is converted, if desired, into its addition salts with a pharmaceutically acceptable acid or base and which is optionally separated into its isomers according to a conventional séparation technique, it being understood thaï at any moment considered appropriate during the course ofthe process described above, some groups (hydroxy. amino...) of the starting reagents or of the synthesis intermediates can be protected, subsequently deprotected and functionalized, as required by the synthesis.
34. Process for the préparation of a compound of formula (I) according to claim l, characterised in that there is used as starting material the compound of formula (ll-b):
7 (Π-b)
Z.4 Z,3
ORIGINAL
- 155wherein Z3 represents iodine, Z4 represents chlorine, hydroxy, and A is as defined for formula (I) in which 1 is linked to the Z4 group and 2 is linked to the Z3 group, which compound of formula (Π-b) is subjected to coupling with a compound of formula (V):
(V) wherein Rb R2, R3, R4 and R3 are as defined for formula (I), and RB| and RB2 represent a hydrogen atom, a linear or branched (C i-C6) alkyl group, or RBI and RB2 form with the oxygen carrying them an optionally methylated ring, to yield the compound of formula (VII):
(Vil) wherein Rb R2, R3 R|, R5 and A are as defined for formula (I), and Z4 is as defined before.
compound of formula (VII) which is further subjected to coupling with compound of formula (III):
ORIGINAL
- 156-
(ΠΙ) wherein Rô, R7, Rf4, W and n are as defined for formula ([), and Alk represents a linear or branched (Ci-C^alkyl group, to yield the compound of formula (VI):
(VI) wherein Rls R2, R3 R_(, R5, R6, R7, Ri4, A, W and n are as defined for formula (I) and Alk is as defined before, the Alk-O-C(O)- ester fonction of which compound of formula (VI) is hydrolyzed to yield the carboxylic acid, which may optionally be reacted with an alcohol of formula 10 Rs'-OH or a chlorinated compound of formula R8’-Cl wherein R8’ represents a linear or branched (C|-C8)alkyl group, a -CHRaRi, group, an ary] group, a heteroaryl group, an aryIalkyl(C|-C6) group, or a heteroarylalkylfCi-Cô) group, Ra and Rb are as defined for formula (I), to yield the compound of formula (l), which may be purified according to a 15 conventional séparation technique, which is converted, if desired, into its addition salts with a pharmaceutically acceptable acid or base and which is optionally separated into its isomers according to a conventional séparation technique.
ORIGINAL
- 157it being understood that at any moment considered appropriate during the course of the process described above, some groups (hydroxy, amino...) of the starting reagents or of the synthesis intermediates can be protected, subsequently deprotected and functionalized, as required by the synthesis.
35. Pharmaceutical composition comprising a compound of formula (I) according to any one of daims l to 32 or an addition sait thereof with a pharmaceutically acceptable acid or base in combination with one or more pharmaceutically acceptable excipients.
36. Pharmaceutical composition according to claim 35 for use as pro-apoptotic agents.
37. Pharmaceutical composition according to claim 36 for use in the treatment of cancers and of auto-immune and immune System diseases.
38. Pharmaceutical composition according to claim 37 for use in the treatment of cancers of the bladder, brain, breast and utérus, chronic lymphoid leukaemias, cancer of the colon, œsophagus and liver, lymphoblastic leukaemias, acute myeloid leukaemias, lymphomas, melanomas, malignant haemopathies, myelomas, ovarian cancer, nonsmall-cell lung cancer, prostate cancer, pancreatîc cancer and small-cell lung cancer.
39. Use of a pharmaceutical composition according to claim 35 in the manufacture of médicaments for use as pro-apoptotic agents.
40. Use of a pharmaceutical composition according to claim 35 in the manufacture of médicaments for use in the treatment of cancers and of auto-immune and immune System diseases.
4L Use of a pharmaceutical composition according to claim 35 in the manufacture of médicaments for use in the treatment of cancers of the bladder, brain, breast and utérus, chronic lymphoid leukaemias, cancer of the colon, œsophagus and liver, lymphoblastic leukaemias, acute myeloid leukaemias, lymphomas, melanomas, malignant haemopathies, myelomas, ovarian cancer, non-small-cell lung cancer,
ORIGINAL
- 158prostate cancer, pancreatic cancer and small-cell lung cancer.
42. Compound of formula (I) according to any one of daims l to 32, or an addition sait thereof with a pharmaceuticaily acceptable acid or base, for use in the treatment of cancers of the bladder, brain, breast and utérus, chronic lymphoid leukaemias, cancer of the colon, œsophagus and iiver, lymphoblastic leukaemias, acute myeloid leukaemias, lymphomas, melanomas, malignant haemopathies, myelomas, ovarian cancer, non-small-cell lung cancer, prostate cancer, pancreatic cancer and small-cell lung cancer.
43. Use of a compound of formula (I) according to any one of daims l to 32, or an addition sait thereof with a pharmaceuticaily acceptable acid or base, in the manufacture of médicaments for use in the treatment of cancers of the bladder, brain, breast and utérus, chronic lymphoid leukaemias, cancer of the colon, œsophagus and liver, lymphoblastic leukaemias, acute myeloid leukaemias, lymphomas, melanomas, malignant haemopathies, myelomas, ovarian cancer, non-small-cell lung cancer, prostate cancer, pancreatic cancer and small-cell lung cancer.
44. Combination of a compound of formula (I) according to any one of daims l to 32 with an anti-cancer agent selected from genotoxic agents, mitotic poisons, anti-metabolites, protéasome inhîbitors, kinase inhibitors and antibodies.
45. Pharmaceutical composition comprising a combination according to daim 44 in combination with one or more pharmaceuticaily acceptable excipients.
46. Combination according to daim 44 for use in the treatment of cancers.
47. Use of a combination according to claim 44 in the manufacture of médicaments for use in the treatment of cancers.
48. Compound of formula (I) according to any one of daims l to 32 for use in the treatment of cancers requiring radiotherapy.
OA1201700513 2015-06-23 2016-06-22 New bicyclic derivatives, a process for their preparation and pharmaceutical compositions containing them. OA18891A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
FR1555750 2015-06-23

Publications (1)

Publication Number Publication Date
OA18891A true OA18891A (en) 2019-09-30

Family

ID=

Similar Documents

Publication Publication Date Title
US10618909B2 (en) Bicyclic derivatives, a process for their preparation and pharmaceutical compositions containing them
CA2990089C (en) New aminoacid derivatives, a process for their preparation and pharmaceutical compositions containing them
AU2016282836B2 (en) New hydroxyester derivatives, a process for their preparation and pharmaceutical compositions containing them
KR102620976B1 (en) Novel hydroxy acid derivatives, methods for producing them and pharmaceutical compositions containing them
JP6871275B2 (en) New ammonium derivative, process for preparing it and pharmaceutical composition containing it
OA18891A (en) New bicyclic derivatives, a process for their preparation and pharmaceutical compositions containing them.
BR112017027470B1 (en) BICYCLIC DERIVATIVES, PROCESS FOR THEIR PREPARATION, PHARMACEUTICAL COMPOSITIONS CONTAINING THEM, COMBINATION AND USE THEREOF
OA18890A (en) New aminoacid derivatives, a process for their preparation and pharmaceutical compositions containing them
OA17201A (en) Tris(hetero)arylpyrazoles and use thereof.
OA19084A (en) New piperidinyl derivatives, a process for their preparation and pharmaceutical compositions containing them.