[go: up one dir, main page]

WO2025219579A1 - Klhdc2 (kelch domain-containing protein 2) ligase ligands - Google Patents

Klhdc2 (kelch domain-containing protein 2) ligase ligands

Info

Publication number
WO2025219579A1
WO2025219579A1 PCT/EP2025/060749 EP2025060749W WO2025219579A1 WO 2025219579 A1 WO2025219579 A1 WO 2025219579A1 EP 2025060749 W EP2025060749 W EP 2025060749W WO 2025219579 A1 WO2025219579 A1 WO 2025219579A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
heterocycloalkyl
compound
unsubstituted
chr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/EP2025/060749
Other languages
French (fr)
Inventor
Michał BIŚTA
Sylvain Cottens
Katarzyna Dudek
Daria GOŁĘBIOWSKA
Arnaud MATHIEU
Ziemowit Pokładek
Bartosz WOŹNIAK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Captor Therapeutics SA
Original Assignee
Captor Therapeutics SA
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 Captor Therapeutics SA filed Critical Captor Therapeutics SA
Publication of WO2025219579A1 publication Critical patent/WO2025219579A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • KLHDC2 KELCH DOMAIN-CONTAINING PROTEIN 2
  • the present invention relates to compounds which can bind to KLHDC2 (Kelch Domain-Containing Protein 2) E3 ubiquitin ligase with high-affinity, and bifunctional degraders containing such compounds.
  • KLHDC2 Kelch Domain-Containing Protein 2
  • E3 ubiquitin ligase with high-affinity and bifunctional degraders containing such compounds.
  • TPD protein degradation
  • CRBN cereblon
  • VHL von Hippel-Lindau
  • CRBN and VHL ligands have sparked the most interest due to the availability of chemical probes with favorable physiochemical properties and their well-studied mechanism of target engagement.
  • both ligases have shown high efficiency and versatility in degrading proteins in the cytoplasm and nucleus.
  • the pharmacological intervention by using the CRBN and VHL ligands is associated with certain risks, such as impairment of developmental processes and hematopoiesis in the case of CRBN and dysregulation of cell survival and angiogenesis in the case of VHL.
  • cells may acquire resistance to CRBN ligands due to mutations in the ligase gene and a broad expression profile of CRBN can lead to unexpected side effects. Therefore, the employment of other E3s for this purpose could expand the therapeutic opportunities offered by TPD by broadening a substrate range, increasing efficacy, and evading drug resistance.
  • KLHDC2 The Kelch-like family member 2
  • DesCEND mechanism A specific degron, or in the other words, destruction motif, recognized by the KLHDC2 contains a diglycine at the C terminus. This kind of sequence has been found in several substrates such as early-terminated selenoproteins (SelK and SelS) and several full-length proteins (Rusnac et al., 2018). Importantly, KLHDC2 emerged as a potent degrader in the proteome-scale induced proximity screens performed by Poirson et al.
  • KLHDC2 AdPROM screen performed by Roth et al. (2022).
  • the utility of KLHDC2 to degrade proteins of interest was shown by using bifunctional compounds based on a degron peptide conjugated with chloroalkane (Roth et al. ,2022) or a promiscuous kinase inhibitor (Kim et al., 2022).
  • a degron peptide was utilized to demonstrate a proof-of-concept, it is poorly applicable to act as an actual drug due to its high molecular weight and related bioavailability issues.
  • Compounds disclosed herein are characterized by a small size with favorable physicochemical properties and can be used as building blocks of bifunctional degraders for the recruitment of KLHDC2 and degradation of the protein of interest. Furthermore, two distinct classes of compounds have been developed: ligands incorporating carboxylic groups and their corresponding prodrugs designed to enhance cellular membrane permeability.
  • X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X 2 is N, then X 3 is CR 1 ; and when X 3 is N, then X 2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH 2 ;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl) 2 , - NH(alkyl), -NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycl
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2, -S(O) 2 NR 9 2, - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH)2, -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 -P(O)(OR P ) 2 , -C(O)O(CH 2 ) P NMe 2 , -C(O)O(CH 2 ) P NHMe, -C(O)OCH 2 CH(OH)CH 2 OH, - C(O)OCH2CH 2 CI ⁇ /le2OH, and ⁇ (OjOCHjCHjSOjIVI
  • a bifunctional protein degrader compound comprising a compound as defined above.
  • a bifunctional protein degrader compound comprising a compound of formula (la'): wherein:
  • X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X 2 is N, then X 3 is CR 1 ; and when X 3 is N, then X 2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH 2 ;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl) 2 , - NH(alkyl),- NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl),
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2, -S(O) 2 NR 9 2, - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 , -P(O)(OR P ) 2 , -C(O)O(CH 2 ) P NMe 2 , -C(O)O(CH 2 ) P NHMe, -C(O)OCH2CH(OH)CH 2 OH, - C(O)OCH2CH 2 CI ⁇ /le2OH, and -C(O)OCH2CH
  • R 19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R 19 ; and wherein:
  • M is O, S or NH, or is absent; indicates attachment to R 18 of the linker;
  • R 11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
  • R 12 is H or Me;
  • L' is H, alkyl, benzyl, acetyl or pivaloyl; or
  • X 4 and X 5 are each independently N or CH;
  • X 6 is N or CH
  • R 30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R 40 , or -C(O)R 40 ;
  • R 31 is H, -OMe, -heteroaryl, -heteroaryl-R 40 or R 40 ;
  • R 32 is H, unsubstituted C1-C6 alkyl
  • R 33 is -N(C1-C6 alkyl) 2 , -NH(C1-C6 alkyl), -NH(aryl), or R 40 ;
  • R 34 is -Me or -C(O)R 40 ;
  • R 40 is a bond connected to R 18 of the linker, wherein the [Target protein binding moiety] contains a single R 40 ; or wherein
  • X 7 is N or CH
  • R 35 is -heterocycloalkyl-R 40 , or R 40 ;
  • R 36 is H or -OMe; and R 40 is a bond connected to R 18 of the linker, wherein the [Target protein binding moiety] contains a single R 40 ; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
  • X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X 2 is N, then X 3 is CR 1 ; and when X 3 is N, then X 2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH 2 ;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl) 2 , - NH(alkyl),- NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl),
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2, -S(O) 2 NR 9 2, - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 , -P(O)(OR P ) 2 , -C(O)O(CH 2 ) P NMe 2 , -C(O)O(CH 2 ) P NHMe, -C(O)OCH2CH(OH)CH 2 OH, - C(O)OCH2CH 2 CI ⁇ /le2OH, and -C(O)OCH 2 CH
  • R 19 is a bond connected to R 14 of the linker, wherein formula (la') contains a single R 19 ; and wherein:
  • R 14 is -Ci-6 alkyl, -C 2.6 alkenyl, -C 2.6 alkynyl, C1-6 alkyl-N(C1-6 alkyl)-, -C(O)-, -SO 2 - or is absent
  • R 15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, Ci.g alkyl-NH-, -Ci.g alkyl-N(Ci-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
  • R 16 is -Ci-6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH 2 -C(O)-, -CH 2 -C(O)-NH-, -CH 2 -C(O)O- or is absent
  • R 17 is -CH 2 (C 2 H4-O)y, (C 2 H 4 -O) X , (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
  • R 18 is -Ci.g alkyl, cycloalkyl
  • R 18 is -C1-6 alkyl-NH-, cycloalkyl-NH, -CH 2 -NH-C(O)-NH-, heterocycloalkyl, heterocycloalkyl-NH, or is absent.
  • the present invention also provides a pharmaceutical composition comprising a compound of the invention.
  • the present invention also provides a compound or pharmaceutical composition of the invention, for use in medicine.
  • the present invention also provides a method of treating a disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or pharmaceutical composition of the invention.
  • alkyl is intended to include both linear and branched alkyl groups, both of which either may be unsubstituted, or may be substituted by one or more additional groups.
  • the alkyl group is an unsubstituted alkyl group.
  • the alkyl group is substituted by one or more groups selected from -OH, -OR W , -NH2, -NHR W , -NR W 2, -SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkyl group is a C1-C12 alkyl, a C1-C10 alkyl, a Ci-Cg alkyl, a Ci-Cg alkyl, or a C1-C4 alkyl group.
  • the alkyl group is a linear alkyl group. In some embodiments the alkyl group is an unsubstituted linear alkyl group.
  • the alkyl group is a linear alkyl group which is substituted by one or more groups selected from -OH, -OR W , -NH2, - NHR W , -NR W 2, -SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkyl group is a branched alkyl group.
  • the alkyl group is an unsubstituted branched alkyl group.
  • the alkyl group is a branched alkyl group which is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • alkenyl is intended to include both unsubstituted alkenyl groups, and alkenyl groups which are substituted by one or more additional groups.
  • the alkenyl group is an unsubstituted alkenyl group.
  • the alkenyl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkenyl group is a C2-C12 alkenyl, a C 2 -CM alkenyl, a C2-C8 alkenyl, a C2-C6 alkenyl, or a C 2 -C 4 alkenyl group.
  • the alkenyl group is a linear alkenyl group.
  • the alkenyl group is an unsubstituted linear alkenyl group.
  • the alkenyl group is a linear alkenyl group which is substituted by one or more groups selected from -OH, - OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkenyl group is a branched alkenyl group.
  • the alkenyl group is an unsubstituted branched alkenyl group.
  • the alkenyl group is a branched alkenyl group which is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • alkynyl is intended to include both unsubstituted alkynyl groups, and alkynyl groups which are substituted by one or more additional groups.
  • the alkynyl group is an unsubstituted alkynyl group.
  • the alkynyl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkynyl group is a C2-C12 alkynyl, a C 2 -Cio alkynyl, a C 2 -Cg alkynyl, a C 2 -Cg alkynyl, or a C 2 -C 4 alkynyl group.
  • the alkynyl group is a linear alkynyl group.
  • the alkynyl group is an unsubstituted linear alkynyl group.
  • the alkynyl group is a linear alkynyl group which is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the alkynyl group is a branched alkynyl group.
  • the alkynyl group is an unsubstituted branched alkynyl group. In some embodiments the alkynyl group is a branched alkynyl group which is substituted by one or more groups selected from -OH, -OR W , -NH2, -NHR W , -NR W 2, -SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • cycloalkyl is intended to include both unsubstituted cycloalkyl groups, and cycloalkyl groups which are substituted by one or more additional groups.
  • cycloalkyl is also intended to include monocyclic and bicyclic ring systems (including spirocyclic ring systems, in which the two rings share a single atom; fused bicyclic ring systems, in which the two rings share two adjacent atoms; and bridged bicyclic ring systems, in which the two rings share three or more atoms).
  • the cycloalkyl group is an unsubstituted cycloalkyl group.
  • the cycloalkyl group is substituted by one or more groups selected from -OH, -OR W , -NH2, -NHR W , -NR W 2, - SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the cycloalkyl group is a C3-C12 cycloalkyl, a C4-C12 cycloalkyl, a C5-C12 cycloalkyl, a C3-C10 cycloalkyl, a C4-C10 cycloalkyl, a C5-C10 cycloalkyl, a C3-C8 cycloalkyl, a C4-C8 cycloalkyl, a C5-C8 cycloalkyl, a C3-C6 cycloalkyl, a C4-C6 cycloalkyl, a C5-C6 cycloalkyl, a C3-C4 cycloalkyl, or a C4-C5 cycloalkyl group.
  • cycloalkenyl is intended to include both unsubstituted cycloalkenyl groups, and cycloalkenyl groups which are substituted by one or more additional groups.
  • the cycloalkenyl group is an unsubstituted cycloalkenyl group.
  • the cycloalkenyl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the cycloalkenyl group is a C4-C12 cycloalkenyl, a C5-C12 cycloalkenyl, a C4-C10 cycloalkenyl, a C5-C10 cycloalkenyl, a C4-C8 cycloalkenyl, a C5-C8 cycloalkenyl, a C4-C6 cycloalkenyl, a C5-C6 cycloalkenyl, or a C4-C5 cycloalkenyl group.
  • heterocycloalkyl is intended to include both unsubstituted heterocycloalkyl groups, and heterocycloalkyl groups which are substituted by one or more additional groups.
  • heterocycloalkyl is also intended to include monocyclic and bicyclic ring systems (including spirocyclic ring systems, in which the two rings share a single atom; fused bicyclic ring systems, in which the two rings share two adjacent atoms; and bridged bicyclic ring systems, in which the two rings share three or more atoms).
  • the heterocycloalkyl group is a monocyclic ring system, a spirocyclic ring system, or a fused bicyclic ring system. In some embodiments, the heterocycloalkyl group is an unsubstituted heterocycloalkyl group.
  • the heterocycloalkyl group is substituted by one or more groups selected from -R w , -OH, -OR W , -NH2, -NHR W , -NR W 2, -SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • one or more -CH 2 - groups of the heterocycloalkyl ring may be replaced with a -C(O)- group
  • the heterocycloalkyl group is a C3-C12 heterocycloalkyl, a C4-C12 heterocycloalkyl, a C5-C12 heterocycloalkyl, a C3-C10 heterocycloalkyl, a C4-C10 heterocycloalkyl, a C5-C10 heterocycloalkyl, a C 3 -C 8 heterocycloalkyl, a C 4 -C 8 heterocycloalkyl, a C 5 -C 8 heterocycloalkyl, a C3-C6 heterocycloalkyl, a C4-C6 heterocycloalkyl, a C5-C6 heterocycloalkyl, a C3-C4 heterocycloalkyl, or a C4-C5 heterocycloalkyl group.
  • aryl is intended to include both unsubstituted aryl groups, and aryl groups which are substituted by one or more additional groups.
  • the aryl group is an unsubstituted aryl group.
  • the aryl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -OC(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • the aryl group is a C6-C10 aryl, a C6-C8 aryl, or a Cg aryl.
  • heteroaryl is intended to include both unsubstituted heteroaryl groups, and heteroaryl groups which are substituted by one or more additional groups.
  • the heteroaryl group is an unsubstituted heteroaryl group.
  • the heteroaryl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • one or more -NH- groups of the heteroaryl ring may be replaced with a -NR W - group.
  • the heteroaryl group is a C6-C10 heteroaryl, a C6-C9 heteroaryl, a C6-C8 heteroaryl, or a Cg heteroaryl.
  • fused heterocycloalkyl-heteroaryl is intended to mean a bicyclic ring system in which one ring is a heterocycloalkyl ring and the other is a heteroaryl ring, and in which the two rings share two adjacent atoms. Of the two adjacent atoms shared by the two rings, both may be carbon atoms; both may be heteroatoms (e. g. independently O, N or S); or one may be a carbon atom and the other a heteroatom (e. g. O, N or S).
  • the fused heterocycloalkyl-heteroaryl may be unsubstituted or may be substituted by one or more additional groups.
  • the fused heterocycloalkylheteroaryl group is an unsubstituted cycloalkenyl group.
  • the fused heterocycloalkyl-heteroaryl group is substituted by one or more groups selected from -OH, -OR W , -NH2, - NHR W , -NR W 2, -SO2R W , -C(O)R W , -CN, and -NO2, wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • benzyl is intended to include both unsubstituted benzyl groups, and benzyl groups which are substituted by one or more additional groups.
  • the benzyl group is an unsubstituted benzyl group.
  • the benzyl group is substituted by one or more groups selected from -OH, -OR W , -NH 2 , -NHR W , -NR W 2 , -SO 2 R W , -C(O)R W , -CN, and -NO 2 , wherein each R w is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
  • all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl, heteroaryl and benzyl groups in the compounds are unsubstituted.
  • the present invention provides a compound of formula (la): wherein:
  • X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X2 is N, then X3 is CR 1 ; and when X3 is N, then X2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl), -NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycl
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2, -S(O) 2 NR 9 2 , - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 -P(O)(OR P ) 2 , -C(O)O(CH 2 ) P NMe 2 , -C(O)O(CH 2 ) P NHMe, -C(O)OCH2CH(OH)CH 2 OH, - C(O)OCH2CH 2 CI ⁇ /le2OH, and -qojOCHjCHj
  • X2 is N. In other embodiments, X3 is N.
  • X2 is CR 3 and X3 is CR 1 .
  • R 1 is H, halogen or methyl.
  • R 3 is H, halogen, unsubstituted alkyl or -CN.
  • R 1 is H. In some such embodiments, R 1 and R 3 are each H. In other embodiments, R 1 and R 2 are each H.
  • R 2 and R 3 are each H.
  • R 3 is H
  • R 1 is halogen or methyl
  • R 2 is -B(OH)2, halogen, -CN, -NR 5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is -B(OH)2, -NR 5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is selected from:
  • R 2 is selected from n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
  • R 2 is selected from
  • n 1, 2 or 3; and wherein m is 0 or 1.
  • R 2 is
  • each R 4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH 2 , - N(alkyl) 2 , -S(O) 2 alkyl, -S(O) 2 aryl, -S(O) 2 N(alkyl) 2 , -CH 2 S(O) 2 NH(alkyl), -S(O) 2 (heterocycloalkyl), - C(O)(heterocycloalkyl), and -C(O)OH; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 .
  • each R 7 is independently selected from -Me, -OMe, -O(CH 2 ) q OMe, -CH 2 OMe, -
  • each R 4 is independently selected from -F, -Cl, - l Bu, -Me, -CF3, -OH, -OMe, -OCF3,
  • each R 6 is independently selected from halogen, haloalkyl, -NH 2 , - C(O)(heterocycloalkyl), -S(O) 2 (heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 .
  • each R 6 is independently selected from -Cl, -CF 3 , -NH 2 , -C(O)piperidine, -C(O)Me
  • R 2 is selected from:
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl); then: C 4 -R 4 is selected from C-alkynyl, C-S(O) 2 alkyl, C-S(O) 2 aryl, C-S(O) 2 N(alkyl) 2 , C-CH 2 S(O) 2 NH(alkyl), C-S(O) 2 (heterocycloalkyl), C-CH 2 heterocycloalkyl, C-CH 2 C(O)NH(alkyl), C-NHC(O)(alkyl), C-SO(alkyl) and C- P(O)(alkyl) 2 ; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 .
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl): then: each R 4 of — ” R4 ) is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH 2 , -N(alkyl) 2 , -S(O) 2 alkyl, -S(O) 2 aryl, -S(O) 2 N(alkyl) 2 , -CH 2 S(O) 2 NH(alkyl), -S(O) 2 (heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH 2 heterocycloalkyl, -CH 2 C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO(alkyl),
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl); then:
  • R 8 is -CH 2 C(O)OH
  • X 2 and X 3 are both CH;
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl); then:
  • R 8 is -CH 2 C(O)OH or -CH ⁇ O ⁇ CH ⁇ CfOfBu;
  • X 2 and X3 are both CH;
  • each R 4 of — ( '” R4 ) is -S(O) 2 (heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 ; or
  • R 1 is methyl
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CH 2 S(O) 2 NR 9 2 , -S(O) 2 NR 9 2 , - CH 2 (heterocycloalkyl), -CH 2 (heteroaryl), -CH 2 B(OH) 2 , -CH 2 P(O)(OH) 2 , -COOR 9 , -B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CH 2 S(O) 2 NR 9 2 , -S(O) 2 NR 9 2 , CH 2 (4- or 5-membered heterocycloalkyl), -CH 2 (5-membered heteroaryl), -CH 2 B(OH) 2 , -CH 2 P(O)(OH) 2 , - COOR 9 , -B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is -CHR 9 C(O)OR 10 .
  • R 8 is selected from -CH 2 C(O)OH, -CHMeC(O)OH, -CH 2 C(O)OMe, -CH 2 C(O)OEt, - CH 2 S(O) 2 NH 2 , -CH ⁇ OjOCfWqorBu, -CH 2 C(O)NH 2 , -CH 2 S(O) 2 NH 2 , -S(O) 2 NH 2 , -CH 2 -oxetane, -CH 2 (1,2,3- triazole), -CH 2 B(OH) 2 , -CH 2 P(O)(OH) 2 , -COOH, -COOMe, -B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is selected from -CH 2 C(O)OCH 2 OC(O) t Bu, -CH 2 C(O)OH and -CHMeC(O)OH.
  • R 8 is -CH2C(O)OH. In other embodiments, R 8 is -CHzCfOjOCHzOCfOfBu.
  • the compound is selected from:
  • the compound is selected from Compound nos. 1, 2, 3, 4, 5, 6, 7 , 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, 19, 20, 21, 23, 24, 26, 28, 30, 31, 33, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 51, 52, 53, 54, 55, 56, 58, 59, 60, 61, 62, 63, 64, 65, 67, 68, 69, 70, 71, 72, 73, 75, 76, 77, 78, 79, 80, 81, 82, 84, 86 and 88.
  • R 1 is H; R 3 is H or methyl; R 2 is aryl substituted with one or more R 4 or is fused bicyclic heteroaryl substituted with one or more R 6 ; and R 8 is -CH2C(O)OH, -CH(Me)C(O)OH or CHR 9 (heteroaryl).
  • each R 4 is independently selected from halogen, unsubstituted alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -NHz, -N(alkyl)?, -NH(CO)alkyl, - C(O)NHalkyl, -C(O)N(alkyl)?, -S(O)?
  • the compound is selected from Compound nos. 1, 2, 3, 8, 10, 11, 12, 13, 14, 15, 19, 22, 24, 25, 27 , 34, 37, 38, 39, 41, 44, 45, 46, 47, 48, 50, 53, 56, 59, 64, 72, 73, 75, 76, 77, 78, 79, 81, 82, 84, 86, 90, 91, 92, 94, 96, 101, 102 and 103.
  • R 3 is H; each R 4 is independently selected from -OH, -NHz, -O(alkyl), -NHC(O)alkyl, -C(O)NHalkyl, - S(O)z(heterocycloalkyl) and -C(O)OH; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 ; and each R 7 is independently selected from -O(alkyl), -O(CHz) q OI ⁇ /le, -(CHz) q OI ⁇ /le and -(OCH2CHz) r NHR 7a ; wherein q is 1 or 2, r is 4 and R 7a is -C(O)Me.
  • the compound is Compound ID no. 2, 24, 37, 45, 46, 47, 73, 76, 77, 79, 82, 84, 86, 91, 96, 102 or 103.
  • a bifunctional protein degrader compound comprising a compound as defined in any of the above embodiments of the first aspect.
  • a bifunctional protein degrader compound comprising a compound of formula (la'): wherein: X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X 2 is N, then X 3 is CR 1 ; and when X 3 is N, then X 2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH 2 ;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl) 2 , - NH(alkyl),- NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl),
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2 , -S(O) 2 NR 9 2 , - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 , -P(O)(OR P ) 2 , -C(O)O(CH 2 )pNMe 2 , -C(O)O(CH 2 ) p NHMe, -C(O)OCH 2 CH(OH)CH 2 OH, - C(O)OCH 2 CH 2 CMe 2 OH, and -C(O)OC
  • R 19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R 19 ; and wherein:
  • the Target protein binding moiety is a moiety which binds to a target protein.
  • the target protein may be a protein which mediates a disease or disorder in a subject. The degradation of the target protein may thus result in a therapeutic effect in a subject treated with the bifunctional protein degrader compound.
  • Proteins susceptible to binding with the Target protein binding moiety (and subsequent degradation by the E3 ligase - KLHDC2 ligase associated with the KLHDC2 ligase binding moiety) encompass a broad spectrum, including any protein or peptide, along with their fragments, analogs, or homologs.
  • Target proteins exhibit diverse biological functions or activities, such as structural, regulatory, hormonal, enzymatic, genetic, immunological, contractile, storage, transportation, and signal transduction roles.
  • these proteins include structural proteins, receptors, enzymes, cell surface proteins, and those integral to overall cell function. This covers proteins involved in catalytic, aromatase, motor, helicase, metabolic, antioxidant, proteolytic, and biosynthetic activities, as well as proteins with kinase, oxidoreductase, transferase, hydrolase, lyase, isomerase, and ligase activities.
  • Other categories involve enzyme regulator, signal transducer, structural molecule, and binding activities, including proteins relevant to cell motility, membrane fusion, cell communication, and the regulation of biological processes, development, cell differentiation, and response to stimuli.
  • target proteins also encompass behavioral proteins, cell adhesion proteins, those involved in cell death, transport proteins, chaperone regulator proteins, nucleic acid binding proteins, transcription regulators, and proteins involved in extracellular organization and biogenesis.
  • the scope extends to proteins from various organisms, including eukaryotes and prokaryotes, such as humans, microbes, viruses, fungi, parasites, and other animals, including domesticated ones.
  • Target protein binding moieties coupled to KLHDC2 ligase binding moieties, as outlined in the present disclosure, and placing/presenting that target protein or polypeptide in proximity to an KLHDC2 ligase may lead to ubiquitination and subsequent degradation of the target protein, allows for the modulation of protein activity, bringing on to therapeutic outcomes.
  • Target proteins of the Target protein binding moiety include, for example, moieties which bind to Human BET Bromodomain-containing proteins, for example BRD4. Further, kinase warhead TL13-87 targeting 183 kinases (Huang et al., 2018) was utilized in the exemplified bifunctional compounds.
  • Table 1 List of kinases targeted by TL13-87 kinase warhead.
  • M is O, S or NH, or is absent; indicates attachment to R 18 of the linker;
  • R 11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
  • R 12 is H or Me
  • L' is H, alkyl, benzyl, acetyl or pivaloyl; or
  • X 4 and X 5 are each independently N or CH;
  • X 6 is N or CH
  • R 30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R 40 , or -C(O)R 40 ;
  • R 31 is H, -OMe, -heteroaryl, -heteroaryl-R 40 or R 40 ;
  • R 32 is H, unsubstituted C1-C6 alkyl, R 33 is -N(C1-C6 alkyl) 2 , -NH(C1-C6 alkyl), -NH(aryl), or R 40 ;
  • R 34 is -Me or -C(O)R 40 ;
  • R 40 is a bond connected to R 18 of the linker, wherein the [Target protein binding moiety] contains a single R 40 ; or wherein
  • X 7 is N or CH
  • R 35 is -heterocycloalkyl-R 40 , or R 40 ;
  • R 36 is H or -OMe
  • R 40 is a bond connected to R 18 of the linker, wherein the [Target protein binding moiety] contains a single R 40 ; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
  • X 2 is N or CR 3 ;
  • X 3 is N or CR 1 ; wherein when X 2 is N, then X 3 is CR 1 ; and when X 3 is N, then X 2 is CR 3 ;
  • R 1 is H, halogen, haloalkyl, methyl, -OH or -NH 2 ;
  • R 3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)z, - NH(alkyl),- NH 2 or -CN;
  • R 2 is H, -B(OH) 2 , halogen, -CN, -NR 5 2 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 ; each R 4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl),
  • R 8 is selected from -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2 , -S(O) 2 NR 9 2 , - CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , -CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 10 , - B(OH) 2 , -P(O)(OH) 2 , -P(O)(OR P ) 2 , -C(O)O(CH 2 )pNMe 2 , -C(O)O(CH 2 ) p NHMe, -C(O)OCH 2 CH(OH)CH 2 OH, - C(O)OCH 2 CH 2 CMe 2 OH, and -C(O)OC
  • R 19 is a bond connected to R 14 of the linker, wherein formula (la') contains a single R 19 ; and wherein:
  • R 14 is -C1-6 alkyl, -C2-6 alkenyl, -C2-6alkynyl, Ci.g alkyl-N(C1-6 alkyl)-, -C(O)-, -SO2- or is absent
  • R 15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, C1-6 alkyl-NH-, -Ci.g alkyl-N(Ci-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
  • R 16 is -C1-6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH 2 -C(O)-, -CH 2 -C(O)-NH-, -CH 2 -C(O)O- or is absent
  • R 17 is -CH2(C2H 4 -O) y , (C 2 H4-O) X , (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
  • X 2 is N. In other embodiments, X 3 is N.
  • X 2 is CR 3 and X 3 is CR 1 .
  • R 1 is H, halogen or methyl.
  • R 3 is H, halogen, unsubstituted alkyl or -CN.
  • R 1 is H. In some embodiments, R 1 and R 3 are each
  • R 1 and R 2 are each H.
  • R 2 and R 3 are each H. In some embodiments of the fourth and fifth aspects,
  • R 3 is H
  • R 1 is halogen or methyl
  • R 2 is -B(OH)2, halogen, -CN, -NR 5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is -B(OH)2, -NR 5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R 19 ; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R 6 .
  • R 2 is selected from
  • R 2 is selected from wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
  • R 2 is selected from wherein n is 1, 2 or 3; and wherein m is 0 or 1. In some embodiments of the fourth and fifth aspects, R 2 is
  • each R 4 is independently selected from halogen, alkyl, -CN, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), - O(heterocycloalkyl), -NH 2 , -N(alkyl) 2 , -S(O) 2 alkyl, -S(O) 2 aryl, -S(O) 2 N(alkyl) 2 , -CH 2 S(O) 2 NH(alkyl), - S(O) 2 (heterocycloalkyl), -C(O)(heterocycloalkyl), -C(O)OH, R 19 and -OR 19 ; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one
  • each R 7 is independently selected from -Me, - OMe, -O(CH 2 ) q OMe, -CH 2 OMe, -C(O)Me, -S(O) 2 Me, R 19 and -OR 19 .
  • each R 4 is independently selected from R 19 , -OR 19 , -NHR 19 -F, -Cl, - l Bu, -Me, -CF 3 , -CN, -OH, -OMe, -OCF 3 , -NH 2 , -NMe 2 , -S(O) 2 Me, -S(O) 2 NMe 2 , - CH 2 S(O) 2 NHMe, cyclopropyl, -C(O)OH,
  • each R 4 is independently selected from R 19 , -OR 19 ,
  • each R 6 is independently selected from haloalkyl, -C(O)(heterocycloalkyl), - S(O) 2 (heterocycloalkyl), -C(O)alkyl, R 19 and NHR 19 ; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 .
  • each R 6 is independently selected from -CF3, -
  • R 2 is selected from:
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl); then
  • C 4 -R 4 is selected from C-alkynyl, C-S(O) 2 alkyl, C-S(O) 2 N(alkyl) 2 , C-CH 2 S(O) 2 NH(alkyl), C- S(O) 2 (heterocycloalkyl), C-S(O) 2 aryl, C-CH 2 heterocycloalkyl, C-CH 2 C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl) 2 ; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R 7 .
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl), n is 1 or 2, then each R 4 of - ( ' ⁇ R4 / ) n is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH2, -N(alkyl)z, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl);
  • R 8 is -CHR 9 C(O)OR 10 , -CHR 9 C(O)NR 9 2 or -CHR 9 (heterocycloalkyl); then:
  • R 8 is -CH 2 C(O)OH or -CH ⁇ O ⁇ CH ⁇ CfOfBu;
  • X 2 and X 3 are both CH;
  • each is -S(O) 2 (heterocycloalkyl), wherein each heterocycloalkyl is
  • R 1 is methyl
  • R 8 is selected from -CHR 9 C(O)OR 10 , - CHR 9 C(O)NR 9 2 , -CHR 9 S(O) 2 NR 9 2 , -S(O) 2 NR 9 2, -CHR 9 (heterocycloalkyl), -CHR 9 (heteroaryl), -CHR 9 B(OH) 2 , - CHR 9 P(O)(OH) 2 , -CHR 9 P(O)(OR P ) 2 , -COOR 9 , -B(OH) 2 , -P(O)(OH) 2 , -P(O)(OR P ) 2 , -C(O)O(CH 2 ) P NMe 2 , - C(O)O(CH 2 ) P NHMe, -C(O)OCH2CH(OH)CH 2 OH, -C(O)OCH2CH 2 CI ⁇ /le2OH
  • R 8 is selected from -CHR 9 C(O)OR 10 , CHR 9 C(O)NR 9 2, -CH 2 S(O)2NR 9 2, -S(O) 2 NR 9 2, -CH 2 (heterocycloalkyl), -CH 2 (heteroaryl), -CH 2 B(OH)2, -CH 2 P(O)(OH)2, -COOR 9 , - B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is selected from -CHR 9 C(O)OR 10 , CHR 9 C(O)NR 9 2, -CH 2 S(O)2NR 9 2, -S(O) 2 NR 9 2, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH 2 B(OH)2, -CH 2 P(O)(OH)2, -COOR 9 , -B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is selected from -CH 2 C(O)OH, -CHMeC(O)OH, - CH 2 C(O)OMe, -CH 2 C(O)OEt, -CH2C(O)OCH 2 OC(O) t Bu, -CH 2 C(O)NH2, -CH 2 S(O)2NH2, -S(O) 2 NH2, -CH 2 - oxetane, -CH 2 (l,2,3-triazole), -CH 2 B(OH)2, -CH 2 P(O)(OH)2, -COOH, -COOMe, -B(OH) 2 and -P(O)(OH) 2 .
  • R 8 is -CHR 9 C(O)OR 10 . In some embodiments of the fourth and fifth aspects, R 8 is selected from -CHjCfOjOCHjOCXOfBu, - CH 2 C(O)OH and -CHMeC(O)OH.
  • R 8 is selected from
  • R 8 is -CH 2 C(O)OH. In other embodiments, R 8 is -CH 2 C(O)OCH 2 OC(O) t Bu.
  • R 1 and R 3 are each H;
  • R 2 is selected from aryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R 4 ; and wherein the fused aryl/heterocycloalkyl is unsubstituted or is substituted with one or more R 6 ; and R 8 is -CHR 9 C(O)OR 10 .
  • R 2 is aryl is substituted with R 4 and R 8 is -CHR 9 C(O)OR 10
  • each R 4 is S(O) 2 (heterocycloalkyl) substituted with R 19 .
  • At least one of R 14 , R 15 , R 16 , R 17 and R 18 is present.
  • R 14 is absent. In other embodiments, R 14 is -C(O)-.
  • R 15 is absent. In other embodiments, R 15 is heterocycloalkyl.
  • R 16 is absent. In other embodiments, R 16 is -Ci.g alkyl. In other embodiments, R 16 is -C(O)-. In other embodiments, R 16 is -C(O)-NH-.
  • R 17 is absent. In other embodiments, R 17 is (C 2 H 4 -O) X . In some embodiments of the fifth aspect, x is 3-9. In some embodiments, x is 3, 5, 7 or 9.
  • R 18 is absent. In other embodiments, R 18 is -Ci-w alkyl or -Ci-io alkyl-NH-. In some embodiments, R 18 is -Ci.g alkyl or -Ci.g alkyl-NH-. In some embodiments, R 18 is - CH 2 CH 2 - or -CH 2 CH 2 -NH-. In other embodiments, R 18 is heterocycloalkyl or heterocycloalkyl-NH-.
  • R 14 , R 15 , R 16 , R 17 and R 18 are each absent.
  • the [Target protein binding moiety] is: wherein
  • M is O, S or NH, or is absent; indicates attachment to R 18 of the linker;
  • R 11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
  • R 12 is H or Me
  • L' is H, alkyl, benzyl, acetyl or pivaloyl.
  • R 12 is Me. In other embodiments, R 12 is H.
  • [Target protein binding moiety] is:
  • [Target protein binding moiety] is:
  • M is absent.
  • L' is H.
  • [Target protein binding moiety] is:
  • [Target protein binding moiety] is: n some embodiments of the fifth aspect, [Target protein binding moiety] is:
  • [Target protein binding moiety] is:
  • [Target protein binding moiety] is:
  • [Target protein binding moiety] is:
  • [Target protein binding moiety] is:
  • k is an integer from 3-8; optionally wherein k is 4, 6, or 8; further optionally wherein k is 6.
  • the compound is selected from:
  • the compound is selected from Compound ID nos. 1001, 1002 and 1003.
  • the compound is selected from Compound ID nos. 1002, 1003, 1005, 1007, 1008, 1010, 1013, 1014, 1016, 1019, 1020, 1022, 1024, 1026, 1028, 1031, 1034, 1036 and 1038.
  • the compound is selected from Compound ID no. 1003, 1008, 1010, 1012, 1014, 1016, 1020, 1022, 1026, 1028, 1034, 1036 and 1038. In some embodiments of the fifth aspect, the compound is selected from Compound ID no. 1005 and 1007.
  • the compound is selected from Compound ID nos. 1008, 1009, 1010, 1011, 1012, 1013, 1014, 1015, 1016, 1017, 1018 and 1019.
  • the compound is selected from Compound ID nos. 1024 and 1025.
  • the compound is selected from Compound ID nos. 1028, 1029, 1030, 1031, 1033, 1034, 1035, 1036 and 1037.
  • the present invention also provides a pharmaceutical composition comprising a compound of the invention.
  • the pharmaceutical composition comprises a bifunctional protein degrader compound of the present invention.
  • the pharmaceutical composition comprises a compound of any of the above embodiments of the fourth or fifth aspects of the present invention.
  • the pharmaceutical composition comprises a compound of any of the above embodiments of the fifth aspect of the present invention.
  • the present invention also provides a compound or pharmaceutical composition of the invention, for use in medicine.
  • the compound is a bifunctional protein degrader compound of the present invention.
  • the compound is a compound of any of the above embodiments of the fourth or fifth aspects of the present invention.
  • the compound is a compound of any of the above embodiments of the fifth aspect of the present invention.
  • the present invention also provides a method of treating a disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or pharmaceutical composition of the invention.
  • the compound is a bifunctional protein degrader compound of the present invention.
  • the compound is a compound of any of the above embodiments of the fourth or fifth aspects of the present invention.
  • the compound is a compound of any of the above embodiments of the fifth aspect of the present invention.
  • the compounds of the present invention were prepared as described below.
  • Preparative HPLC was performed using Waters auto purification instrument equipped with Gemini C18 column (100 x 19 mm, 5m), YMC-Actus C18 (250 x 20 mm, 5m), YMC-Triart C18 (250 x 20 mm, 5m), Hydrosphere C18 (250 x 20 mm, 5m).
  • PdXphosG3 (2-Dicyclohexylphosphino-2',4',6'-triisopropyl-l,r-biphenyl)[2-
  • the ester (1 equiv) was taken in appropriate solvent or in mixture of solvents (e.g. MeOH, THF) and lithium hydroxide monohydrate (1-5 equiv) or sodium hydroxide (1-5 equiv) or cesium carbonate (1-5 equiv) in water was added at temperature between 0°C and RT. The mixture was stirred at temperature between RT and 90°C for 1-18 h. After completion the solvent was removed under reduced pressure, the residue was diluted with water and washed with ether. The aqueous layer was cooled to 0°C, acidified with IM HCI and extracted with ethyl acetate. The organic layer was dried over anhydrous NajSCU and evaporated. The product was purified by flash column chromatography and/or preparative HPLC unless otherwise stated.
  • solvents e.g. MeOH, THF
  • Reaction Scheme 10 Buchwald-Hartwig cross coupling The suspension of appropriate aryl iodide (1 equiv), amine (0.8-2.5 equiv), CS2CO3 (2-2.5 equiv), Xantphos (0.1-0.2 equiv) and Pd2(dba)3 (0.08-0.12 equiv) in DMF was stirred at 90-110°C for 5-18 h. The volatiles were removed under reduced pressure and the product was purified by flash column chromatography unless otherwise stated.
  • Step 1 Ethyl 5-(2-methoxyphenyl)pyrazolo[l,5-a]pyridine-3-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (60% yield), using ethyl 5- bromopyrazolo[l,5-a]pyridine-3-carboxylate [commercial] (300 mg, 1.11 mmol, 1 equiv) and (2- methoxyphenyl)boronic acid [commercial] (1.2 equiv) as starting materials, K3PO4 (5 equiv) as base, PdXphosG3 as catalyst (0.1 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated. The product was purified by flash column chromatography.
  • Step 2 (5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7, above (49% yield), using ethyl 5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridine-3-carboxylate (290 mg, 1.03 mmol, 1 equiv) as starting material and LAH (1.2 equiv, IM in THF) as reducing agent.
  • Step 3 2-(5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (48% yield), using (5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)methanol (100 mg, 0.4 mmol, 1 equiv) as starting material.
  • Step 4 2-(5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (27% yield), using 2-(5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile (50 mg, 0.19 mmol, 1 equiv) as starting material. After extraction, the residue was triturated with 50% ether in pentane to afford pure product.
  • Step 1 Methyl 2-(4-bromopyridin-2-yl)acetate [commercial] (5.6 g, 24.3 mmol, 1 equiv) and DMF-DMA [commercial] (46 mL) were stirred at 80°C for 16 h under nitrogen. After consumption of the starting material the reaction mixture was evaporated under reduced pressure and the residue was triturated with ether to afford crude methyl (Z)-2-(4-bromopyridin-2-yl)-3-(dimethylamino)acrylate (5.0 g) which was forwarded directly for the next step.
  • Step 2 To a solution of methyl (Z)-2-(4-bromopyridin-2-yl)-3-(dimethylamino)acrylate (5.0 g, 17.5 mmol, 1 equiv, crude after Step 1) in DCM (50 mL) was added O-(mesitylsulfonyl)hydroxylamine [commercial] (4.53 g, 21 mmol, 1.2 equiv) and the reaction mixture was stirred at RT for 16 h. After completion the reaction was quenched with water and extracted with ethyl acetate. The combined organic fractions were washed with brine and evaporated. Methyl 5-bromopyrazolo[l,5-o]pyridine-3-carboxylate (2.2 g, 8.63 mmol, 35% yield over two steps) was purified by flash column chromatography.
  • Step 3 (5-Bromopyrazolo[l,5-a]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7 , above (56% yield), using methyl 5- bromopyrazolo[l,5-a]pyridine-3-carboxylate (1.0 g, 3.92 mmol, 1 equiv) as starting material and DIBAL (15.6 mL, 15.6 mmol, 4 equiv, IM solution in toluene) as reducing agent.
  • Step 4 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (49% yield) using (5-bromopyrazolo[l,5- o]pyridin-3-yl)methanol (250 mg, 1.1 mmol, 1 equiv) as starting material.
  • Step 5 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (61% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetonitrile (75 mg, 0.32 mmol) as a starting material. After completion, the acidified reaction mixture was extracted with ethyl acetate, the combined organic fractions were dried over Na 2 SO 4 and evaporated. The residue was triturated with 50% (v/v) mixture of ether and pentane to afford the target compound.
  • Step 6 [TLS-364, step 1] To a solution of 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (1.7 g, 6.67 mmol, 1 equiv) in MeOH (20 mL) was added concentrated H 2 SO 4 (0.5 mL) and the mixture was refluxed for 6 h. After completion the volatiles were removed under reduced pressure. The residue was taken up in ethyl acetate, washed with water, saturated NaHCOs solution, dried over Na 2 SO 4 and evaporated to afford methyl 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetate (1.4 g, 5.2 mmol, 78% yield).
  • Step 8 Methyl 2-(5-(2-((/V-methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (258.3 mg, 0.817 mmol, 1.3 equiv) and l-(2-bromophenyl)-/V-methylmethanesulfonamide [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.2 equiv). After completion of the reaction the solution was filtered, the volatiles were removed under reduced pressure and the resulting crude was used in the next step without purification.
  • Step 9 2-(5-(2-((/V-Methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (59% yield over two steps), using methyl 2-(5-(2-((/V-methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting material.
  • Example 1-003 Synthesis of 2-(5-(2-chlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 69 )
  • Step 1 2-(5-(2-Chlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetonitrile (35 mg, 0.15 mmol, 1 equiv) and (2-chlorophenyl)boronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv). After completion the solution was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated to
  • Step 2 2-(5-(2-Chlorophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 1, above (46% yield over two steps), using 2-(5-(2-chlorophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetonitrile (200 mg, 0.75 mmol) as a starting material.
  • Step 1 2-(5-(2,6-Dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile (150 mg, 0.63 mmol, 1 equiv) and (2,6- dichlorophenyl)boronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over Na2SO 4 and evaporated to give crude product which was used in the next step without further purification.
  • Step 1 2-(5-(m-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5-o]pyridin-3- yl)acetonitrile (20 mg, 0.08 mmol, 1 equiv) and m-tolylboronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.11 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated to give crude product which was used in the next step without further purification.
  • Step 2 2-(5-(m-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (8% yield over two steps), using 2-(5-(m- tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile (100 mg, 0.4 mmol) as a starting material.
  • Step 1 2-(5-(2-(Methylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (62% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (80 mg, 0.315 mmol, 1 equiv) and 4,4,5,5-tetramethyl-2-(2- (methylsulfonyl)phenyl)-l,3,2-dioxaborolane (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(4-(l-Methylpiperidin-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (61% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (50 mg, 0.197 mmol, 1 equiv) and (4-(l- methylpiperidin-4-yl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dppf)CI 2 as catalyst (0.1 equiv). LCMS (ESI+) m/z 350.2 [M+H] +
  • Step 1 2-(5-Benzylpyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (55% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetic acid (60 mg, 0.236 mmol, 1 equiv) and 2-benzyl-4,4,5,5-tetramethyl-l,3,2- dioxaborolane [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv).
  • Step 1 Methyl 2-(5-(2-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (171 mg, 0.541 mmol, 1.3 equiv) and l-(2-bromophenyl)piperidine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.2 equiv). After completion the solvents were removed under reduced pressure and the obtained crude was used in the next step without further purification.
  • Step 2 2-(5-(2-(Piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (34% yield over two steps), using methyl 2-(5-(2-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(2,3-Dihydrobenzo[b][l,4]dioxin-5-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (27% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.394 mmol, 1 equiv) and (2,3- dihydrobenzo[b][l,4]dioxin-5-yl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 Methyl 2-(5-(2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (135 mg, 0.43 mmol, 1.3 equiv) and l-((2-bromophenyl)sulfonyl)piperidine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.15 equiv). After completion the solvents were removed under reduced pressure and the resulting crude was used in the next step without purification.
  • Step 2 2-(5-(2-(Piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (13% yield over two steps), using methyl 2-(5-(2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(2,6-Dimethoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (24% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (100 mg, 0.39 mmol, 1 equiv) and (2,6- dimethoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.2 equiv).
  • Step 1 2-(5-(2-(Trifluoromethoxy)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (31% yield), using 2- (5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (60 mg, 0.23 mmol, 1 equiv) and (2- (trifluoromethoxy)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(2-Methoxy-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (32% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.39 mmol, 1 equiv) and (2- methoxy-6-(trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Ck as catalyst (0.1 equiv).
  • Step 1 2-(5-(4-(4-Methylpiperazin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (60 mg, 0.23 mmol, 1 equiv) and l-methyl-4-(4-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)piperazine [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 To a solution of 4-bromo-2,3-dimethylpyridine (5.0 g, 26.9 mmol, 1 equiv) in THF (75 mL), cooled to -78°C, was added LDA (41 mL, 80.6 mmol, 3 equiv, 2M solution in THF) and the resulting mixture was stirred at that temperature for 1 h. Dimethyl carbonate (5 mL, 53.8 mmol, 2 equiv) was added and the reaction was stirred at -78°C for 1 h. The reaction was then quenched with saturated NH 4 CI solution and extracted with ethyl acetate.
  • LDA 41 mL, 80.6 mmol, 3 equiv, 2M solution in THF
  • Step 2 Methyl 2-(4-bromo-3-methylpyridin-2-yl)acetate (4.0 g, 16.46 mmol, 1 equiv) and DMF-DMA (40 mL) were stirred at 140°C for 16 h under nitrogen. After consumption of the starting material the reaction mixture was evaporated under reduced pressure and the residue was triturated with ether to afford crude methyl (Z)-2-(4-bromo-3-methylpyridin-2-yl)-3-(dimethylamino)acrylate which was forwarded directly for the next step.
  • Step 3 To a solution of methyl (Z)-2-(4-bromo-3-methylpyridin-2-yl)-3-(dimethylarriino)acrylate (5.0 g, 16.7 mmol, 1 equiv, crude after Step 2) in DCM (40 mL) was added O-(mesitylsulfonyl)hydroxylamine (4.3 g, 20.04 mmol, 1.2 equiv) and the reaction mixture was stirred at RT for 16 h. After completion, the reaction was quenched with water and extracted with ethyl acetate. The combined organic fractions were washed with brine and evaporated. Methyl 5-bromo-4-methylpyrazolo[l,5-a]pyridine-3-carboxylate (2.5 g, 9.3 mmol, 57% yield over two steps) was purified by flash column chromatography.
  • Step 4 (5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7, above (58% yield), using methyl 5-bromo- 4-methylpyrazolo[l,5-o]pyridine-3-carboxylate (2.5 g, 9.3 mmol, 1 equiv) as starting material and DIBAL (2 equiv) as reducing agent.
  • Step 5 2-(5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (38% yield), using (5-bromo-4- methylpyrazolo[l,5-o]pyridin-3-yl)methanol (1.0 g, 4.15 mmol, 1 equiv) as starting material.
  • Step 6 2-(5-(2-Methoxyphenyl)-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (67% yield), using 2- (5-bromo-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile (200 mg, 0.8 mmol, 1 equiv) and (2- methoxyphenyl)boronic acid (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 4-(4-Bromo-3,5-dichlorophenyl)-3,6-dihydro-2H-pyran was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using 2-bromo-l,3- dichloro-5-iodobenzene [commercial] (250 mg, 0.715 mmol, l.l equiv) and 2-(3,6-dihydro-2H-pyran-4- yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(dppf)CL as catalyst (0.11 equiv).
  • Step 2 Methyl 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (45% yield), using 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydro-2H-pyran (130 mg, 0.41 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv). After completion, the reaction mixture was quenched with water and extracted with e
  • Step 3 2-(5-(2,6-Dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using methyl 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (120 mg, 0.288 mmol) as starting material and lithium hydroxide as base.
  • reaction mixture was diluted with water and washed with 30% ethyl acetate in hexanes.
  • the aqueous layer was acidified with IM HCI and extracted with ethyl acetate, dried over Na 2 SO 4 and evaporated to yield crude product which was used to the next step without additional purification.
  • Step 4 2-(5-(2,6-Dichloro-4-(tetrahydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (10% yield over two steps), using 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid as starting material.
  • Step 1 To a stirred solution of piperidin-l-yl(lH-pyrrol-2-yl)methanone (70 mg, 0.393mmol, 1 equiv) in DMSO (2 mL) was added 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (1 equiv) and the suspension was purged with argon for 10 min. CuO (0.2 equiv) and K2CO3 (3 equiv) were added, the reaction mixture was purged with argon for 5 min and stirred at 140°C for 16 h.
  • Piperidin-l-yl(lH-pyrrol-2-yl)methanone was synthesized as described in Gao, S. et al., J. Org. Chem. 2018, 83, 9250.
  • Step 2 2-(5-(2,6-Dichlorophenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 1, above (37% yield), using 2- (5-(2,6-dichlorophenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetonitrile (15 mg, 0.047 mmol). After completion the cooled solution was diluted with water, washed with ether, acidified with IM HCI. The precipitation was filtered and dried to give the pure product.
  • Example 1-020 Synthesis of 2-(5-(4-(l-acetylpiperidin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 41)
  • Step 1 tert-Butyl 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (93% yield), using 2-bromo-l,3-dichloro-5-iodobenzene (500 mg, 1.42 mmol, l.l equiv) and tert-butyl 4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-l(2H)-carboxylate [commercial] (1 equiv) as starting materials
  • Step 2 4-(4-Bromo-3,5-dichlorophenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (73% yield), using tert-butyl 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate (400 mg, 0.988 mmol) as starting material.
  • Step 3 l-(4-(4-Bromo-3,5-dichlorophenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (89% yield), using 4-(4-bromo-3,5-dichlorophenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (300 mg, 0.72 mmol, 1 equiv) and acetic anhydride (2.7 equiv) as starting materials.
  • Step 4 Methyl 2-(5-(4-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l- one (150 mg, 0.43 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Ck
  • Step 6 2-(5-(4-(l-Acetylpiperidin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (22 mg, 10% yield over three steps), using 2-(5-(4-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6- dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting material.
  • Step 1 To a stirred solution of l,4-dioxaspiro[4.5]decan-8-ol [commercial] (500 mg, 3.163 mmol, 1 equiv) in dry THF (10 mL) was added sodium hydride (60% suspension in mineral oil, 380 mg, 9.488 mmol, 3 equiv) at 0°C and stirred for 10 min. l-Bromo-2-methoxyethane (1.6 mL, 15.814 mmol, 5 equiv) was added at 0°C and the reaction mixture was stirred at RT for 16 h. After completion, the reaction mixture was cooled, quenched with cold water and extracted with ethyl acetate.
  • Step 2 To a stirred solution of 8-(2-methoixyethoxy)-l,4-dioxaspiro[4.5]decane (500 mg, 2.313 mmol, 1 equiv) in THF (12 mL) was added 5M HCI (6 mL, 30 mmol, 13 equiv) at 0°C. Then the reaction mixture was stirred at RT for 5 h. After completion, the reaction mixture was quenched with cold water and extracted with ethyl acetate. The organic layer was washed with brine, dried over NazSCU and evaporated to obtain 4-(2-methoxyethoxy)cyclohiexan-l-one (250 mg, 62% yield).
  • Step 3 To a stirred solution of 4-(2-methoxyethoxy)cyclohexan-l-one (250 mg, 1.453 mmol, 1 equiv) in dry THF (10 mL) was added 2M LDA in THF (1.45 ml, 2.907 mmol, 2 equiv) at -78°C and stirred at that temperature for 1 h. Bis(trifluoromethanesulfonyl)aniline (624 mg, 1.744 mmol, 2 equiv) in THF was added and stirred at -78°C for 1 h, then at RT for 16 h. After completion, the reaction mixture was quenched with saturated NH 4 CI and extracted with ethyl acetate.
  • Step 4 2-(4-(2-Methoxyethoxy)cyclohex-l-en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above, using 4-(2-methoxyethoxy)cyclohex-l-en-l-yl trifluoromethanesulfonate (380 mg, 1.25 mmol) as starting material. After completion, the reaction mixture was filtered through Celite®, evaporated and the obtained crude was used directly in the next step.
  • Step 5 4'-Bromo-3',5'-dichloro-4-(2-methoxyethoxy)-2,3,4,5-tetrahydro-l,l'-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (12% yield over two steps), using 2-bromo-l,3-dichloro-5-iodobenzene (291 mg, 1.029 mmol, 1 equiv) and 2-(4-(2- methoxyethoxy)cyclohex-l-en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (1.11 equiv) as starting materials, KaPO 4 (3.33 equiv) as base and Pd(dppf)CI 2 as catalyst (0.14 equiv).
  • Step 6 Methyl 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4'-bromo-3',5'-dichloro-4-(2-methoxyethoxy)-2, 3,4,5- tetrahydro-l,l'-biphenyl (40 mg, 0.106 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.11 equiv) as starting materials, K3PO4 (3.33 equiv) as base and
  • Step 7 2-(5-(3,5-Dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using mixture of methyl 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'- tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(3,5-dichloro-4'-(2- methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo
  • Step 8 2-(5-(2,6-Dichloro-4-(4-(2-methoxyethoxy)cyclohexyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (17% yield over three steps), using 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro- [l,l'-biphenyl]-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
  • Step 1 4'-Bromo-3',5'-dichloro-4-methoxy-2,3,4,5-tetrahydro-l,r-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (42% yield), using methyl 2-bromo-l,3-dichloro-5-iodobenzene (100 mg, 0.286 mmol, 1.11 equiv) and 2-(4-methoxycyclohex-l-en- l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K3PO4 (2.3 equiv) as base, Pd(dppf)CL as catalyst (0.12 equiv). Reaction was quenched with water and extracted with ethyl acetate, organic layer was washed with brine, dried over NajSCU and concentrated under reduced pressure. The product was purified by flash column chromatography.
  • Step 2 2-(5-(3,5-Dichloro-4'-methoxy-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4'-bromo-3',5'-dichloro-4-methoxy-2,3,4,5-tetrahydro-l,l'-biphenyl (100 mg, 0.299 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1 equiv) as starting materials, K3PO4 (3.5 equiv) as base, Pd(dppf)CL as catalyst (0.1 equiv
  • Step 3 2-(5-(2,6-Dichloro-4-(4-methoxycyclohexyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (29% yield over two steps), using methyl 2-(5-(3,5-dichloro-4'-methoxy-2',3',4',5'-tetrahydro-[l,r-biphenyl]-4- yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
  • Step 1 2-(5-(2-(Trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid [commercial] (150 mg, 0.591 mmol, 1 equiv) and (2- (trifluoromethyl)phenyl)boronic acid (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(2-Chloro-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (2-chloro-6- methoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 tert-Butyl 4-(4-bromo-3,5-dichlorophenyl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (85% yield), using 2- bromo-l,3-dichloro-5-iodobenzene (500 mg, 1.429 mmol, 1.25 equiv) and tert-butyl piperazine-1- carboxylate [commercial] (1 equiv) as starting materials. After completion the solution was filtered through Celite®, evaporated and the resulting crude was used in the next step.
  • Step 3 l-(4-(4-Bromo-3,5-dichlorophenyl)piperazin-l-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (93% yield over two steps), using l-(4-bromo-3,5-dichlorophenyl)piperazine trifluoroacetate and acetic anhydride as starting materials.
  • Step 4 Methyl 2-(5-(4-(4-acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(4-bromo-3,5-dichlorophenyl)piperazin-l-yl)ethan-l-one (80 mg, 0.229 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated
  • Step 5 2-(5-(4-(4-Acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (10 mg, 10% yield two steps), using methyl 2-(5-(4-(4-acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(pyridin-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.317 mmol, 1 equiv) and 4-bromopyridine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
  • Step 2 2-(5-(Pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (7 mg, 11% yield over two steps), using methyl 2-(5-(pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(lH-benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (150 mg, 0.765 mmol, 1 equiv) and 4-bromo-lH-benzo[d]imidazole [commercial] (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
  • Step 2 2-(5-(lH-Benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (20 mg, 9% yield over two steps), using methyl 2-(5-(lH-benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material. After completion the solvents were removed under reduced pressure and the residue was taken up in water, washed with ethyl acetate, acidified with IM HCI and extracted with ethyl acetate. The organic layer was washed with water, brine, dried over Na2SO 4 and evaporated to obtain pure product.
  • Step 1 Methyl 2-(5-(2-(/V,/V-dimethylsulfamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (180 mg, 0.57 mmol, 1.5 equiv) and 2-bromo-/V,/V-dimethylbenzenesulfonamide [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv). After completion the reaction mixture was evaporated to give crude product which was used directly in the next step.
  • Step 1 2-(5-(2-Chloro-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (14% yield), using 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (50 mg, 0.197 mmol, 1 equiv) and (2-chloro-6- (trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 To the stirred solution of 2-bromo-l-chloro-3-iodobenzene [commercial] (120 mg, 0.342 mmol, 1.11 equiv), Xantphos (0.22 equiv), Pd?(dba)3 (0.011 equiv) and DIPEA (3.33 equiv) in dioxane (2 mL) was added (4-methoxyphenyl)methanethiol (0.044 mL, 0.308 mmol, 1 equiv). The suspension was purged with argon for 15 min and the reaction mixture was stirred at 90°C for 5 h in a sealed tube. After completion of the reaction, the solvent was evaporated and (2-bromo-3-chlorophenyl)(4-methoxybenzyl)sulfane (90 mg, 69% yield) was purified by flash column chromatography.
  • Step 2 To a stirred solution of (2-bromo-3-chlorophenyl)(4-methoxybenzyl)sulfane (420 mg, 1.22 mmol, 1 equiv) in ACN (15 mL), cooled in an ice-water bath, were added acetic acid (0.5 mL) and water (0.5 mL). l,3-Dichloro-5,5-dimethylimidazolidine-2, 4-dione (3 equiv) was added and the resulting mixture was stirred at 0°C for 1 h. After completion, the reaction was quenched with cold water and extracted with DCM. The organic fraction was washed with brine, dried over NajSCU and evaporated to afford crude 2- bromo-3-chlorobenzenesulfonyl chloride (400 mg) which was used directly in the next step.
  • Step 3 l-((2-Bromo-3-chlorophenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (43% yield over two steps), using 2-bromo-3- chlorobenzenesulfonyl chloride (781 mg, 2.69 mmol, 1 equiv) and piperidine (1.1 equiv) as starting materials.
  • Step 4 Methyl 2-(5-(2-chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-3-chlorophenyl)sulfonyl)piperidine (100 mg, 0.295 mmol, 1.11 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs 2 CO 3 (4.4 equiv) as base and Pd(dtbpf)CI 2 as catalyst (0.36 equiv). After completion of the reaction, the solvent was evaporated to give crude product which was used directly in the next step.
  • Step 5 2-(5-(2-Chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over three steps), using methyl 2-(5-(2-chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate.
  • Example 1-031 Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 47)
  • Step 1 l-((2-Bromophenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (46% yield), using 2-bromobenzenesulfonyl chloride (532 mg, 2.09 mmol, 1 equiv) and 4-methoxypiperidine (1.25 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (100 mg, 0.3 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion of the reaction, solvent was evaporated, quenched with IM HCI and extracted with dichloromethane to
  • Step 3 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (13% yield over two steps), using methyl 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromophenyl)sulfonyl)-3-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (46% yield), using 2-bromobenzenesulfonyl chloride (1.1 g, 4.17 mmol, 1 equiv) and 3-methoxypiperidine (1.25 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-((3-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-3-methoxypiperidine (100 mg, 0.3 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was acidified with IM HCI and extracted with ethyl acetate to give crude
  • Step 3 2-(5-(2-((3-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (28% yield over two steps), using methyl 2-(5-(2-((3-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromophenyl)sulfonyl)-4-(2-methoxyethoxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (28% yield), using 2- bromobenzenesulfonyl chloride (385 mg, 1.50 mmol, 1 equiv) and 4-(2-methoxyethoxy)piperidine [commercial] (1.25 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-((4-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-(2-methoxyethoxy)piperidine (100 mg, 0.265 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base and Pd(dppf)CI 2 as a catalyst (0.1 equiv). After completion of the reaction, solvent was evaporated, quenched with
  • Step 3 2-(5-(2-((4-(2-Methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over two steps), using methyl 2-(5-(2-((4-(2-methoxyethoxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromophenyl)sulfonyl)-4-(methoxymethyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (59% yield), using 2- bromobenzenesulfonyl chloride (300 mg, 1.176 mmol, 1 equiv) and 4-(methoxymethyl)piperidine [commercial] (1.25 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-((4-(methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-(methoxymethyl)piperidine (88 mg, 0.253 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, Cs 2 CO 3 (5 equiv) as base and Pd(dtbpf)CI 2 as a catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product
  • Step 3 2-(5-(2-((4-(Methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (6% yield over two steps), using methyl 2-(5-(2-((4-(methoxymethyl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(4-(Piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (36% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (60 mg, 0.236 mmol, 1 equiv) and l-(4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)phenyl)piperidine [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Example 1-036 Synthesis of 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 34)
  • Step 1 l-(4-Bromo-3,5-dichlorophenyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (38% yield), using 2-bromo-l,3-dichloro-5- iodobenzene (300 mg, 0.853 mmol, 1 equiv) and piperidine [commercial] (1.6 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-bromo-3,5-dichlorophenyl)piperidine (100 mg, 0.324 mmol, 1 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
  • Step 3 2-(5-(2,6-Dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (7% yield over two steps), using methyl 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(3-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (36% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (3- methoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(3-(Trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (40% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (3- (trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 l-(4-Bromo-3,5-dichlorophenyl)-3-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (62% yield), using 2-bromo-l,3- dichloro-5-iodobenzene (500 mg, 1.421 mmol, 1 equiv) and 3-methoxypiperidine [commercial] (1.6 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2,6-dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-bromo-3,5-dichlorophenyl)-3-methoxypiperidine (200 mg, 0.59 mmol, 1 equiv) and methyl 2- (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
  • Step 3 2-(5-(2,6-Dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (10% yield over two steps), using methyl 2-(5-(2,6-dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(6-bromo-3,4-dihydroisoquinolin-2(lH)-yl)ethan-l-one [commercial] (80 mg, 0.316 mmol, 1.0 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was
  • Step 2 2-(5-(2-Acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (24% yield over two steps), using methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Example 1-041 Synthesis of 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 30) Step 1: Methyl 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2- (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (100 mg, 0.316 mmol, 1.25 equiv) and (3-bromophenyl)(piperidin-l-yl)methanone [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbp
  • Step 2 2-(5-(3-(Piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(4-(tert-Butyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (44% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.394 mmol, 1 equiv) and (4-(tert- butyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(4-Cyclopropylphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (63% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (4- cyclopropylphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (28% yield), using 2-bromo-3- methoxybenzenesulfonyl chloride [commercial] (400 mg, 1.404 mmol, 1 equiv) and piperidine (2 equiv) as starting materials.
  • Step 2 2-(5-(2-Methoxy-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (29% yield), using l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidine (120 mg, 0.36 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 4-Bromo-3,5-dichloro-/V,/V-dimethylaniline was synthesized using the general procedure shown in
  • Step 2 2-(5-(2,6-Dichloro-4-(dimethylamino)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (39% yield), using 4-bromo-3,5-dichloro-/V,/ ⁇ /-dimethylaniline (60 mg, 0.3 mmol, 1.33 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv).
  • Step 1 2-(5-(4-(Prop-l-yn-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (27% yield), using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.309 mmol, 1.03 equiv) and l-bromo-4-(prop-l-yn-l-yl)benzene (1 equiv) as starting materials, Cs 2 CO 3 (4.1 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv). l-Bromo-4-(prop-l-yn-l-yl)benzene was synthesized as described in Fujihara, T. et
  • Step 1 4-(2-Bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(2- bromo-3-(trifluoromethyl)phenoxy)piperidine-l-carboxylate (600 mg, 1.415 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was forwarded into the next step. tert-Butyl 4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidine-l-carboxylate was prepared according to the procedure described in WO2018217809A1.
  • Step 2 l-(4-(2-Bromo-3-(trifluoromethyl)phenoxy)piperidin-l-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (46% yield over two steps), using 4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate (300 mg, 0.68 mmol, 1 equiv) and acetyl chloride (1.5 equiv) as starting materials.
  • Step 3 Methyl 2-(5-(2-((l-acetylpiperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidin-l-yl)ethan-l-one (110 mg, 0.328 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base and Pd(dtbpf)CI 2 as catalyst (0.11 equiv). After completion of the reaction the solvent was evaporated to give crude
  • Step 4 2-(5-(2-((l-Acetylpiperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(2-((l-acetylpiperidin-4-yl)oxy)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Example 1-048 Synthesis of 2-(5-(2-((4-methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 17)
  • Step 1 To a stirred solution of 2-bromo-3-(trifluoromethyl)phenol [commercial] (600 mg, 2.5 mmol, 1 equiv) in THF were added 4-methoxycyclohexan-l-ol [commercial] (391 mg, 3.0 mmol, 1.2 equiv) and PPha (980 mg, 3.75 mmol, 1.5 equiv) at 0°C and the suspension was stirred for 10 min.
  • Step 2 Methyl 2-(5-(2-((4-methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-l-((4-methoxycyclohexyl)oxy)-3-(trifluoromethyl)benzene (55 mg, 0.156 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)CI 2 as catalyst (0.1 equiv). After completion, the reaction mixture was quenched with water and extracted with ethy
  • Step 3 2-(5-(2-((4-Methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (31% yield over two steps), using methyl 2-(5-(2-((4-methoxycyclohexyl)oxy)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 4-(2-Bromo-3-(trifluoromethyl)phenoxy)-l-(methylsulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (64% yield), using 4-(2- bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate (300 mg, 0.69 mmol, 1 equiv) and methanesulfonyl chloride (2 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-((l-(methylsulfonyl)piperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4-(2-bromo-3-(trifluoromethyl)phenoxy)-l-(methylsulfonyl)piperidine (130 mg, 0.323 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated to give crude product which
  • Step 3 2-(5-(2-((l-(Methylsulfonyl)piperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin- 3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (16% yield over two steps), using methyl 2-(5-(2-((l-(methylsulfonyl)piperidin-4-yl)oxy)- 6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(5-methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-4-methoxy-l-(trifluoromethyl)benzene [commercial] (150 mg, 0.588 mmol, 1.0 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite®, the solvent was evaporated and the crude product was used in the next step without additional purification.
  • Step 2 2-(5-(5-Methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(5-methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(4-(trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 3-bromo- 4-(trifluoromethyl)pyridine [commercial] (150 mg, 0.664 mmol, 1 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, Cs 2 CO 3 (2.7 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
  • Step 2 2-(5-(4-(Trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (20% yield over two steps), using methyl 2-(5-(4-(trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 tert-Butyl 4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridine-l(2H)-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (86% yield), using 2-bromo-4-iodo-l-(trifluoromethyl)benzene [commercial] (2.5 g, 7.143 mmol, l.ll equiv) and tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-l(2H)-carboxylate (1 equiv) as starting materials, K 3 PO 4 (3.33 equiv) as base and Pd(PPh 3 ) 2 Cl2 as catalyst (0.11 equiv).
  • Step 2 4-(3-Bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridine-l(2H)-carboxylate (500 mg, 1.232 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure, the residue was washed with ether and dried to afford crude product which was directly forwarded into the next step.
  • Step 3 l-(4-(3-Bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (70% yield over two steps), using 4-(3-bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (350 mg, 0.835 mmol, 1 equiv) and acetyl anhydride (1.5 equiv) as staring materials.
  • Step 4 Methyl 2-(5-(5-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridin-l(2H)- yl)ethan-l-one (110 mg, 0.316 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base and Pd(dtbpf)CI
  • Step 5 2-(5-(5-(l-Acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (57% yield over two steps), using methyl 2-(5-(5-(l-acetyl-l,2,3,6- tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.219 mmol) as starting material. After completion the solvents were removed under reduced pressure, the residue was triturated with diluted HCI and dried to afford pure product.
  • Step 6 2-(5-(5-(l-Acetylpiperidin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (7% yield), using 2-(5-(5-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (80 mg, 0.181 mmol) as starting material.
  • Step 1 4-(3-Bromo-4-(trifluoromethyl)phenyl)-l-(methylsulfonyl)-l,2,3,6-tetrahydropyridine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (64% yield), using 4-(3-bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (350 mg,
  • Step 2 Methyl 2-(5-(5-(l-(methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2-
  • Step 3 2-(5-(5-(l-(Methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2-
  • Step 4 2-(5-(5-(l-(Methylsulfonyl)piperidin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (27% yield), using 2-(5-(5-(l-(methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2- (trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (80 mg, 0.167 mmol) as starting material.
  • Example 1-054 Synthesis of 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 19)
  • Step 1 Methyl 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (135 mg, 0.425 mmol, 1.1 equiv) and 2-bromo-4-chloro-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(d
  • Step 2 2-(5-(5-Chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (25% yield over 2 steps), using methyl 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as a starting material.
  • Step 1 2-(5-(5-Fluoro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (65% yield), using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (79 mg, 0.247 mmol, 1 equiv) and 2-bromo-4-fluoro-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv). LCMS (ESI+) m/z 339.0 [M+H]
  • Step 1 Methyl 2-(5-(5-methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.315 mmol, 1 equiv) and 2-bromo-4-methyl-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
  • Step 2 2-(5-(5-Methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (15% yield over two steps), using methyl 2-(5-(5-methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as starting material.
  • Step 1 Methyl 2-(5-(3-(trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (105 mg, 0.332 mmol, 1 equiv) and 4-bromo-3-(trifluoromethyl)pyridine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the crude product was used in the next step without additional purification.
  • Step 2 2-(5-(3-(Trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (22% yield over two steps), using methyl 2-(5-(3-(trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as a starting material.
  • Step 1 3'-Bromo-4-methoxy-4'-(trifluoromethyl)-2,3,4,5-tetrahydro-l,l'-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (41% yield), using 2- bromo-4-iodo-l-(trifluoromethyl)benzene (280 mg, 0.8 mmol, 1.11 equiv) and 2-(4-methoxycyclohex-l- en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K 3 PO 4 (3.33 equiv) as base and Pd(PPh 3 ) 2 Cl2 as catalyst (0.11 equiv).
  • Step 2 Methyl 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-3- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 3'-bromo-4-methoxy-4'-(trifluoromethyl)-2, 3,4,5- tetrahydro-l,l'-biphenyl (120 mg, 0.359 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Ch as
  • Step 4 2-(5-(5-(4-Methoxycyclohexyl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (16% yield over three steps), using 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,r- biphenyl]-3-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
  • Step 1 To a stirred solution of 2-bromo-3-(trifluoromethyl)phenol (200 mg, 0.83 mmol, 1 equiv) in THF (3 mL), cooled in an ice-water bath, were added PPh 3 (1.5 equiv), DIAD (1.5 equiv) and cyclohexanol (1 equiv) and the resulting mixture was stirred at RT for 16 h. After completion, the reaction was quenched with saturated NaHCO 3 solution and extracted with ethyl acetate. The combined organic fractions were washed with brine, dried over NajSCU and evaporated to give crude product. 2-Bromo-l-(cyclohexyloxy)-3- (trifluoromethyl)benzene (160 mg, 0.49 mmol, 59% yield) was purified by flash column chromatography.
  • Step 2 Methyl 2-(5-(2-(cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-l-(cyclohexyloxy)-3-(trifluoromethyl)benzene (100 mg, 0.311 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (3 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the solvents were evaporated, the residue was quenched with water, extracted with ethyl acetate and dried over
  • Step 3 2-(5-(2-(Cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(2-(cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 To a stirred solution of 2-bromo-l,4-diiodobenzene (1.0 g, 2.45 mmol, 1 equiv) in dioxane (5 mL) were added (4-methoxyphenyl)methanethiol (1.9 equiv) and DIPEA (6 equiv). The mixture was then bubbled with argon for 10 min and XantPhos (0.2 equiv) and Pd?(dba)3 (0.1 equiv) were added and the mixture was further bubbled with argon for 5 min and stirred in a sealed tube at 90°C for 3 h.
  • Step 2 To a stirred solution of (2-bromo-l,4-phenylene)bis((4-methoxybenzyl)sulfane) (340 mg, 0.74 mmol, 1 equiv) in ACN (5 mL) were added AcOH (0.3 mL), water (0.3 mL) and the solution was cooled to 0°C. After 10 min l,3-dichloro-5,5-dimethylhydantoin (6 equiv) was added and the reaction was stirred at RT for 1 h. After completion, the reaction was quenched with water and extracted with DCM. The combined organic fractions were washed with brine, dried over NajSCU and evaporated to afford crude 2- bromobenzene-l,4-disulfonyl dichloride, which was forwarded directly for the next step.
  • Step 3 l,l'-(2-Bromo-l,4-phenylenedisulfonyl)bis(4-methoxypiperidine) was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (43% yield over two steps), using 2-bromobenzene-l,4-disulfonyl dichloride (260 mg, 0.739 mmol, 1 equiv) and 4-methoxypiperidine (37 equiv) as starting materials.
  • Step 1 2-(5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (73% yield), using 2-(5-bromo-4- methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile (700 mg, 2.789 mmol) as starting material. After completion the reaction mixture was diluted with cold water, washed with ether and acidified with IM HCI. The precipitated product was filtered and dried to afford pure product.
  • Step 3 Methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (46% yield), using methyl 2-(5-bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetate (500 mg, 1.943 mmol, 1 equiv) as starting material.
  • Step 4 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (3.8% yield), using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (100 mg, 0.299 mmol, 1 equiv) and methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst.
  • Step 1 2-(5-(2,5-Bis((4-methoxypiperidin-l-yl)sulfonyl)phenyl)-4-methylpyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (26% yield), using l,l'-(2-bromo-l,4-phenylenedisulfonyl)bis(4-methoxypiperidine) (120 mg, 0.235 mmol, 1 equiv) and methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 1 Methyl 2-(5-(2,6-bis(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo- l,3-bis(trifluoromethyl)benzene [commercial] (100 mg, 0.316 mmol, 1.25 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.23 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
  • Step 2 2-(5-(2,6-Bis(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield over 2 steps), using methyl 2-(5-(2,6-bis(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 2-(5-(p-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and p-tolylboronic acid [commercial] (1.5 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base, Pd(dtbpf)CI 2 as catalyst (0.3 equiv).
  • Step 1 Methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (100 mg, 0.316 mmol, 1.25 equiv) and l-(7-bromo-3,4-dihydroisoquinolin-2(lH)-yl)ethan-l-one [commercial] (1 equiv) as starting materials, CS2CO3 (5 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.125 equiv). After completion the solvents were evaporated, the residue was quenched with
  • Step 2 2-(5-(2-Acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (43% yield over two steps), using methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromo-3-(trifluoromethyl)phenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (47% yield), using 2-bromo-3- (trifluoromethyl)benzenesulfonyl chloride [commercial] (185 mg, 0.572 mmol, 1 equiv) and piperidine (3 equiv) as starting materials.
  • Step 2 Methyl 2-(5-(2-(piperidin-l-ylsulfonyl)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-3-(trifluoromethyl)phenyl)sulfonyl)piperidine (50 mg, 0.135 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
  • Step 3 2-(5-(2-(Piperidin-l-ylsulfonyl)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over two steps), using methyl 2-(5-(2-(piperidin-l-ylsulfonyl)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromo-5-methylphenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (67% yield), using 2-bromo-5- methylbenzenesulfonyl chloride [commercial] (150 mg, 0.557 mmol, 1 equiv) and piperidine (2 equiv) as starting materials. After completion the reaction mixture was diluted with cold water and extracted with DCM. The organic fraction was dried over NajSCU and evaporated to give crude product.
  • Step 2 Methyl 2-(5-(4-methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-5-methylphenyl)sulfonyl)piperidine (60 mg, 0.189 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dppf)Ck as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
  • Step 3 2-(5-(4-Methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (44% yield over two steps), using methyl 2-(5-(4-methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
  • Step 1 To a stirred solution of tert-butyl 3-hydroxypiperidine-l-carboxylate (1.0 g, 4.97 mmol, 1 equiv) in THF (6 mL), cooled in an ice-water bath, was added NaH (2 equiv, 60% suspension in mineral oil) followed by l-bromo-2-methoxyethane (1.2 equiv). The reaction mixture was stirred at RT for 3 h, quenched with cold water and extracted with DCM. Combined organic fractions were dried over NajSCU and evaporated. tert-Butyl 3-(2-methoxyethoxy)piperidine-l-carboxylate (800 mg, 3.1 mmol, 62% yield) was purified by flash column chromatography.
  • Step 2 3-(2-Methoxyethoxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (47% yield), using tert-butyl 3-(2- methoxyethoxy)piperidine-l-carboxylate (300 mg, 1.15 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure to afford crude product which was directly forwarded to the next step.
  • Step 3 l-((2-Bromophenyl)sulfonyl)-3-(2-methoxyethoxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (76% yield), using 3-(2- methoxyethoxy)piperidine trifluoroacetate (700 mg, 2.76 mmol, 1 equiv) and 2-bromobenzenesulfonyl chloride (1.2 equiv) as starting materials.
  • Step 4 Methyl 2-(5-(2-((3-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-3-(2-methoxyethoxy)piperidine (100 mg, 0.265 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dppf)Ck as catalyst (0.1 equiv). After completion the reaction was quenched with IM HCI and the volatile
  • Step 5 2-(5-(2-((3-(2-Methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield over two steps), using methyl 2-(5-(2-((3-(2-methoxyethoxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
  • Step 1 l-((2-Bromophenyl)sulfonyl)-3-(methoxymethyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (51% yield), using 2- bromobenzenesulfonyl chloride (200 mg, 0.784 mmol, 1 equiv) and 3-(methoxymethyl)piperidine (1.2 equiv) as starting materials.
  • Step 2 2-(5-(2-((3-(Methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (32% yield), using l-((2-bromophenyl)sulfonyl)-3-(methoxymethyl)piperidine (88 mg, 0.253 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.25 equiv) as starting materials, CS2CO3 (5 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.126 equiv).
  • Step 1 4-Methoxy-l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (98% yield), using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (3 g, 9.009 mmol, 1 equiv) as starting material.
  • Step 2 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (37% yield), using 4-methoxy-l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine (100 mg, 0.262 mmol, 1 equiv) and 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
  • Step 3 To a stirred solution of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin- 3-yl)acetonitrile (40 mg, 0.097 mmol, 1 equiv) in DMF (1 mL) were added sodium azide (20 mg, 0.292 mmol, 3 equiv) and zinc bromide (65 mg, 0.292 mmol, 3 equiv) and the reaction mixture was stirred at 140°C for 16 h.
  • Example 1-071 Synthesis of 2-(5-(2-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1002)
  • Step 1 2-(5-(2-((4-(tert-Butoxycarbonyl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using tert-butyl 4-((2-bromophenyl)sulfonyl)piperazine-l-carboxylate (500 mg, 1.235 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into
  • Step 3 To a stirred solution of 2-(5-(2-(piperazin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid trifluoroacetate (200 mg, 0.39 mmol, 1 equiv) in DMSO (2 mL) were added 2-(2,6-dioxopiperidin-3- yl)-5,6-difluoroisoindoline-l, 3-dione (1.1 equiv) and DIPEA (2 equiv) at RT. The reaction mixture was stirred at 110°C for 6 h.
  • Example 1-072 Synthesis of 2-(5-(2-((4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1003)
  • Step 1 tert-Butyl 4-(l-((2-bromophenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (68% yield), using 2-bromobenzenesulfonyl chloride (500 mg, 1.961 mmol, 1 equiv) and tert-butyl 4-(piperidin-4- yl)piperazine-l-carboxylate [commercial] (1.1 equiv) as starting materials.
  • Step 2 tert-Butyl 4-(l-((2-(3-(2-methoxy-2-oxoethyl)pyrazolo[l,5-o]pyridin-5- yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using tert-butyl 4-(l-((2- bromophenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate (150 mg, 0.308 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf
  • Step 3 2-(5-(2-((4-(4-(tert-Butoxycarbonyl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using tert-butyl 4-(l-((2-(3-(2-methoxy-2-oxoethyl)pyrazolo[l,5-o]pyridin-5- yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate as starting material.
  • Step 4 2-(5-(2-((4-(Piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using 2-(5-(2-((4-(4-(tert-butoxycarbonyl)piperazin-l-yl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
  • Step 5 To a stirred solution of 2-(5-(2-((4-(piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid trifluoroacetate (130 mg, 0.218 mmol, 1.11 equiv) in DMSO (3 mL) were added 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-l, 3-dione (1 equiv) and DIPEA (3 equiv) at RT. The reaction mixture was stirred at 110°C for 2 h.
  • Example 1-073 Synthesis of (2-(5-(2-((4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate
  • Step 1 To a stirred solution of 2-(5-(2-((4-(4-(tert-butoxycarbonyl)piperazin-l-yl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (150 mg, 0.257 mmol, 1 equiv) in DMF (4 mL), cooled in an ice-water bath, were added chloromethyl pivalate (1.5 equiv) and KHCO 3 (2 equiv). The reaction mixture was stirred at RT for 2 h. After completion the solution was diluted with cold water and extracted with ethyl acetate.
  • Step 2 (2-(5-(2-((4-(Piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(l-((2-(3-(2-oxo-2- ((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-o]pyridin-5-yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l- carboxylate as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
  • Step 3 To a stirred solution of (2-(5-(2-((4-(piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate trifluoroacetate (130 mg, 0.183 mmol, 1.11 equiv) in DMSO (3 mL) were added 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-l, 3-dione (1 equiv) and DIPEA (3 equiv) at RT. The reaction mixture was stirred at 110°C for 2 h.
  • Example 1-074 Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)propanoic acid (Compound 49, S (left)
  • Step 1 To a stirred solution of 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetonitrile (500 mg, 2.1 mmol, 1 equiv) in THF (5 mL), cooled to -78°C, was added NaHMDS (3.2 mL, 3.2 mmol, 1.5 equiv, IM solution in THF) and the reaction was stirred for 40 min at the same temperature, lodomethane (1.5 equiv) was added and the reaction mixture was stirred for 1 h at RT, then quenched with water and extracted with ethyl acetate.
  • Step 2 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)propanoic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (86% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)propanenitrile (150 mg, 0.6 mmol) as a starting material.
  • Step 3 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)propanoic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)propanoic acid (150 mg, 0.56 mmol, 1 equiv) and 4-methoxy- l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine (1 equiv) as starting materials, Cs 2 CO 3 (4 equiv) as base and Pd(dtbpf)CI 2 as catalyst (0.1 equiv). The product (mixture of stereoisomers) was purified by preparative HPLC.
  • Absolute configuration of the stereoisomers was assigned based on crystal structure.
  • Example 1-075 Synthesis of 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin- l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1007)
  • Step 1 To a stirred suspension of sodium hydride (173 mg, 60% in mineral oil, 7.2 mmol, 1.5 equiv) in THF (3 mL) and DCM (3 mL), cooled to 0°C, was added l-phenyl-2,5,8,ll,14,17,20-heptaoxadocosan-22-ol [commercial] (2 g, 4.802 mmol, 1 equiv) and the resulting mixture was stirred at RT for 30 min. 1-Chloro- 6-iodohexane (2.36 g, 9.604 mmol, 2 equiv) was added at 0°C and the reaction mixture was stirred at RT for 16 h.
  • Step 2 To a solution of 29-chloro-l-phenyl-2,5,8,ll,14,17,20,23-octaoxanonacosane (2 g, 3.73 mmol) in methanol (5 mL) was added Pd/C (280 mg, 10% wt.) and the reaction mixture was stirred at RT under hydrogen atmosphere for 16 h. After completion the solution was filtered through Celite® and evaporated to yield 27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosan-l-ol (1.58 g, 94% yield).
  • Step 3 To a solution of 27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosan-l-ol (180 mg, 0.405 mmol, 1 equiv) in DCM (2 mL), cooled in an ice-water bath, were added TEA (0.17 mL, 1.216 mmol, 3 equiv), DMAP (catalytic) and 4-methylbenzene-l-sulfonyl chloride (115 mg, 0.608 mmol, 1.5 equiv) and the resulting solution was stirred at RT for 3 h. After completion the reaction was diluted with water and extracted with DCM. The organic fraction was dried over Na 2 SO 4 and evaporated to give crude product. 27-Chloro- 3,6,9,12,15,18,21-heptaoxaheptacosyl 4-methylbenzenesulfonate (220 mg, 90% yield) was purified by flash column chromatography.
  • Step 4 To a suspension of sodium hydride (10 mg, 60% in mineral oil, 0.417 mmol, 2.5 equiv) in DMF (2 mL), cooled in an ice-water bath, was added tert-butyl 4-hydroxypiperidine-l-carboxylate [commercial] (100 mg, 0.167 mmol, 1 equiv) and the mixture was stirred at 0°C for 30 min. 27-Chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl 4-methylbenzenesulfonate (100 mg, 0.167 mmol, 1 equiv) was added and the reaction mixture was stirred at RT for 16 h.
  • Step 5 4-((27-Chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (97% yield), using tert-butyl 4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine-l-carboxylate (1 g, 1.59 mmol). After completion the volatiles were removed under reduced pressure and the product was purified by multiple azeotropic evaporation with toluene.
  • Step 6 l-((2-Bromophenyl)sulfonyl)-4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (56% yield), using 4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine trifluoroacetate
  • Step 7 Methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-((27- chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine (200 mg, 0.268 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials,
  • Step 8 2-(5-(2-((4-((27-Chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (51 mg, 22% yield over two steps), using methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (250 mg, 0.292 mmol) as starting material.
  • Example 1-076 Synthesis of methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (Compound 1006)
  • Step 1 To a stirred solution of 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (35 mg, 0.042 mmol, 1 equiv) in methanol (1 mL) and ethyl acetate (1 mL) was added trimethylsilyldiazomethane (2M solution in hexanes, 5 equiv) at -10°C and stirred at RT for 1 h.
  • Example 1-077 Synthesis of methyl 2-(5-(2-((4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin- l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (Compound 1004) and 2-(5-(2-((4-((21-chloro- 3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
  • Step 1 To a solution of 21-chloro-3,6,9,12,15-pentaoxahenicosan-l-ol [commercial] (700 mg, 1.96 mmol, 1 equiv) in DCM (12 mL), cooled in an ice-water bath, were added TEA (794 mg, 7.86 mmol, 4 equiv), DMAP (catalytic) and 4-methylbenzene-l-sulfonyl chloride (1.3 g, 6.69 mmol, 3.4 equiv) and the reaction mixture was stirred at RT for 3 h. After completion the solution was diluted with water and extracted with ethyl acetate. The organic fraction was dried over Na 2 SO 4 and evaporated to give crude product. 21- Chloro-3,6,9,12,15-pentaoxahenicosyl 4-methylbenzenesulfonate (370 mg, 37% yield) was purified by flash column chromatography.
  • Step 3 4-((21-Chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine-l-carboxylate (300 mg, 0.55 mmol). After completion the volatiles were removed under reduced pressure and the residue was triturated with pentane to provide crude product which was used directly in the next step.
  • Step 4 l-((2-Bromophenyl)sulfonyl)-4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (38% yield over three steps), using 4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine trifluoroacetate (300 mg, 0.54 mmol, 1 equiv) and 2-bromobenzenesulfonyl chloride (1.18 equiv) as starting materials.
  • Step 5 Methyl 2-(5-(2-((4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(2-((4-((21-chloro-3,6,9,12,15- pentaoxahenicosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid were synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine (120 mg, 0.18 mmol,
  • Step 1 2-(5-(2-Cyanophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (30% yield), using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (15 mg, 0.047 mmol, 1 equiv) and 2-bromobenzonitrile [commercial] (1 equiv) as starting materials, PDFPPhah (0.1 equiv) as catalyst and sodium carbonate (2 equiv) as base.
  • Step 1 l-((2-Bromo-5-methoxyphenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (59% yield), using 2-bromo-5- methoxybenzenesulfonyl chloride [commercial] (8 g, 28.07 mmol, 1 equiv) and 4-methoxypiperidine (1 equiv) as starting materials.
  • Step 2 To a stirred solution of l-((2-bromo-5-methoxyphenyl)sulfonyl)-4-methoxypiperidine (1.5 g, 4.12 mmol, 1 equiv) in DMF (15 mL) was added LiCI (10 equiv) and the reaction mixture was stirred at 150°C for 30 h. The reaction was cooled, quenched with ice-water and extracted with ethyl acetate. The organic fractions were combined, washed with brine, dried over NajSCU and concentrated under reduced pressure. 4-Bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)phenol (700 mg, 2 mmol, 48% yield) was purified by flash column chromatography.
  • Step 3 (TLS-561, lnt-8, step 5A) To a stirred solution of 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (6.5 g, 25.59 mmol, 1 equiv) in DMF (5 mL) were added KHCO 3 (2 equiv) and chloromethyl pivalate (1.5 equiv) and the resulting solution was stirred at RT for 16 h.
  • reaction mixture was quenched with ice-water and extracted with ethyl acetate to provide (2-(5-bromopyrazolo[l,5-o]pyridin- 3-yl)acetoxy)methyl pivalate (7.5 g, 20.3 mmol, 79% yield).
  • Step 4 (TLS-561, lnt-8, step 6A) (2-(5-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (86% yield), using (2-(5-bromopyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate (8.2 g, 22.22 mmol, 1 equiv) as starting material.
  • Step 5 (2-(5-(4-Hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (15% yield), using 4-bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)phenol (400 mg, 1.14 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate (1.2 equiv) as starting materials, Pd(dtbpf)CI 2 (0.1 equiv) as catalyst and Cs 2 CO 3 (2 equiv) as base.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention relates to compounds which can bind to KLHDC2 (Kelch Domain-Containing Protein 2) E3 ubiquitin ligase with high-affinity, and bifunctional degraders containing such compounds.

Description

KLHDC2 (KELCH DOMAIN-CONTAINING PROTEIN 2) LIGASE LIGANDS
FIELD OF THE INVENTION
The present invention relates to compounds which can bind to KLHDC2 (Kelch Domain-Containing Protein 2) E3 ubiquitin ligase with high-affinity, and bifunctional degraders containing such compounds.
BACKGROUND
In recent years targeted protein degradation (TPD) has emerged as a new and promising pharmaceutical modality beyond the established occupancy-driven paradigm intrinsic for protein inhibitors. Among others, TPD uses bifunctional molecules consisting of E3 ubiquitin ligase ligand connected to a warhead for degradation of medically relevant proteins. TPD holds great promise to address so far incurable diseases, nevertheless, a paucity of known E3 ligase ligands is a major impediment to advancing the field. From about 600 known E3s expressed in human cells, only a handful have been exploited for TPD applications. Among them, the cereblon (CRBN) and von Hippel-Lindau (VHL) E3 ubiquitin ligases have sparked the most interest due to the availability of chemical probes with favorable physiochemical properties and their well-studied mechanism of target engagement. Besides, both ligases have shown high efficiency and versatility in degrading proteins in the cytoplasm and nucleus. However, the pharmacological intervention by using the CRBN and VHL ligands is associated with certain risks, such as impairment of developmental processes and hematopoiesis in the case of CRBN and dysregulation of cell survival and angiogenesis in the case of VHL. In addition, cells may acquire resistance to CRBN ligands due to mutations in the ligase gene and a broad expression profile of CRBN can lead to unexpected side effects. Therefore, the employment of other E3s for this purpose could expand the therapeutic opportunities offered by TPD by broadening a substrate range, increasing efficacy, and evading drug resistance.
The Kelch-like family member 2 (KLHDC2) is a BC-box protein and a substrate receptor of the CRL2 E3 complex that is implicated in the regulation of protein stability by destruction via C-end degrons (DesCEND mechanism). A specific degron, or in the other words, destruction motif, recognized by the KLHDC2 contains a diglycine at the C terminus. This kind of sequence has been found in several substrates such as early-terminated selenoproteins (SelK and SelS) and several full-length proteins (Rusnac et al., 2018). Importantly, KLHDC2 emerged as a potent degrader in the proteome-scale induced proximity screens performed by Poirson et al. (2022) and AdPROM screen performed by Roth et al. (2022). Moreover, the utility of KLHDC2 to degrade proteins of interest was shown by using bifunctional compounds based on a degron peptide conjugated with chloroalkane (Roth et al. ,2022) or a promiscuous kinase inhibitor (Kim et al., 2022). Although a degron peptide was utilized to demonstrate a proof-of-concept, it is poorly applicable to act as an actual drug due to its high molecular weight and related bioavailability issues. Recently, the recruitment of the CRL2 KLHDC ligase complex using bifunctional compounds composed of low molecular weight KLHDC2 ligands leading to degradation of model proteins BRD4 and STAT3 (WO2023192578A1) as well as BRD4 and androgen receptor (Hickey et al., 2024) were demonstrated.
Compounds disclosed herein are characterized by a small size with favorable physicochemical properties and can be used as building blocks of bifunctional degraders for the recruitment of KLHDC2 and degradation of the protein of interest. Furthermore, two distinct classes of compounds have been developed: ligands incorporating carboxylic groups and their corresponding prodrugs designed to enhance cellular membrane permeability.
References:
1. Rusnac D.V., Lin H.C., Canzani D., Tien K.X., Hinds T.R., Tsue A.F., Bush M.F., Yen H.C.S. and Zheng N., 2018. Recognition of the diglycine C-end degron by CRL2KLHDC2 ubiquitin ligase. Mol. cell, 72(5), 813-822.
2. Poirson J., Dhillon A., Cho H., Lam M.H.Y., Alerasool N., Lacoste J., Mizan L. and Taipale M., 2022. Proteome-scale induced proximity screens reveal highly potent protein degraders and stabilizers. bioRxiv.
3. Roth S., Carton B., Sathyamurthi P.S., Watt M., Macartney T.J., Chan K.H., Isidro-Llobet A., Konopacka A., Queisser M.A. and Sapkota G., Screening of E3 Ligases Uncovers KLHDC2 as an Efficient Proximity-Induced Degrader of K-RAS, STK33, 0-catenin and FoxP3. STK33, 0-catenin and FoxP3.
4. Kim Y., Seo C., Jeon E., You I., Hwang K., Kim N., Choi H.S., Hinshaw S.M., Gray N.S. and Sim T., 2022. Targeted Kinase Degradation via the KLHDC2 Ubiquitin E3 Ligase. bioRxiv.
5. Hickey Ch.M., Digianantonio K.M., Zimmermann K., Harbin A., Quinn C., Patel A., Gareiss P., Chapman A., Tiber! B., Dobrodziej J., Corradi J., Cacace A.M., Longley D.R., Bekes M., 2023. Coopting the E3 ligase KLHDC2 for targeted protein degradation by small molecules. Nat. Struct. Mol. Biol., 31, 311-322. SUMMARY OF INVENTION
In accordance with a first aspect of the invention, there is provided a compound of formula (la): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl), -NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), and -P(O)(alkyl)2; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl or heteroaryl, each of which is independently unsubstituted or is substituted with at least one of -OH, -NH2, -NH(alkyl) or -N (al kyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CHzJqOMe, -(OCHzCFbJrNHR73, -C(O)alkyl and -S(O)2alkyl; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or - C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2 -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and ^(OjOCHjCHjSOjIVIe; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkyl)2, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen. In accordance with a second aspect of the invention, there is provided the use of a compound as defined above in a bifunctional protein degrader compound.
In accordance with a third aspect of the invention, there is provided a bifunctional protein degrader compound comprising a compound as defined above.
In accordance with a fourth aspect of the invention, there is provided a bifunctional protein degrader compound comprising a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)z; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOI\/le, -(CH2)qOI\/le, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19 and -OR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and -C(O)OCH2CH2SO2l\/le; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
In accordance with a fifth aspect of the invention, there is provided a compound of formula (II)
[KLHDC2 ligase binding moiety] - linker - [Target protein binding moiety]
CD or a salt, solvate, hydrate or isomer thereof, wherein [Target protein binding moiety] is: wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl; R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl; or
(b) wherein indicates attachment to R18 of the linker; and k is an integer from 1-10;
(c)
wherein
X4 and X5 are each independently N or CH;
X6 is N or CH;
R30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R40, or -C(O)R40;
R31 is H, -OMe, -heteroaryl, -heteroaryl-R40 or R40;
R32 is H, unsubstituted C1-C6 alkyl,
R33 is -N(C1-C6 alkyl)2, -NH(C1-C6 alkyl), -NH(aryl), or R40;
R34 is -Me or -C(O)R40; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; or wherein
X7 is N or CH;
R35 is -heterocycloalkyl-R40, or R40;
R36 is H or -OMe; and R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CH2)q0Me, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19, -OR19 and -(OCH2CH2)rR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and -C(O)OCH2CH2SO2Me; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alkylhcycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connected to R14 of the linker, wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen; and wherein [linker] has the following formula wherein
R14 is -Ci-6 alkyl, -C2.6 alkenyl, -C2.6alkynyl, C1-6 alkyl-N(C1-6 alkyl)-, -C(O)-, -SO2- or is absent
R15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, Ci.g alkyl-NH-, -Ci.g alkyl-N(Ci-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
R16 is -Ci-6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH2-C(O)-, -CH2-C(O)-NH-, -CH2-C(O)O- or is absent
R17 is -CH2(C2H4-O)y, (C2H4-O)X, (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
(a) when [Target protein binding moiety] is
, then R18 is -Ci.g alkyl, cycloalkyl,
CH2-NH-C(0)-, heterocycloalkyl, or is absent; and
(b) when [Target protein binding moiety] is
then R18 is -C1-6 alkyl-NH-, cycloalkyl-NH, -CH2-NH-C(O)-NH-, heterocycloalkyl, heterocycloalkyl-NH, or is absent.
The present invention also provides a pharmaceutical composition comprising a compound of the invention.
The present invention also provides a compound or pharmaceutical composition of the invention, for use in medicine.
The present invention also provides a method of treating a disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or pharmaceutical composition of the invention.
As used herein the term "alkyl" is intended to include both linear and branched alkyl groups, both of which either may be unsubstituted, or may be substituted by one or more additional groups. In some embodiments, the alkyl group is an unsubstituted alkyl group. In some embodiments, the alkyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the alkyl group is a C1-C12 alkyl, a C1-C10 alkyl, a Ci-Cg alkyl, a Ci-Cg alkyl, or a C1-C4 alkyl group. In some embodiments the alkyl group is a linear alkyl group. In some embodiments the alkyl group is an unsubstituted linear alkyl group. In some embodiments the alkyl group is a linear alkyl group which is substituted by one or more groups selected from -OH, -ORW, -NH2, - NHRW, -NRW2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments the alkyl group is a branched alkyl group. In some embodiments the alkyl group is an unsubstituted branched alkyl group. In some embodiments the alkyl group is a branched alkyl group which is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
As used herein the term "alkenyl" is intended to include both unsubstituted alkenyl groups, and alkenyl groups which are substituted by one or more additional groups. In some embodiments, the alkenyl group is an unsubstituted alkenyl group. In some embodiments, the alkenyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the alkenyl group is a C2-C12 alkenyl, a C2-CM alkenyl, a C2-C8 alkenyl, a C2-C6 alkenyl, or a C2-C4 alkenyl group. In some embodiments the alkenyl group is a linear alkenyl group. In some embodiments the alkenyl group is an unsubstituted linear alkenyl group. In some embodiments the alkenyl group is a linear alkenyl group which is substituted by one or more groups selected from -OH, - ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments the alkenyl group is a branched alkenyl group. In some embodiments the alkenyl group is an unsubstituted branched alkenyl group. In some embodiments the alkenyl group is a branched alkenyl group which is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
As used herein the term "alkynyl" is intended to include both unsubstituted alkynyl groups, and alkynyl groups which are substituted by one or more additional groups. In some embodiments, the alkynyl group is an unsubstituted alkynyl group. In some embodiments, the alkynyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the alkynyl group is a C2-C12 alkynyl, a C2-Cio alkynyl, a C2-Cg alkynyl, a C2-Cg alkynyl, or a C2-C4 alkynyl group. In some embodiments the alkynyl group is a linear alkynyl group. In some embodiments the alkynyl group is an unsubstituted linear alkynyl group. In some embodiments the alkynyl group is a linear alkynyl group which is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments the alkynyl group is a branched alkynyl group. In some embodiments the alkynyl group is an unsubstituted branched alkynyl group. In some embodiments the alkynyl group is a branched alkynyl group which is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
As used herein the term "cycloalkyl" is intended to include both unsubstituted cycloalkyl groups, and cycloalkyl groups which are substituted by one or more additional groups. The term "cycloalkyl" is also intended to include monocyclic and bicyclic ring systems (including spirocyclic ring systems, in which the two rings share a single atom; fused bicyclic ring systems, in which the two rings share two adjacent atoms; and bridged bicyclic ring systems, in which the two rings share three or more atoms). In some embodiments, the cycloalkyl group is an unsubstituted cycloalkyl group. In some embodiments, the cycloalkyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW2, - SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the cycloalkyl group is a C3-C12 cycloalkyl, a C4-C12 cycloalkyl, a C5-C12 cycloalkyl, a C3-C10 cycloalkyl, a C4-C10 cycloalkyl, a C5-C10 cycloalkyl, a C3-C8 cycloalkyl, a C4-C8 cycloalkyl, a C5-C8 cycloalkyl, a C3-C6 cycloalkyl, a C4-C6 cycloalkyl, a C5-C6 cycloalkyl, a C3-C4 cycloalkyl, or a C4-C5 cycloalkyl group.
As used herein the term "cycloalkenyl" is intended to include both unsubstituted cycloalkenyl groups, and cycloalkenyl groups which are substituted by one or more additional groups. In some embodiments, the cycloalkenyl group is an unsubstituted cycloalkenyl group. In some embodiments, the cycloalkenyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the cycloalkenyl group is a C4-C12 cycloalkenyl, a C5-C12 cycloalkenyl, a C4-C10 cycloalkenyl, a C5-C10 cycloalkenyl, a C4-C8 cycloalkenyl, a C5-C8 cycloalkenyl, a C4-C6 cycloalkenyl, a C5-C6 cycloalkenyl, or a C4-C5 cycloalkenyl group.
As used herein the term "heterocycloalkyl" is intended to include both unsubstituted heterocycloalkyl groups, and heterocycloalkyl groups which are substituted by one or more additional groups. The term "heterocycloalkyl" is also intended to include monocyclic and bicyclic ring systems (including spirocyclic ring systems, in which the two rings share a single atom; fused bicyclic ring systems, in which the two rings share two adjacent atoms; and bridged bicyclic ring systems, in which the two rings share three or more atoms). In some embodiments, the heterocycloalkyl group is a monocyclic ring system, a spirocyclic ring system, or a fused bicyclic ring system. In some embodiments, the heterocycloalkyl group is an unsubstituted heterocycloalkyl group. In some embodiments, the heterocycloalkyl group is substituted by one or more groups selected from -Rw, -OH, -ORW, -NH2, -NHRW, -NRW2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, one or more -CH2- groups of the heterocycloalkyl ring may be replaced with a -C(O)- group, In some embodiments, the heterocycloalkyl group is a C3-C12 heterocycloalkyl, a C4-C12 heterocycloalkyl, a C5-C12 heterocycloalkyl, a C3-C10 heterocycloalkyl, a C4-C10 heterocycloalkyl, a C5-C10 heterocycloalkyl, a C3-C8 heterocycloalkyl, a C4-C8 heterocycloalkyl, a C5-C8 heterocycloalkyl, a C3-C6 heterocycloalkyl, a C4-C6 heterocycloalkyl, a C5-C6 heterocycloalkyl, a C3-C4 heterocycloalkyl, or a C4-C5 heterocycloalkyl group.
As used herein the term "aryl" is intended to include both unsubstituted aryl groups, and aryl groups which are substituted by one or more additional groups. In some embodiments, the aryl group is an unsubstituted aryl group. In some embodiments, the aryl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -OC(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, the aryl group is a C6-C10 aryl, a C6-C8 aryl, or a Cg aryl.
As used herein the term "heteroaryl" is intended to include both unsubstituted heteroaryl groups, and heteroaryl groups which are substituted by one or more additional groups. In some embodiments, the heteroaryl group is an unsubstituted heteroaryl group. In some embodiments, the heteroaryl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl. In some embodiments, one or more -NH- groups of the heteroaryl ring may be replaced with a -NRW- group. In some embodiments, the heteroaryl group is a C6-C10 heteroaryl, a C6-C9 heteroaryl, a C6-C8 heteroaryl, or a Cg heteroaryl.
As used herein the term "fused heterocycloalkyl-heteroaryl" is intended to mean a bicyclic ring system in which one ring is a heterocycloalkyl ring and the other is a heteroaryl ring, and in which the two rings share two adjacent atoms. Of the two adjacent atoms shared by the two rings, both may be carbon atoms; both may be heteroatoms (e. g. independently O, N or S); or one may be a carbon atom and the other a heteroatom (e. g. O, N or S). The fused heterocycloalkyl-heteroaryl may be unsubstituted or may be substituted by one or more additional groups. In some embodiments, the fused heterocycloalkylheteroaryl group is an unsubstituted cycloalkenyl group. In some embodiments, the fused heterocycloalkyl-heteroaryl group is substituted by one or more groups selected from -OH, -ORW, -NH2, - NHRW, -NRW2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
As used herein the term "benzyl" is intended to include both unsubstituted benzyl groups, and benzyl groups which are substituted by one or more additional groups. In some embodiments, the benzyl group is an unsubstituted benzyl group. In some embodiments, the benzyl group is substituted by one or more groups selected from -OH, -ORW, -NH2, -NHRW, -NRW 2, -SO2RW, -C(O)RW, -CN, and -NO2, wherein each Rw is unsubstituted and is independently alkyl, cycloalkyl, alkenyl, cycloalkenyl, aryl, heteroaryl, or benzyl.
In some embodiments of any of the above aspects of the invention, all alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, aryl, heteroaryl and benzyl groups in the compounds are unsubstituted.
DETAILED DESCRIPTION OF THE INVENTION
As discussed above, in a first aspect the present invention provides a compound of formula (la): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl), -NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)z, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), and -P(O)(alkyl)2; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl or heteroaryl, each of which is independently unsubstituted or is substituted with at least one of -OH, -NH2, -NH(alkyl) or -N (alkyl >2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CH2)q0Me, -(OCH2CH2)rNHR7a, -C(O)alkyl and -S(O)2alkyl; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or - C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR9 2, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2 -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and -qojOCHjCHjSOjMe; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)( C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH( C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(0)(0H)2, - 0P(0)(0H)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyl)morpholinium cation, and N-(unsubstituted C1-C6alkyl)imidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
In some embodiments, X2 is N. In other embodiments, X3 is N.
In other embodiments, X2 is CR3 and X3 is CR1.
In certain embodiments, R1 is H, halogen or methyl.
In some embodiments, R3 is H, halogen, unsubstituted alkyl or -CN.
In certain embodiments, R1 is H. In some such embodiments, R1 and R3 are each H. In other embodiments, R1 and R2 are each H.
In certain embodiments, R2 and R3 are each H.
In certain embodiments,
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In certain embodiments, R2 is -B(OH)2, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In certain embodiments, R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In some embodiments, R2 is selected from:
n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
In some embodiments, R2 is selected from n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
In certain embodiments, R2 is selected from
wherein n is 1, 2 or 3; and wherein m is 0 or 1.
In some embodiments, R2 is
In some embodiments, each R4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), and -C(O)OH; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments, each R7 is independently selected from -Me, -OMe, -O(CH2)qOMe, -CH2OMe, -
C(O)Me and -S(O)2Me. In some embodiments, each R4 is independently selected from -F, -Cl, -lBu, -Me, -CF3, -OH, -OMe, -OCF3,
-CN, -NH2, -NMe2, , -NHC(O)Me, -S(O)2Me, -S(O)2NMe2, -CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
In some embodiments, each R6 is independently selected from halogen, haloalkyl, -NH2, - C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments, each R6 is independently selected from -Cl, -CF3, -NH2, -C(O)piperidine, -C(O)Me
In some embodiments, R2 is selected from:
In some embodiments, when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then: C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2aryl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C-S(O)2(heterocycloalkyl), C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C-SO(alkyl) and C- P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments, when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl): then: each R4 of — R4) is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments, when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH;
X2 and X3 are both CH; and
In some embodiments, when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH or -CH^O^CH^CfOfBu;
X2 and X3 are both CH; and
(i) each R4 of — ( '”R4) is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; or
In certain embodiments, R1 is methyl.
In some embodiments, -
In some embodiments, R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, - CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2. In some such embodiments, R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), -CH2B(OH)2, -CH2P(O)(OH)2, - COOR9, -B(OH)2 and -P(O)(OH)2. In some embodiments, R8 is -CHR9C(O)OR10.
In some embodiments, R8 is selected from -CH2C(O)OH, -CHMeC(O)OH, -CH2C(O)OMe, -CH2C(O)OEt, - CH2S(O)2NH2, -CH^OjOCfWqorBu, -CH2C(O)NH2, -CH2S(O)2NH2, -S(O)2NH2, -CH2-oxetane, -CH2(1,2,3- triazole), -CH2B(OH)2, -CH2P(O)(OH)2, -COOH, -COOMe, -B(OH)2 and -P(O)(OH)2. In some such embodiments, R8 is selected from -CH2C(O)OCH2OC(O)tBu, -CH2C(O)OH and -CHMeC(O)OH.
In some embodiments, R8 is -CH2C(O)OH. In other embodiments, R8 is -CHzCfOjOCHzOCfOfBu.
In some embodiments, the compound is selected from:
In some embodiments, the compound is selected from Compound nos. 1, 2, 3, 4, 5, 6, 7 , 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, 19, 20, 21, 23, 24, 26, 28, 30, 31, 33, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 51, 52, 53, 54, 55, 56, 58, 59, 60, 61, 62, 63, 64, 65, 67, 68, 69, 70, 71, 72, 73, 75, 76, 77, 78, 79, 80, 81, 82, 84, 86 and 88.
In some embodiments, R1 is H; R3 is H or methyl; R2 is aryl substituted with one or more R4 or is fused bicyclic heteroaryl substituted with one or more R6; and R8 is -CH2C(O)OH, -CH(Me)C(O)OH or CHR9(heteroaryl). In some such embodiments, each R4 is independently selected from halogen, unsubstituted alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -NHz, -N(alkyl)?, -NH(CO)alkyl, - C(O)NHalkyl, -C(O)N(alkyl)?, -S(O)? N(alkyl)2,-S(0)z(aryl), -S(O)2(heterocycloalkyl) and -C(O)OH; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R6 is -S(O)z(heterocycloalkyl), alkyl or -NH?; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments, the compound is selected from Compound nos. 1, 2, 3, 8, 10, 11, 12, 13, 14, 15, 19, 22, 24, 25, 27 , 34, 37, 38, 39, 41, 44, 45, 46, 47, 48, 50, 53, 56, 59, 64, 72, 73, 75, 76, 77, 78, 79, 81, 82, 84, 86, 90, 91, 92, 94, 96, 101, 102 and 103.
In some embodiments:
R3 is H; each R4 is independently selected from -OH, -NHz, -O(alkyl), -NHC(O)alkyl, -C(O)NHalkyl, - S(O)z(heterocycloalkyl) and -C(O)OH; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from -O(alkyl), -O(CHz)qOI\/le, -(CHz)qOI\/le and -(OCH2CHz)rNHR7a; wherein q is 1 or 2, r is 4 and R7a is -C(O)Me.
In some embodiments, the compound is Compound ID no. 2, 24, 37, 45, 46, 47, 73, 76, 77, 79, 82, 84, 86, 91, 96, 102 or 103.
In accordance with a second aspect of the invention, there is provided the use of a compound as defined in any of the above embodiments of the first aspect in a bifunctional protein degrader compound.
In accordance with a third aspect of the invention, there is provided a bifunctional protein degrader compound comprising a compound as defined in any of the above embodiments of the first aspect.
In accordance with a fourth aspect of the invention, there is provided a bifunctional protein degrader compound comprising a compound of formula (la'): wherein: X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19 and -OR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR9 2, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)pNMe2, -C(O)O(CH2)pNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CMe2OH, and -C(O)OCH2CH2SO2Me; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkyl)2, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alkyl)2cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
In the fourth embodiment, the Target protein binding moiety is a moiety which binds to a target protein. The target protein may be a protein which mediates a disease or disorder in a subject. The degradation of the target protein may thus result in a therapeutic effect in a subject treated with the bifunctional protein degrader compound. Proteins susceptible to binding with the Target protein binding moiety (and subsequent degradation by the E3 ligase - KLHDC2 ligase associated with the KLHDC2 ligase binding moiety) encompass a broad spectrum, including any protein or peptide, along with their fragments, analogs, or homologs. Target proteins exhibit diverse biological functions or activities, such as structural, regulatory, hormonal, enzymatic, genetic, immunological, contractile, storage, transportation, and signal transduction roles. Specifically, within certain embodiments, these proteins include structural proteins, receptors, enzymes, cell surface proteins, and those integral to overall cell function. This covers proteins involved in catalytic, aromatase, motor, helicase, metabolic, antioxidant, proteolytic, and biosynthetic activities, as well as proteins with kinase, oxidoreductase, transferase, hydrolase, lyase, isomerase, and ligase activities. Other categories involve enzyme regulator, signal transducer, structural molecule, and binding activities, including proteins relevant to cell motility, membrane fusion, cell communication, and the regulation of biological processes, development, cell differentiation, and response to stimuli.
These target proteins also encompass behavioral proteins, cell adhesion proteins, those involved in cell death, transport proteins, chaperone regulator proteins, nucleic acid binding proteins, transcription regulators, and proteins involved in extracellular organization and biogenesis. The scope extends to proteins from various organisms, including eukaryotes and prokaryotes, such as humans, microbes, viruses, fungi, parasites, and other animals, including domesticated ones.
Employing these diverse target proteins in screening processes by utilizing target protein binding moieties, coupled to KLHDC2 ligase binding moieties, as outlined in the present disclosure, and placing/presenting that target protein or polypeptide in proximity to an KLHDC2 ligase may lead to ubiquitination and subsequent degradation of the target protein, allows for the modulation of protein activity, bringing on to therapeutic outcomes.
Target proteins of the Target protein binding moiety according to the present invention include, for example, moieties which bind to Human BET Bromodomain-containing proteins, for example BRD4. Further, kinase warhead TL13-87 targeting 183 kinases (Huang et al., 2018) was utilized in the exemplified bifunctional compounds.
Table 1. List of kinases targeted by TL13-87 kinase warhead.
In accordance with a fifth aspect of the invention, there is provided a compound of formula (II)
[KLHDC2 ligase binding moiety] - linker - [Target protein binding moiety] CD or a salt, solvate, hydrate or isomer thereof, wherein [Target protein binding moiety] is:
wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl; or
(b) wherein indicates attachment to R18 of the linker; and k is an integer from 1-10; or
(c)
wherein
X4 and X5 are each independently N or CH;
X6 is N or CH;
R30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R40, or -C(O)R40;
R31 is H, -OMe, -heteroaryl, -heteroaryl-R40 or R40;
R32 is H, unsubstituted C1-C6 alkyl, R33 is -N(C1-C6 alkyl)2, -NH(C1-C6 alkyl), -NH(aryl), or R40;
R34 is -Me or -C(O)R40; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; or wherein
X7 is N or CH;
R35 is -heterocycloalkyl-R40, or R40;
R36 is H or -OMe; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2; R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)z, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19, -OR19 and -(OCH2CH2)rR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR9 2, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)pNMe2, -C(O)O(CH2)pNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CMe2OH, and -C(O)OCH2CH2SO2Me; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(0)NMe2;
R19 is a bond connected to R14 of the linker, wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen; and wherein [linker] has the following formula
R14.R15.R16.R17.R18 wherein
R14 is -C1-6 alkyl, -C2-6 alkenyl, -C2-6alkynyl, Ci.g alkyl-N(C1-6 alkyl)-, -C(O)-, -SO2- or is absent
R15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, C1-6 alkyl-NH-, -Ci.g alkyl-N(Ci-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
R16 is -C1-6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH2-C(O)-, -CH2-C(O)-NH-, -CH2-C(O)O- or is absent
R17 is -CH2(C2H4-O)y, (C2H4-O)X, (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
(a) when [Target protein binding moiety] is cycloalkyl, CH2-NH-C(O)-, heterocycloalkyl, or is absent; and.
(b) when [Target protein binding moiety] is
C(O)-NH-, heterocycloalkyl, heterocycloalkyl-NH, or is absent.
In some embodiments of the fourth and fifth aspects, X2 is N. In other embodiments, X3 is N.
In other embodiments of the fourth and fifth aspects, X2 is CR3 and X3 is CR1.
In some embodiments of the fourth and fifth aspects, R1 is H, halogen or methyl.
In some embodiments of the fourth and fifth aspects, R3 is H, halogen, unsubstituted alkyl or -CN.
In some embodiments of the fourth and fifth aspects, R1 is H. In some embodiments, R1 and R3 are each
H. In other embodiments, R1 and R2 are each H.
In some embodiments of the fourth and fifth aspects, R2 and R3 are each H. In some embodiments of the fourth and fifth aspects,
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In some embodiments of the fourth and fifth aspects, R2 is -B(OH)2, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In some embodiments of the fourth and fifth aspects, R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
In some embodiments of the fourth and fifth aspects, R2 is selected from
wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
In some embodiments of the fourth and fifth aspects, R2 is selected from wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
In some embodiments of the fourth and fifth aspects, R2 is selected from wherein n is 1, 2 or 3; and wherein m is 0 or 1. In some embodiments of the fourth and fifth aspects, R2 is
In some embodiments of the fourth and fifth aspects, each R4 is independently selected from halogen, alkyl, -CN, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), - O(heterocycloalkyl), -NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), - S(O)2(heterocycloalkyl), -C(O)(heterocycloalkyl), -C(O)OH, R19 and -OR19; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments of the fourth and fifth aspects, each R7 is independently selected from -Me, - OMe, -O(CH2)qOMe, -CH2OMe, -C(O)Me, -S(O)2Me, R19 and -OR19.
In some embodiments of the fourth and fifth aspects, each R4 is independently selected from R19, -OR19, -NHR19 -F, -Cl, -lBu, -Me, -CF3, -CN, -OH, -OMe, -OCF3, -NH2, -NMe2, -S(O)2Me, -S(O)2NMe2, - CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
In some embodiments of the fourth and fifth aspects, each R4 is independently selected from R19, -OR19,
In some embodiments, each R6 is independently selected from haloalkyl, -C(O)(heterocycloalkyl), - S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7. In some embodiments of the fourth and fifth aspects, each R6 is independently selected from -CF3, -
C(O)piperidine, -
In some embodiments of the fourth and fifth aspects, R2 is selected from:
In some embodiments of the fourth and fifth aspects: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then
C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C- S(O)2(heterocycloalkyl), C-S(O)2aryl, C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments of the fourth and fifth aspects: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl), n is 1 or 2, then each R4 of - ( '~R4 /)n is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH2, -N(alkyl)z, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
In some embodiments of the fourth and fifth aspects: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl);
In some embodiments of the fourth and fifth aspects: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH or -CH^O^CH^CfOfBu;
X2 and X3 are both CH; and
(i) each is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is
In some embodiments of the fourth and fifth aspects, R1 is methyl.
In some embodiments of the fourth and fifth aspects, -
In some embodiments of the fourth and fifth aspects, R8 is selected from -CHR9C(O)OR10, - CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR92, -CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, - CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR9, -B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, - C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, -C(O)OCH2CH2CI\/le2OH, and -C(O)OCH2CH2SO2Me
In some embodiments of the fourth and fifth aspects, R8 is selected from -CHR9C(O)OR10, CHR9C(O)NR92, -CH2S(O)2NR92, -S(O)2NR92, -CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, - B(OH)2 and -P(O)(OH)2.
In some embodiments of the fourth and fifth aspects, R8 is selected from -CHR9C(O)OR10, CHR9C(O)NR92, -CH2S(O)2NR92, -S(O)2NR92, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2.
In some embodiments of the fourth and fifth aspects, R8 is selected from -CH2C(O)OH, -CHMeC(O)OH, - CH2C(O)OMe, -CH2C(O)OEt, -CH2C(O)OCH2OC(O)tBu, -CH2C(O)NH2, -CH2S(O)2NH2, -S(O)2NH2, -CH2- oxetane, -CH2(l,2,3-triazole), -CH2B(OH)2, -CH2P(O)(OH)2, -COOH, -COOMe, -B(OH)2 and -P(O)(OH)2.
In some embodiments of the fourth and fifth aspects, R8 is -CHR9C(O)OR10. In some embodiments of the fourth and fifth aspects, R8 is selected from -CHjCfOjOCHjOCXOfBu, - CH2C(O)OH and -CHMeC(O)OH.
In some embodiments of the fourth and fifth aspects, R8 is selected from
In some embodiments, R8 is -CH2C(O)OH. In other embodiments, R8 is -CH2C(O)OCH2OC(O)tBu.
In some embodiments of the fourth and fifth aspects, R1 and R3 are each H; R2 is selected from aryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the fused aryl/heterocycloalkyl is unsubstituted or is substituted with one or more R6; and R8 is -CHR9C(O)OR10. In some such embodiments, R2 is aryl is substituted with R4 and R8 is -CHR9C(O)OR10
In some embodiments of the fourth and fifth aspects, each R4 is S(O)2(heterocycloalkyl) substituted with R19.
In some embodiments of the fifth aspect of the invention, at least one of R14, R15, R16, R17 and R18 is present.
In some embodiments of the fifth aspect, R14 is absent. In other embodiments, R14 is -C(O)-.
In some embodiments of the fifth aspect, R15 is absent. In other embodiments, R15 is heterocycloalkyl.
In some embodiments of the fifth aspect, R16 is absent. In other embodiments, R16 is -Ci.g alkyl. In other embodiments, R16 is -C(O)-. In other embodiments, R16 is -C(O)-NH-.
In some embodiments of the fifth aspect, R17 is absent. In other embodiments, R17 is (C2H4-O)X. In some embodiments of the fifth aspect, x is 3-9. In some embodiments, x is 3, 5, 7 or 9.
In some embodiments of the fifth aspect, R18 is absent. In other embodiments, R18 is -Ci-w alkyl or -Ci-io alkyl-NH-. In some embodiments, R18 is -Ci.g alkyl or -Ci.g alkyl-NH-. In some embodiments, R18 is - CH2CH2- or -CH2CH2-NH-. In other embodiments, R18 is heterocycloalkyl or heterocycloalkyl-NH-.
In some embodiments of the fifth aspect, R14, R15, R16, R17 and R18 are each absent.
In some embodiments of the fifth aspect, [linker] is
rT' n 1 rvT wherei indicates attachment to [KLHDC2 ligase binding moiety] and 2 indicates attachment to [Target protein binding moiety].
In some embodiments of the fifth aspect, the [Target protein binding moiety] is: wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl.
In some embodiments of the fifth aspect, R12 is Me. In other embodiments, R12 is H.
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments, M is absent.
In some embodiments, L' is H.
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is: n some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
In some embodiments of the fifth aspect, [Target protein binding moiety] is:
_,1 wherein indicates attachment to [KLHDC2 ligase binding moiety] indicates attachment to [Target protein binding moiety]. In some embodiments, k is an integer from 3-8; optionally wherein k is 4, 6, or 8; further optionally wherein k is 6.
In some embodiments of the fifth aspect, the compound is selected from:
In some embodiments of the fifth aspect, the compound is selected from Compound ID nos. 1001, 1002 and 1003.
In some embodiments of the fifth aspect, the compound is selected from Compound ID nos. 1002, 1003, 1005, 1007, 1008, 1010, 1013, 1014, 1016, 1019, 1020, 1022, 1024, 1026, 1028, 1031, 1034, 1036 and 1038.
In some embodiments of the fifth aspect, the compound is selected from Compound ID no. 1003, 1008, 1010, 1012, 1014, 1016, 1020, 1022, 1026, 1028, 1034, 1036 and 1038. In some embodiments of the fifth aspect, the compound is selected from Compound ID no. 1005 and 1007.
In some embodiments of the fifth aspect, the compound is selected from Compound ID nos. 1008, 1009, 1010, 1011, 1012, 1013, 1014, 1015, 1016, 1017, 1018 and 1019.
In some embodiments of the fifth aspect, the compound is selected from Compound ID nos. 1024 and 1025.
In some embodiments of the fifth aspect, the compound is selected from Compound ID nos. 1028, 1029, 1030, 1031, 1033, 1034, 1035, 1036 and 1037.
The present invention also provides a pharmaceutical composition comprising a compound of the invention. In some embodiments, the pharmaceutical composition comprises a bifunctional protein degrader compound of the present invention. In some embodiments, the pharmaceutical composition comprises a compound of any of the above embodiments of the fourth or fifth aspects of the present invention. In some embodiments, the pharmaceutical composition comprises a compound of any of the above embodiments of the fifth aspect of the present invention.
The present invention also provides a compound or pharmaceutical composition of the invention, for use in medicine. In some embodiments, the compound is a bifunctional protein degrader compound of the present invention. In some embodiments, the compound is a compound of any of the above embodiments of the fourth or fifth aspects of the present invention. In some embodiments, the compound is a compound of any of the above embodiments of the fifth aspect of the present invention.
The present invention also provides a method of treating a disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound or pharmaceutical composition of the invention. In some embodiments, the compound is a bifunctional protein degrader compound of the present invention. In some embodiments, the compound is a compound of any of the above embodiments of the fourth or fifth aspects of the present invention. In some embodiments, the compound is a compound of any of the above embodiments of the fifth aspect of the present invention.
EXAMPLES
The compounds of the present invention were prepared as described below.
The reagents and solvents were used as received from the commercial sources. Proton nuclear magnetic resonance (NMR) spectra were recorded on Bruker Avance-lll 400 MHz and Bruker Avance Neo 400 MHz spectrometers. The spectra are reported in terms of chemical shift (6 [ppm]), multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, p = quintet, m= multiplet), coupling constant (J [Hz]), and integration. Chemical shifts are reported in ppm relative to dimethyl sulfoxide-dg (6 2.50), chloroform-d (6 7.26) or methanol-c/4 (6 3.34) (as indicated in NMR spectra data. The samples were prepared by dissolving a dry sample (0.2 - 2 mg) in an appropriate deuterated solvent (0.7-1 mL).
GCMS were performed on Agilent Model 7890B GC system with 5977B MSD.
LCMS were collected using Waters SQD2 or API 2000 Mass Spectrometers. All masses reported are the m/z of the protonated parent ions unless otherwise stated.
Preparative HPLC was performed using Waters auto purification instrument equipped with Gemini C18 column (100 x 19 mm, 5m), YMC-Actus C18 (250 x 20 mm, 5m), YMC-Triart C18 (250 x 20 mm, 5m), Hydrosphere C18 (250 x 20 mm, 5m).
The chemical names were generated using ChemDraw Professional v. 18.2.0.48 from PerkinElmer Informatics, Inc.
Abbreviations used in the following examples are presented below in the alphabetical order:
ACN Acetonitrile
AcOH Acetic acid
Bpin Boronic acid pinacol ester
BOC tert-Butoxycarbonyl m-CPBA 3-Chlorobenzoperoxoic acid
DBU l,8-Diazabicyclo[5.4.0]undec-7-ene
DCM Dichloromethane DIAD Diisopropyl azodicarboxylate
DIBAL Diisobutylaluminium hydride
DIPEA /V,/V-Diisopropyl ethylamine
DMAP Dimethyl(pyridin-4-yl)azane
DME Dimethoxyethane
DMF /V,/V-Dimethylformamide
DMF-DMA l,l-dimethoxy-/V,/V-dimethylmethanamine
DMSO Dimethyl sulfoxide
EtOH Ethanol
FA Formic acid
GCMS Gas chromatography-mass spectrometry
HATU l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5- b]pyridinium 3-oxide hexafluorophosphate
HPLC High performance liquid chromatography
LAH Lithium aluminium hydride
LDA Lithium diisopropylamide
LCMS Liquid chromatography-mass spectrometry
M molarity
Me Methyl
MeOH Methanol
NaHMDS Sodium l7l,l-trimethyl-/V-(trimethylsilyl)silanaminide
NMR Nuclear magnetic resonance
Pd2(dba)3 Tris(dibenzylideneacetone)dipalladium
Pd(dppf)CI2 [l,r-Bis(diphenylphosphino)ferrocene]palladium(ll) dichloride
Pd(dtbpf)CI2 [l,r-Bis(di-tert-butylphosphino)ferrocene]dichloropalladium(ll)
Pd(PPh3)4 Tetrakis(triphenylphosphine)palladium(0)
Pd(PPh3)2CI2 Bis(triphenylphosphine)palladium(ll) dichloride
PdXphosG3 (2-Dicyclohexylphosphino-2',4',6'-triisopropyl-l,r-biphenyl)[2-
(2,2'-amino-l,r-biphenyl)]palladium(ll) methanesulfonate
RT Room temperature
TBAI Tetrabutylammonium iodide
TEA Triethylamine
TFA Trifluoroacetic acid TFAA Trifluoroacetic anhydride
THF Tetrahydrofuran
XantPhos (9,9-Dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphane)
General procedures
The synthesis of the compounds can be summarized in the following general procedures as set out below:
Example method 1: Ester hydrolysis
Reaction Scheme 1: Ester hydrolysis
The ester (1 equiv) was taken in appropriate solvent or in mixture of solvents (e.g. MeOH, THF) and lithium hydroxide monohydrate (1-5 equiv) or sodium hydroxide (1-5 equiv) or cesium carbonate (1-5 equiv) in water was added at temperature between 0°C and RT. The mixture was stirred at temperature between RT and 90°C for 1-18 h. After completion the solvent was removed under reduced pressure, the residue was diluted with water and washed with ether. The aqueous layer was cooled to 0°C, acidified with IM HCI and extracted with ethyl acetate. The organic layer was dried over anhydrous NajSCU and evaporated. The product was purified by flash column chromatography and/or preparative HPLC unless otherwise stated.
Example method 2: Nitrile hydrolysis
R^^CN
Reaction Scheme 2: Nitrile hydrolysis
The appropriate nitrile was taken up in IM NaOH and THF and refluxed for 1-16 h. The reaction mixture was cooled, acidified with IM HCI and extracted with ethyl acetate. The organic fraction was washed with water, brine, dried over NajSCU and evaporated. The resulting product was purified by flash column chromatography and/or preparative HPLC unless otherwise stated.
Example method 3: Suzuki reaction
The solution of appropriate halide (i.e. bromide or iodide) (1-1.6 equiv) in mixture of 1,4-dioxane/water or DME/water or THF/water system was added boronic acid (1-1.6 equiv) or boronic ester (1-1.6 equiv), base (e.g. K3PO4, CS2CO3) (3.5-5.5 equiv), palladium catalyst (0.1-0.4 equiv). The resulting mixture was bubbled with argon or nitrogen for 15 min and stirred at 60-100°C for 4-16 h. The volatiles were removed under reduced pressure and the product was purified by flash column chromatography and/or preparative HPLC unless otherwise stated.
Example method 4: Synthesis of nitrile from alcohol
To a stirred solution of triphenylphosphine (1-1.3 equiv) in DCM, cooled in an ice-water bath, was added carbononitridic bromide (1.4-1.9 equiv). The reaction was stirred for 15-30 min at 0°C and solution of appropriate alcohol (0.7-1 equiv) ) in DCM or THF was added slowly and the reaction was stirred at 0°C for 1 h. After full consumption of the starting material DBU (1.5-2 equiv) was added dropwise at 0°C and the resulting solution was stirred at RT for 12-16 h. After completion of the reaction the volatiles were removed under reduced pressure and the product was purified by flash column chromatography.
Example method 5: BOC removal
Reaction Scheme 5: BOC removal
To the ice-cold solution of appropriate BOC-protected amine in DCM was added TFA and the resulting solution was stirred at RT for 1-18 h. Once the reaction was completed the volatiles were removed under reduced pressure and the crude product was used into the next step without additional purification unless otherwise stated.
Example method 6: Reduction of C=C double bond
Reaction Scheme 6: Reduction of C=C double bond
To a solution of the substrate in appropriate solvent or in mixture of solvents (e.g. EtOH, THF) was added palladium on activated carbon (10% wt.) and the reaction mixture was stirred at RT for 1-16 h in Parr shaker under hydrogen atmosphere. After completion of the reaction the solid particles were filtered off, the solvent was removed under reduced pressure. The product was purified by flash column chromatography and/or preparative HPLC unless otherwise stated.
Example method 7: Reduction of ester to alcohol
Reaction Scheme 7: Reduction of ester to alcohol
To a solution of an ester (1 equiv) in THF was slowly added LAH (1.2 equiv) in THF or DIBAL (2 equiv) in THF with cooling between -15°C to -5°C. The reaction mixture was stirred for 1-3 h at temperature between -10°C to RT. After completion the reaction was quenched with Rochelle salt solution and extracted with ethyl acetate. The organic layer was washed with water, brine, dried over anhydrous NajSCU and evaporated. The product was purified by flash column chromatography. Example method 8: Sulfonamide synthesis
Reaction Scheme 8: Sulfonamide synthesis
To a solution of sulfonyl chloride (1 equiv) in DCM, cooled in an ice-water bath, was added TEA (2-4 equiv) and appropriate amine or amine trifluoroacetate (0.5-3 equiv) and the reaction mixture was stirred at RT for 1-4 h. After completion, the volatiles were removed under reduced pressure and the product was purified by flash column chromatography unless otherwise stated.
Example method 9: Miyaura borylation reaction
R'X
X = Br, OTf
Reaction Scheme 9: Miyaura borylation reaction
The solution of appropriate bromide or triflate (1 equiv), potassium acetate (2.5-3.5 equiv) and bis(pinacolato)diboron (1.5-2.5 equiv) in 1,4-dioxane was bubbled with argon for 15 min. Pd(dppf)CI2 (0.08-0.12 equiv) was added and the reaction mixture was stirred at 80-100°C for 4-18 h in a sealed tube. The volatiles were removed under reduced pressure and the product was purified by flash column chromatography.
Example method 10: Buchwald-Hartwig cross coupling
R^ R'N'R2
Reaction Scheme 10: Buchwald-Hartwig cross coupling The suspension of appropriate aryl iodide (1 equiv), amine (0.8-2.5 equiv), CS2CO3 (2-2.5 equiv), Xantphos (0.1-0.2 equiv) and Pd2(dba)3 (0.08-0.12 equiv) in DMF was stirred at 90-110°C for 5-18 h. The volatiles were removed under reduced pressure and the product was purified by flash column chromatography unless otherwise stated.
Example method 11: Acylation of amine
Reaction Scheme 11: Acylation of amine
To a solution of appropriate amine trifluoroacetate (1 equiv) in DCM, cooled in an ice-water bath, was added TEA (2-4 equiv) followed by acetyl anhydride (1.5-3 equiv) or acetyl chloride (1.2-1.7 equiv). The resulting solution was stirred at RT for 1-4 h and the volatiles were removed under reduced pressure. The product was purified by flash column chromatography unless otherwise stated.
Example 1-001: Synthesis of 2-(5-(2-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound
71)
Step 1: Ethyl 5-(2-methoxyphenyl)pyrazolo[l,5-a]pyridine-3-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (60% yield), using ethyl 5- bromopyrazolo[l,5-a]pyridine-3-carboxylate [commercial] (300 mg, 1.11 mmol, 1 equiv) and (2- methoxyphenyl)boronic acid [commercial] (1.2 equiv) as starting materials, K3PO4 (5 equiv) as base, PdXphosG3 as catalyst (0.1 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated. The product was purified by flash column chromatography.
LCMS (ESI+) m/z 297.2 [M+H]+
Step 2: (5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7, above (49% yield), using ethyl 5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridine-3-carboxylate (290 mg, 1.03 mmol, 1 equiv) as starting material and LAH (1.2 equiv, IM in THF) as reducing agent.
LCMS (ESI+) m/z 255.1 [M+H]+
Step 3: 2-(5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (48% yield), using (5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)methanol (100 mg, 0.4 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 264.2 [M+H]+
Step 4: 2-(5-(2-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (27% yield), using 2-(5-(2- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile (50 mg, 0.19 mmol, 1 equiv) as starting material. After extraction, the residue was triturated with 50% ether in pentane to afford pure product.
LCMS (ESI+) m/z 283.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.93 - 11.79 (m, 1H), 8.60 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.70 (s, 1H), 7.45 - 7.33 (m, 2H), 7.15 (d, J = 8.3 Hz, 1H), 7.07 (t, J = 7.2 Hz, 1H), 7.00 (dd, J = 1.9, 7.3 Hz, 1H), 3.80 (s, 3H), 3.73 (s, 2H). Example 1-002: Synthesis of 2-(5-(2-((/V-methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 67)
Step 1: Methyl 2-(4-bromopyridin-2-yl)acetate [commercial] (5.6 g, 24.3 mmol, 1 equiv) and DMF-DMA [commercial] (46 mL) were stirred at 80°C for 16 h under nitrogen. After consumption of the starting material the reaction mixture was evaporated under reduced pressure and the residue was triturated with ether to afford crude methyl (Z)-2-(4-bromopyridin-2-yl)-3-(dimethylamino)acrylate (5.0 g) which was forwarded directly for the next step.
LCMS (ESI+) m/z 284.8 [M+H]+
Step 2: To a solution of methyl (Z)-2-(4-bromopyridin-2-yl)-3-(dimethylamino)acrylate (5.0 g, 17.5 mmol, 1 equiv, crude after Step 1) in DCM (50 mL) was added O-(mesitylsulfonyl)hydroxylamine [commercial] (4.53 g, 21 mmol, 1.2 equiv) and the reaction mixture was stirred at RT for 16 h. After completion the reaction was quenched with water and extracted with ethyl acetate. The combined organic fractions were washed with brine and evaporated. Methyl 5-bromopyrazolo[l,5-o]pyridine-3-carboxylate (2.2 g, 8.63 mmol, 35% yield over two steps) was purified by flash column chromatography.
GCMS (ESI+) m/z 254.0 [M+H] Step 3: (5-Bromopyrazolo[l,5-a]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7 , above (56% yield), using methyl 5- bromopyrazolo[l,5-a]pyridine-3-carboxylate (1.0 g, 3.92 mmol, 1 equiv) as starting material and DIBAL (15.6 mL, 15.6 mmol, 4 equiv, IM solution in toluene) as reducing agent.
LCMS (ESI+) m/z 227.0, 229.0 [M+H]+
Step 4: 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (49% yield) using (5-bromopyrazolo[l,5- o]pyridin-3-yl)methanol (250 mg, 1.1 mmol, 1 equiv) as starting material.
GCMS (ESI+) m/z 235.0 [M+H]+
Step 5: 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (61% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetonitrile (75 mg, 0.32 mmol) as a starting material. After completion, the acidified reaction mixture was extracted with ethyl acetate, the combined organic fractions were dried over Na2SO4 and evaporated. The residue was triturated with 50% (v/v) mixture of ether and pentane to afford the target compound.
LCMS (ESI+) m/z 254.9 [M+H]+
Step 6: [TLS-364, step 1] To a solution of 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (1.7 g, 6.67 mmol, 1 equiv) in MeOH (20 mL) was added concentrated H2SO4 (0.5 mL) and the mixture was refluxed for 6 h. After completion the volatiles were removed under reduced pressure. The residue was taken up in ethyl acetate, washed with water, saturated NaHCOs solution, dried over Na2SO4 and evaporated to afford methyl 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetate (1.4 g, 5.2 mmol, 78% yield).
LCMS (ESI+) m/z 270.9 [M+H] Step 7: Methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (42% yield), using methyl 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetate (700 mg, 2.612 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 317.3 [M+H]+
Step 8: Methyl 2-(5-(2-((/V-methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (258.3 mg, 0.817 mmol, 1.3 equiv) and l-(2-bromophenyl)-/V-methylmethanesulfonamide [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.2 equiv). After completion of the reaction the solution was filtered, the volatiles were removed under reduced pressure and the resulting crude was used in the next step without purification.
LCMS (ESI+) m/z 374.2 [M+H]+
Step 9: 2-(5-(2-((/V-Methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (59% yield over two steps), using methyl 2-(5-(2-((/V-methylsulfamoyl)methyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting material.
LCMS (ESI+) m/z 360.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.1 Hz, 1H), 7.89 (s, 1H), 7.76 (s, 1H), 7.62 - 7.55 (m, 1H), 7.49 - 7.41 (m, 2H), 7.43 - 7.36 (m, 1H), 6.88 (d, J = 7.0 Hz, 1H), 4.37 (s, 2H), 3.60 (s, 2H), 2.42 (s, 3H).
Example 1-003: Synthesis of 2-(5-(2-chlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 69 ) Step 1: 2-(5-(2-Chlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetonitrile (35 mg, 0.15 mmol, 1 equiv) and (2-chlorophenyl)boronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv). After completion the solution was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated to give crude product which was forwarded to the next step without purification.
Step 2: 2-(5-(2-Chlorophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 1, above (46% yield over two steps), using 2-(5-(2-chlorophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetonitrile (200 mg, 0.75 mmol) as a starting material.
LCMS (ESI+) m/z 287.1 [M+H]+
'H NMR (400 MHz, DMSO-d6) δ 8.65 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.68 (s, 1H), 7.64 - 7.55 (m, 1H), 7.54- 7.49 (m, 1H), 7.47 - 7.40 (m, 2H), 6.90 (d, J = 7.1 Hz, 1H), 3.60 (s, 2H).
Example 1-004: Synthesis of 2-(5-(2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound
70)
Step 1: 2-(5-(2,6-Dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile (150 mg, 0.63 mmol, 1 equiv) and (2,6- dichlorophenyl)boronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over Na2SO4 and evaporated to give crude product which was used in the next step without further purification.
LCMS (ESI+) m/z 301.9 [M+H]+ Step 2: 2-(5-(2,6-Dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (17% yield over two steps), using 2-(5-(2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile (200 mg, 0.66 mmol) as a starting material.
LCMS (ESI+) m/z 321.0 [M+H]+
!H NMR (400 MHz, DMSO-d6) δ 8.69 (d, J = 7.1 Hz, 1H), 7.92 (s, 1H), 7.63 (m, 1H), 7.60 (m, 2H), 7.52 - 7.43 (m, 1H), 6.71 (dd, J = 1.9, 7.1 Hz, 1H), 3.58 (s, 2H).
Example 1-005: Synthesis of 2-(5-(m-tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 68)
Step 1: 2-(5-(m-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5-o]pyridin-3- yl)acetonitrile (20 mg, 0.08 mmol, 1 equiv) and m-tolylboronic acid [commercial] (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.11 equiv). After completion the reaction mixture was diluted with ethyl acetate, washed with water, brine, dried over NajSCU and evaporated to give crude product which was used in the next step without further purification.
LCMS (ESI+) m/z 248.2 [M+H]+
Step 2: 2-(5-(m-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (8% yield over two steps), using 2-(5-(m- tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile (100 mg, 0.4 mmol) as a starting material.
LCMS (ESI+) m/z 267.1 [M+H]+
!H NMR (400 MHz, DMSO-d6) δ 13.21 - 11.52 (m, 1H), 8.67 (d, J = 7.3 Hz, 1H), 7.97 (d, J = 2.0 Hz, 1H), 7.91 (s, 1H), 7.63 (s, 1H), 7.59 (d, J = 7.7 Hz, 1H), 7.39 (t, J = 7.7 Hz, 1H), 7.26 - 7.16 (m, 2H), 3.78 (s, 2H), 2.40 (s, 3H). Example 1-006: Synthesis of 2-(5-(2-(methylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 65)
Step 1: 2-(5-(2-(Methylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (62% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (80 mg, 0.315 mmol, 1 equiv) and 4,4,5,5-tetramethyl-2-(2- (methylsulfonyl)phenyl)-l,3,2-dioxaborolane (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 331.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.75 - 11.90 (s, 1H), 8.63 (d, J = 7.1 Hz, 1H), 8.11 (d, J = 7.8 Hz, 1H), 7.95 (s, 1H), 7.80 (t, J = 7.4 Hz, 1H), 7.72 (t, J = 7.6 Hz, 1H), 7.66 (s, 1H), 7.49 (d, J = 7.3 Hz, 1H), 6.86 (d, J = 7.0 Hz, 1H), 3.68 (s, 2H), 3.06 (s, 3H).
Example 1-007: Synthesis of 2-(5-(4-(l-methylpiperidin-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 66)
Step 1: 2-(5-(4-(l-Methylpiperidin-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (61% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (50 mg, 0.197 mmol, 1 equiv) and (4-(l- methylpiperidin-4-yl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dppf)CI2 as catalyst (0.1 equiv). LCMS (ESI+) m/z 350.2 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.3 Hz, 1H), 7.94 (s, 1H), 7.91 (s, 1H), 7.73 (d, J = 8.0 Hz, 2H), 7.37 (d, J = 8.0 Hz, 2H), 7.19 (d, J = 6.9 Hz, 1H), 3.77 (s, 2H), 2.90 (d, J = 11.4 Hz, 2H), 2.22 (s, 3H), 2.01 (t, J = 11.1 Hz, 2H), 1.86 - 1.61 (m, 4H).
Example 1-008: Synthesis of 2-(5-benzylpyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 63)
Step 1: 2-(5-Benzylpyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (55% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetic acid (60 mg, 0.236 mmol, 1 equiv) and 2-benzyl-4,4,5,5-tetramethyl-l,3,2- dioxaborolane [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv).
LCMS (ESI+) m/z 267.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.47 (d, J = 7.1 Hz, 1H), 7.81 (s, 1H), 7.48 (s, 1H), 7.35 - 7.24 (m, 4H), 7.25 - 7.16 (m, 1H), 6.63 (dd, J = 2.0, 7.2 Hz, 1H), 3.96 (s, 2H), 3.55 (s, 2H).
Example 1-009: Synthesis of 2-(5-(2-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 60)
Step 1: Methyl 2-(5-(2-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (171 mg, 0.541 mmol, 1.3 equiv) and l-(2-bromophenyl)piperidine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.2 equiv). After completion the solvents were removed under reduced pressure and the obtained crude was used in the next step without further purification.
LCMS (ESI+) m/z 350.3 [M+H]+
Step 2: 2-(5-(2-(Piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (34% yield over two steps), using methyl 2-(5-(2-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 336.2 [M+H]+
1H NMR (400 MHz, DMSO-d6) δ 12.66 - 11.93 (s, 1H), 8.61 (d, J = 7.2 Hz, 1H), 7.89 (s, 1H), 7.83 (s, 1H), 7.37 - 7.24 (m, 3H), 7.15 - 7.05 (m, 2H), 3.73 (s, 2H), 2.84 - 2.69 (m, 4H), 1.58 - 1.34 (m, 6H).
Example 1-010: Synthesis of 2-(5-(2,3-dihydrobenzo[b][l,4]dioxin-5-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 62)
Step 1: 2-(5-(2,3-Dihydrobenzo[b][l,4]dioxin-5-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (27% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.394 mmol, 1 equiv) and (2,3- dihydrobenzo[b][l,4]dioxin-5-yl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 311.1 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 12.33 (s, 1H), 8.62 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.71 (s, 1H), 7.03 (dd, J = 1.3, 7.2 Hz, 1H), 7.02 - 6.88 (m, 3H), 4.29 (m, 4H), 3.74 (s, 2H). Example 1-011: Synthesis of 2-(5-(2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 64)
Step 1: Methyl 2-(5-(2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (135 mg, 0.43 mmol, 1.3 equiv) and l-((2-bromophenyl)sulfonyl)piperidine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.15 equiv). After completion the solvents were removed under reduced pressure and the resulting crude was used in the next step without purification.
LCMS (ESI+) m/z 414.0 [M+H]+
Step 2: 2-(5-(2-(Piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (13% yield over two steps), using methyl 2-(5-(2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 400.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.0 Hz, 1H), 7.98 (d, J = 7.5 Hz, 1H), 7.92 (s, 1H), 7.74 (t, J = 6.9 Hz, 1H), 7.66 (t, J = 7.0 Hz, 1H), 7.57 (s, 1H), 7.46 (d, J = 7.4 Hz, 1H), 6.83 (d, J = 7.1 Hz, 1H), 3.65 (s, 2H), 2.91 - 2.71 (m, 4H), 1.45 - 1.16 (m, 6H).
Example 1-012: Synthesis of 2-(5-(2,6-dimethoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 61)
Step 1: 2-(5-(2,6-Dimethoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (24% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (100 mg, 0.39 mmol, 1 equiv) and (2,6- dimethoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.2 equiv).
LCMS (ESI+) m/z 313.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.28 (s, 1H), 8.56 (d, J = 7.2 Hz, 1H), 7.88 (s, 1H), 7.44 (s, 1H), 7.35 (t, J = 8.3 Hz, 1H), 6.77 (d, J = 8.4 Hz, 2H), 6.68 (dd, J = 1.2, 7.0 Hz, 1H), 3.69 (m, 8H).
Example 1-013: Synthesis of 2-(5-(2-(trifluoromethoxy)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 58)
Step 1: 2-(5-(2-(Trifluoromethoxy)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (31% yield), using 2- (5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (60 mg, 0.23 mmol, 1 equiv) and (2- (trifluoromethoxy)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 337.1 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 12.54 - 12.15 (s, 1H), 8.71 (d, J = 7.1 Hz, 1H), 7.97 (s, 1H), 7.79 (s, 1H), 7.66
(dd, J = 2.3, 7.0 Hz, 1H), 7.63 - 7.49 (m, 3H), 6.97 (d, J = 7.1 Hz, 1H), 3.76 (s, 2H). Example 1-014: Synthesis of 2-(5-(2-methoxy-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 59)
Step 1: 2-(5-(2-Methoxy-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (32% yield), using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.39 mmol, 1 equiv) and (2- methoxy-6-(trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Ck as catalyst (0.1 equiv).
(2-Methoxy-6-(trifluoromethyl)phenyl)boronic acid was synthesized according to procedure described in Kopka, I.E. et al., Biomol
LCMS (ESI+) m/z 351.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.76 - 11.89 (s, 1H), 8.62 (d, J = 7.0 Hz, 1H), 7.93 (s, 1H), 7.64 (t, J = 8.0 Hz, 1H), 7.49 - 7.40 (m, 3H), 6.67 (d, J = 7.0 Hz, 1H), 3.74 (s, 3H), 3.69 (s, 2H).
Example 1-015: Synthesis of 2-(5-pp(4-(4-methylpiperazin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 57)
Step 1: 2-(5-(4-(4-Methylpiperazin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (60 mg, 0.23 mmol, 1 equiv) and l-methyl-4-(4-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)piperazine [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 351.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 10.57 (s, 1H), 8.62 (d, J = 7.3 Hz, 1H), 7.89 (d, J = 4.8 Hz, 2H), 7.74 (d, J = 8.4 Hz, 2H), 7.24 - 7.16 (m, 1H), 7.13 (d, J = 8.3 Hz, 2H), 3.95 (d, J = 10.7 Hz, 2H), 3.78 (s, 2H), 3.51 (d, J = 9.6 Hz, 2H), 3.14 (m, 4H), 2.88 - 2.79 (m, 3H).
Example 1-016: Synthesis of 2-(5-(2-methoxyphenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 9)
Step 1: To a solution of 4-bromo-2,3-dimethylpyridine (5.0 g, 26.9 mmol, 1 equiv) in THF (75 mL), cooled to -78°C, was added LDA (41 mL, 80.6 mmol, 3 equiv, 2M solution in THF) and the resulting mixture was stirred at that temperature for 1 h. Dimethyl carbonate (5 mL, 53.8 mmol, 2 equiv) was added and the reaction was stirred at -78°C for 1 h. The reaction was then quenched with saturated NH4CI solution and extracted with ethyl acetate. The combined organic fractions were washed with brine, dried over Na2SO4 and evaporated to yield crude product. Methyl 2-(4-bromo-3-methylpyridin-2-yl)acetate (4.0 g, 16.4 mmol, 61% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 244.0, 246.0 [M+H]+
Step 2: Methyl 2-(4-bromo-3-methylpyridin-2-yl)acetate (4.0 g, 16.46 mmol, 1 equiv) and DMF-DMA (40 mL) were stirred at 140°C for 16 h under nitrogen. After consumption of the starting material the reaction mixture was evaporated under reduced pressure and the residue was triturated with ether to afford crude methyl (Z)-2-(4-bromo-3-methylpyridin-2-yl)-3-(dimethylamino)acrylate which was forwarded directly for the next step. LCMS (ESI+) m/z 299.2 [M+H]
Step 3: To a solution of methyl (Z)-2-(4-bromo-3-methylpyridin-2-yl)-3-(dimethylarriino)acrylate (5.0 g, 16.7 mmol, 1 equiv, crude after Step 2) in DCM (40 mL) was added O-(mesitylsulfonyl)hydroxylamine (4.3 g, 20.04 mmol, 1.2 equiv) and the reaction mixture was stirred at RT for 16 h. After completion, the reaction was quenched with water and extracted with ethyl acetate. The combined organic fractions were washed with brine and evaporated. Methyl 5-bromo-4-methylpyrazolo[l,5-a]pyridine-3-carboxylate (2.5 g, 9.3 mmol, 57% yield over two steps) was purified by flash column chromatography.
LCMS (ESI+) m/z 269.2, 271.2 [M+H]+
Step 4: (5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)methanol was synthesized using the general procedure shown in Reaction Scheme 7 and Example Method 7, above (58% yield), using methyl 5-bromo- 4-methylpyrazolo[l,5-o]pyridine-3-carboxylate (2.5 g, 9.3 mmol, 1 equiv) as starting material and DIBAL (2 equiv) as reducing agent.
LCMS (ESI+) m/z 241.2, 243.2 [M+H]+
Step 5: 2-(5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 4 and Example Method 4, above (38% yield), using (5-bromo-4- methylpyrazolo[l,5-o]pyridin-3-yl)methanol (1.0 g, 4.15 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 250.2 [M+H]+
Step 6: 2-(5-(2-Methoxyphenyl)-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (67% yield), using 2- (5-bromo-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile (200 mg, 0.8 mmol, 1 equiv) and (2- methoxyphenyl)boronic acid (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 278.4 [M+H]+ Step 7: 2-(5-(2-Methoxyphenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 1, above (48% yield), using 2-(5-(2- methoxyphenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetonitrile (75 mg, 0.32 mmol) as starting material. After completion, the reaction mixture was cooled and washed with ether. Aqueous layer was separated and slowly acidified with IM HCI. Solid was precipitated and filtered and triturated with 50% ether in pentane to obtain the product.
LCMS (ESI+) m/z 297.2 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 12.48 - 12.15 (s, 1H), 8.44 (d, J = 7.0 Hz, 1H), 7.86 (s, 1H), 7.46 - 7.36 (m, 1H), 7.20 - 7.10 (m, 2H), 7.05 (t, J = 7.3 Hz, 1H), 6.59 (d, J = 7.1 Hz, 1H), 3.88 (s, 2H), 3.73 (s, 3H), 2.25 (s, 3H).
Example 1-017: Synthesis of 2-(5-(2,6-dichloro-4-(tetrahydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 50)
Step 1: 4-(4-Bromo-3,5-dichlorophenyl)-3,6-dihydro-2H-pyran was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using 2-bromo-l,3- dichloro-5-iodobenzene [commercial] (250 mg, 0.715 mmol, l.l equiv) and 2-(3,6-dihydro-2H-pyran-4- yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(dppf)CL as catalyst (0.11 equiv). After completion, the reaction mixture was quenched with ice-cold water and extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2SO4 and evaporated under reduced pressure. The crude product was purified by flash column chromatography.
GCMS (ESI+) m/z 307.9 [M+H]+
Step 2: Methyl 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (45% yield), using 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydro-2H-pyran (130 mg, 0.41 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.1 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv). After completion, the reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2SO4and evaporated. The resulting crude was purified by flash column chromatography.
LCMS (ESI+) m/z 416.9, 418.8 [M+H]+
Step 3: 2-(5-(2,6-Dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using methyl 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (120 mg, 0.288 mmol) as starting material and lithium hydroxide as base. After completion, the reaction mixture was diluted with water and washed with 30% ethyl acetate in hexanes. The aqueous layer was acidified with IM HCI and extracted with ethyl acetate, dried over Na2SO4 and evaporated to yield crude product which was used to the next step without additional purification.
LCMS (ESI+) m/z 403.0 [M+H]+
Step 4: 2-(5-(2,6-Dichloro-4-(tetrahydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (10% yield over two steps), using 2-(5-(2,6-dichloro-4-(3,6-dihydro-2H-pyran-4-yl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid as starting material.
LCMS (ESI-) m/z 403.1 [M-H]’ 1H NMR (400 MHz, DMSO-d6) δ 12.81 - 11.92 (s, 1H), 8.71 (d, J = 7.0 Hz, 1H), 7.97 (s, 1H), 7.62 (s, 1H), 7.52 (s, 2H), 6.75 (d, J = 7.8 Hz, 1H), 3.97 (d, J = 10.4 Hz, 2H), 3.72 (s, 2H), 3.49 - 3.39 (m, 2H), 3.03 - 2.80 (m, 1H), 1.85 - 1.60 (m, 4H).
Example 1-018: Synthesis of 2-(5-(2-(piperidine-l-carbonyl)-lH-pyrrol-l-yl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 4)
Step 1: To a stirred solution of piperidin-l-yl(lH-pyrrol-2-yl)methanone (70 mg, 0.393mmol, 1 equiv) in DMSO (2 mL) was added 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (1 equiv) and the suspension was purged with argon for 10 min. CuO (0.2 equiv) and K2CO3 (3 equiv) were added, the reaction mixture was purged with argon for 5 min and stirred at 140°C for 16 h. After completion, the solvent was removed under reduced pressure and the crude was purified by preparative HPLC affording 2-(5-(2-(piperidine-l- carbonyl)-lH-pyrrol-l-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (10 mg, 7% yield).
Piperidin-l-yl(lH-pyrrol-2-yl)methanone was synthesized as described in Gao, S. et al., J. Org. Chem. 2018, 83, 9250.
LCMS (ESI-) m/z 351.1 [M-H]’
XH NMR (400 MHz, DMSO-d6, 373K) 6 8.61 (d, J = 7.6 Hz, 1H), 7.93 (s, 1H), 7.48 (d, J = 2.0 Hz, 1H), 7.20 (dd, J = 2.0, 2.8 Hz, 1H), 6.80 (dd, J = 2.4, 7.6 Hz, 1H), 6.46 (dd, J = 1.6, 4.0 Hz, 1H), 6.30 (t, J = 3.2 Hz, 1H), 3.67 (s, 2H), 3.55 - 3.43 (m, 4H), 1.61 - 1.53 (m, 2H), 1.50 - 1.39 (m, 4H).
Example 1-019: Synthesis of 2-(5-(2,6-dichlorophenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 54) Step 1: 2-(5-(2,6-Dichlorophenyl)-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (31% yield), using 2- (5-bromo-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile (20 mg, 0.08 mmol, 1 equiv) and (2,6- dichlorophenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 316.1 [M+H]+
Step 2: 2-(5-(2,6-Dichlorophenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 1, above (37% yield), using 2- (5-(2,6-dichlorophenyl)-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetonitrile (15 mg, 0.047 mmol). After completion the cooled solution was diluted with water, washed with ether, acidified with IM HCI. The precipitation was filtered and dried to give the pure product.
LCMS (ESI+) m/z 335.1, 337.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.40 (s, 1H), 8.55 (d, J = 7.1 Hz, 1H), 7.92 (s, 1H), 7.64 (d, J = 8.0 Hz, 2H), 7.50 (t, J = 8.0 Hz, 1H), 6.58 (d, J = 7.0 Hz, 1H), 3.90 (s, 2H), 2.21 (s, 3H).
Example 1-020: Synthesis of 2-(5-(4-(l-acetylpiperidin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 41) Step 1: tert-Butyl 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (93% yield), using 2-bromo-l,3-dichloro-5-iodobenzene (500 mg, 1.42 mmol, l.l equiv) and tert-butyl 4- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-l(2H)-carboxylate [commercial] (1 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(dppf)CL as catalyst (0.11 equiv).
LCMS (ESI+) m/z 305.4 [M-BOC+H]+
Step 2: 4-(4-Bromo-3,5-dichlorophenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (73% yield), using tert-butyl 4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridine-l(2H)-carboxylate (400 mg, 0.988 mmol) as starting material.
LCMS (ESI+) m/z 307.9, 309.9 [M+H]+
Step 3: l-(4-(4-Bromo-3,5-dichlorophenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (89% yield), using 4-(4-bromo-3,5-dichlorophenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (300 mg, 0.72 mmol, 1 equiv) and acetic anhydride (2.7 equiv) as starting materials.
LCMS (ESI+) m/z 347.9, 350.0 [M+H]+
Step 4: Methyl 2-(5-(4-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(4-bromo-3,5-dichlorophenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l- one (150 mg, 0.43 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Ck as catalyst (0.1 equiv). After completion the solvents were evaporated, the residue was quenched with IM HCI and extracted with DCM. The organic layer was evaporated to afford crude product (220 mg) which was used directly in the next step.
LCMS (ESI+) m/z 458.3 [M+H] Step 5: 2-(5-(4-(l-Acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example
Method 1, above, using methyl 2-(5-(4-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6- dichlorophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate. After completion, the crude product was used directly in the next step.
LCMS (ESI+) m/z 444.1 [M+H]+
Step 6: 2-(5-(4-(l-Acetylpiperidin-4-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (22 mg, 10% yield over three steps), using 2-(5-(4-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2,6- dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting material.
LCMS (ESI+) m/z 446.0, 448.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.68 (d, J = 7.0 Hz, 1H), 7.93 (s, 1H), 7.60 (s, 1H), 7.52 (s, 2H), 6.72 (d, J = 7.0 Hz, 1H), 4.55 (d, J = 12.3 Hz, 1H), 3.94 (d, J = 14.4 Hz, 1H), 3.62 (s, 2H), 3.12 (t, J = 12.4 Hz, 1H), 2.99 - 2.81 (m, 1H), 2.64 - 2.54 (m, 1H), 2.03 (s, 3H), 1.90 - 1.74 (m, 2H), 1.78 - 1.58 (m, 1H), 1.64 - 1.37 (m, 1H), 1.24 - 1.13 (m, 1H).
Example 1-021: Synthesis of 2-(5-(2,6-dichloro-4-(4-(2-methoxyethoxy)cyclohexyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 10)
Step 1: To a stirred solution of l,4-dioxaspiro[4.5]decan-8-ol [commercial] (500 mg, 3.163 mmol, 1 equiv) in dry THF (10 mL) was added sodium hydride (60% suspension in mineral oil, 380 mg, 9.488 mmol, 3 equiv) at 0°C and stirred for 10 min. l-Bromo-2-methoxyethane (1.6 mL, 15.814 mmol, 5 equiv) was added at 0°C and the reaction mixture was stirred at RT for 16 h. After completion, the reaction mixture was cooled, quenched with cold water and extracted with ethyl acetate. Organic layer was washed with brine, dried over Na2SO4 and evaporated. 8-(2-Methoxyethoxy)-l,4-dioxaspiro[4.5]decane (300 mg, 43% yield) was purified by flash column chromatography.
NMR (400 MHz, Chloroform-d) 6 4.00 - 3.83 (m, 4H), 3.58 (dd, J = 5.8, 3.4 Hz, 4H), 3.46 - 3.24 (m, 4H), 1.91 - 1.60 (m, 6H), 1.60 - 1.41 (m, 2H).
Step 2: To a stirred solution of 8-(2-methoixyethoxy)-l,4-dioxaspiro[4.5]decane (500 mg, 2.313 mmol, 1 equiv) in THF (12 mL) was added 5M HCI (6 mL, 30 mmol, 13 equiv) at 0°C. Then the reaction mixture was stirred at RT for 5 h. After completion, the reaction mixture was quenched with cold water and extracted with ethyl acetate. The organic layer was washed with brine, dried over NazSCU and evaporated to obtain 4-(2-methoxyethoxy)cyclohiexan-l-one (250 mg, 62% yield).
GCMS (ESI+) m/z 172.2 [M+H] NMR (400 MHz, Chloroform-d) 6 3.75 (tt, J = 6.0, 3.1 Hz, 1H), 3.66 (dd, J = 5.8, 3.5 Hz, 2H), 3.57 (dd, J =
5.8, 3.5 Hz, 2H), 3.40 (s, 3H), 2.58 (ddd, J = 15.2, 9.9, 5.7 Hz, 2H), 2.25 (dt, J = 14.6, 6.0 Hz, 2H), 2.09 (q, J =
6.3, 5.8 Hz, 2H), 2.02 - 1.89 (m, 2H).
Step 3: To a stirred solution of 4-(2-methoxyethoxy)cyclohexan-l-one (250 mg, 1.453 mmol, 1 equiv) in dry THF (10 mL) was added 2M LDA in THF (1.45 ml, 2.907 mmol, 2 equiv) at -78°C and stirred at that temperature for 1 h. Bis(trifluoromethanesulfonyl)aniline (624 mg, 1.744 mmol, 2 equiv) in THF was added and stirred at -78°C for 1 h, then at RT for 16 h. After completion, the reaction mixture was quenched with saturated NH4CI and extracted with ethyl acetate. The organic layer was dried over Na2SO4 and evaporated. 4-(2-Methoxyethoxy)cyclohex-l-en-l-yl trifluoromethanesulfonate (150 mg, 34% yield) was purified by flash column chromatography.
XH NMR (400 MHz, Chloroform-d) 6 5.64 (s, 1H), 3.75 - 3.58 (m, 3H), 3.57 - 3.45 (m, 2H), 3.39 (s, 3H), 2.54 - 2.11 (m, 4H), 2.12 - 1.78 (m, 2H).
Step 4: 2-(4-(2-Methoxyethoxy)cyclohex-l-en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above, using 4-(2-methoxyethoxy)cyclohex-l-en-l-yl trifluoromethanesulfonate (380 mg, 1.25 mmol) as starting material. After completion, the reaction mixture was filtered through Celite®, evaporated and the obtained crude was used directly in the next step.
Step 5: 4'-Bromo-3',5'-dichloro-4-(2-methoxyethoxy)-2,3,4,5-tetrahydro-l,l'-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (12% yield over two steps), using 2-bromo-l,3-dichloro-5-iodobenzene (291 mg, 1.029 mmol, 1 equiv) and 2-(4-(2- methoxyethoxy)cyclohex-l-en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (1.11 equiv) as starting materials, KaPO4 (3.33 equiv) as base and Pd(dppf)CI2 as catalyst (0.14 equiv).
XH NMR (400 MHz, Chloroform-d) 6 7.36 (s, 2H), 6.10 - 6.02 (m, 1H), 3.74 - 3.61 (m, 3H), 3.61 - 3.52 (m, 2H), 3.40 (s, 3H), 2.62 - 2.44 (m, 2H) 2.42 - 2.31 (m, 1H), 2.29 - 2.19 (m, 1H), 2.12 - 2.00 (m, 1H), 1.90 - 1.74 (m, 1H). Step 6: Methyl 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4'-bromo-3',5'-dichloro-4-(2-methoxyethoxy)-2, 3,4,5- tetrahydro-l,l'-biphenyl (40 mg, 0.106 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.11 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(dppf)CI2 as catalyst (0.12 equiv). After completion the obtained crude mixture of methyl 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro- [l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was used for the next step.
LCMS (ESI+) m/z 489.1, 491.0 [M+H]+
Step 7: 2-(5-(3,5-Dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using mixture of methyl 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'- tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(3,5-dichloro-4'-(2- methoxyethoxy)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting materials. The crude product was used for the next step.
LCMS (ESI+) m/z 475.0 [M+H]
Step 8: 2-(5-(2,6-Dichloro-4-(4-(2-methoxyethoxy)cyclohexyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (17% yield over three steps), using 2-(5-(3,5-dichloro-4'-(2-methoxyethoxy)-2',3',4',5'-tetrahydro- [l,l'-biphenyl]-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
LCMS (ESI+) m/z 477.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.2 Hz, 1H), 7.89 (s, 1H), 7.56 (s, 1H), 7.40 (s, 2H), 6.68 (d, J = 7.0 Hz, 1H), 3.65 - 3.55 (m, 2H), 3.54 - 3.44 (m, 5H), 3.27 (s, 3H), 2.76 - 2.58 (m, 1H), 1.99 - 1.88 (m, 2H), 1.76 - 1.63 (m, 2H), 1.62 - 1.55 (m, 2H), 1.54 - 1.44 (m, 2H). Example 1-022: Synthesis of 2-(5-(2,6-dichloro-4-(4-methoxycyclohexyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 27)
Step 1: 4'-Bromo-3',5'-dichloro-4-methoxy-2,3,4,5-tetrahydro-l,r-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (42% yield), using methyl 2-bromo-l,3-dichloro-5-iodobenzene (100 mg, 0.286 mmol, 1.11 equiv) and 2-(4-methoxycyclohex-l-en- l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K3PO4 (2.3 equiv) as base, Pd(dppf)CL as catalyst (0.12 equiv). Reaction was quenched with water and extracted with ethyl acetate, organic layer was washed with brine, dried over NajSCU and concentrated under reduced pressure. The product was purified by flash column chromatography.
GCMS (ESI+) m/z 333.9, 335.9 [M+H]+
Step 2: 2-(5-(3,5-Dichloro-4'-methoxy-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-4-yl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4'-bromo-3',5'-dichloro-4-methoxy-2,3,4,5-tetrahydro-l,l'-biphenyl (100 mg, 0.299 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1 equiv) as starting materials, K3PO4 (3.5 equiv) as base, Pd(dppf)CL as catalyst (0.1 equiv). Reaction was quenched with water and extracted with ethyl acetate. Organic layer was washed with brine, dried over Na?SO4 and concentrated under reduced pressure to obtain crude which was used directly in the next step. LCMS (ESI+) m/z 431.1, 433.1 [M+H]
Step 3: 2-(5-(2,6-Dichloro-4-(4-methoxycyclohexyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (29% yield over two steps), using methyl 2-(5-(3,5-dichloro-4'-methoxy-2',3',4',5'-tetrahydro-[l,r-biphenyl]-4- yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
LCMS (ESI+) m/z 433.0, 435.0 [M+H]+
1H NMR (400 MHz, DMSO-d6) δ 12.77 - 11.88 (s, 1H), 8.70 (d, J = 7.0 Hz, 1H), 7.97 (s, 1H), 7.62 (s, 1H), 7.43 (s, 2H), 6.75 (d, J = 7.0 Hz, 1H), 3.72 (s, 2H), 3.48 (s, 1H), 3.25 (s, 3H), 2.06 - 1.88 (m, 2H), 1.74 - 1.54 (m, 4H), 1.51 - 1.47 (m, 2H).
Example 1-023: Synthesis of 2-(5-(2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 55)
Step 1: 2-(5-(2-(Trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid [commercial] (150 mg, 0.591 mmol, 1 equiv) and (2- (trifluoromethyl)phenyl)boronic acid (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 321.2 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 8.62 (d, J = 6.9 Hz, 1H), 7.92 - 7.84 (m, 2H), 7.76 (t, J = 7.7 Hz, 1H), 7.66 (t, J = 7.6 Hz, 1H), 7.57 (s, 1H), 7.51 (d, J = 7.7 Hz, 1H), 6.75 (d, J = 7.2 Hz, 1H), 3.48 (s, 2H).
Example 1-024: Synthesis of 2-(5-(2-chloro-6-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 56)
Step 1: 2-(5-(2-Chloro-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (2-chloro-6- methoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 317.1, 319.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.62 (d, J = 7.2 Hz, 1H), 7.89 (s, 1H), 7.50 (s, 1H), 7.41 (t, J = 8.2 Hz, 1H), 7.18 (d, J = 8.0 Hz, 1H), 7.13 (d, J = 8.3 Hz, 1H), 6.66 (d, J = 7.0 Hz, 1H), 3.72 (s, 3H), 3.60 (s, 2H).
Example 1-025: Synthesis of 2-(5-(4-(4-acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 15)
Step 1: tert-Butyl 4-(4-bromo-3,5-dichlorophenyl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (85% yield), using 2- bromo-l,3-dichloro-5-iodobenzene (500 mg, 1.429 mmol, 1.25 equiv) and tert-butyl piperazine-1- carboxylate [commercial] (1 equiv) as starting materials. After completion the solution was filtered through Celite®, evaporated and the resulting crude was used in the next step.
LCMS (ESI+) m/z 410.9, 412.9 [M+H]+ Step 2: l-(4-Bromo-3,5-dichlorophenyl)piperazine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(4-bromo-3,5- dichlorophenyl)piperazine-l-carboxylate (500 mg, 1.21 mmol) as starting material. After completion the volatiles were removed under reduced pressure and the crude product was used directly in the next step.
Step 3: l-(4-(4-Bromo-3,5-dichlorophenyl)piperazin-l-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (93% yield over two steps), using l-(4-bromo-3,5-dichlorophenyl)piperazine trifluoroacetate and acetic anhydride as starting materials.
LCMS (ESI+) m/z 350.8, 352.8 [M+H]+
Step 4: Methyl 2-(5-(4-(4-acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(4-bromo-3,5-dichlorophenyl)piperazin-l-yl)ethan-l-one (80 mg, 0.229 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 461.2 [M+H]+
Step 5: 2-(5-(4-(4-Acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (10 mg, 10% yield two steps), using methyl 2-(5-(4-(4-acetylpiperazin-l-yl)-2,6-dichlorophenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 447.1, 449.1 [M+H]+
NMR (400 MHz, DMSO-d6, 373K) 6 8.60 (d, J = 7.2 Hz, 1H), 7.92 (s, 1H), 7.50 (s, 1H), 7.08 (s, 2H), 6.67 (d, J = 7.2 Hz, 1H), 3.67 (s, 2H), 3.61 (t, J = 5.6 Hz, 4H), 3.33 (t, J = 5.4 Hz, 4H), 2.05 (s, 3H). Example 1-026: Synthesis of 2-(5-(pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 26)
Step 1: Methyl 2-(5-(pyridin-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.317 mmol, 1 equiv) and 4-bromopyridine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 268.0 [M+H]+
Step 2: 2-(5-(Pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (7 mg, 11% yield over two steps), using methyl 2-(5-(pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 254.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 12.73 - 12.10 (s, 1H), 8.76 (d, J = 7.2 Hz, 1H), 8.71 (d, J = 4.6 Hz, 2H), 8.29 (s, 1H), 7.99 (s, 1H), 7.92 (d, J = 5.2 Hz, 2H), 7.34 (d, J = 7.3 Hz, 1H), 3.84 (s, 2H).
Example 1-027: Synthesis of 2-(5-(lH-benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 51)
Step 1: Methyl 2-(5-(lH-benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (150 mg, 0.765 mmol, 1 equiv) and 4-bromo-lH-benzo[d]imidazole [commercial] (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 307.0 [M+H]+
Step 2: 2-(5-(lH-Benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (20 mg, 9% yield over two steps), using methyl 2-(5-(lH-benzo[d]imidazol-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material. After completion the solvents were removed under reduced pressure and the residue was taken up in water, washed with ethyl acetate, acidified with IM HCI and extracted with ethyl acetate. The organic layer was washed with water, brine, dried over Na2SO4 and evaporated to obtain pure product.
LCMS (ESI+) m/z 293.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.2 Hz, 1H), 8.56 - 8.35 (m, 1H), 8.33 (s, 1H), 7.89 (s, 1H), 7.74 - 7.47 (m, 3H), 7.32 (t, J = 7.8 Hz, 1H), 3.65 (s, 2H).
Example 1-028: Synthesis of 2-(5-(2-(/V,/V-dimethylsulfamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 53)
Step 1: Methyl 2-(5-(2-(/V,/V-dimethylsulfamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (180 mg, 0.57 mmol, 1.5 equiv) and 2-bromo-/V,/V-dimethylbenzenesulfonamide [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dppf)Ck as catalyst (0.1 equiv). After completion the reaction mixture was evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 374.1 [M+H]+ Step 2: 2-(5-(2-(/V,/V-Dimethylsulfamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (35 mg, 25% yield over two steps), using methyl 2-(5-(2-(/V,/V-dimethylsulfamoyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting material.
LCMS (ESI+) m/z 360.1 [M+H]+
TH NMR (400 MHz, Methanol-d4) 6 8.42 (d, J = 7.0 Hz, 1H), 8.04 (d, J = 7.9 Hz, 1H), 7.91 (s, 1H), 7.69 (t, J = 7.5 Hz, 1H), 7.65 - 7.56 (m, 2H), 7.47 (d, J = 7.4 Hz, 1H), 6.86 (d, J = 7.2 Hz, 1H), 3.58 (s, 2H), 2.48 (s, 6H).
Example 1-029: Synthesis of 2-(5-(2-chloro-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 42)
Step 1: 2-(5-(2-Chloro-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (14% yield), using 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (50 mg, 0.197 mmol, 1 equiv) and (2-chloro-6- (trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 355.0, 357.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.0 Hz, 1H), 7.95 (d, J = 9.0 Hz, 2H), 7.88 (d, J = 8.0 Hz, 1H), 7.69 (t, J = 8.2 Hz, 1H), 7.54 (s, 1H), 6.70 (d, J = 7.0 Hz, 1H), 3.52 (s, 2H).
Example 1-030: Synthesis of 2-(5-(2-chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 39)
Step 1: To the stirred solution of 2-bromo-l-chloro-3-iodobenzene [commercial] (120 mg, 0.342 mmol, 1.11 equiv), Xantphos (0.22 equiv), Pd?(dba)3 (0.011 equiv) and DIPEA (3.33 equiv) in dioxane (2 mL) was added (4-methoxyphenyl)methanethiol (0.044 mL, 0.308 mmol, 1 equiv). The suspension was purged with argon for 15 min and the reaction mixture was stirred at 90°C for 5 h in a sealed tube. After completion of the reaction, the solvent was evaporated and (2-bromo-3-chlorophenyl)(4-methoxybenzyl)sulfane (90 mg, 69% yield) was purified by flash column chromatography.
GCMS (ESI+) m/z 344.0 [M+H]+
Step 2: To a stirred solution of (2-bromo-3-chlorophenyl)(4-methoxybenzyl)sulfane (420 mg, 1.22 mmol, 1 equiv) in ACN (15 mL), cooled in an ice-water bath, were added acetic acid (0.5 mL) and water (0.5 mL). l,3-Dichloro-5,5-dimethylimidazolidine-2, 4-dione (3 equiv) was added and the resulting mixture was stirred at 0°C for 1 h. After completion, the reaction was quenched with cold water and extracted with DCM. The organic fraction was washed with brine, dried over NajSCU and evaporated to afford crude 2- bromo-3-chlorobenzenesulfonyl chloride (400 mg) which was used directly in the next step.
Step 3: l-((2-Bromo-3-chlorophenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (43% yield over two steps), using 2-bromo-3- chlorobenzenesulfonyl chloride (781 mg, 2.69 mmol, 1 equiv) and piperidine (1.1 equiv) as starting materials.
LCMS (ESI+) m/z 338.0, 339.8 [M+H]+
Step 4: Methyl 2-(5-(2-chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-3-chlorophenyl)sulfonyl)piperidine (100 mg, 0.295 mmol, 1.11 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs2CO3 (4.4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.36 equiv). After completion of the reaction, the solvent was evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 448.0 [M+H]+
Step 5: 2-(5-(2-Chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over three steps), using methyl 2-(5-(2-chloro-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate.
LCMS (ESI+) m/z 434.0 [M+H]+
'H NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.2 Hz, 1H), 8.00 - 7.85 (m, 3H), 7.67 (t, J = 8.0 Hz, 1H), 7.45 (s, 1H), 6.67 (d, J = 7.2 Hz, 1H), 3.62 (d, J = 3.6 Hz, 2H), 2.93 - 2.70 (m, 4H), 1.40 - 1.34 (m, 6H).
Example 1-031: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 47) Step 1: l-((2-Bromophenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (46% yield), using 2-bromobenzenesulfonyl chloride (532 mg, 2.09 mmol, 1 equiv) and 4-methoxypiperidine (1.25 equiv) as starting materials.
LCMS (ESI+) m/z 334.0 [M+H]+
Step 2: Methyl 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (100 mg, 0.3 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion of the reaction, solvent was evaporated, quenched with IM HCI and extracted with dichloromethane to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 443.7 [M+H]+
Step 3: 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (13% yield over two steps), using methyl 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 430.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.1 Hz, 1H), 7.99 (d, J = 8.0 Hz, 1H), 7.92 (s, 1H), 7.75 (t, J = 6.6 Hz, 1H), 7.67 (t, J = 7.6 Hz, 1H), 7.56 (s, 1H), 7.47 (d, J = 6.9 Hz, 1H), 6.81 (d, J = 7.4 Hz, 1H), 3.65 (s, 2H), 3.28 - 3.17 (m, 1H), 3.14 (s, 3H), 3.06 - 2.94 (m, 2H), 2.79 - 2.63 (m, 2H), 1.72 - 1.48 (m, 2H), 1.43 - 1.14 (m, 2H).
Example 1-032: Synthesis of 2-(5-(2-((3-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 44)
Step 1: l-((2-Bromophenyl)sulfonyl)-3-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (46% yield), using 2-bromobenzenesulfonyl chloride (1.1 g, 4.17 mmol, 1 equiv) and 3-methoxypiperidine (1.25 equiv) as starting materials.
LCMS (ESI+) m/z 333.9, 335.9 [M+H]+
Step 2: Methyl 2-(5-(2-((3-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-3-methoxypiperidine (100 mg, 0.3 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was acidified with IM HCI and extracted with ethyl acetate to give crude product which was used directly for the next step.
LCMS (ESI+) m/z 444.2 [M+H]+
Step 3: 2-(5-(2-((3-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (28% yield over two steps), using methyl 2-(5-(2-((3-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 430.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.2 Hz, 1H), 8.01 (d, J = 7.9 Hz, 1H), 7.91 (s, 1H), 7.74 (t, J = 7.4 Hz, 1H), 7.66 (t, J = 7.5 Hz, 1H), 7.58 (s, 1H), 7.46 (d, J = 7.4 Hz, 1H), 6.83 (d, J = 7.0 Hz, 1H), 3.60 (s, 2H), 3.14 - 3.07 (m, 1H), 3.05 (s, 3H), 2.98 - 2.82 (m, 2H), 2.77 - 2.57 (m, 1H), 2.58 - 2.51 (m, 1H), 1.72 - 1.60 (m, 1H), 1.56 - 1.46 (m, 1H), 1.28 - 1.13 (m, 2H). Example 1-033: Synthesis of 2-(5-(2-((4-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 45)
Step 1: l-((2-Bromophenyl)sulfonyl)-4-(2-methoxyethoxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (28% yield), using 2- bromobenzenesulfonyl chloride (385 mg, 1.50 mmol, 1 equiv) and 4-(2-methoxyethoxy)piperidine [commercial] (1.25 equiv) as starting materials.
LCMS (ESI+) m/z 378.0, 380.0 [M+H]+
Step 2: Methyl 2-(5-(2-((4-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-(2-methoxyethoxy)piperidine (100 mg, 0.265 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base and Pd(dppf)CI2 as a catalyst (0.1 equiv). After completion of the reaction, solvent was evaporated, quenched with IM HCI and extracted with dichloromethane to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 488.0 [M+H]+
Step 3: 2-(5-(2-((4-(2-Methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over two steps), using methyl 2-(5-(2-((4-(2-methoxyethoxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 474.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.58 (d, J = 7.1 Hz, 1H), 7.99 (d, J = 7.9 Hz, 1H), 7.90 (s, 1H), 7.75 (t, J = 7.3 Hz, 1H), 7.66 (t, J = 7.8 Hz, 1H), 7.56 (s, 1H), 7.46 (d, J = 7.9 Hz, 1H), 6.79 (d, J = 6.7 Hz, 1H), 3.57 (s, 2H), 3.46 - 3.34 (m, 5H), 3.20 (s, 3H), 3.09 - 2.96 (m, 2H), 2.76 - 2.59 (m, 2H), 1.76 - 1.51 (m, 2H), 1.40 - 1.16 (m, 2H).
Example 1-034: Synthesis of 2-(5-(2-((4-(methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 37)
Step 1: l-((2-Bromophenyl)sulfonyl)-4-(methoxymethyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (59% yield), using 2- bromobenzenesulfonyl chloride (300 mg, 1.176 mmol, 1 equiv) and 4-(methoxymethyl)piperidine [commercial] (1.25 equiv) as starting materials.
LCMS (ESI+) m/z 348.0, 350.2 [M+H]+
Step 2: Methyl 2-(5-(2-((4-(methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-(methoxymethyl)piperidine (88 mg, 0.253 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, Cs2CO3 (5 equiv) as base and Pd(dtbpf)CI2 as a catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 458.1 [M+H]+
Step 3: 2-(5-(2-((4-(Methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (6% yield over two steps), using methyl 2-(5-(2-((4-(methoxymethyl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 444.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.62 (d, J = 7.2 Hz, 1H), 7.99 (dd, J = 7.9, 1.4 Hz, 1H), 7.95 (s, 1H), 7.75 (td, J = 7.5, 1.4 Hz, 1H), 7.67 (td, J = 7.7, 1.5 Hz, 1H), 7.58 (d, J = 1.9 Hz, 1H), 7.47 (dd, J = 7.6, 1.4 Hz, 1H), 3.71 (s, 2H), 3.32 (s, 4H), 3.27 - 3.23 (m, 1H), 3.16 (s, 3H), 3.02 (d, J = 6.0 Hz, 2H), 2.38 (td, J = 12.3, 1.8 Hz, 2H), 1.50 (dd, J = 9.7, 5.6 Hz, 3H), 0.82 (q, J = 12.5 Hz, 2H).
Example 1-035: Synthesis of 2-(5-(4-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 29)
Step 1: 2-(5-(4-(Piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (36% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (60 mg, 0.236 mmol, 1 equiv) and l-(4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)phenyl)piperidine [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 336.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.57 (d, J = 7.4 Hz, 1H), 7.83 (s, 2H), 7.64 (d, J = 8.9 Hz, 2H), 7.13 (dd, J = 2.0, 7.3 Hz, 1H), 7.01 (d, J = 9.0 Hz, 2H), 3.66 (s, 2H), 3.22 (t, J = 5.6 Hz, 4H), 1.74 - 1.48 (m, 6H).
Example 1-036: Synthesis of 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 34) Step 1: l-(4-Bromo-3,5-dichlorophenyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (38% yield), using 2-bromo-l,3-dichloro-5- iodobenzene (300 mg, 0.853 mmol, 1 equiv) and piperidine [commercial] (1.6 equiv) as starting materials.
GCMS (ESI+) m/z 307.9 [M+H]+
Step 2: Methyl 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-bromo-3,5-dichlorophenyl)piperidine (100 mg, 0.324 mmol, 1 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 418.0, 419.9 [M+H]+
Step 3: 2-(5-(2,6-Dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (7% yield over two steps), using methyl 2-(5-(2,6-dichloro-4-(piperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 404.2, 406.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.1 Hz, 1H), 7.91 (s, 1H), 7.52 (s, 1H), 7.08 (s, 2H), 6.67 (dd, J = 1.6, 7.0 Hz, 1H), 3.64 (s, 2H), 3.31 - 3.12 (m, 4H), 1.58 (m, 6H).
Example 1-037: Synthesis of 2-(5-(3-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound
33) Step 1: 2-(5-(3-Methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (36% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (3- methoxyphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 283.1[M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.67 (dd, J = 7.3, 0.9 Hz, 1H), 8.01 (dd, J = 2.1, 0.9 Hz, 1H), 7.92 (s, 1H), 7.42 (t, J = 7.8 Hz, 1H), 7.37 (dt, J = 7.7, 1.4 Hz, 1H), 7.34 (t, J = 2.1 Hz, 1H), 7.22 (dd, J = 7.3, 2.1 Hz, 1H), 6.99 (ddd, J = 8.0, 2.6, 1.2 Hz, 1H), 3.85 (s, 3H), 3.80 (s, 2H).
Example 1-038: Synthesis of 2-(5-(3-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 31)
Step 1: 2-(5-(3-(Trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (40% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (3- (trifluoromethyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 321.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.68 (d, J = 7.2 Hz, 1H), 8.16 - 8.07 (m, 3H), 7.90 (s, 1H), 7.80 - 7.68 (m, 2H), 7.26 (dd, J = 2.0, 7.3 Hz, 1H), 3.66 (s, 2H).
Example 1-039: Synthesis of 2-(5-(2,6-dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-
3-yl)acetic acid (Compound 13)
Step 1: l-(4-Bromo-3,5-dichlorophenyl)-3-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 10 and Example Method 10, above (62% yield), using 2-bromo-l,3- dichloro-5-iodobenzene (500 mg, 1.421 mmol, 1 equiv) and 3-methoxypiperidine [commercial] (1.6 equiv) as starting materials.
NMR (400 MHz, Chloroform-d) 6 6.96 - 6.78 (s, 2H), 3.56 - 3.47 (m, 1H), 3.44 - 3.38 (s, 3H), 3.40 - 3.27 (m, 2H), 3.00 - 2.79 (m, 2H), 2.05 - 1.95 (m, 1H), 1.92 - 1.81 (m, 1H), 1.65 - 1.50 (m, 2H).
Step 2: Methyl 2-(5-(2,6-dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-bromo-3,5-dichlorophenyl)-3-methoxypiperidine (200 mg, 0.59 mmol, 1 equiv) and methyl 2- (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and evaporated to give crude product which was used directly in the next step.
Step 3: 2-(5-(2,6-Dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (10% yield over two steps), using methyl 2-(5-(2,6-dichloro-4-(3-methoxypiperidin-l-yl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 434.3, 436.3 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.2 Hz, 1H), 7.91 (s, 1H), 7.48 (s, 1H), 7.06 (s, 2H), 6.67 (d, J = 7.0 Hz, 1H), 3.67 - 3.56 (m, 2H), 3.49 - 3.34 (m, 1H), 3.28 (s, 3H), 3.12 - 2.84 (m, 2H), 2.00 - 1.81 (m, 2H), 1.79 - 1.66 (m, 2H), 1.59 - 1.32 (m, 2H).
Example 1-040: Synthesis of 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 28)
Step 1: Methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(6-bromo-3,4-dihydroisoquinolin-2(lH)-yl)ethan-l-one [commercial] (80 mg, 0.316 mmol, 1.0 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 364.3 [M+H]+
Step 2: 2-(5-(2-Acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (24% yield over two steps), using methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-6-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 350.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.2 Hz, 1H), 7.96 (s, 1H), 7.87 (s, 1H), 7.72 - 7.53 (m, 2H), 7.30 (d, J = 7.7 Hz, 1H), 7.17 (d, J = 6.9 Hz, 1H), 4.69 (s, 1H), 4.63 (s, 1H), 3.73 - 3.63 (m, 4H), 2.99 - 2.92 (m, 1H), 2.90 - 2.80 (m, 1H), 2.10 (s, 3H).
Example 1-041: Synthesis of 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 30) Step 1: Methyl 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2- (5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (100 mg, 0.316 mmol, 1.25 equiv) and (3-bromophenyl)(piperidin-l-yl)methanone [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 378.1 [M+H]+
Step 2: 2-(5-(3-(Piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(3-(piperidine-l-carbonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 364.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.67 (d, J = 7.3 Hz, 1H), 8.05 (s, 1H), 7.90 (s, 1H), 7.87 (d, J = 7.8 Hz, 1H), 7.78 (s, 1H), 7.56 (t, J = 7.7 Hz, 1H), 7.38 (d, J = 7.6 Hz, 1H), 7.22 (d, J = 7.2 Hz, 1H), 3.72 (s, 2H), 3.67 - 3.56 (m, 2H), 3.53 - 3.11 (m, 2H), 1.71 - 1.35 (m, 6H).
Example 1-042: Synthesis of 2-(5-(4-(tert-butyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 36)
Step 1: 2-(5-(4-(tert-Butyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (44% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (100 mg, 0.394 mmol, 1 equiv) and (4-(tert- butyl)phenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 309.2 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.2 Hz, 1H), 7.92 (s, 1H), 7.88 (s, 1H), 7.71 (d, J = 8.0 Hz, 2H), 7.50 (d, J = 8.0 Hz, 2H), 7.16 (d, J = 7.2 Hz, 1H), 3.68 (s, 2H), 1.32 (s, 9H).
Example 1-043: Synthesis of 2-(5-(4-cyclopropylphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound
Step 1: 2-(5-(4-Cyclopropylphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (63% yield), using 2-(5- bromopyrazolo[l,5-a]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and (4- cyclopropylphenyl)boronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 293.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.61 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.84 (s, 1H), 7.66 (d, J = 8.1 Hz, 2H), 7.18 (d, J = 8.0 Hz, 2H), 7.14 (d, J = 7.3 Hz, 1H), 3.60 (s, 2H), 2.03 - 1.91 (m, 1H), 1.04 - 0.94 (m, 2H), 0.77 - 0.68 (m, 2H).
Example 1-044: Synthesis of 2-(5-(2-methoxy-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 24)
Step 1: l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (28% yield), using 2-bromo-3- methoxybenzenesulfonyl chloride [commercial] (400 mg, 1.404 mmol, 1 equiv) and piperidine (2 equiv) as starting materials.
LCMS (ESI+) m/z 334.0 [M+H]+
Step 2: 2-(5-(2-Methoxy-6-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (29% yield), using l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidine (120 mg, 0.36 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 430.1 [M+H]+
NMR (400 MHz, DMSO-d6, 373K) 6 8.51 (d, J = 7.2 Hz, 1H), 7.89 (s, 1H), 7.66 - 7.55 (m, 2H), 7.50 - 7.31 (m, 2H), 6.63 (d, J = 7.2 Hz, 1H), 3.74 (s, 3H), 3.64 (s, 2H), 2.91 - 2.76 (m, 4H), 1.54 - 1.30 (m, 6H).
Example 1-045: Synthesis of 2-(5-(2,6-dichloro-4-(dimethylamino)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 22)
Step 1: 4-Bromo-3,5-dichloro-/V,/V-dimethylaniline was synthesized using the general procedure shown in
Reaction Scheme 10 and Example Method 10, above (43% yield), using 2-bromo-l,3-dichloro-5- iodobenzene (300 mg, 0.853 mmol, 1 equiv) and dimethylamine (2.5 equiv, 2M solution in THF) as starting materials.
GCMS (ESI+) m/z 267.9 [M+H]+
Step 2: 2-(5-(2,6-Dichloro-4-(dimethylamino)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (39% yield), using 4-bromo-3,5-dichloro-/V,/\/-dimethylaniline (60 mg, 0.3 mmol, 1.33 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 364.0, 366.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.63 (d, J = 7.2 Hz, 1H), 7.91 (s, 1H), 7.51 (s, 1H), 6.84 (s, 2H), 6.66 (d, J = 6.9 Hz, 1H), 3.63 (s, 2H), 2.97 (s, 6H).
Example 1-046: Synthesis of 2-(5-(4-(prop-l-yn-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 23)
Step 1: 2-(5-(4-(Prop-l-yn-l-yl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (27% yield), using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.309 mmol, 1.03 equiv) and l-bromo-4-(prop-l-yn-l-yl)benzene (1 equiv) as starting materials, Cs2CO3 (4.1 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv). l-Bromo-4-(prop-l-yn-l-yl)benzene was synthesized as described in Fujihara, T. et al., Angew. Chem. Int. Ed. 2012, 51, 11487.
LCMS (ESI+) m/z 291.1 [M+H]+ NMR (400 MHz, DMSO-d6) δ 8.65 (d, J = 7.2 Hz, 1H), 8.01 (s, 1H), 7.90 (s, 1H), 7.79 (d, J = 8.0 Hz, 2H),
7.49 (d, J = 7.6 Hz, 2H), 7.20 (d, J = 7.2 Hz, 1H), 3.70 (s, 2H), 2.08 (s, 3H).
Example 1-047: Synthesis of 2-(5-(2-((l-acetylpiperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 6)
Step 1: 4-(2-Bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(2- bromo-3-(trifluoromethyl)phenoxy)piperidine-l-carboxylate (600 mg, 1.415 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was forwarded into the next step. tert-Butyl 4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidine-l-carboxylate was prepared according to the procedure described in WO2018217809A1.
LCMS (ESI+) m/z 323.6, 325.8 [M+H]+
Step 2: l-(4-(2-Bromo-3-(trifluoromethyl)phenoxy)piperidin-l-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (46% yield over two steps), using 4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate (300 mg, 0.68 mmol, 1 equiv) and acetyl chloride (1.5 equiv) as starting materials. LCMS (ESI+) m/z 366.2 [M+H]
Step 3: Methyl 2-(5-(2-((l-acetylpiperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(2-bromo-3-(trifluoromethyl)phenoxy)piperidin-l-yl)ethan-l-one (110 mg, 0.328 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs2CO3 (4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.11 equiv). After completion of the reaction the solvent was evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 476.0 [M+H]+
Step 4: 2-(5-(2-((l-Acetylpiperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(2-((l-acetylpiperidin-4-yl)oxy)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 462.1 [M+H]+
NMR (400 MHz, DMSO-d6, 373K) 6 8.57 (d, J = 7.6 Hz, 1H), 7.90 (s, 1H), 7.60 (t, J = 8.0 Hz, 1H), 7.50 (d, J = 8.0 Hz, 1H), 7.43 (d, J = 7.2 Hz, 2H), 6.68 (d, J = 7.2 Hz, 1H), 4.72 - 4.58 (m, 1H), 3.65 (s, 2H), 3.41 - 3.16 (m, 4H), 1.90 (s, 3H), 1.86 - 1.67 (m, 2H), 1.58 - 1.36 (m, 2H).
Example 1-048: Synthesis of 2-(5-(2-((4-methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 17) Step 1: To a stirred solution of 2-bromo-3-(trifluoromethyl)phenol [commercial] (600 mg, 2.5 mmol, 1 equiv) in THF were added 4-methoxycyclohexan-l-ol [commercial] (391 mg, 3.0 mmol, 1.2 equiv) and PPha (980 mg, 3.75 mmol, 1.5 equiv) at 0°C and the suspension was stirred for 10 min. DIAD (1 mL, 5.0 mmol, 2 equiv) was added and the reaction mixture was stirred at RT for 16 h. After completion, the resulting mixture was quenched with water, extracted with ethyl acetate, then organic layer was washed with brine, dried over Na2SO4 and evaporated under reduced pressure. The crude product was purified by flash column chromatography to obtain 2-bromo-l-((4-methoxycyclohexyl)oxy)-3-(trifluoromethyl)benzene (400 mg, 45% yield).
TH NMR (400 MHz, Chloroform-d) 6 7.40 - 7.26 (m, 2H), 7.07 (t, J = 6.9 Hz, 1H), 4.52 - 4.39 (m, 1H), 3.36 (s, 3H), 2.12 - 1.96 (m, 4H), 1.87 (t, J = 9.4 Hz, 1H), 1.75 - 1.64 (m, 4H).
Step 2: Methyl 2-(5-(2-((4-methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-l-((4-methoxycyclohexyl)oxy)-3-(trifluoromethyl)benzene (55 mg, 0.156 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.1 equiv). After completion, the reaction mixture was quenched with water and extracted with ethyl acetate. The organic layer was washed with brine, dried over Na2SO4 and concentrated to afford crude product which was used directly in the next step.
LCMS (ESI+) m/z 463.2 [M+H]+
Step 3: 2-(5-(2-((4-Methoxycyclohexyl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (31% yield over two steps), using methyl 2-(5-(2-((4-methoxycyclohexyl)oxy)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 449.6 [M+H]+
'H NMR (400 MHz, DMSO-d6) δ 8.60 (d, J = 7.0 Hz, 1H), 7.92 (d, J = 2.0 Hz, 1H), 7.58 (t, J = 8.0 Hz, 1H), 7.52 - 7.44 (m, 1H), 7.46 - 7.36 (m, 2H), 6.67 (d, J = 6.9 Hz, 1H), 4.58 - 4.38 (m, 1H), 3.64 (s, 2H), 3.25 - 2.97 (m, 5H), 1.92 - 1.73 (m, 1H), 1.70 - 1.42 (m, 3H), 1.42 - 1.18 (m, 3H). Example 1-049: Synthesis of 2-(5-(2-((l-(methylsulfonyl)piperidin-4-yl)oxy)-6-
(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 7)
Step 1: 4-(2-Bromo-3-(trifluoromethyl)phenoxy)-l-(methylsulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (64% yield), using 4-(2- bromo-3-(trifluoromethyl)phenoxy)piperidine trifluoroacetate (300 mg, 0.69 mmol, 1 equiv) and methanesulfonyl chloride (2 equiv) as starting materials.
LCMS (ESI+) m/z 401.9, 403.8 [M+H]+
Step 2: Methyl 2-(5-(2-((l-(methylsulfonyl)piperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4-(2-bromo-3-(trifluoromethyl)phenoxy)-l-(methylsulfonyl)piperidine (130 mg, 0.323 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 512.2 [M+H]+
Step 3: 2-(5-(2-((l-(Methylsulfonyl)piperidin-4-yl)oxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin- 3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (16% yield over two steps), using methyl 2-(5-(2-((l-(methylsulfonyl)piperidin-4-yl)oxy)- 6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 498.0 [M+H]+ XH NMR (400 MHz, DMSO-d6, 373K) 6 8.60 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.61 (t, J = 8.0 Hz, 1H), 7.50 (d, J = 8.4 Hz, 1H), 7.47 - 7.42 (m, 2H), 6.72 (d, J = 7.6 Hz, 1H), 4.86 - 4.50 (m, 1H), 3.68 (s, 2H), 3.20 - 3.02 (m, 2H), 2.81 - 2.67 (m, 2H), 2.45 (s, 3H), 1.93 - 1.53 (m, 4H).
Example 1-050: Synthesis of 2-(5-(5-methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 12)
Step 1: Methyl 2-(5-(5-methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-4-methoxy-l-(trifluoromethyl)benzene [commercial] (150 mg, 0.588 mmol, 1.0 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite®, the solvent was evaporated and the crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 365.2 [M+H]+
Step 2: 2-(5-(5-Methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(5-methoxy-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting material.
LCMS (ESI+) m/z 351.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.2 Hz, 1H), 7.92 (s, 1H), 7.78 (d, J = 8.8 Hz, 1H), 7.59 (s, 1H), 7.17 (dd, J = 2.6, 8.8 Hz, 1H), 7.01 (d, J = 2.4 Hz, 1H), 6.78 (d, J = 7.2 Hz, 1H), 3.87 (s, 3H), 3.57 (s, 2H). Example 1-051: Synthesis of 2-(5-(4-(trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 20)
Step 1: Methyl 2-(5-(4-(trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 3-bromo- 4-(trifluoromethyl)pyridine [commercial] (150 mg, 0.664 mmol, 1 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, Cs2CO3 (2.7 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 336.3 [M+H]+
Step 2: 2-(5-(4-(Trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (20% yield over two steps), using methyl 2-(5-(4-(trifluoromethyl)pyridin-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 322.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.92 (d, J = 5.1 Hz, 1H), 8.79 (s, 1H), 8.72 (d, J = 7.2 Hz, 1H), 7.99 (s, 1H), 7.90 (d, J = 5.1 Hz, 1H), 7.72 (s, 1H), 6.87 (d, J = 7.3 Hz, 1H), 3.69 (s, 2H).
Example 1-052: Synthesis of 2-(5-(5-(l-acetylpiperidin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 11)
Step 1: tert-Butyl 4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridine-l(2H)-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (86% yield), using 2-bromo-4-iodo-l-(trifluoromethyl)benzene [commercial] (2.5 g, 7.143 mmol, l.ll equiv) and tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-3,6-dihydropyridine-l(2H)-carboxylate (1 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(PPh3)2Cl2 as catalyst (0.11 equiv).
TH NMR (400 MHz, Chloroform-d) 6 7.69 (s, 1H), 7.63 (d, J = 8.3 Hz, 1H), 7.37 (d, J = 8.3 Hz, 1H), 6.22 - 6.07 (m, 1H), 4.15 - 4.07 (m, 2H), 3.64 (t, J = 5.7 Hz, 2H), 2.55 - 2.43 (m, 2H), 1.49 (s, 9H).
Step 2: 4-(3-Bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridine-l(2H)-carboxylate (500 mg, 1.232 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure, the residue was washed with ether and dried to afford crude product which was directly forwarded into the next step.
LCMS (ESI+) m/z 306.0, 308.0 [M+H]+
Step 3: l-(4-(3-Bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridin-l(2H)-yl)ethan-l-one was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (70% yield over two steps), using 4-(3-bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (350 mg, 0.835 mmol, 1 equiv) and acetyl anhydride (1.5 equiv) as staring materials.
LCMS (ESI+) m/z 347.9 [M+H]+
Step 4: Methyl 2-(5-(5-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-(4-(3-bromo-4-(trifluoromethyl)phenyl)-3,6-dihydropyridin-l(2H)- yl)ethan-l-one (110 mg, 0.316 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan- 2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Cs2CO3 (4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.1 equiv). After completion of the reaction the solvent was evaporated to give crude product which was used directly in the next step.
LCMS (ESI+) m/z 458.2 [M+H]+
Step 5: 2-(5-(5-(l-Acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (57% yield over two steps), using methyl 2-(5-(5-(l-acetyl-l,2,3,6- tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.219 mmol) as starting material. After completion the solvents were removed under reduced pressure, the residue was triturated with diluted HCI and dried to afford pure product.
LCMS (ESI+) m/z 444.2 [M+H]+
Step 6: 2-(5-(5-(l-Acetylpiperidin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (7% yield), using 2-(5-(5-(l-acetyl-l,2,3,6-tetrahydropyridin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (80 mg, 0.181 mmol) as starting material.
LCMS (ESI+) m/z 446.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.2 Hz, 1H), 7.96 (s, 1H), 7.80 (d, J = 8.4 Hz, 1H), 7.60 (s, 1H), 7.53 (d, J = 8.0 Hz, 1H), 7.39 (s, 1H), 6.80 (d, J = 6.8 Hz, 1H), 4.69 - 4.37 (m, 1H), 4.00 - 3.82 (m, 1H), 3.69 (s, 2H), 3.21 - 2.99 (m, 1H), 3.01 - 2.81 (m, 1H), 2.64 - 2.51 (m, 1H), 2.01 (s, 3H), 1.93 - 1.76 (m, 2H), 1.74
- 1.59 (m, 1H), 1.58 - 1.41 (m, 1H).
Example 1-053: Synthesis of 2-(5-(5-(l-(methylsulfonyl)piperidin-4-yl)-2-
(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 8)
Step 1: 4-(3-Bromo-4-(trifluoromethyl)phenyl)-l-(methylsulfonyl)-l,2,3,6-tetrahydropyridine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (64% yield), using 4-(3-bromo-4-(trifluoromethyl)phenyl)-l,2,3,6-tetrahydropyridine trifluoroacetate (350 mg,
1.144 mmol, 1 equiv) and methanesulfonyl chloride (2 equiv) as starting materials.
LCMS (ESI+) m/z 382.0, 384.0 [M+H]+
Step 2: Methyl 2-(5-(5-(l-(methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2-
(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 4-(3-bromo-4-(trifluoromethyl)phenyl)- l-(methylsulfonyl)-l,2,3,6-tetrahydropyridine (120 mg, 0.313 mmol, 1 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion of the reaction the solvent was evaporated to give crude product which was used in the next step.
LCMS (ESI+) m/z 494.1 [M+H]+
Step 3: 2-(5-(5-(l-(Methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2-
(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (53% yield over two steps), using methyl 2-(5-(5-(l-(methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2-
(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material. After completion the solvents were removed under reduced pressure, the residue was triturated with diluted HCI and dried to afford pure product.
LCMS (ESI+) m/z 480.1 [M+H]+
Step 4: 2-(5-(5-(l-(Methylsulfonyl)piperidin-4-yl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (27% yield), using 2-(5-(5-(l-(methylsulfonyl)-l,2,3,6-tetrahydropyridin-4-yl)-2- (trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (80 mg, 0.167 mmol) as starting material.
LCMS (ESI+) m/z 482.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.64 (d, J = 7.2 Hz, 1H), 7.93 (s, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.62 - 7.53 (m, 2H), 7.40 (s, 1H), 6.79 (d, J = 7.2 Hz, 1H), 3.69 (d, J = 10.8 Hz, 2H), 3.60 (s, 2H), 2.90 (s, 3H), 2.81 (t, J = 11.2 Hz, 3H), 2.02 - 1.89 (m, 2H), 1.86 - 1.65 (m, 2H).
Example 1-054: Synthesis of 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 19) Step 1: Methyl 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (135 mg, 0.425 mmol, 1.1 equiv) and 2-bromo-4-chloro-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 369.2 [M+H]+
Step 2: 2-(5-(5-Chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (25% yield over 2 steps), using methyl 2-(5-(5-chloro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as a starting material.
LCMS (ESI+) m/z 355.1, 357.1 [M+H]+
'H NMR (400 MHz, DMSO-d6) δ 8.67 (d, J = 7.3 Hz, 1H), 7.96 (s, 1H), 7.90 (d, J = 8.4 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 7.65 (d, J = 9.6 Hz, 2H), 6.82 (d, J = 8.0 Hz, 1H), 3.66 (s, 2H).
Example 1-055: Synthesis of 2-(5-(5-fluoro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 21)
Step 1: 2-(5-(5-Fluoro-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (65% yield), using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (79 mg, 0.247 mmol, 1 equiv) and 2-bromo-4-fluoro-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv). LCMS (ESI+) m/z 339.0 [M+H]
1H NMR (400 MHz, DMSO-cfe) 6 8.64 (d, J = 7.0 Hz, 1H), 7.99 - 7.89 (m, 2H), 7.62 (s, 1H), 7.51 (t, J = 8.6 Hz, 1H), 7.43 (d, J = 8.7 Hz, 1H), 6.78 (d, J = 6.8 Hz, 1H), 3.49 (s, 2H).
Example 1-056: Synthesis of 2-(5-(5-methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 18)
Step 1: Methyl 2-(5-(5-methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (100 mg, 0.315 mmol, 1 equiv) and 2-bromo-4-methyl-l-(trifluoromethyl)benzene [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 349.2 [M+H]+
Step 2: 2-(5-(5-Methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (15% yield over two steps), using methyl 2-(5-(5-methyl-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate as starting material.
LCMS (ESI-) m/z 333.1 [M-H]’
TH NMR (400 MHz, DMSO-d6, 373K) 6 8.61 (d, J = 7.2 Hz, 1H), 7.94 (s, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.54 (s, 1H), 7.46 (d, J = 8.4 Hz, 1H), 7.32 (s, 1H), 6.78 (d, J = 6.8 Hz, 1H), 3.71 (s, 2H), 2.44 (s, 3H). Example 1-057: Synthesis of 2-(5-(3-(trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid
(Compound 16)
Step 1: Methyl 2-(5-(3-(trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (105 mg, 0.332 mmol, 1 equiv) and 4-bromo-3-(trifluoromethyl)pyridine [commercial] (1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 336.1 [M+H]+
Step 2: 2-(5-(3-(Trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (22% yield over two steps), using methyl 2-(5-(3-(trifluoromethyl)pyridin-4-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate as a starting material.
LCMS (ESI-) m/z 320.0 [M-H]’
NMR (400 MHz, DMSO-d6, 373K) 69.03 (s, 1H), 8.92 (d, J = 5.2 Hz, 1H), 8.67 (d, J = 7.2 Hz, 1H), 7.98 (s, 1H), 7.67 (s, 1H), 7.57 (d, J = 5.2 Hz, 1H), 6.83 (d, J = 6.8 Hz, 1H), 3.70 (s, 2H).
Example 1-058: Synthesis of 2-(5-(5-(4-methoxycyclohexyl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 14)
Step 1: 3'-Bromo-4-methoxy-4'-(trifluoromethyl)-2,3,4,5-tetrahydro-l,l'-biphenyl was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (41% yield), using 2- bromo-4-iodo-l-(trifluoromethyl)benzene (280 mg, 0.8 mmol, 1.11 equiv) and 2-(4-methoxycyclohex-l- en-l-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane [commercial] (1 equiv) as starting materials, K3PO4 (3.33 equiv) as base and Pd(PPh3)2Cl2 as catalyst (0.11 equiv).
GCMS (ESI+) m/z 335.9 [M+H]+
Step 2: Methyl 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-3- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 3'-bromo-4-methoxy-4'-(trifluoromethyl)-2, 3,4,5- tetrahydro-l,l'-biphenyl (120 mg, 0.359 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Ch as catalyst (0.1 equiv). After completion of the reaction the solvent was evaporated to give crude mixture of product and 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,l'- biphenyl]-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid which was used directly in the next step.
LCMS (ESI+) m/z 445.1 [M+H] Step 3: 2-(5-(4'-Methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-3-yl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using mixture of methyl 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'- tetrahydro-[l,l'-biphenyl]-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(4'-methoxy-4- (trifluoromethyl)-2',3',4',5'-tetrahydro-[l,l'-biphenyl]-3-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting materials. After completion the solvents were removed under reduced pressure and crude product was used directly in the next step.
LCMS (ESI+) m/z 431.2 [M+H]+
Step 4: 2-(5-(5-(4-Methoxycyclohexyl)-2-(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 6 and Example Method 6, above (16% yield over three steps), using 2-(5-(4'-methoxy-4-(trifluoromethyl)-2',3',4',5'-tetrahydro-[l,r- biphenyl]-3-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material.
LCMS (ESI+) m/z 433.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.2 Hz, 1H), 7.96 (s, 1H), 7.78 (d, J = 8.2 Hz, 1H), 7.60 (s, 1H), 7.49 (d, J = 8.2 Hz, 1H), 7.30 (s, 1H), 6.81 (d, J = 7.1 Hz, 1H), 3.71 (s, 2H), 3.51 - 3.44 (m, 1H), 3.23 (s, 3H), 2.82 - 2.58 (m, 1H), 2.01 - 1.92 (m, 2H), 1.79 - 1.45 (m, 6H).
Example 1-059: Synthesis of 2-(5-(2-(cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 5)
Step 1: To a stirred solution of 2-bromo-3-(trifluoromethyl)phenol (200 mg, 0.83 mmol, 1 equiv) in THF (3 mL), cooled in an ice-water bath, were added PPh3 (1.5 equiv), DIAD (1.5 equiv) and cyclohexanol (1 equiv) and the resulting mixture was stirred at RT for 16 h. After completion, the reaction was quenched with saturated NaHCO3 solution and extracted with ethyl acetate. The combined organic fractions were washed with brine, dried over NajSCU and evaporated to give crude product. 2-Bromo-l-(cyclohexyloxy)-3- (trifluoromethyl)benzene (160 mg, 0.49 mmol, 59% yield) was purified by flash column chromatography.
NMR (400 MHz, CDCI3) 6 7.36 - 7.22 (m, 2H), 7.07 (d, J = 8.1 Hz, 1H), 4.44 - 4.28 (m, 1H), 1.91 (d, J = 10.8 Hz, 2H), 1.87 (m, 2H), 1.76 - 1.62 (m, 2H), 1.41 (d, J = 8.4 Hz, 4H).
Step 2: Methyl 2-(5-(2-(cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo-l-(cyclohexyloxy)-3-(trifluoromethyl)benzene (100 mg, 0.311 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (3 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion, the solvents were evaporated, the residue was quenched with water, extracted with ethyl acetate and dried over Na2SO4. The organic layer was concentrated to afford crude product which was used directly in the next step.
LCMS (ESI+) m/z 432.8 [M+H]+
Step 3: 2-(5-(2-(Cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (23% yield over two steps), using methyl 2-(5-(2-(cyclohexyloxy)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 419.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.61 (d, J = 7.1 Hz, 1H), 7.91 (s, 1H), 7.58 (t, J = 8.0 Hz, 1H), 7.47 (d, J = 8.3 Hz, 1H), 7.43 (s, 1H), 7.39 (d, J = 7.8 Hz, 1H), 6.67 (dd, J = 1.2, 7.1 Hz, 1H), 4.49 - 4.38 (m, 1H), 3.65 (s, 2H), 1.92 - 1.59 (m, 2H), 1.53 - 1.04 (m, 8H).
Example 1-060: Synthesis of 2-(5-(2,5-bis((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 2)
Step 1: To a stirred solution of 2-bromo-l,4-diiodobenzene (1.0 g, 2.45 mmol, 1 equiv) in dioxane (5 mL) were added (4-methoxyphenyl)methanethiol (1.9 equiv) and DIPEA (6 equiv). The mixture was then bubbled with argon for 10 min and XantPhos (0.2 equiv) and Pd?(dba)3 (0.1 equiv) were added and the mixture was further bubbled with argon for 5 min and stirred in a sealed tube at 90°C for 3 h. After completion the volatiles were removed under reduced pressure and (2-bromo-l,4-phenylene)bis((4- methoxybenzyl)sulfane) (400 mg, 0.87 mmol, 35% yield) was purified by flash column chromatography.
GCMS (ESI+) m/z 462.1 [M+H]+
Step 2: To a stirred solution of (2-bromo-l,4-phenylene)bis((4-methoxybenzyl)sulfane) (340 mg, 0.74 mmol, 1 equiv) in ACN (5 mL) were added AcOH (0.3 mL), water (0.3 mL) and the solution was cooled to 0°C. After 10 min l,3-dichloro-5,5-dimethylhydantoin (6 equiv) was added and the reaction was stirred at RT for 1 h. After completion, the reaction was quenched with water and extracted with DCM. The combined organic fractions were washed with brine, dried over NajSCU and evaporated to afford crude 2- bromobenzene-l,4-disulfonyl dichloride, which was forwarded directly for the next step.
Step 3: l,l'-(2-Bromo-l,4-phenylenedisulfonyl)bis(4-methoxypiperidine) was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (43% yield over two steps), using 2-bromobenzene-l,4-disulfonyl dichloride (260 mg, 0.739 mmol, 1 equiv) and 4-methoxypiperidine (37 equiv) as starting materials.
LCMS (ESI+) m/z 511.0, 513.0 [M+H]+ Step 4: 2-(5-(2,5-Bis((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using l,l'-(2-bromo-l,4-phenylenedisulfonyl)bis(4-methoxypiperidine) (100 mg, 0.196 mmol,
1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 607.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.1 Hz, 1H), 8.22 (d, J = 8.3 Hz, 1H), 8.08 - 7.90 (m, 2H), 7.69 (d, J = 13.2 Hz, 2H), 6.89 (d, J = 6.9 Hz, 1H), 3.73 (s, 2H), 3.29-3.11 (m, 10H), 3.14 - 2.99 (m, 2H), 2.88 (t, J = 4.9 Hz, 2H), 2.80 - 2.69 (m, 2H), 1.95 - 1.76 (m, 2H), 1.72 - 1.43 (m, 4H), 1.38 - 1.20 (m, 2H).
Example 1-061: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)-4-methylpyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 3)
Step 1: 2-(5-Bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (73% yield), using 2-(5-bromo-4- methylpyrazolo[l,5-o]pyridin-3-yl)acetonitrile (700 mg, 2.789 mmol) as starting material. After completion the reaction mixture was diluted with cold water, washed with ether and acidified with IM HCI. The precipitated product was filtered and dried to afford pure product.
LCMS (ESI+) m/z 269.1 [M+H]+ Step 2: To a stirred solution of 2-(5-bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetic acid (550 mg, 2.157 mmol) in methanol (10 mL) was added H2SO4 (0.5 mL) at RT and the solution was heated at 85°C for 6 h. After completion the solvent was evaporated and the residue was taken up in water and extracted with ethyl acetate. The organic layer was dried over NazSCU and concentrated to give pure methyl 2-(5-bromo- 4-methylpyrazolo[l,5-o]pyridin-3-yl)acetate (520 mg, 85% yield).
LCMS (ESI+) m/z 283.2, 285.0 [M+H]+
Step 3: Methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (46% yield), using methyl 2-(5-bromo-4-methylpyrazolo[l,5-a]pyridin-3-yl)acetate (500 mg, 1.943 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 331.33 [M+H]+
Step 4: 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)-4-methylpyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (3.8% yield), using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (100 mg, 0.299 mmol, 1 equiv) and methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst.
LCMS (ESI+) m/z 444.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.41 (d, J = 7.1 Hz, 1H), 7.98 (d, J = 7.9 Hz, 1H), 7.83 (s, 1H), 7.78 - 7.70 (m, 1H), 7.70 - 7.62 (m, 1H), 7.33 (dd, J = 7.4, 1.4 Hz, 1H), 6.56 (d, J = 7.0 Hz, 1H), 3.73 (s, 2H), 3.28 - 3.20 (m, 1H), 3.17 (s, 3H), 3.08 - 2.95 (m, 2H), 2.76 - 2.64 (m, 2H), 2.18 (s, 3H), 1.77 - 1.64 (m, 2H), 1.44 - 1.28 (m, 2H).
Example 1-062: Synthesis of 2-(5-(2,5-bis((4-methoxypiperidin-l-yl)sulfonyl)phenyl)-4- methylpyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1)
Step 1: 2-(5-(2,5-Bis((4-methoxypiperidin-l-yl)sulfonyl)phenyl)-4-methylpyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (26% yield), using l,l'-(2-bromo-l,4-phenylenedisulfonyl)bis(4-methoxypiperidine) (120 mg, 0.235 mmol, 1 equiv) and methyl 2-(4-methyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 621.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.48 (d, J = 6.9 Hz, 1H), 8.23 (d, J = 8.1 Hz, 1H), 8.00 (d, J = 8.1 Hz, 1H), 7.89 (s, 1H), 7.60 (s, 1H), 6.65 (d, J = 7.1 Hz, 1H), 3.82 (s, 2H), 3.28 - 3.23 (m, 2H), 3.18 - 3.14 (m, 8H), 3.08 - 2.99 (m, 2H), 2.94 - 2.83 (m, 2H), 2.78 - 2.69 (m, 2H), 2.19 (s, 3H), 1.85 - 1.79 (m, 2H), 1.75 - 1.69 (m, 2H), 1.53 - 1.44 (m, 2H), 1.39 - 1.32 (m, 2H).
Example 1-063: Synthesis of 2-(5-(2,6-bis(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 43)
Step 1: Methyl 2-(5-(2,6-bis(trifluoromethyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-bromo- l,3-bis(trifluoromethyl)benzene [commercial] (100 mg, 0.316 mmol, 1.25 equiv) and methyl 2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (5 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.23 equiv). After completion, the reaction mixture was filtered through Celite® and solvent was evaporated. The crude product was used in the next step without additional purification.
LCMS (ESI+) m/z 403.2 [M+H]+
Step 2: 2-(5-(2,6-Bis(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield over 2 steps), using methyl 2-(5-(2,6-bis(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 389.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.66 (d, J = 7.1 Hz, 1H), 8.21 (d, J = 8.0 Hz, 2H), 7.97 (s, 1H), 7.91 (t, J = 7.9 Hz, 1H), 7.61 (s, 1H), 6.80 (d, J = 6.9 Hz, 1H), 3.66 (s, 2H).
Example 1-064: Synthesis of 2-(5-(p-tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 35)
Step 1: 2-(5-(p-Tolyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (48% yield), using 2-(5-bromopyrazolo[l,5- o]pyridin-3-yl)acetic acid (70 mg, 0.276 mmol, 1 equiv) and p-tolylboronic acid [commercial] (1.5 equiv) as starting materials, Cs2CO3 (4 equiv) as base, Pd(dtbpf)CI2 as catalyst (0.3 equiv).
LCMS (ESI+) m/z 267.2 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.61 (d, J = 7.3 Hz, 1H), 7.92 (s, 1H), 7.85 (s, 1H), 7.68 (d, J = 7.9 Hz, 2H), 7.30 (d, J = 7.8 Hz, 2H), 7.14 (d, J = 6.3 Hz, 1H), 3.58 (s, 2H), 2.36 (s, 3H). Example 1-065: Synthesis of 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 52)
Step 1: Methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (100 mg, 0.316 mmol, 1.25 equiv) and l-(7-bromo-3,4-dihydroisoquinolin-2(lH)-yl)ethan-l-one [commercial] (1 equiv) as starting materials, CS2CO3 (5 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.125 equiv). After completion the solvents were evaporated, the residue was quenched with IM HCI and extracted with DCM. The organic layer was evaporated to afford crude product which was used directly in the next step.
LCMS (ESI+) m/z 364.0 [M+H]+
Step 2: 2-(5-(2-Acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (43% yield over two steps), using methyl 2-(5-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-7-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 350.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.64 (dd, J = 7.3, 5.6 Hz, 1H), 7.97 (dd, J = 8.9, 2.0 Hz, 1H), 7.87 (s, 1H), 7.69 - 7.64 (m, 1H), 7.61 (dt, J = 8.1, 2.6 Hz, 1H), 7.29 (d, J = 8.0 Hz, 1H), 7.17 (ddd, J = 7.4, 5.3, 2.0 Hz, 1H), 4.73 (s, 1H), 4.69 (s, 1H), 3.68 (t, J = 6.0 Hz, 2H), 3.63 (d, J = 6.8 Hz, 2H), 2.91 (t, J = 6.0 Hz, 1H), 2.79 (t, J = 6.0 Hz, 1H), 2.10 (d, J = 3.5 Hz, 3H).
Example 1-066: Synthesis of 2-(5-(2-(piperidin-l-ylsulfonyl)-6-(trifluoromethyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 40)
Step 1: l-((2-Bromo-3-(trifluoromethyl)phenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (47% yield), using 2-bromo-3- (trifluoromethyl)benzenesulfonyl chloride [commercial] (185 mg, 0.572 mmol, 1 equiv) and piperidine (3 equiv) as starting materials.
GCMS (ESI+) m/z 372.0 [M+H]+
Step 2: Methyl 2-(5-(2-(piperidin-l-ylsulfonyl)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-3-(trifluoromethyl)phenyl)sulfonyl)piperidine (50 mg, 0.135 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
LCMS (ESI+) m/z 482.3 [M+H]+
Step 3: 2-(5-(2-(Piperidin-l-ylsulfonyl)-6-(trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (8% yield over two steps), using methyl 2-(5-(2-(piperidin-l-ylsulfonyl)-6- (trifluoromethyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 468.0 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.41 - 12.17 (s, 1H), 8.60 (d, J = 7.2 Hz, 1H), 8.30 (d, J = 7.9 Hz, 1H), 8.17 (t, J = 7.2 Hz, 1H), 7.95 (s, 1H), 7.89 (t, J = 8.0 Hz, 1H), 7.51 (d, J = 11.8 Hz, 1H), 6.76 (d, J = 6.5 Hz, 1H), 3.69 (s, 2H), 2.90 - 2.75 (m, 4H), 1.40 (s, 6H). Example 1-067: Synthesis of 2-(5-(4-methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (Compound 25)
Step 1: l-((2-Bromo-5-methylphenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (67% yield), using 2-bromo-5- methylbenzenesulfonyl chloride [commercial] (150 mg, 0.557 mmol, 1 equiv) and piperidine (2 equiv) as starting materials. After completion the reaction mixture was diluted with cold water and extracted with DCM. The organic fraction was dried over NajSCU and evaporated to give crude product.
LCMS (ESI+) m/z 317.9 [M+H]+
Step 2: Methyl 2-(5-(4-methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-5-methylphenyl)sulfonyl)piperidine (60 mg, 0.189 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dppf)Ck as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
LCMS (ESI+) m/z 427.7 [M+H]+
Step 3: 2-(5-(4-Methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (44% yield over two steps), using methyl 2-(5-(4-methyl-2-(piperidin-l-ylsulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 414.1 [M+H] NMR (400 MHz, DMSO-d6) δ 8.58 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.79 (s, 1H), 7.54 (d, J = 9.3 Hz, 2H), 7.34 (d, J = 7.7 Hz, 1H), 6.79 (d, J = 7.2 Hz, 1H), 3.61 (s, 2H), 2.78 (d, J = 6.2 Hz, 4H), 2.46 (s, 3H), 1.53 - 1.16 (m, 6H).
Example 1-068: Synthesis of 2-(5-(2-((3-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 46)
Step 1: To a stirred solution of tert-butyl 3-hydroxypiperidine-l-carboxylate (1.0 g, 4.97 mmol, 1 equiv) in THF (6 mL), cooled in an ice-water bath, was added NaH (2 equiv, 60% suspension in mineral oil) followed by l-bromo-2-methoxyethane (1.2 equiv). The reaction mixture was stirred at RT for 3 h, quenched with cold water and extracted with DCM. Combined organic fractions were dried over NajSCU and evaporated. tert-Butyl 3-(2-methoxyethoxy)piperidine-l-carboxylate (800 mg, 3.1 mmol, 62% yield) was purified by flash column chromatography.
XH NMR (400 MHz, DMSO-d6) δ 4.81 (d, J = 4.2 Hz, 1H), 4.03 (qd, J = 7.1, 1.9 Hz, 1H), 3.74 (s, 1H), 3.64 - 3.54 (m, 1H), 3.37 (d, J = 7.2 Hz, 1H), 2.77 (s, 1H), 2.61 (s, 2H), 1.98 (d, J = 2.0 Hz, 3H), 1.80 (q, J = 5.8 Hz, 1H), 1.67 - 1.55 (m, 1H), 1.38 (d, J = 2.1 Hz, 9H), 1.27 (dq, J = 10.5, 6.7 Hz, 2H), 1.17 (td, J = 7.1, 2.0 Hz, 1H). Step 2: 3-(2-Methoxyethoxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (47% yield), using tert-butyl 3-(2- methoxyethoxy)piperidine-l-carboxylate (300 mg, 1.15 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure to afford crude product which was directly forwarded to the next step.
Step 3: l-((2-Bromophenyl)sulfonyl)-3-(2-methoxyethoxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (76% yield), using 3-(2- methoxyethoxy)piperidine trifluoroacetate (700 mg, 2.76 mmol, 1 equiv) and 2-bromobenzenesulfonyl chloride (1.2 equiv) as starting materials.
LCMS (ESI+) m/z 378.0, 380.0 [M+H]+
Step 4: Methyl 2-(5-(2-((3-(2-methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-3-(2-methoxyethoxy)piperidine (100 mg, 0.265 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.5 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dppf)Ck as catalyst (0.1 equiv). After completion the reaction was quenched with IM HCI and the volatiles were removed under reduced pressure. The obtained crude product was forwarded into the next step.
LCMS (ESI+) m/z 487.9 [M+H]+
Step 5: 2-(5-(2-((3-(2-Methoxyethoxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield over two steps), using methyl 2-(5-(2-((3-(2-methoxyethoxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 474.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.59 (d, J = 7.2 Hz, 1H), 8.01 (dd, J = 7.9, 1.4 Hz, 1H), 7.90 (s, 1H), 7.75 (td, J = 7.5, 1.4 Hz, 1H), 7.66 (td, J = 7.7, 1.5 Hz, 1H), 7.58 (d, J = 2.0 Hz, 1H), 7.46 (dd, J = 7.5, 1.4 Hz, 1H), 6.83 (dd, J = 7.2, 2.0 Hz, 1H), 3.56 (s, 2H), 3.32 - 3.31 (m, 4H), 3.17 (s, 3H), 3.17 - 3.11 (m, 1H), 3.10 - 3.02 (m, 2H), 2.91 (d, J = 12.3 Hz, 1H), 2.64 - 2.56 (m, 1H), 1.78 - 1.65 (m, 1H), 1.65 - 1.49 (m, 1H), 1.30 - 1.12 (m,
2H).
Example 1-069: Synthesis of 2-(5-(2-((3-(methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 38)
Step 1: l-((2-Bromophenyl)sulfonyl)-3-(methoxymethyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (51% yield), using 2- bromobenzenesulfonyl chloride (200 mg, 0.784 mmol, 1 equiv) and 3-(methoxymethyl)piperidine (1.2 equiv) as starting materials.
LCMS (ESI+) m/z 348.0, 349.8 [M+H]+
Step 2: 2-(5-(2-((3-(Methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (32% yield), using l-((2-bromophenyl)sulfonyl)-3-(methoxymethyl)piperidine (88 mg, 0.253 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.25 equiv) as starting materials, CS2CO3 (5 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.126 equiv).
LCMS (ESI+) m/z 444.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.61 (d, J = 7.2 Hz, 1H), 7.97 (d, J = 6.7 Hz, 1H), 7.93 (s, 1H), 7.78 - 7.72 (m, 1H), 7.70 - 7.65 (m, 1H), 7.57 (s, 1H), 7.47 (d, J = 7.5 Hz, 1H), 6.83 (dd, J = 7.2, 1.9 Hz, 1H), 3.67 (s, 2H), 3.26 - 3.15 (m, 3H), 3.10 (s, 3H), 3.02 - 2.93 (m, 1H), 2.40 - 2.32 (m, 1H), 2.22 - 2.12 (m, 1H), 1.51 (m, 3H), 1.23 - 1.09 (m, 1H), 0.96 - 0.89 (m, 1H). Example 1-070: Synthesis of 3-((lH-tetrazol-5-yl)methyl)-5-(2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a] pyridine (Compound 72)
Step 1: 4-Methoxy-l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (98% yield), using l-((2-bromophenyl)sulfonyl)-4-methoxypiperidine (3 g, 9.009 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 382.1 [M+H]+
Step 2: 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetonitrile was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (37% yield), using 4-methoxy-l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine (100 mg, 0.262 mmol, 1 equiv) and 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetonitrile (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 411.1 [M+H]+
Step 3: To a stirred solution of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin- 3-yl)acetonitrile (40 mg, 0.097 mmol, 1 equiv) in DMF (1 mL) were added sodium azide (20 mg, 0.292 mmol, 3 equiv) and zinc bromide (65 mg, 0.292 mmol, 3 equiv) and the reaction mixture was stirred at 140°C for 16 h. After completion, 3-((lH-tetrazol-5-yl)methyl)-5-(2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridine (24 mg, 54% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 454.2 [M+H] NMR (400 MHz, DMSO-d6) δ 8.58 (s, 1H), 7.97 (d, J = 8.0 Hz, 2H), 7.74 (s, 1H), 7.67 (d, J = 7.4 Hz, 1H), 7.55 (s, 1H), 7.43 (d, J = 7.5 Hz, 1H), 6.80 (d, J = 7.1 Hz, 1H), 4.27 (s, 2H), 3.14 (m, 4H), 3.02 - 2.88 (m, 2H), 2.61 (d, J = 10.2 Hz, 2H), 1.59 (s, 2H), 1.26 (d, J = 13.7 Hz, 2H).
Example 1-071: Synthesis of 2-(5-(2-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1002)
Step 1: 2-(5-(2-((4-(tert-Butoxycarbonyl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using tert-butyl 4-((2-bromophenyl)sulfonyl)piperazine-l-carboxylate (500 mg, 1.235 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetate (1.1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
LCMS (ESI+) m/z 501.0 [M+H]+ Step 2: 2-(5-(2-(Piperazin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using 2-(5-(2-((4-(tert-butoxycarbonyl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
LCMS (ESI+) m/z 401.2 [M+H]+
Step 3: To a stirred solution of 2-(5-(2-(piperazin-l-ylsulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid trifluoroacetate (200 mg, 0.39 mmol, 1 equiv) in DMSO (2 mL) were added 2-(2,6-dioxopiperidin-3- yl)-5,6-difluoroisoindoline-l, 3-dione (1.1 equiv) and DIPEA (2 equiv) at RT. The reaction mixture was stirred at 110°C for 6 h. After completion 2-(5-(2-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3- dioxoisoindolin-5-yl)piperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (30 mg, 3.6% yield over three steps) was purified by preparative HPLC.
LCMS (ESI-) m/z 673.0 [M-H]’
NMR (400 MHz, DMSO-d6) δ 13.31 - 11.63 (s, 1H), 11.30 - 10.81 (s, 1H), 8.63 (d, J = 7.1 Hz, 1H), 8.03 (d, J = 7.9 Hz, 1H), 7.94 (s, 1H), 7.84 - 7.67 (m, 3H), 7.64 (s, 1H), 7.51 (d, J = 7.4 Hz, 1H), 7.39 (d, J = 7.2 Hz, 1H), 6.88 (d, J = 7.2 Hz, 1H), 5.10 (dd, J = 5.4, 12.8 Hz, 1H), 3.70 (s, 2H), 3.21 - 2.96 (m, 8H), 2.95 - 2.81 (m, 1H), 2.74 - 2.54 (m, 2H), 2.10 - 1.99 (m, 1H).
Example 1-072: Synthesis of 2-(5-(2-((4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1003)
Step 1: tert-Butyl 4-(l-((2-bromophenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (68% yield), using 2-bromobenzenesulfonyl chloride (500 mg, 1.961 mmol, 1 equiv) and tert-butyl 4-(piperidin-4- yl)piperazine-l-carboxylate [commercial] (1.1 equiv) as starting materials.
NMR (400 MHz, DMSO-d6) δ 8.05 - 7.95 (m, 1H), 7.88 (d, J = 7.2 Hz, 1H), 7.59 (d, J = 9.0 Hz, 2H), 3.71 - 3.67 (m, 2H), 3.27-3.22 (m, 4H) 2.79 - 2.61 (m, 2H), 2.41 - 2.30 (m, 6H), 1.79 - 1.69 (m, 2H), 1.38 (s, 9H).
Step 2: tert-Butyl 4-(l-((2-(3-(2-methoxy-2-oxoethyl)pyrazolo[l,5-o]pyridin-5- yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using tert-butyl 4-(l-((2- bromophenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate (150 mg, 0.308 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (4 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the solvent was evaporated and the resulting crude was forwarded into the next step.
LCMS (ESI+) m/z 598.4 [M+H]+
Step 3: 2-(5-(2-((4-(4-(tert-Butoxycarbonyl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above, using tert-butyl 4-(l-((2-(3-(2-methoxy-2-oxoethyl)pyrazolo[l,5-o]pyridin-5- yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate as starting material.
LCMS (ESI+) m/z 584.2 [M+H]+
Step 4: 2-(5-(2-((4-(Piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using 2-(5-(2-((4-(4-(tert-butoxycarbonyl)piperazin-l-yl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
LCMS (ESI+) m/z 484.6 [M+H]+
Step 5: To a stirred solution of 2-(5-(2-((4-(piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid trifluoroacetate (130 mg, 0.218 mmol, 1.11 equiv) in DMSO (3 mL) were added 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-l, 3-dione (1 equiv) and DIPEA (3 equiv) at RT. The reaction mixture was stirred at 110°C for 2 h. After completion 2-(5-(2-((4-(4-(2-(2,6-dioxopiperidin-3-yl)- 6-fluoro-l,3-dioxoisoindolin-5-yl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetic acid (16 mg, 6.85% yield over four steps) was purified by preparative HPLC.
LCMS (ESI+) m/z 758.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 11.10 (s, 1H), 8.64 (d, J = 7.2 Hz, 1H), 8.03 (d, J = 7.9 Hz, 1H), 7.96 (s, 1H), 7.82 - 7.64 (m, 3H), 7.60 (s, 1H), 7.50 (d, J = 7.2 Hz, 1H), 7.42 (d, J = 7.3 Hz, 1H), 6.89 (dd, J = 1.5, 7.1 Hz, 1H), 5.10 (dd, J = 5.3, 12.8 Hz, 1H), 3.72 (s, 2H), 3.22 - 3.09 (m, 4H), 3.15 - 2.74 (m, 4H), 2.63 - 2.51 (m, 1H), 2.47 - 2.39 (m, 6H), 2.35 - 2.29 (m, 1H), 2.10 - 1.95 (m, 1H), 1.66 - 1.57 (m, 2H), 1.18 - 0.93 (m, 2H). Example 1-073: Synthesis of (2-(5-(2-((4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5- yl)piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate
(Compound 1001)
Step 1: To a stirred solution of 2-(5-(2-((4-(4-(tert-butoxycarbonyl)piperazin-l-yl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (150 mg, 0.257 mmol, 1 equiv) in DMF (4 mL), cooled in an ice-water bath, were added chloromethyl pivalate (1.5 equiv) and KHCO3 (2 equiv). The reaction mixture was stirred at RT for 2 h. After completion the solution was diluted with cold water and extracted with ethyl acetate. The organic fraction was dried over NajSCU and evaporated to give crude tert-butyl 4-(l-((2-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-o]pyridin-5- yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l-carboxylate which was used in the next step.
LCMS (ESI+) m/z 698.3 [M+H]+
Step 2: (2-(5-(2-((4-(Piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-(l-((2-(3-(2-oxo-2- ((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-o]pyridin-5-yl)phenyl)sulfonyl)piperidin-4-yl)piperazine-l- carboxylate as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
LCMS (ESI+) m/z 598.4 [M+H]+
Step 3: To a stirred solution of (2-(5-(2-((4-(piperazin-l-yl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate trifluoroacetate (130 mg, 0.183 mmol, 1.11 equiv) in DMSO (3 mL) were added 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-l, 3-dione (1 equiv) and DIPEA (3 equiv) at RT. The reaction mixture was stirred at 110°C for 2 h. After completion of the reaction (2-(5-(2-((4-(4-(2- (2,6-dioxopiperidin-3-yl)-6-fluoro-l,3-dioxoisoindolin-5-yl)piperazin-l-yl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (12 mg, 5.4% yield over three steps) was purified by preparative HPLC.
LCMS (ESI+) m/z 872.2 [M+H]+
'H NMR (400 MHz, DMSO-d6) δ 11.33 - 10.97 (m, 1H), 8.66 (d, J = 7.0 Hz, 1H), 8.03 (d, J = 7.8 Hz, 1H), 7.96 (s, 1H), 7.77 (t, J = 7.4 Hz, 1H), 7.73 - 7.65 (m, 2H), 7.60 (s, 1H), 7.50 (d, J = 7.2 Hz, 1H), 7.41 (d, J = 7.0 Hz, 1H), 6.92 (d, J = 7.2 Hz, 1H), 5.71 (s, 2H), 5.14 - 5.06 (m, 1H), 3.91 (s, 2H), 3.30 - 3.28 (m, 5H), 3.19 - 3.14 (m, 4H), 3.00 - 2.78 (m, 1H), 2.78 - 2.59 (m, 1H), 2.48 - 2.37 (m, 4H), 2.37 - 2.24 (m, 1H), 2.12 - 1.97 (m, 2H), 1.98 - 1.88 (m, 1H), 1.71 - 1.56 (m, 2H), 1.21 - 0.96 (s, 9H).
Example 1-074: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)propanoic acid (Compound 49, S (left) | | | Compound 48, R (right)) Step 1: To a stirred solution of 2-(5-bromopyrazolo[l,5-a]pyridin-3-yl)acetonitrile (500 mg, 2.1 mmol, 1 equiv) in THF (5 mL), cooled to -78°C, was added NaHMDS (3.2 mL, 3.2 mmol, 1.5 equiv, IM solution in THF) and the reaction was stirred for 40 min at the same temperature, lodomethane (1.5 equiv) was added and the reaction mixture was stirred for 1 h at RT, then quenched with water and extracted with ethyl acetate. 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)propanenitrile (150 mg, 0.6 mmol, 28% yield) was purified by flash column chromatography.
TH NMR (400 MHz, DMSO) δ 8.69 (dd, J = 13.7, 7.2 Hz, 1H), 8.12 (d, J = 19.5 Hz, 2H), 7.10 (dd, J = 15.4, 7.7 Hz, 1H), 1.79 (s, 3H), 1.63 (d, J = 7.2 Hz, 1H).
Step 2: 2-(5-Bromopyrazolo[l,5-o]pyridin-3-yl)propanoic acid was synthesized using the general procedure shown in Reaction Scheme 2 and Example Method 2, above (86% yield), using 2-(5- bromopyrazolo[l,5-o]pyridin-3-yl)propanenitrile (150 mg, 0.6 mmol) as a starting material.
LCMS (ESI+) m/z 267.0 [M+H]+
Step 3: 2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)propanoic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)propanoic acid (150 mg, 0.56 mmol, 1 equiv) and 4-methoxy- l-((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)sulfonyl)piperidine (1 equiv) as starting materials, Cs2CO3 (4 equiv) as base and Pd(dtbpf)CI2 as catalyst (0.1 equiv). The product (mixture of stereoisomers) was purified by preparative HPLC.
Separation of the stereoisomers by preparative HPLC: Chiral separation was done on Agilent 1200 series instrument. Column name : CHIRALPAK IG (250 x 21 mm) 5p. Operating at ambient temperature and flow rate 21.0 mL/min. Mobile phase: mixture of 60% hexane, 20% DCM and 20% ethanol.
Absolute configuration of the stereoisomers was assigned based on crystal structure.
(S)-2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)propanoic acid
(Compound 49) (7 mg, 2.8% yield) LCMS (ESI+) m/z 444.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 12.83 - 11.77 (s, 1H), 8.62 (d, J = 7.1 Hz, 1H), 8.03 - 7.94 (m, 2H), 7.76 (t, J = 7.4 Hz, 1H), 7.68 (t, J = 7.6 Hz, 1H), 7.60 (s, 1H), 7.48 (d, J = 7.4 Hz, 1H), 6.84 (d, J = 7.0 Hz, 1H), 3.93 (q, J = 7.2 Hz, 1H), 3.23 - 3.14 (m, 1H), 3.13 (s, 3H), 3.04 - 2.94 (m, 2H), 2.73 - 2.62 (m, 2H), 1.65 - 1.53 (m, 2H), 1.46 (d, J = 7.0 Hz, 3H), 1.38 - 1.17 (m, 2H).
(R)-2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)propanoic acid (Compound 48) (6 mg, 2.4% yield)
LCMS (ESI+) m/z 444.1 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 12.82 - 11.81 (s, 1H), 8.62 (d, J = 7.1 Hz, 1H), 8.03 - 7.94 (m, 2H), 7.76 (t, J = 7.4 Hz, 1H), 7.68 (t, J = 7.4 Hz, 1H), 7.60 (s, 1H), 7.48 (d, J = 7.4 Hz, 1H), 6.84 (d, J = 7.0 Hz, 1H), 3.93 (q, J = 7.2 Hz, 1H), 3.19 (dt, J = 4.2, 8.0 Hz, 1H), 3.14 (s, 3H), 3.04 - 2.94 (m, 2H), 2.74 - 2.62 (m, 2H), 1.68 - 1.52 (m, 2H), 1.46 (d, J = 7.1 Hz, 3H), 1.38 - 1.17 (m, 2H).
Example 1-075: Synthesis of 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin- l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1007)
Step 1: To a stirred suspension of sodium hydride (173 mg, 60% in mineral oil, 7.2 mmol, 1.5 equiv) in THF (3 mL) and DCM (3 mL), cooled to 0°C, was added l-phenyl-2,5,8,ll,14,17,20-heptaoxadocosan-22-ol [commercial] (2 g, 4.802 mmol, 1 equiv) and the resulting mixture was stirred at RT for 30 min. 1-Chloro- 6-iodohexane (2.36 g, 9.604 mmol, 2 equiv) was added at 0°C and the reaction mixture was stirred at RT for 16 h. After completion, the mixture was quenched with cold water, extracted with ethyl acetate, the organic fraction was dried over Na2SO4 and evaporated. 29-Chloro-l-phenyl-2,5,8,ll,14,17,20,23- octaoxanonacosane (1.1 g, 42% yield) was purified by flash column chromatography.
Step 2: To a solution of 29-chloro-l-phenyl-2,5,8,ll,14,17,20,23-octaoxanonacosane (2 g, 3.73 mmol) in methanol (5 mL) was added Pd/C (280 mg, 10% wt.) and the reaction mixture was stirred at RT under hydrogen atmosphere for 16 h. After completion the solution was filtered through Celite® and evaporated to yield 27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosan-l-ol (1.58 g, 94% yield). Step 3: To a solution of 27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosan-l-ol (180 mg, 0.405 mmol, 1 equiv) in DCM (2 mL), cooled in an ice-water bath, were added TEA (0.17 mL, 1.216 mmol, 3 equiv), DMAP (catalytic) and 4-methylbenzene-l-sulfonyl chloride (115 mg, 0.608 mmol, 1.5 equiv) and the resulting solution was stirred at RT for 3 h. After completion the reaction was diluted with water and extracted with DCM. The organic fraction was dried over Na2SO4 and evaporated to give crude product. 27-Chloro- 3,6,9,12,15,18,21-heptaoxaheptacosyl 4-methylbenzenesulfonate (220 mg, 90% yield) was purified by flash column chromatography.
!H NMR (400 MHz, Chloroform-d) 6 7.85 - 7.75 (m, 2H), 7.34 (d, J = 8.0 Hz, 2H), 4.20 - 4.11 (m, 2H), 3.71 - 3.54 (m, 26H), 3.53 (t, J = 6.7 Hz, 2H), 3.45 (t, J = 6.6 Hz, 2H), 2.45 (s, 3H), 1.77 (p, J = 6.9 Hz, 2H), 1.58 (q, J = 7.1 Hz, 2H), 1.49 - 1.35 (m, 4H).
Step 4: To a suspension of sodium hydride (10 mg, 60% in mineral oil, 0.417 mmol, 2.5 equiv) in DMF (2 mL), cooled in an ice-water bath, was added tert-butyl 4-hydroxypiperidine-l-carboxylate [commercial] (100 mg, 0.167 mmol, 1 equiv) and the mixture was stirred at 0°C for 30 min. 27-Chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl 4-methylbenzenesulfonate (100 mg, 0.167 mmol, 1 equiv) was added and the reaction mixture was stirred at RT for 16 h. After completion, cold water was added and the solution was extracted with ethyl acetate, the organic fraction was dried over Na2SO4 and evaporated to yield tert-butyl 4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine-l-carboxylate (52 mg, 49% yield).
Step 5: 4-((27-Chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (97% yield), using tert-butyl 4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine-l-carboxylate (1 g, 1.59 mmol). After completion the volatiles were removed under reduced pressure and the product was purified by multiple azeotropic evaporation with toluene.
LCMS (ESI+) m/z 528.3, 530.2 [M+H]+
Step 6: l-((2-Bromophenyl)sulfonyl)-4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (56% yield), using 4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine trifluoroacetate
(70 mg, 0.109 mmol, 1 equiv) and 2-bromobenzenesulfonyl chloride (1.2 equiv) as starting materials.
1H NMR (400 MHz, Chloroform-d) 6 8.09 (dd, J = 7.8, 1.8 Hz, 1H), 7.74 (dd, J = 7.8, 1.4 Hz, 1H), 7.44 (td, J = 7.6, 1.4 Hz, 1H), 7.38 (td, J = 7.6, 1.8 Hz, 1H), 3.69 - 3.54 (m, 24H), 3.51 - 3.41 (m, 11H), 3.21-3.1 (m, 2H), 1.92-1.83 (m, 2H), 1.81-1.73 (m, 2H), 1.61 - 1.53 (m, 4H), 1.49 - 1.31 (m, 4H).
Step 7: Methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-((27- chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidine (200 mg, 0.268 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, CS2CO3 (3 equiv) as base and Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction was diluted with cold water, extracted with ethyl acetate, the organic layer was dried over Na2SO4 and evaporated. The crude product was directly used into the next step.
LCMS (ESI+) m/z 856.0 [M+H]+
Step 8: 2-(5-(2-((4-((27-Chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (51 mg, 22% yield over two steps), using methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (250 mg, 0.292 mmol) as starting material.
LCMS (ESI+) m/z 842.3 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 12.08 - 11.10 (s, 1H), 8.55 (d, J = 6.8 Hz, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.92 (s, 1H), 7.73 (t, J = 7.6 Hz, 1H), 7.65 (t, J = 7.6 Hz, 1H), 7.55 (s, 1H), 7.46 (d, J = 7.2 Hz, 1H), 6.83 (d, J = 7.2 Hz, 1H), 3.70 (s, 2H), 3.60 (t, J = 6.4 Hz, 2H), 3.56 - 3.44 (m, 28H), 3.44 - 3.34 (m, 3H), 3.17 - 3.05 (m, 2H), 2.82 - 2.70 (m, 2H), 1.81 - 1.69 (m, 2H), 1.69 - 1.58 (m, 2H), 1.58 - 1.29 (m, 8H). Example 1-076: Synthesis of methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (Compound 1006)
Step 1: To a stirred solution of 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21- heptaoxaheptacosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (35 mg, 0.042 mmol, 1 equiv) in methanol (1 mL) and ethyl acetate (1 mL) was added trimethylsilyldiazomethane (2M solution in hexanes, 5 equiv) at -10°C and stirred at RT for 1 h. After completion the reaction was quenched with cold water, extracted with ethyl acetate, the organic layer was dried over NajSCU and evaporated. Methyl 2-(5-(2-((4-((27-chloro-3,6,9,12,15,18,21-heptaoxaheptacosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (14 mg, 39% yield) was purified by preparative TLC.
LCMS (ESI+) m/z 856.6 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.57 (d, J = 6.8 Hz, 1H), 8.01 (d, J = 8.0 Hz, 1H), 7.93 (s, 1H), 7.73 (t, J = 7.6 Hz, 1H), 7.67 (t, J = 7.6 Hz, 1H), 7.56 (s, 1H), 7.47 (d, J = 7.2 Hz, 1H), 6.84 (d, J = 7.6 Hz, 1H), 3.80 (s, 2H), 3.66 - 3.54 (m, 4H), 3.56 - 3.44 (m, 22H), 3.44 - 3.31 (m, 3H), 3.14 - 3.01 (m, 6H), 2.79 - 2.68 (m, 2H), 1.80 - 1.69 (m, 2H), 1.68 - 1.59 (m, 3H), 1.57 - 1.45 (m, 2H), 1.47 - 1.22 (m, 8H).
Example 1-077: Synthesis of methyl 2-(5-(2-((4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin- l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (Compound 1004) and 2-(5-(2-((4-((21-chloro- 3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 1005)
Step 1: To a solution of 21-chloro-3,6,9,12,15-pentaoxahenicosan-l-ol [commercial] (700 mg, 1.96 mmol, 1 equiv) in DCM (12 mL), cooled in an ice-water bath, were added TEA (794 mg, 7.86 mmol, 4 equiv), DMAP (catalytic) and 4-methylbenzene-l-sulfonyl chloride (1.3 g, 6.69 mmol, 3.4 equiv) and the reaction mixture was stirred at RT for 3 h. After completion the solution was diluted with water and extracted with ethyl acetate. The organic fraction was dried over Na2SO4 and evaporated to give crude product. 21- Chloro-3,6,9,12,15-pentaoxahenicosyl 4-methylbenzenesulfonate (370 mg, 37% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 511.2, 513.1 [M+H]+ Step 2: To a solution of tert-butyl 4-hydroxypiperidine-l-carboxylate (124 mg, 0.62 mmol, 1 equiv) in DMF (4 mL), cooled in an ice-water bath, were added sodium hydride (25 mg, 60% suspension in mineral oil, 1.02 mmol, 1.65 equiv) and the mixture was stirred at 0°C for 30 min. 21-Chloro-3,6,9,12,15- pentaoxahenicosyl 4-methylbenzenesulfonate (350 mg, 0.67 mmol, 1.08 equiv) was added at 0°C and the reaction mixture was stirred at RT for 3 h. The solution was diluted with cold water and extracted with ethyl acetate. The organic fraction was evaporated to dryness and the crude tert-butyl 4-((21-chloro- 3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine-l-carboxylate (300 mg) was forwarded into the next step.
LCMS (ESI+) m/z 540.1 [M+H]+
Step 3: 4-((21-Chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine-l-carboxylate (300 mg, 0.55 mmol). After completion the volatiles were removed under reduced pressure and the residue was triturated with pentane to provide crude product which was used directly in the next step.
LCMS (ESI+) m/z 440.0 [M+H]+
Step 4: l-((2-Bromophenyl)sulfonyl)-4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (38% yield over three steps), using 4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine trifluoroacetate (300 mg, 0.54 mmol, 1 equiv) and 2-bromobenzenesulfonyl chloride (1.18 equiv) as starting materials.
LCMS (ESI+) m/z 658.4, 660.2 [M+H]+
Step 5: Methyl 2-(5-(2-((4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate and 2-(5-(2-((4-((21-chloro-3,6,9,12,15- pentaoxahenicosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid were synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromophenyl)sulfonyl)-4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidine (120 mg, 0.18 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin- 3-yl)acetate (1.2 equiv) as starting materials, K3PO4 (2 equiv) as base and Pd(dppf)CL as catalyst (0.1 equiv). After completion the products were separated using preparative HPLC.
Methyl 2-(5-(2-((4-((21-chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate (Compound 1004) (24 mg, 17% yield)
LCMS (ESI+) m/z 768.4, 770.2 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.62 (d, J = 7.1 Hz, 1H), 7.99 (d, J = 7.8 Hz, 1H), 7.96 (s, 1H), 7.76 (t, J = 7.4 Hz, 1H), 7.68 (t, J = 7.7 Hz, 1H), 7.58 (s, 1H), 7.47 (d, J = 7.4 Hz, 1H), 6.84 (d, J = 7.1 Hz, 1H), 3.82 (s, 2H), 3.66 - 3.57 (m, 4H), 3.55 - 3.40 (m, 20H), 3.40 - 3.27 (m, 3H), 3.06 - 2.96 (m, 2H), 2.72 - 2.62 (m, 2H), 1.77 - 1.57 (m, 4H), 1.53 - 1.43 (m, 2H), 1.43 - 1.20 (m, 7H).
2-(5-(2-((4-((21-Chloro-3,6,9,12,15-pentaoxahenicosyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 1005) (40 mg, 29% yield)
LCMS (ESI+) m/z 754.4, 756.2 [M+H]+
XH NMR (400 MHz, DMSO-d6) δ 13.02 - 11.66 (s, 1H), 8.61 (d, J = 7.1 Hz, 1H), 7.99 (d, J = 7.8 Hz, 1H), 7.94 (s, 1H), 7.75 (t, J = 7.5 Hz, 1H), 7.70 - 7.66 (m, 1H), 7.56 (s, 1H), 7.47 (d, J = 7.4 Hz, 1H), 6.83 (dd, J = 1.8, 7.2 Hz, 1H), 3.71 (s, 2H), 3.61 (t, J = 6.6 Hz, 2H), 3.54 - 3.41 (m, 20H), 3.40 - 3.31 (m, 3H), 3.09 - 2.98 (m, 2H), 2.71 - 2.60 (m, 2H), 1.75 - 1.55 (m, 4H), 1.53 - 1.41 (m, 2H), 1.43 - 1.21 (m, 6H).
Example 1-078: Synthesis of 2-(5-(2-cyanophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 88)
Step 1: 2-(5-(2-Cyanophenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (30% yield), using methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (15 mg, 0.047 mmol, 1 equiv) and 2-bromobenzonitrile [commercial] (1 equiv) as starting materials, PdfPPhah (0.1 equiv) as catalyst and sodium carbonate (2 equiv) as base. Once the reaction was completed, IM LiOH aqueous solution (0.95 mL, 0.95 mmol, 20 equiv) was added and the reaction mixture was stirred at RT for 2 h. The solution was neutralized by addition of IM HCI and the product was purified by preparative HPLC.
LCMS (ESI+) m/z 278.0 [M+H]+
NMR (500 MHz, DMSO-d6) δ 8.77 (dd, J = 7.2, 1.0 Hz, 1H), 7.99 (d, J = 6.9 Hz, 2H), 7.92 - 7.87 (m, 1H), 7.84 (td, J = 7.7, 1.4 Hz, 1H), 7.73 (dd, J = 7.9, 1.2 Hz, 1H), 7.64 (td, J = 7.7, 1.3 Hz, 1H), 7.08 (dd, J = 7.2, 2.1 Hz, 1H), 3.76 (s, 2H).
Example 1-079: Synthesis of (2-(5-(4-hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 87)
Step 1: l-((2-Bromo-5-methoxyphenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (59% yield), using 2-bromo-5- methoxybenzenesulfonyl chloride [commercial] (8 g, 28.07 mmol, 1 equiv) and 4-methoxypiperidine (1 equiv) as starting materials.
LCMS (ESI+) m/z 364.0, 366.0 [M+H]+
Step 2: To a stirred solution of l-((2-bromo-5-methoxyphenyl)sulfonyl)-4-methoxypiperidine (1.5 g, 4.12 mmol, 1 equiv) in DMF (15 mL) was added LiCI (10 equiv) and the reaction mixture was stirred at 150°C for 30 h. The reaction was cooled, quenched with ice-water and extracted with ethyl acetate. The organic fractions were combined, washed with brine, dried over NajSCU and concentrated under reduced pressure. 4-Bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)phenol (700 mg, 2 mmol, 48% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 350.1 [M+H]+
Step 3: (TLS-561, lnt-8, step 5A) To a stirred solution of 2-(5-bromopyrazolo[l,5-o]pyridin-3-yl)acetic acid (6.5 g, 25.59 mmol, 1 equiv) in DMF (5 mL) were added KHCO3 (2 equiv) and chloromethyl pivalate (1.5 equiv) and the resulting solution was stirred at RT for 16 h. After completion, the reaction mixture was quenched with ice-water and extracted with ethyl acetate to provide (2-(5-bromopyrazolo[l,5-o]pyridin- 3-yl)acetoxy)methyl pivalate (7.5 g, 20.3 mmol, 79% yield).
LCMS (ESI+) m/z 368.8, 371.0 [M+H]+
Step 4: (TLS-561, lnt-8, step 6A) (2-(5-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 9 and Example Method 9, above (86% yield), using (2-(5-bromopyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate (8.2 g, 22.22 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 417.2 [M+H]+
Step 5: (2-(5-(4-Hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (15% yield), using 4-bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)phenol (400 mg, 1.14 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate (1.2 equiv) as starting materials, Pd(dtbpf)CI2 (0.1 equiv) as catalyst and Cs2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 560.1 [M+H]+
NMR (400 MHz, DMSO-d6) δ 10.33 (s, 1H), 8.58 (d, J = 7.2 Hz, 1H), 7.92 (s, 1H), 7.50 (s, 1H), 7.36 (s, 1H), 7.27 (d, J = 8.3 Hz, 1H), 7.10 (d, J = 8.1 Hz, 1H), 6.81 (d, J = 7.2 Hz, 1H), 5.70 (s, 2H), 3.88 (s, 2H), 3.15 (m, 4H), 3.02 - 2.94 (m, 2H), 2.73 - 2.63 (m, 2H), 1.60 (s, 2H), 1.33 - 1.26 (m, 2H), 1.05 (s, 9H). Example 1-080: Synthesis of 2-(5-(4-hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 86)
Step 1: 2-(5-(4-Hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (83% yield), using (2-(5-(44iydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (17.5 mg, 0.031 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 446.2 [M+H]+
NMR (500 MHz, DMSO-d6) δ 10.39 (s, 1H), 8.56 (d, J = 7.1 Hz, 1H), 7.91 (s, 1H), 7.50 (d, J = 1.9 Hz, 1H), 7.36 (d, J = 2.6 Hz, 1H), 7.26 (d, J = 8.2 Hz, 1H), 7.10 (dd, J = 8.4, 2.6 Hz, 1H), 6.78 (dd, J = 7.2, 1.9 Hz, 1H), 3.69 (s, 2H), 3.19 (tt, J = 7.5, 3.5 Hz, 1H), 3.15 (s, 3H), 2.99 (ddd, J = 11.6, 7.1, 3.8 Hz, 2H), 2.67 (ddd, J = 12.2, 8.3, 3.5 Hz, 2H), 1.60 (ddt, J = 13.8, 7.2, 3.6 Hz, 2H), 1.29 (dtd, J = 12.1, 8.0, 3.7 Hz, 2H).
Example 1-081: Synthesis of 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-
((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-5-yl)benzoic acid (Compound 85)
Step 1: Methyl 4-bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)benzoate was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (93% yield), using methyl 4-bromo- 3-(chlorosulfonyl)benzoate [commercial] (2.5 g, 7.97 mmol, 1 equiv) and 4-methoxypiperidine (1.2 equiv) as starting materials.
LCMS (ESI+) m/z 392.0, 394.1 [M+H]+ Step 2: 4-Bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)benzoic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (98% yield), using methyl 4-bromo- 3-((4-methoxypiperidin-l-yl)sulfonyl)benzoate (2 g, 1.51 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 376.0, 378.0 [M+H]+
Step 3: 3-((4-Methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5- o]pyridin-5-yl)benzoic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (35% yield), using 4-bromo-3-((4-methoxypiperidin-l-yl)sulfonyl)benzoic acid (550 mg, 1.45 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (1 equiv) as starting materials, Pd(dtbpf)Ck (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 588.3 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.68 (d, J = 7.2 Hz, 1H), 8.48 (d, J = 1.8 Hz, 1H), 8.25 (dd, J = 7.9, 1.7 Hz, 1H), 7.97 (s, 1H), 7.63 (d, J = 8.4 Hz, 2H), 6.89 (dd, J = 7.2, 1.9 Hz, 1H), 5.70 (s, 2H), 3.91 (s, 2H), 3.25 - 3.15 (m, 1H), 3.14 (s, 3H), 3.04 - 2.93 (m, 2H), 2.73 - 2.62 (m, 2H), 1.60 (m, J = 8.8 Hz, 2H), 1.34 - 1.20 (m, 2H), 1.04 (s, 9H).
Example 1-082: Synthesis of 4-(3-(carboxymethyl)pyrazolo[l,5-a]pyridin-5-yl)-3-((4-methoxypiperidin-l- yl)sulfonyl)benzoic acid (Compound 84)
Step 1: 4-(3-(Carboxymethyl)pyrazolo[l,5-a]pyridin-5-yl)-3-((4-methoxypiperidin-l-yl)sulfonyl)benzoic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (83% yield), using 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2- ((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-o]pyridin-5-yl)benzoic acid (10 mg, 0.017 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 474.1 [M+H]+
NMR (500 MHz, DMSO-d6) δ 12.35 (s, 1H), 8.61 (d, J = 7.2 Hz, 1H), 8.46 (d, J = 1.6 Hz, 1H), 8.16 (dd, J = 7.9, 1.3 Hz, 1H), 7.94 (s, 1H), 7.60 (d, J = 1.9 Hz, 1H), 7.46 (d, J = 7.8 Hz, 1H), 6.84 (dd, J = 7.2, 1.9 Hz, 1H), 6.73 (s, 1H), 3.72 (s, 2H), 3.20 - 3.18 (m, 1H), 3.14 (s, 3H), 2.99 (ddd, J = 11.6, 7.1, 3.8 Hz, 2H), 2.67 (ddd, J = 12.3, 8.4, 3.7 Hz, 2H), 1.60 (ddt, J = 13.6, 7.1, 3.6 Hz, 2H), 1.29 (dp, J = 12.8, 4.0 Hz, 2H).
Example 1-083: Synthesis of (2-(5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 83)
Step 1: l-((2-Bromo-5-nitrophenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (78% yield), using 2-bromo-5- nitrobenzenesulfonyl chloride [commercial] (3.5 g, 11.66 mmol, 1 equiv) and 4-methoxypiperidine (1.5 equiv) as starting materials.
LCMS (ESI+) m/z 379.0, 380.8 [M+H]+
Step 2: (2-(5-(2-((4-Methoxypiperidin-l-yl)sulfonyl)-4-nitrophenyl)pyrazolo[l,5-o]pyridin-3- yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (50% yield), using l-((2-bromo-5-nitrophenyl)sulfonyl)-4-methoxypiperidine (400 mg, 1.06 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (1.82 equiv) as starting materials, Pd(dtbpf)Ck (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 589.3 [M+H] Step 3: To a stirred solution of (2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)-4-nitrophenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (250 mg, 0.42 mmol, 1 equiv) in THF/water system (5/2, v/v, 1.4 mL) were added zinc dust (5 equiv) and NH4CI (5 equiv) and the reaction mixture was stirred at 70°C for 2 h. After completion, the solid particles were filtered off and the filtrate was concentrated under reduced pressure. (2-(5-(4-Amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (80 mg, 0.14 mmol, 34% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 559.2 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.54 (d, J = 7.3 Hz, 1H), 7.90 (s, 1H), 7.45 (s, 1H), 7.18 (d, J = 2.5 Hz, 1H), 7.08 (d, J = 8.3 Hz, 1H), 6.87 - 6.76 (m, 2H), 5.70 (s, 2H), 3.87 (s, 2H), 3.21 (d, J = 26.2 Hz, 1H), 3.15 (s, 3H), 2.97 (m, 2H), 2.65 (d, J = 13.6 Hz, 2H), 1.60 (m, 2H), 1.29 (m, 2H), 1.05 (s, 9H).
Example 1-084: Synthesis of 2-(5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 82)
Step 1: 2-(5-(4-Amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (75% yield), using (2-(5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (12 mg, 0.021 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 445.1 [M+H]+
TH NMR (500 MHz, DMSO-d6) δ 12.32 (s, 1H), 8.54 (d, J = 7.1 Hz, 1H), 7.91 (s, 1H), 7.49 - 7.45 (m, 1H), 7.20 (d, J = 2.3 Hz, 1H), 7.12 - 7.06 (m, 1H), 6.85 (dd, J = 8.3, 2.4 Hz, 1H), 6.78 (dd, J = 7.2, 1.9 Hz, 1H), 5.79 (s, 2H), 3.69 (s, 2H), 3.21 - 3.18 (m, 1H), 3.17 (s, 3H), 3.00 (ddd, J = 11.6, 7.4, 3.9 Hz, 2H), 2.68 (ddd, J = 12.3, 8.3, 3.5 Hz, 2H), 1.60 (ddt, J = 13.7, 7.1, 3.5 Hz, 2H), 1.29 (dtd, J = 12.0, 8.0, 3.8 Hz, 2H). Example 1-085: Synthesis of 2-(5-(2-methoxy-6-((4-methoxy-4-(methoxymethyl)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 77)
Step 1: To a suspension of NaH (30 mg, 0.81 mmol, 2 equiv, 60% in mineral oil) in DMF (2 mL), cooled in an ice-water bath, was added tert-butyl 4-hydroxy-4-(methoxymethyl)piperidine-l-carboxylate (100 mg, 0.407 mmol, 1 equiv) and stirred for 30 min. lodomethane (1.2 equiv) was added and the reaction mixture was stirred at RT for 1 h. After completion, the reaction was quenched with cold water and extracted with ethyl acetate. The organic phase was dried over NajSCU and evaporated. tert-Butyl 4-methoxy-4- (methoxymethyl)piperidine-l-carboxylate (80 mg, 0.308 mmol, 76% yield) was purified by flash column chromatography.
TH NMR (400 MHz, Chloroform-d) 6 3.78 (d, J = 12.9 Hz, 2H), 3.37 (s, 3H), 3.31 (s, 2H), 3.25 (s, 3H), 3.14 - 3.03 (m, 2H), 1.76 (dd, J = 12.3, 3.4 Hz, 2H), 1.45 (m, 11H).
Step 2: 4-Methoxy-4-(methoxymethyl)piperidine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (84% yield), using tert-butyl 4- methoxy-4-(methoxymethyl)piperidine-l-carboxylate (80 mg, 0.173 mmol) as starting material.
LCMS (ESI+) m/z 160.0 [M+H]+
Step 3: l-((2-Bromo-3-methoxyphenyl)sulfonyl)-4-methoxy-4-(rnethoxymethyl)piperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (41% yield), using 4-methoxy-4-(methoxymethyl)piperidine trifluoroacetate (80 mg, 0.293 mmol, 1 equiv) and 2-bromo-3-methoxybenzenesulfonyl chloride [commercial] (1.1 equiv) as starting materials.
LCMS (ESI+) m/z 408.0, 410.0 [M+H] Step 4: 2-(5-(2-Methoxy-6-((4-methoxy-4-(methoxymethyl)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (9% yield), using l-((2-bromo-3-methoxyphenyl)sulfonyl)-4-methoxy-4- (methoxymethyl)piperidine (50 mg, 0.123 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, Pd(dtbpf)CI2 (0.1 equiv) as catalyst and Cs2CO3 (4 equiv) as base.
LCMS (ESI+) m/z 504.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.54 (d, J = 7.2 Hz, 1H), 7.87 (s, 1H), 7.63 (t, J = 8.0 Hz, 1H), 7.56 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.40 (d, J = 1.9 Hz, 1H), 6.60 (dd, J = 7.2, 1.9 Hz, 1H), 3.72 (s, 3H), 3.62 - 3.50 (m, 2H), 3.22 (s, 3H), 3.19 (s, 2H), 3.07 - 2.94 (m, 2H), 3.03 (s, 3H), 2.72 - 2.54 (m, 2H), 1.61 (t, J = 12.4 Hz, 2H), 1.36 - 1.14 (m, 2H).
Example 1-086: Synthesis of 2-(5-(7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 73)
Step 1: To a stirred solution of 8-bromoquinolin-7-amine [commercial] (500 mg, 2.2 mmol, 1 equiv) in acetonitrile (5 mL) was added p-toluenesulfonic acid monohydrate (3.1 equiv) and the mixture was cooled in ice-water bath. The solutions of NaNO2 (2.05 equiv) in water (1.2 mL) and KI (2.55 equiv) in water (1.2 mL) were slowly added over the period of 5 min. The reaction mixture was stirred for 30 min at RT, then diluted with water (10 mL), neutralized with saturated NaHCOs solution and extracted with ethyl acetate. The combined organic fractions were washed with saturated NaHCOs solution, then 10% Na2S2C>3 and brine. After drying over Na2SO4 the solvent was evaporated and 8-bromo-7-iodoquinoline (550 mg, 1.65 mmol, 75% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 333.6, 335.7 [M+H]+
Step 2: To a stirred solution of 8-bromo-7-iodoquinoline (550 mg, 1.65 mmol, 1 equiv) in dioxane were added (4-methoxyphenyl)methanethiol (0.91 equiv) and DIPEA (3 equiv). The mixture was purged with argon for 5 min, followed by addition of XantPhos (0.2 equiv.) and Pd2(dba)3 (0.1 equiv). The reaction was stirred at 90°C for 16 h. After completion, the solid particles were filtered off and the filtrate was evaporated. 8-Bromo-7-((4-methoxybenzyl)thio)quinoline (490 mg, 1.36 mmol, 82% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 359.8, 361.8 [M+H]+
Step 3: To a stirred solution of 8-bromo-7-((4-methoxybenzyl)thio)quinoline (200 mg, 0.56 mmol, 1 equiv) in ACN (4 mL) were added AcOH (0.1 mL), water (0.2 mL) and the solution was cooled to 0°C. After 10 min l,3-dichloro-5,5-dimethylhydantoin (2 equiv) was added and the reaction was stirred at 0°C for 1 h. After completion, the reaction was quenched with water and extracted with DCM. The combined organic fractions were washed with brine, dried over Na2SO4 and evaporated to afford crude 8-bromoquinoline-
7-sulfonyl chloride, which was forwarded directly for the next step.
Step 4: 8-Bromo-7-((4-methoxypiperidin-l-yl)sulfonyl)quinoline was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (55% yield over two steps), using
8-bromoquinoline-7-sulfonyl chloride (170 mg, 0.56 mmol, 1 equiv) and 4-methoxypiperidine (1.5 equiv) as starting materials.
LCMS (ESI+) m/z 385.0, 387.0 [M+H]+
Step 5: 2-(5-(7-((4-Methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (20% yield), using 8-bromo-7-((4-methoxypiperidin-l-yl)sulfonyl)quinoline (100 mg, 0.26 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (1.2 equiv) as starting materials, Pd(dtbpf)CL (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 481.3 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.95-8.89 (m, 1H), 8.58 (dd, J = 7.0, 12.4 Hz, 2H), 8.29 (d, J = 8.7 Hz, 1H), 8.12 (d, J = 8.7 Hz, 1H), 7.92 (s, 1H), 7.74-7.66 (m, 1H), 7.49 (s, 1H), 6.75 (d, J = 6.9 Hz, 1H), 3.68-3.58 (m, 2H), 3.26-3.19 (m, 1H), 3.16 (s, 3H), 3.12-3.02 (m, 2H), 2.91-2.70 (m, 2H), 1.86-1.59 (m, 2H), 1.56-1.16 (m, 2H).
Example 1-087: Synthesis of (2-(5-(7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 74)
Step 1: To a stirred solution of 2-(5-(7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (10 mg, 0.021 mmol, 1 equiv) in dry ACN (0.54 mL) were added chloromethyl pivalate (1.5 equiv) and TEA (3 equiv) and the reaction was carried at 55°C for 24 h. (2-(5-(7-((4- Methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (7.0 mg, 0.012 mmol, 56% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 595.2 [M+H]+
XH NMR (500 MHz, DMSO-d6) δ 8.92 (dd, J = 4.1, 1.8 Hz, 1H), 8.62 (dd, J = 7.1, 1.0 Hz, 1H), 8.56 (dd, J = 8.4, 1.8 Hz, 1H), 8.29 (d, J = 8.8 Hz, 1H), 8.12 (d, J = 8.9 Hz, 1H), 7.94 (s, 1H), 7.70 (dd, J = 8.3, 4.1 Hz, 1H), 7.49 (dd, J = 1.9, 1.0 Hz, 1H), 6.79 (dd, J = 7.1, 1.9 Hz, 1H), 5.68 (s, 2H), 3.92 - 3.81 (m, 2H), 3.23 (dq, J = 7.4, 3.7 Hz, 1H), 3.16 (s, 3H), 3.12 (dd, J = 7.1, 4.1 Hz, 1H), 3.06 (ddd, J = 11.5, 6.7, 3.7 Hz, 1H), 2.82 (ddd, J = 12.1, 8.4, 3.5 Hz, 1H), 2.76 (ddt, J = 12.3, 8.5, 4.1 Hz, 1H), 1.72 (ddt, J = 9.9, 6.2, 3.4 Hz, 2H), 1.39 (tdd, J = 17.0, 8.3, 4.1 Hz, 2H), 1.00 (s, 9H). Example 1-088: Synthesis of 2-(5-(2-methoxy-6-((4-methoxyphenyl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 75)
Step 1: To a stirred solution of 2-bromo-l-iodo-3-methoxybenzene [commercial] (500 mg, 1.6 mmol, 1 equiv) in dioxane (30 mL) were added 4-methoxybenzenethiol (3 equiv) and DIPEA (4.9 equiv). The mixture was purged with argon for 10 min, followed by addition of XantPhos (0.2 equiv.) and Pd2(dba)3 (0.1 equiv). The reaction was stirred at 90°C for 16 h. After completion, the volatiles were evaporated and (2-bromo-3-methoxyphenyl)(4-methoxyphenyl)sulfane (200 mg, 0.615 mmol, 38% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 325.0, 326.8 [M+H]+
Step 2: To a stirred solution of (2-bromo-3-methoxyphenyl)(4-methoxyphenyl)sulfane (200 mg, 0.615 mmol, 1 equiv) in DCM (5 mL) was added m-CPBA (2 equiv) and the reaction was carried at RT for 5 h. After completion, the reaction was quenched with ice-cold water and extracted with DCM. The combined organic fractions were washed with saturated aqueous NaHCO3 solution, dried over Na2SO4 and evaporated to afford 2-bromo-l-methoxy-3-((4-methoxyphenyl)sulfonyl)benzene (110 mg, 0.308 mmol, 50% yield) as white solid.
LCMS (ESI+) m/z 356.9, 358.8 [M+H]+
Step 3: 2-(5-(2-Methoxy-6-((4-methoxyphenyl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (41% yield), using 2-bromo-l-methoxy-3-((4-methoxyphenyl)sulfonyl)benzene (100 mg, 0.281 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.1 equiv) as starting materials, Pd(dtbpf)CI2 (0.1 equiv) as catalyst and Cs2CO3 (4 equiv) as base.
LCMS (ESI+) m/z 453.1 [M+H] 'H NMR (400 MHz, DMSO-d6) δ 8.43 (d, J = 7.2 Hz, 1H), 7.94 - 7.84 (m, 2H), 7.70 (t, J = 8.1 Hz, 1H), 7.47 (d, J = 8.2 Hz, 1H), 7.23 (d, J = 8.9 Hz, 2H), 6.85 - 6.75 (m, 3H), 6.30 (dd, J = 1.6, 7.1 Hz, 1H), 3.76 (s, 3H), 3.69 (s, 3H), 3.58 - 3.54 (m, 1H), 3.42 - 3.38 (m, 1H).
Example 1-089: Synthesis of 2-(5-(2-methoxy-6-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetic acid (Compound 76)
Step 1: l-((2-Bromo-3-methoxyphenyl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above, using 2-bromo-3- methoxybenzenesulfonyl chloride (100 mg, 0.35 mmol, 1 equiv) and 4-methoxypiperidine (5 equiv) as starting materials. After completion, the reaction mixture was evaporated and the resulting crude was directly forwarded into the next step.
LCMS (ESI+) m/z 364.0, 365.8 [M+H]+
Step 2: Methyl 2-(5-(2-methoxy-6-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using l-((2-bromo-3-methoxyphenyl)sulfonyl)-4-methoxypiperidine (100 mg, 0.27 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1.12 equiv) as starting materials, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base. After completion, the volatiles were removed under reduced pressure and the resulting crude was forwarded directly into the next step.
LCMS (ESI+) m/z 473.8 [M+H]+
Step 3: 2-(5-(2-Methoxy-6-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (14% yield over three steps), using methyl 2-(5-(2-methoxy-6-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate (50 mg, 0.106 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 460.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.50 (d, J = 7.1 Hz, 1H), 7.82 (s, 1H), 7.61 (t, J = 8.0 Hz, 1H), 7.56 (d, J = 7.8 Hz, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.37 (s, 1H), 6.55 (d, J = 7.0 Hz, 1H), 3.72 (s, 3H), 3.39 - 3.36 (m, 2H), 3.16 (s, 3H), 3.07 - 2.86 (m, 2H), 2.77 - 2.59 (m, 2H), 1.81 - 1.59 (m, 2H), 1.48 - 1.09 (m, 2H).
Example 1-090: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetamide (Compound 80)
Step 1: To a stirred solution of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin- 3-yl)acetic acid (9.7 mg, 0.023 mmol, 1 equiv) in dry DMF (1 mL) were added HATU (1.5 equiv), DIPEA (5 equiv) and NH4CI (5 equiv). The reaction mixture was stirred at RT for 30 min and 2-(5-(2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetamide (6.5 mg, 0.015 mmol, 67% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 429.1 [M+H]+
XH NMR (500 MHz, DMSO-d6) δ 8.64 - 8.59 (m, 1H), 8.01 (dd, J = 8.0, 1.3 Hz, 1H), 7.93 (s, 1H), 7.78 (td, J = 7.5, 1.4 Hz, 1H), 7.70 (td, J = 7.7, 1.4 Hz, 1H), 7.62 - 7.61 (m, 1H), 7.49 (dd, J = 7.6, 1.4 Hz, 1H), 7.42 (br s, 1H, NH), 6.86 (br s, 1H, NH), 6.83 (dd, J = 7.2, 1.9 Hz, 1H), 3.52 (s, 2H), 3.23 (tt, J = 7.4, 3.5 Hz, 1H), 3.17 (s, 3H), 3.02 (ddd, J = 11.7, 7.1, 3.8 Hz, 2H), 2.71 (ddd, J = 12.1, 8.3, 3.5 Hz, 2H), 1.64 (ddt, J = 13.7, 7.1, 3.6 Hz, 2H), 1.33 (dtd, J = 12.0, 8.0, 3.7 Hz, 2H). Example 1-091: Synthesis of (S)-2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1008)
Step 1: l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidin-4-ol was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (57% yield), using 2-bromo-3- methoxybenzenesulfonyl chloride (425 mg, 1.5 mmol, 1 equiv) and piperidin-4-ol (1.5 equiv) as starting materials.
LCMS (ESI+) m/z 349.6, 351.9 [M+H]+
Step 2: To a solution of l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4-ol (300 mg, 0.86 mmol, 1 equiv) in DMF (3 mL), cooled in an ice-water bath, was added NaH (52 mg, 1.29 mmol, 1.5 equiv, 60% suspension in mineral oil) and the reaction mixture was stirred at RT for 30 min, then again cooled in an ice-water bath. 2,2-Dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl 4-methylbenzenesulfonate (1.28 equiv) was added and the reaction mixture was stirred at RT for 16 h. After completion, the reaction was diluted with water and extracted with ethyl acetate. The combined organic fractions were evaporated and tert-butyl (2-(2-(2-(2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (200 mg, 0.32 mmol, 37% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 625.2 [M+H]+
Step 3: Methyl 2-(5-(2-((4-((2,2-dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl)oxy)piperidin- l-yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (51% yield), using tert-butyl (2-(2- (2-(2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (250 mg, 0.4 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 735.3 [M+H]+
Step 4: Methyl 2-(5-(2-((4-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (50% yield), using methyl 2-(5-(2- ((4-((2,2-dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl)oxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (30 mg, 0.04 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 635.4 [M+H]+
Step 5: To a solution of methyl 2-(5-(2-((4-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethoxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate trifluoroacetate (28 mg, 0.038 mmol, 1 equiv) in DMF (1 mL) were added HATU (1.5 equiv) and DIPEA (2.9 equiv) and the resulting mixture was stirred at RT for 30 min. (S)-2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- o][l,4]diazepin-6-yl)acetic acid (1.2 equiv) in DMF (3 mL) and DIPEA (2.9 equiv) was added and the reaction mixture was stirred at RT for 1 h. After completion, the reaction was diluted with water and extracted with ethyl acetate. The combined organic fractions were dried over NajSCU and evaporated. The crude methyl (S)-2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate was forwarded directly into the next step.
LCMS (ESI+) m/z 1017.6 [M+H]+
Step 6: (S)-2-(5-(2-((4-((l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (50% yield over two steps), using methyl (S)- 2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate (50 mg, crude) as starting material.
LCMS (ESI+) m/z 1003.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.57 (d, J = 7.1 Hz, 1H), 8.27 (t, J = 5.6 Hz, 1H), 7.91 (s, 1H), 7.68-7.53 (m, 2H), 7.52-7.39 (m, 6H), 6.63 (dd, J = 1.6, 7.1 Hz, 1H), 4.66-4.40 (m, 1H), 3.72 (s, 3H), 3.67 (d, J = 5.3 Hz, 2H), 3.55-3.42 (m, 16H), 3.30-3.15 (m, 3H), 3.10-2.95 (m, 2H), 2.72-2.60 (m, 3H), 2.60 (s, 3H), 2.42 (s, 3H), 1.75-1.55 (m, 5H), 1.37-1.22 (m, 2H).
Example 1-092: Synthesis of (S)-(2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1009)
Step 1: To a stirred solution of (S)-2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (60 mg, 0.06 mmol, 1 equiv) in DMF (1 mL) were added KHCO3 (3 equiv) and chloromethyl pivalate (1.5 equiv) at 0°C. The resulting solution was stirred at RT for 2 h. After completion the reaction was quenched with ice-water and extracted with ethyl acetate. The combined organic fractions were dried over NajSCU and evaporated. The residue was triturated with pentane/ether (3/1, v/v) to afford (S)-(2-(5-(2-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl- 6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14- yl)oxy)piperidin-l-yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (30 mg, 0.027 mmol, 44% yield) in form of white solid.
LCMS (ESI+) m/z 1117.2, 1119.1 [M+H]+
TH NMR (400 MHz, DMSO-d6) δ 8.58 (d, J = 7.1 Hz, 1H), 8.26 (t, J = 5.6 Hz, 1H), 7.90 (s, 1H), 7.63 (t, J = 8.2 Hz, 1H), 7.56 (d, J = 7.4 Hz, 1H), 7.47 (t, J = 5.0 Hz, 2H), 7.41 (d, J = 8.2 Hz, 3H), 6.65 (dd, J = 1.8, 7.1 Hz, 1H), 5.70 (s, 2H), 4.50 (dd, J = 6.1, 8.0 Hz, 1H), 3.94 - 3.79 (m, 2H), 3.71 (s, 3H), 3.54 - 3.40 (m, 13H), 3.30 - 3.14 (m, 5H), 3.11 - 2.91 (m, 3H), 2.67 - 2.59 (m, 1H), 2.59 (s, 3H), 2.40 (s, 3H), 1.75 - 1.62 (m, 2H), 1.61 (s, 3H), 1.36 - 1.21 (m, 3H), 1.13 (s, 1H), 1.05 (s, 9H).
Example 1-093: Synthesis of 2-(5-(2-((4-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethoxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 78)
Step 1: 2-(5-(2-((4-((2,2-Dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl)oxy)piperidin-l- yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield), using methyl 2-(5-(2- ((4-((2,2-dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl)oxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetate (150 mg, 0.204 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 721.4 [M+H]+
Step 2: 2-(5-(2-((4-(2-(2-(2-(2-Aminoethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (as trifluoroacetic acid salt) was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (95% yield), using 2- (5-(2-((4-((2,2-dimethyl-4-oxo-3,8,ll,14-tetraoxa-5-azahexadecan-16-yl)oxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (30 mg, 0.04 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 621.4 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.57 (d, J = 7.1 Hz, 1H), 7.91 (s, 1H), 7.87-7.68 (m, 2H), 7.63 (t, J = 8.0 Hz, 1H), 7.57 (d, J = 7.4 Hz, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.40 (s, 1H), 6.63 (dd, J = 1.7, 7.1 Hz, 1H), 3.72 (s, 3H), 3.68 (d, J = 5.0 Hz, 2H), 3.61-3.52 (m, 6H), 3.49 (s, 4H), 3.45 (s, 4H), 3.39 - 3.28 (m, 1H), 3.11-2.91 (m, 4H), 2.67 - 2.50 (m, 2H), 1.72-1.61 (m, 2H), 1.38-1.21 (m, 2H).
Example 1-094: Synthesis of 2-(5-(2-methoxy-6-((4-((2-oxo-6,9,12-trioxa-3-azatetradecan-14- yl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 79)
Step 1: 2-(2-(2-(2-((l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethan-l-amine trifluoroacetate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl (2-(2-(2-(2-((l-((2- bromo-3-methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (70 mg, 0.12 mmol) as starting material. After completion, the volatiles were removed under reduced pressure. The residue was twice co-evaporated with DCM and then lyophilized to afford crude material, which was directly forwarded into the next step.
LCMS (ESI+) m/z 524.9 [M+H]+
Step 2: /V-(2-(2-(2-(2-((l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)acetamide was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above, using 2-(2-(2-(2-((l-((2-bromo-3- methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethan-l-amine trifluoroacetate (90 mg, 0.14 mmol, 1 equiv) as starting material. After completion, the reaction was quenched with ice-cold water and extracted with DCM. The combined organic fractions were dried over NajSCU, evaporated and the resulting crude was used directly for the next step.
LCMS (ESI+) m/z 566.9, 569.0 [M+H]+
Step 3: 2-(5-(2-Methoxy-6-((4-((2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (4% yield over three steps), using /V-(2-(2-(2- (2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)acetamide (70 mg, 0.124 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- o]pyridin-3-yl)acetate (1.2 equiv) as starting materials, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 663.2 [M+H]+
NMR (400 MHz, DMSO-d6) δ 8.56 (d, J = 7.1 Hz, 1H), 8.07-7.84 (m, 2H), 7.63 (t, J = 8.0 Hz, 1H), 7.57 (d, J = 7.6 Hz, 1H), 7.45 (d, J = 8.1 Hz, 1H), 7.40 (s, 1H), 6.62 (dd, J = 1.3, 7.1 Hz, 1H), 3.72 (s, 3H), 3.71-3.57 (m, 2H), 3.54-3.43 (m, 13H), 3.38 (t, J = 5.9 Hz, 2H), 3.17 (q, J = 5.7 Hz, 2H), 3.10-2.94 (m, 2H), 2.72-2.58 (m, 2H), 1.78 (s, 3H), 1.71-1.61 (m, 2H), 1.40-1.20 (m, 2H).
Example 1-095: Synthesis of (S)-(2-(5-(4-(l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-amido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1011)
Step 1: To a stirred solution of (S)-l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-oic acid (12 mg, 0.019 mmol, 1 equiv), (2-(5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (1 equiv) and pyridine (5 equiv) in THF (1 mL), cooled to 0°C, was added solution of 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (0.056 mL, 50% in THF, 5 equiv) and the reaction mixture was stirred at 65°C for 18 h. After completion, the volatiles were removed under reduced pressure and (S)-(2-(5-(4-(l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-amido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (11 mg, 0.006 mmol, 30% yield) was purified by preparative HPLC.
(S)-l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo- 6,9,12,15-tetraoxa-3-azaoctadecan-18-oic acid was prepared as described in Chen, H. et al., J. Am. Chem. Soc. 2021, 143, 15073.
LCMS (ESI+) m/z 1188.2 [M+H]+
TH NMR (500 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.62 (dd, J = 7.3, 1.0 Hz, 1H), 8.32 (d, J = 2.2 Hz, 1H), 8.28 (t, J = 5.7 Hz, 1H), 7.98 - 7.92 (m, 2H), 7.56 (dd, J = 2.0, 1.0 Hz, 1H), 7.52 - 7.48 (m, 2H), 7.46 - 7.43 (m, 2H), 7.41 (s, 1H), 6.85 (dd, J = 7.2, 2.0 Hz, 1H), 5.72 (s, 2H), 4.52 (dd, J = 8.1, 6.0 Hz, 1H), 3.91 (s, 2H), 3.74 (t, J = 6.2 Hz, 2H), 3.57 - 3.51 (m, 12H), 3.47 (t, J = 5.9 Hz, 2H), 3.38 (s, 2H), 3.30 (s, 2H), 3.26 - 3.18 (m, 1H), 3.17 (s, 3H), 3.03 (ddd, J = 11.6, 7.0, 3.7 Hz, 2H), 2.72 (ddd, J = 12.2, 8.2, 3.4 Hz, 2H), 2.63 (t, J = 6.2 Hz, 2H), 2.61 (s, 3H), 2.42 (d, J = 1.0 Hz, 3H), 1.68 - 1.60 (m, 5H), 1.37 - 1.24 (m, 2H), 1.06 (s, 9H).
Example 1-096: Synthesis of (S)-2-(5-(4-(l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-amido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1010)
Step 1: (S)-2-(5-(4-(l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaoctadecan-18-amido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (30% yield), using (S)-(2-(5-(4-(l-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12,15- tetraoxa-3-azaoctadecan-18-amido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (10 mg, 0.008 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1074.2 [M+H]+
TH NMR (500 MHz, DMSO-d6) δ 10.34 (s, 1H), 8.50 (d, J = 7.1 Hz, 1H), 8.25 - 8.22 (m, 1H), 8.21 (t, J = 5.7 Hz, 1H), 7.88 - 7.83 (m, 2H), 7.46 (d, J = 1.9 Hz, 1H), 7.43 - 7.39 (m, 2H), 7.37 - 7.33 (m, 2H), 7.32 (d, J = 8.4 Hz, 1H), 6.72 (dd, J = 7.1, 2.0 Hz, 1H), 4.43 (dd, J = 8.1, 6.0 Hz, 1H), 3.65 (t, J = 6.2 Hz, 2H), 3.58 (s, 2H), 3.47 - 3.41 (m, 12H), 3.38 (t, J = 5.9 Hz, 2H), 3.17 - 3.10 (m, 5H), 3.07 (s, 3H), 2.98 - 2.91 (m, 3H), 2.62 (ddd, J = 12.2, 8.2, 3.5 Hz, 2H), 2.54 (t, J = 6.3 Hz, 2H), 2.52 (s, 3H), 2.33 (s, 3H), 1.58 - 1.49 (m, 5H), 1.22 (dtd, J = 12.1, 8.0, 3.7 Hz, 2H).
Example 1-097: Synthesis of (S)-(2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)carbamoyl)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1013)
Step 1: 3-((4-Methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5- o]pyridin-5-yl)benzoic acid (34.9 mg, 0.059 mmol, 1.1 equiv) and HATU (1.2 equiv) were dissolved in dry DMF (0.25mL) and stirred at RT for 10 min. Then (S)-/V-(2-(2-(2-(2-aminoethoxy)ethoxy)ethoxy)ethyl)-2- (4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamide hydrochloride (33 mg, 0.054 mmol, 1 equiv) was added followed by DIPEA (2 equiv) and the reaction mixture was stirred at RT for 3 h. After completion, the solvent was removed in vacuo and (S)-(2-(5-(4-((l- (4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo- 6,9,12-trioxa-3-azatetradecan-14-yl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (31 mg, 0.026 mmol, 49% yield) was purified by preparative HPLC.
(S)-/V-(2-(2-(2-(2-Aminoethoxy)ethoxy)ethoxy)ethyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H- thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamide hydrochloride was prepared as described in WO2019238816A1.
LCMS (ESI+) m/z 1144.3 [M+H]+
1H NMR (500 MHz, DMSO-d6) δ 8.93 (t, J = 5.6 Hz, 1H), 8.65 (dd, J = 7.3, 0.9 Hz, 1H), 8.45 (d, J = 1.9 Hz, 1H), 8.26 (t, J = 5.7 Hz, 1H), 8.19 (dd, J = 8.0, 1.9 Hz, 1H), 7.96 (s, 1H), 7.61 (dd, J = 2.0, 1.0 Hz, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.5 Hz, 2H), 7.45 - 7.39 (m, 2H), 6.86 (dd, J = 7.1, 1.9 Hz, 1H), 5.70 (s, 2H), 4.50 (dd, J = 8.0, 6.1 Hz, 1H), 3.90 (s, 2H), 3.61 - 3.50 (m, 10H), 3.46 (dt, J = 13.7, 5.9 Hz, 4H), 3.29 - 3.15 (m, 6H), 3.13 (s, 3H), 2.99 (ddd, J = 15.2, 6.8, 3.8 Hz, 2H), 2.66 (ddd, J = 12.4, 8.5, 3.6 Hz, 2H), 2.59 (s, 3H), 2.40 (s, 3H), 1.61 (s, 3H), 1.60 - 1.56 (m, 1H), 1.32 - 1.22 (m, 2H), 1.04 (s, 9H).
Example 1-098: Synthesis of (S)-2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)carbamoyl)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1012) Step 1: (S)-2-(5-(4-((l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (88% yield), using (S)-(2-(5-(4-((l-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12- trioxa-3-azatetradecan-14-yl)carbamoyl)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- o]pyridin-3-yl)acetoxy)methyl pivalate (15 mg, 0.013 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1030.3 [M+H]+
'H NMR (500 MHz, DMSO-d6) δ 8.93 (t, J = 5.6 Hz, 1H), 8.63 (d, J = 7.2 Hz, 1H), 8.45 (d, J = 1.8 Hz, 1H), 8.26 (t, J = 5.7 Hz, 1H), 8.18 (dd, J = 7.9, 1.9 Hz, 1H), 7.95 (s, 1H), 7.62 - 7.59 (m, 1H), 7.57 (d, J = 8.0 Hz, 1H), 7.48 (d, J = 8.5 Hz, 2H), 7.45 - 7.39 (m, 2H), 6.83 (dd, J = 7.2, 1.9 Hz, 1H), 4.50 (dd, J = 8.0, 6.1 Hz, 1H), 3.70 (s, 2H), 3.61 - 3.50 (m, 10H), 3.46 (dt, J = 12.3, 5.8 Hz, 6H), 3.30 - 3.14 (m, 4H), 3.13 (s, 3H), 2.99 (ddd, J = 11.5, 6.9, 3.8 Hz, 2H), 2.70 - 2.60 (m, 2H), 2.59 (s, 3H), 2.40 (s, 3H), 1.61 (s, 3H), 1.57 (dq, J = 10.7, 3.6 Hz, 2H), 1.31 - 1.19 (m, 2H).
Example 1-099: Synthesis of 2-(5-(2-((4-((14-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)-3,6,9,12- tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1024)
Step 1: To a stirred solution of 14-(piperidin-4-yloxy)-3,6,9,12-tetraoxatetradecan-l-ol hydrochloride (600 mg, 1.67 mmol, 1 equiv) in DCM (10 mL), cooled to 0°C, were added 2-bromobenzenesulfonyl chloride (3 equiv) and TEA (6.45 equiv) and the reaction mixture was stirred at RT for 16 h. After completion the reaction was quenched with cold water and extracted with DCM. The combined organic fractions were washed with brine, dried over NajSCU and evaporated. 14-((l-((2-Bromophenyl)sulfonyl)piperidin-4- yl)oxy)-3,6,9,12-tetraoxatetradecyl 2-bromobenzenesulfonate (400 mg, 0.53 mmol, 31% yield) was purified by flash column chromatography.
14-(Piperidin-4-yloxy)-3,6,9,12-tetraoxatetradecan-l-ol hydrochloride was prepared as described in W02021077010A1 and US2014031559A1.
LCMS (ESI+) m/z 761.8 [M+H]
Step 2: To a solution of 2,5-dichloro-/V-(2-(isopropylsulfonyl)phenyl)pyrimidin-4-amine (2.5 g, 7.2 mmol, 1 equiv) in isopropanol (25 mL) and 6M HCI (25 mL) was added tert-butyl 4-(4-aminophenyl)piperazine-l- carboxylate (1.2 equiv) and stirred at 95°C for 16 h. The volatiles were removed under reduced pressure, the residue was triturated with ether, taken up in saturated NaHCOs solution and extracted with ethyl acetate. The organic fraction was then evaporated and 5-chloro-/V4-(2-(isopropylsulfonyl)phenyl)-/V2-(4- (piperazin-l-yl)phenyl)pyrimidine-2,4-diamine (1.0 g, 2.05 mmol, 28% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 487.0, 489.0 [M+H]+
Step 3: To a solution of 5-chloro-/V4-(2-(isopropylsulfonyl)phenyl)-/V2-(4-(piperazin-l- yl)phenyl)pyrimidine-2,4-diamine (35 mg, 0.071 mmol, 1.2 equiv) in DMF (3 mL) was added K2CO3 (2.5 equiv) at RT and the resulting mixture was stirred at the same temperature for 30 min. Then 14-((l-((2- bromophenyl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl 2-bromobenzenesulfonate (45 mg, 0.059 mmol, 1 equiv) was added and the reaction mixture was stirred at 70°C for 16 h. After completion, the reaction was diluted with water and extracted with ethyl acetate. /V2-(4-(4-(14-((l-((2- Bromophenyl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl)piperazin-l-yl)phenyl)-5-chloro- /V4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (30 mg, 0.03 mmol, 49% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 1008.1, 1010.3 [M+H]+
Step 4: 2-(5-(2-((4-((14-(4-(4-((5-Chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)-3,6,9,12-tetraoxatetradecyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (9% yield), using /V2-(4-(4-(14-((l-((2- bromophenyl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl)piperazin-l-yl)phenyl)-5-chloro- /V4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (50 mg, 0.05 mmol, 1 equiv) and methyl 2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, Pd(dppf)CI2 (0.1 equiv) as catalyst and K3PO4 (3.5 equiv) as base.
LCMS (ESI+) m/z 1104.7 [M+H]+
4H NMR (400 MHz, DMSO-d6) δ 9.49 (s, 1H), 9.43 - 9.27 (m, 1H), 8.77 - 8.64 (m, 1H), 8.61 (d, J = 6.8 Hz, 1H), 8.24 (s, 1H), 7.98 (d, J = 8.6 Hz, 1H), 7.94 (s, 1H), 7.84 (d, J = 6.4 Hz, 1H), 7.78 - 7.71 (m, 2H), 7.66 (t, J = 7.2 Hz, 1H), 7.56 (s, 1H), 7.46 (d, J = 7.5 Hz, 3H), 7.36 (t, J = 8.3 Hz, 1H), 7.00 - 6.85 (m, 2H), 6.82 (d, J = 7.7 Hz, 1H), 3.90 - 3.66 (m, 6H), 3.65 - 3.33 (m, 20H), 3.18 - 2.90 (m, 8H), 2.80 - 2.58 (m, 2H), 1.68 - 1.54 (m, 2H), 1.38 - 1.21 (m, 2H), 1.16 (d, J = 6.8 Hz, 6H). Example 1-100: Synthesis of (2-(5-(2-((4-((14-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)-3,6,9,12- tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1025)
Step 1: (2-(5-(2-((4-((14-(4-(4-((5-Chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)-3,6,9,12-tetraoxatetradecyl)oxy)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (4.5% yield), using /V2-(4-(4-(14- ((l-((2-bromophenyl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl)piperazin-l-yl)phenyl)-5- chloro-/V4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (140 mg, 0.139 mmol, 1 equiv) and (2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (1.1 equiv) as starting materials, Pd(dtbpf)Cl2 (0.1 equiv) as catalyst and CS2CO3 (2 equiv) as base.
LCMS (ESI+) m/z 1218.4, 1220.3 [M+H]+
4H NMR (400 MHz, DMSO-d6) δ 9.49 (s, 1H), 9.31 (s, 1H), 8.62 (d, J = 7.2 Hz, 1H), 8.23 (s, 1H), 8.06 - 7.92 (m, 2H), 7.83 (d, J = 8.0 Hz, 1H), 7.74 (q, J = 7.7 Hz, 2H), 7.67 (t, J = 7.7 Hz, 1H), 7.56 (s, 1H), 7.52 - 7.40 (m, 3H), 7.36 (t, J = 7.6 Hz, 1H), 6.95 - 6.79 (m, 3H), 5.70 (s, 2H), 3.89 (s, 2H), 3.49 (q, J = 19.6, 16.2 Hz, 22H), 3.03 (d, J = 12.4 Hz, 7H), 2.66 (d, J = 9.4 Hz, 3H), 1.60 (s, 2H), 1.28 (dd, J = 25.4, 14.8 Hz, 4H), 1.16 (d, J = 6.8 Hz, 6H), 1.03 (s, 9H). Example 1-101: Synthesis of (S)-(2-(5-(4-(l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azapentadecan-15-amido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1017)
Step 1: To the solution of (S)-l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azapentadecan-15-oic acid (19 mg, 0.031 mmol, 1 equiv), (2- (5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (1.02 equiv) and pyridine (2 equiv) in THF (0.32 mL) was added solution of 2,4,6-tripropyl- 1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (0.028 mL, 50% in THF, 1.5 equiv) and the reaction mixture was stirred at 60°C for 24 h. After completion, the volatiles were removed under reduced pressure. The residue was taken up in ethyl acetate, washed with water, saturated NaHCOs, brine, dried over MgSO4 and evaporated. (S)-(2-(5-(4-(l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azapentadecan-15-amido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (6.5 mg, 0.006 mmol, 17% yield) was purified by preparative HPLC.
(S)-l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-
6,9,12-trioxa-3-azapentadecan-15-oic acid was prepared as described in Chen, H. et al., J. Am. Chem. Soc.
2021, 143, 15073. LCMS (ESI+) m/z 1144.3 [M+H]+
NMR (500 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.60 (dd, J = 7.2, 1.0 Hz, 1H), 8.29 (d, J = 2.3 Hz, 1H), 8.25 (t, J = 5.6 Hz, 1H), 7.93 (s, 1H), 7.93 (dd, J = 8.4, 2.2 Hz, 1H), 7.54 (dd, J = 2.0, 1.0 Hz, 1H), 7.49 - 7.46 (m, 2H), 7.44 - 7.38 (m, 3H), 6.83 (dd, J = 7.2, 2.0 Hz, 1H), 5.70 (d, J = 1.4 Hz, 2H), 4.50 (dd, J = 8.0, 6.1 Hz, 1H), 3.88 (s, 2H), 3.73 (t, J = 6.2 Hz, 2H), 3.56 - 3.49 (m, 6H), 3.45 (q, J = 6.1 Hz, 2H), 3.29 - 3.16 (m, 5H), 3.14 (s, 3H), 3.01 (ddd, J = 11.5, 7.0, 3.7 Hz, 2H), 2.78 - 2.65 (m, 2H), 2.61 (t, J = 6.2 Hz, 2H), 2.59 (s, 3H), 2.40 (d, J = 0.9 Hz, 3H), 1.65 - 1.59 (m, 4H), 1.29 (dtd, J = 12.1, 8.0, 3.7 Hz, 3H), 1.24 (s, 2H), 1.04 (s, 9H).
Example 1-102: Synthesis of (S)-2-(5-(4-(l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azapentadecan-15-amido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1016)
Step 1: (S)-2-(5-(4-(l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azapentadecan-15-amido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (75% yield), using (S)-(2-(5-(4-(l-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12- trioxa-3-azapentadecan-15-amido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (4 mg, 0.003 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1030.3 [M+H]+ 'H NMR (500 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.56 (d, J = 7.2 Hz, 1H), 8.32 - 8.25 (m, 2H), 7.92 (dd, J = 8.4, 2.3 Hz, 1H), 7.90 (s, 1H), 7.55 - 7.51 (m, 1H), 7.50 - 7.45 (m, 2H), 7.44 - 7.40 (m, 2H), 7.39 (d, J = 8.4 Hz, 1H), 6.78 (dd, J = 7.2, 2.0 Hz, 1H), 4.50 (dd, J = 8.0, 6.1 Hz, 1H), 3.73 (t, J = 6.2 Hz, 2H), 3.60 (s, 2H), 3.57 - 3.48 (m, 8H), 3.44 (t, J = 5.9 Hz, 2H), 3.28 - 3.16 (m, 4H), 3.14 (s, 3H), 3.02 (ddd, J = 11.8, 7.0, 3.6 Hz, 2H), 2.69 (ddd, J = 12.1, 8.2, 3.5 Hz, 2H), 2.61 (t, J = 6.2 Hz, 2H), 2.59 (s, 3H), 2.40 (s, 3H), 1.65 - 1.51 (m, 5H), 1.34 - 1.25 (m, 2H), 1.25 - 1.22 (m, 2H).
Example 1-103: Synthesis of (S)-(2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1018)
Step 1: To a solution of (S)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetic acid (50 mg, 0.125 mmol, 1 equiv) and HATU (1.5 equiv) in dry DMF (2.5 mL) was added DIPEA (3 equiv) and the mixture was stirred at RT for 15 min. 2-(2-(2-(2- Aminoethoxy)ethoxy)ethoxy)ethan-l-ol (3 equiv) was added and the reaction mixture was stirred at RT for 20 h. After completion, the solution was concentrated under reduced pressure, diluted with saturated NajCOs aqueous solution and extracted with DCM. The combined organic fractions were washed with water, brine, dried over Na2SO4 and concentrated. (S)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H- thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-/V-(2-(2-(2-(2- hydroxyethoxy)ethoxy)ethoxy)ethyl)acetamide (35 mg, 0.06 mmol, 47% yield) was purified by flash column chromatography. LCMS (ESI+) m/z 576.2 [M+H]
Step 2: To a solution of (S)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-/V-(2-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)ethyl)acetamide (30 mg, 0.052 mmol, 1 equiv), 4-methylbenzenesulfonyl chloride (1.5 equiv) and DMAP (0.1 equiv) in DCM (0.52 mL) was added TEA (4 equiv) and the resulting mixture was stirred at RT for 24 h. Then the reaction was diluted with pyridine (1 mL) and cut filter paper was added to remove excess of 4-methylbenzenesulfonyl chloride. The reaction mixture was sonicated for 1 h and filtered. The filtrate was diluted with 10% KHSO4 aqueous solution and extracted with DCM. The combined organic fractions were washed with water, brine, dried over NajSCU and concentrated. (S)-l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl 4- methylbenzenesulfonate (12 mg, 0.016 mmol, 30% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 730.0 [M+H]+
Step 3: To a stirred solution of (S)-l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl 4- methylbenzenesulfonate (11.5 mg, 0.016 mmol, 1 equiv) and (2-(5-(4-hydroxy-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (1.05 equiv) in ACN (0.16 mL) was added K2CO3 (5 equiv) and the reaction mixture was stirred at 60°C for 24 h. The volatiles were then removed in vacuo and (S)-(2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (9 mg, 0.008 mmol, 50% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 1117.3 [M+H]+
NMR (500 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.60 (dd, J = 7.2, 1.0 Hz, 1H), 8.29 (d, J = 2.3 Hz, 1H), 8.25 (t, J = 5.6 Hz, 1H), 7.93 (s, 1H), 7.93 (dd, J = 8.4, 2.2 Hz, 1H), 7.54 (dd, J = 2.0, 1.0 Hz, 1H), 7.49 - 7.45 (m, 2H), 7.45 - 7.37 (m, 3H), 6.83 (dd, J = 7.2, 2.0 Hz, 1H), 5.70 (d, J = 1.4 Hz, 2H), 4.50 (dd, J = 8.0, 6.1 Hz, 1H), 3.88 (s, 2H), 3.73 (t, J = 6.2 Hz, 2H), 3.56 - 3.49 (m, 8H), 3.44 (t, J = 5.9 Hz, 2H), 3.27 - 3.16 (m, 4H), 3.14 (s, 3H), 3.01 (ddd, J = 11.5, 7.0, 3.7 Hz, 2H), 2.69 (ddd, J = 12.1, 8.2, 3.5 Hz, 2H), 2.61 (t, J = 6.2 Hz, 2H), 2.59 (s, 3H), 2.40 (d, J = 0.9 Hz, 3H), 1.66 - 1.60 (m, 4H), 1.29 (dtd, J = 12.1, 8.0, 3.7 Hz, 1H), 1.24 (s, 2H), 1.04 (s, 9H). Example 1-104: Synthesis of (S)-2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1019)
Step 1: (S)-2-(5-(4-((l-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (44% yield), using (S)-(2-(5-(4-((l-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12- trioxa-3-azatetradecan-14-yl)oxy)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (5 mg, 0.004 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1003.3 [M+H]+
NMR (500 MHz, DMSO-d6) δ 12.29 (s, 1H), 8.57 (dd, J = 7.1, 0.9 Hz, 1H), 8.25 (t, J = 5.7 Hz, 1H), 7.92 (s, 1H), 7.52 (dd, J = 2.0, 1.0 Hz, 1H), 7.50 - 7.45 (m, 2H), 7.45 - 7.39 (m, 3H), 7.36 (d, J = 8.5 Hz, 1H), 7.32 (dd, J = 8.5, 2.6 Hz, 1H), 6.78 (dd, J = 7.2, 2.0 Hz, 1H), 4.51 (dd, J = 8.1, 6.0 Hz, 1H), 4.26 - 4.20 (m, 2H), 3.82 - 3.77 (m, 2H), 3.69 (s, 2H), 3.65 - 3.58 (m, 2H), 3.60 - 3.52 (m, 2H), 3.55 (s, 3H), 3.46 (t, J = 5.9 Hz, 2H), 3.24 (dt, J = 13.4, 5.3 Hz, 2H), 3.21 - 3.12 (m, 1H), 3.14 (s, 3H), 3.00 (ddd, J = 11.7, 7.2, 3.8 Hz, 2H), 2.67 (td, J = 8.6, 4.2 Hz, 2H), 2.58 (s, 3H), 2.40 (s, 3H), 1.62 - 1.61 (m, 3H), 1.59 (dd, J = 6.6, 3.2 Hz, 1H), 1.32 - 1.24 (m, 3H), 1.26 - 1.22 (m, 3H). Example 1-105: Synthesis of (S)-(2-(5-(4-(2-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)ethoxy)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1015)
Step 1: (2-(5-(4-Hydroxy-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (25 mg, 0.045 mmol, 1 equiv) and K2CO3 (5 equiv) were stirred in ACN (5 mL) at RT for 5 min. (S)-2-(2-(2-(2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- o][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (1.1 equiv) was added and the reaction mixture was stirred at 70°C for 10 h. (S)-(2-(5-(4-(2-(2-(2-(2-(4-(4-Chlorophenyl)-2,3,9- trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)ethoxy)-2- ((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (4.5 mg, 0.004 mmol, 9.4% yield) was purified by preparative HPLC.
(S)-2-(2-(2-(2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6- yl)acetamido)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate was synthesized according to procedure described in W02017011590A1.
LCMS (ESI+) m/z 1073.3 [M+H]+
NMR (500 MHz, DMSO-d6) δ 8.59 (dd, J = 7.2, 0.9 Hz, 1H), 8.27 (t, J = 5.7 Hz, 1H), 7.93 (s, 1H), 7.52 (dd, J = 2.0, 1.0 Hz, 1H), 7.50 - 7.46 (m, 2H), 7.44 - 7.41 (m, 3H), 7.37 (t, J = 8.4 Hz, 1H), 7.33 (dd, J = 8.5, 2.6 Hz, 1H), 6.81 (dd, J = 7.2, 1.9 Hz, 1H), 5.70 (s, 2H), 4.51 (dd, J = 8.1, 6.0 Hz, 1H), 4.27 - 4.21 (m, 2H), 3.88 (s, 2H), 3.83 - 3.79 (m, 2H), 3.66 - 3.62 (m, 2H), 3.61 - 3.57 (m, 2H), 3.48 (t, J = 5.9 Hz, 2H), 3.28 - 3.16 (m, 5H), 3.13 (s, 3H), 2.99 (ddd, J = 11.3, 6.7, 3.7 Hz, 2H), 2.66 (ddd, J = 12.4, 8.6, 3.5 Hz, 2H), 2.58 (s, 3H), 2.40 (s, 3H), 1.63 - 1.52 (m, 5H), 1.30 - 1.25 (m, 2H), 1.04 (s, 9H).
Example 1-106: Synthesis of (S)-2-(5-(4-(2-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)ethoxy)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1014)
Step 1: (S)-2-(5-(4-(2-(2-(2-(2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)ethoxy)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (27% yield), using (S)-(2-(5-(4-(2-(2-(2-(2-(4- (4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)ethoxy)ethoxy)ethoxy)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (4.5 mg, 0.004 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 959.2 [M+H]+
NMR (500 MHz, DMSO-d6) δ 8.58 (d, J = 7.1 Hz, 1H), 8.30 (t, J = 5.7 Hz, 1H), 7.93 (s, 1H), 7.53 (d, J = 1.7 Hz, 1H), 7.51 - 7.48 (m, 2H), 7.46 - 7.42 (m, 3H), 7.37 (d, J = 8.4 Hz, 1H), 7.33 (dd, J = 8.5, 2.6 Hz, 1H), 6.78 (dd, J = 7.2, 2.0 Hz, 1H), 4.53 (dd, J = 8.1, 6.0 Hz, 1H), 4.27 - 4.23 (m, 2H), 3.84 - 3.81 (m, 2H), 3.67 - 3.64 (m, 4H), 3.62 - 3.60 (m, 2H), 3.50 (t, J = 5.8 Hz, 2H), 3.27 - 3.23 (m, 5H), 3.15 (s, 3H), 3.04 - 2.99 (m, 2H), 2.68 (ddd, J = 12.4, 8.5, 3.6 Hz, 2H), 2.60 (s, 3H), 2.42 (s, 3H), 1.63 (s, 3H), 1.63 - 1.57 (m, 2H), 1.29 (ddd, J = 12.7, 8.3, 4.0 Hz, 2H). Example 1-107: Synthesis of (S)-(2-(5-(4-((10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)decyl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1023)
Step 1: (ALMA-273-01) The solution of (S)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetic acid (30 mg, 0.075 mmol, 1 equiv) and HATU (1.5 equiv) in DMF (1.1 mL) was stirred at RT for 10 min. tert-Butyl (lO-aminodecyl)carbamate (1.2 equiv) was added followed by DIPEA (5 equiv) and the reaction mixture was stirred at RT for 3 h, until complete conversion was indicated by LCMS. tert-Butyl (S)-(10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)decyl)carbamate (32.9 mg, 0.049 mmol, 66% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 655.3 [M+H]+
Step 2: (ALMA-274-01) (S)-/V-(10-Aminodecyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide hydrochloride was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (100% yield), using tert-butyl (S)- (10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)decyl)carbamate (32 mg, 0.049 mmol) as starting material.
LCMS (ESI+) m/z 554.8 [M+H]+ Step 3: (ALMA-276-01) The solution of 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2- ((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-5-yl)benzoic acid (34.9 mg, 0.059 mmol, 1.1 equiv) and HATU (1.2 equiv) in DMF (0.25 mL) was stirred at RT for 10 min. (S)-/V-(10-Aminodecyl)-2-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide hydrochloride (31.6 mg, 0.054 mmol, 1 equiv) and DIPEA (2 equiv) were added and the reaction was stirred at RT for 3 h, until full conversion was indicated by LCMS. (S)-(2-(5-(4-((10-(2-(4-(4-chlorophenyl)-2,3,9- trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)decyl)carbamoyl)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (12.5 mg, 0.011 mmol, 20% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 1124.4 [M+H]+
1H NMR (500 MHz, DMSO-d6) δ 8.81 (t, J = 5.6 Hz, 1H), 8.66 (dd, J = 7.1, 1.0 Hz, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.18 (dd, J = 8.0, 1.9 Hz, 1H), 8.14 (t, J = 5.7 Hz, 1H), 7.97 (s, 1H), 7.61 (dd, J = 2.0, 1.0 Hz, 1H), 7.58 (d, J = 7.8 Hz, 1H), 7.47 (d, J = 8.7 Hz, 2H), 7.45 - 7.40 (m, 2H), 6.87 (dd, J = 7.2, 1.9 Hz, 1H), 5.71 (s, 2H), 4.50 (dd, J = 8.4, 5.8 Hz, 1H), 3.90 (s, 2H), 3.28 (s, 3H), 3.25 (m, 2H), 3.24 - 3.08 (m, 2H), 3.14 (s, 3H), 3.06 (p, J = 6.4 Hz, 2H), 3.01 - 2.95 (m, 2H), 2.70 - 2.63 (m, 2H), 2.59 (s, 3H), 2.42 - 2.39 (m, 3H), 1.62 (s, 3H), 1.61 - 1.51 (m, 4H), 1.45 - 1.40 (m, 2H), 1.27 (s, 12H), 1.04 (s, 9H).
Example 1-108: Synthesis of (S)-2-(5-(4-((10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)decyl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid (Compound 1022)
Step 1: (ALMA-277-01) (S)-2-(5-(4-((10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)decyl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (60% yield), using (S)-(2-(5-(4-((10-(2-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)decyl)carbamoyl)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (8 mg, 0.007 mmol) as starting material.
LCMS (ESI+) m/z 1010.3 [M+H]+
'H NMR (500 MHz, DMSO-d6) δ 8.80 (t, J = 5.6 Hz, 1H), 8.60 (d, J = 7.2 Hz, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.17 (s, 1H), 8.17 (dd, J = 8.0, 1.9 Hz, 1H), 7.91 (s, 1H), 7.60 (d, J = 2.1 Hz, 1H), 7.56 (d, J = 7.9 Hz, 1H), 7.49 - 7.46 (m, 2H), 7.44 - 7.41 (m, 2H), 6.79 (dd, J = 7.1, 2.0 Hz, 1H), 4.50 (dd, J = 8.4, 5.7 Hz, 1H), 3.53 (s, 2H), 3.24 - 3.16 (m, 4H), 3.14 (s, 3H), 3.05 (td, J = 13.0, 6.8 Hz, 1H), 2.99 (dd, J = 9.7, 4.9 Hz, 2H), 2.72 - 2.63 (m, 2H), 2.59 (s, 3H), 2.41 (s, 3H), 1.62 (s, 3H), 1.60 - 1.51 (m, 4H), 1.43 (d, J = 7.6 Hz, 2H), 1.30 - 1.24 (m, 16H).
Example 1-109: Synthesis of (S)-(2-(5-(4-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)octanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1021)
Step 1: (ALMA-280-01) To a solution of (S)-8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- /][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)octanoic acid (30 mg, 0.055 mmol, 1 equiv), (2-(5-(4- amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-o]pyridin-3-yl)acetoxy)methyl pivalate (1.02 equiv) and pyridine (2 equiv) in THF (0.55 mL) was added 2,4,6-tripropyl-l,3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide (0.05 mL, 50% in THF, 1.5 equiv). The reaction mixture was stirred at 60°C for 20 h until complete conversion was indicated by LCMS. The volatiles were removed in vacuo, the residue was taken up in ethyl acetate and washed with water, NaHCOs saturated solution and brine. The organic phase was dried over MgSO4 and evaporated. (S)-(2-(5-(4-(8-(2-(4-(4-chlorophenyl)-2,3,9- trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6-yl)acetamido)octanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (11.5 mg, 0.011 mmol, 19% yield) was purified by preparative HPLC.
(S)-8-(2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-o][l,4]diazepin-6- yl)acetamido)octanoic acid was prepared as described in W02017197051A1.
LCMS (ESI+) m/z 1082.3 [M+H]+
TH NMR (500 MHz, DMSO-d6) δ 10.33 (s, 1H), 8.63 - 8.58 (m, 1H), 8.27 (d, J = 2.2 Hz, 1H), 8.16 (t, J = 5.7 Hz, 1H), 7.95 (d, J = 7.8 Hz, 2H), 7.53 (dd, J = 2.0, 0.9 Hz, 1H), 7.51 - 7.46 (m, 2H), 7.45 - 7.38 (m, 2H), 7.39 (d, J = 8.4 Hz, 1H), 6.83 (dd, J = 7.1, 2.0 Hz, 1H), 5.70 (s, 2H), 4.50 (dd, J = 8.1, 6.0 Hz, 1H), 3.89 (s, 2H), 3.29 - 3.16 (m, 2H), 3.15 (s, 3H), 3.17 - 3.04 (m, 2H), 3.00 (ddd, J = 11.7, 7.0, 3.8 Hz, 2H), 2.69 (ddd, J = 12.2, 8.3, 3.6 Hz, 2H), 2.59 (s, 3H), 2.40 (d, J = 1.0 Hz, 3H), 2.37 - 2.32 (m, 2H), 1.64 - 1.58 (m, 6H), 1.47 - 1.42 (m, 2H), 1.33 - 1.30 (m, 6H), 1.30 - 1.22 (m, 4H), 1.04 (s, 9H).
Example 1-110: Synthesis of (S)-2-(5-(4-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)octanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1020)
Step 1: (ALMA-281-01) (S)-2-(5-(4-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-
/][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)octanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (93% yield), using (S)-(2-(5-(4-(8-(2-(4-(4- chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-/][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)octanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (8 mg, 0.007 mmol) as starting material.
LCMS (ESI+) m/z 968.3 [M+H]+
TH NMR (500 MHz, DMSO-d6) δ 12.33 (s, 1H), 10.33 (s, 1H), 8.59 (dd, J = 7.1, 0.9 Hz, 1H), 8.28 (d, J = 2.2 Hz, 1H), 8.16 (t, J = 5.6 Hz, 1H), 7.97 - 7.91 (m, 1H), 7.93 (s, 1H), 7.53 (dd, J = 2.0, 1.0 Hz, 1H), 7.48 (d, J = 8.8 Hz, 2H), 7.49 - 7.39 (m, 2H), 7.38 (d, J = 8.3 Hz, 1H), 6.80 (dd, J = 7.2, 2.0 Hz, 1H), 4.50 (dd, J = 8.1, 6.0 Hz, 1H), 3.68 (s, 2H), 3.26 - 3.17 (m, 2H), 3.15 (s, 3H), 3.10 (dq, J = 13.0, 6.3 Hz, 1H), 3.01 (td, J = 7.3, 3.5 Hz, 2H), 2.69 (ddd, J = 12.1, 8.4, 3.6 Hz, 2H), 2.59 (s, 3H), 2.40 (s, 3H), 2.37 - 2.32 (m, 2H), 1.62 (s, 3H), 1.59 (m, 2H), 1.45 (s, 2H), 1.36 - 1.20 (m, 12H).
Example 1-111: Synthesis of 2-(5-(2-((4-(2-(2-acetamidoethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 81)
Step 1: To a solution of l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4-ol (215 mg, 0.616 mmol, 1 equiv) in DMF (2 mL), cooled in an ice-water bath, was added NaH (2 equiv, 60% suspension in mineral oil) and the resulting mixture was stirred for 15 min. 2-(2-Azidoethoxy)ethyl 4-methylbenzenesulfonate (1 equiv) was then added and the reaction mixture was stirred at RT for 16 h, quenched with ice-cold water and extracted with diethyl ether. The combined organic fractions were dried over NajSCU and evaporated yielding crude 4-(2-(2-azidoethoxy)ethoxy)-l-((2-bromo-3- methoxyphenyl)sulfonyl)piperidine (300 mg) which was directly forwarded to the next step. 2-(2-Azidoethoxy)ethyl 4-methylbenzenesulfonate was prepared as described in Abellan-Flos, M. et al., Org. Biomol. Chem. 2015, 27, 7445
LCMS (ESI+) m/z 465.0 [M+H]+
Step 2: To a stirred solution of crude 4-(2-(2-azidoethoxy)ethoxy)-l-((2-bromo-3- methoxyphenyl)sulfonyl)piperidine (300 mg) in THF (5 mL) and water (1 mL) was added triphenylphosphine (340 mg, 1.296 mmol) and the reaction mixture was stirred at 60°C for 4 h and quenched with water. The resulting mixture was extracted with DCM, the combined organic fractions were dried over Na2SO4 and evaporated. 2-(2-((l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethan-l-amine (160 mg, 0.37 mmol, 59% yield over two steps) was purified by flash column chromatography.
LCMS (ESI+) m/z 439.0 [M+H]+
Step 3: /V-(2-(2-((l-((2-Bromo-3-methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethyl)acetamide was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above, using 2-(2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethan-l-amine (70 mg, 0.161 mmol) as starting material. After completion the crude product was directly forwarded into the next step.
LCMS (ESI+) m/z 481.0 [M+H]+
Step 4: Methyl 2-(5-(2-((4-(2-(2-acetamidoethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using /V-(2-(2-((l-((2-bromo-3- methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethyl)acetamide (70 mg, 0.146 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-o]pyridin-3-yl)acetate (1 equiv) as starting materials, CS2CO3 (2 equiv) as base, Pd(dtbpf)Cl2 as catalyst (0.1 equiv). After completion the reaction mixture was filtered through Celite®, the filtrate was evaporated to give crude product which was used in the next step without further purification.
LCMS (ESI+) m/z 589.4 [M+H] Step 5: 2-(5-(2-((4-(2-(2-Acetamidoethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-o]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (4% yield over three steps), using methyl 2- (5-(2-((4-(2-(2-acetamidoethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5- o]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 575.2 [M+H]+
TH NMR (400 MHz, DMSO-d6, 100°C) 68.51 (d, J = 7.2 Hz, 1H), 7.89 (s, 1H), 7.64 - 7.57 (m, 2H), 7.46 - 7.38 (m, 2H), 6.66 - 6.59 (m, 1H), 3.74 (s, 3H), 3.65 (s, 2H), 3.51 - 3.46 (m, 3H), 3.48 - 3.38 (m, 4H), 3.24 - 2.98 (m, 4H), 2.76 - 2.67 (m, 2H), 1.80 (s, 3H), 1.69 - 1.64 (m, 2H), 1.39 - 1.35 (m, 2H).
Example 1-112: Synthesis of (S)-2-(5-(2-((4-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1041)
Step 1: To a stirred solution of 2-(2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethan-l-amine (70 mg, 0.161mmol, 1 equiv) in DMF (1.0 mL) were added (S)-2-(4-(4- chlorophenyl)-2,3-dimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetic acid
[commercial] (64.0 mg, 0.161 mmol), HATU (92.0 mg, 0.241mmol, 1.5 equiv) and DIPEA (0.08 mL, 0.482 mmol, 3 equiv). The resulting mixture was stirred at RT for 20 min and after completion, reaction was quenched with ice water and extracted with ethyl acetate. Organic fraction was evaporated under reduced pressure to afford crude (S)-N-(2-(2-((l-((2-bromo-3-methoxyphenyl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamide, which was forwarded to the next step without purification.
LCMS (ESI+) m/z 819.2, 821.2 [M+H]+ Step 2: (S)-2-(5-(2-((4-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (27% yield over two steps) using (S)-N-(2-(2-((l-((2-bromo-3- methoxyphenyl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H- thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide (120 mg, 0.146 mmol, 1 equiv) and methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (46mg, 0.146 mmol, 1 equiv) as starting materials, Pd-118 (0.1 equiv) as catalyst and Cs2CO3 (2 equiv) as a base.
LCMS (ESI+) m/z 915.2 [M+H]+
THNMR (400MHz, DMSO-d6) 6 8.56 (d, J = 7.2 Hz, 1H), 8.26 (t, J = 5.7 Hz, 1H), 7.90 (s, 1H), 7.66 - 7.58 (m, 1H), 7.58 - 7.52 (m, 1H), 7.51 - 7.37 (m, 6H), 6.62 (dd, J = 1.8, 7.2 Hz, 1H), 4.49 (dd, J = 6.2, 8.0 Hz, 1H), 3.72 (s, 3H), 3.68 - 3.63 (m, 2H), 3.52 - 3.46 (m, 4H), 3.43 (t, J = 5.8 Hz, 2H), 3.32 - 3.14 (m, 5H), 3.11 - 2.95 (m, 2H), 2.70 - 2.59 (m, 2H), 2.59 (s, 3H), 2.41 (s, 3H), 1.71 - 1.63 (m, 2H), 1.61 (s, 3H), 1.37 - 1.22 (m, 2H).
Example 1-113: Synthesis of (S)-(2-(5-(2-((4-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1040)
Step 1: To a stirred solution of (S)-2-(5-(2-((4-(2-(2-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6- methoxyphenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (35 mg, 0.038 mmol, 1 equiv) in DMF (1 mL) were added KHC03 (2 equiv) and chloromethyl pivalate (1.5 equiv) at 0°C. The resulting solution was stirred at RT for 2 h. After completion the volatiles were removed under reduced pressure. (S)-(2-(5-(2-((4-(2-(2-(2- (4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)-6-methoxyphenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (5 mg, 12% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 1029.2 [M+H]+
NMR (401 MHZ, DMSO-d6) 6 8.58 (d, J = 7.2 Hz, 1H), 8.24 (t, J = 5.7 Hz, 1H), 7.90 (s, 1H), 7.66 - 7.58 (m, 1H), 7.58 - 7.52 (m, 1H), 7.51 - 7.38 (m, 6H), 6.65 (dd, J = 1.9, 7.2 Hz, 1H), 5.70 (s, 2H), 4.53 - 4.45 (m, 1H), 3.94 - 3.79 (m, 2H), 3.71 (s, 3H), 3.51 - 3.45 (m, 4H), 3.43 (t, J = 5.9 Hz, 2H), 3.32 - 3.14 (m, 5H), 3.07 - 2.93 (m, 2H), 2.70 - 2.60 (m, 2H), 2.59 (s, 3H), 2.41 (s, 3H), 1.71 - 1.62 (m, 2H), 1.61 (s, 3H), 1.37 - 1.26 (m, 2H) 1.05 (s, 9H).
Example 1-114: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)naphthalen-l-yl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid (Compound 90)
Step 1: To a solution of l-bromonaphthalen-2-amine [commercial] (2.5 g, 11.257 mmol, 1 equiv) in acetonitrile (25 mL), cooled to 0 °C, was added acetic acid (10 mL) and concentrated hydrochloric acid, 32% (5 mL) and reaction was stirred for 5 min. An aqueous solution of sodium nitrite (1.052 g, 12.383 mmol, 1.1 equiv) in water (15 mL) was added over 10 min and the reaction was stirred for another 30 min at 0°C. The resulting mixture was added to a solution of thionyl chloride (3.875 mL, 53.122 mmol, 4.75 equiv) in water (20 mL), followed by addition of copper chloride (1.810 g, 13.509 mmol, 1.2 equiv) in water (15 mL). Reaction mixture was stirred at RT for 1 h and, after completion, extracted with ethyl acetate. The combined organic fraction was dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford crude l-bromonaphthalene-2-sulfonyl chloride, which was forwarded directly for the next step.
Step 2: l-((l-bromonaphthalen-2-yl)sulfonyl)-4-methoxypiperidine was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (23% yield over 2 steps), using 1- bromonaphthalene-2-sulfonyl chloride (2.0 g, 6.54 mmol, 1 equiv) and 4-methoxypiperidine (1.2 equiv) as starting materials.
LCMS (ESI+) m/z 383.9 [M+H]+
Step 3: 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)naphthalen-l-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (6% yield), using 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (200 mg, 0.66 mmol, 1 equiv) and l-((l-bromonaphthalen-2-yl) sulfonyl)-4-methoxypiperidine (1.2 equiv) as starting materials, K3PO4 (3.0 equiv) as base, Pd(dppf)CI2 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 480.3 [M+H]+
NMR (400 MHz, DMSO) δ 12.30 (s, 1H), 8.69 (dd, J = 7.1, 0.9 Hz, 1H), 8.24 (d, J = 8.8 Hz, 1H), 8.13 (d, J = 8.2 Hz, 1H), 8.01 (d, J = 8.9 Hz, 1H), 7.98 (s, 1H), 7.72 (ddd, J = 8.1, 6.8, 1.2 Hz, 1H), 7.61 - 7.53 (m, 2H), 7.44 (dd, J = 8.6, 1.1 Hz, 1H), 6.79 (dd, J = 7.1, 1.9 Hz, 1H), 3.77 - 3.62 (m, 2H), 3.27 - 3.19 (m, 1H), 3.16 (s, 3H), 3.19 - 3.10 (m, 1H), 3.15 - 3.02 (m, 1H), 2.87 - 2.77 (m, 1H), 2.80 - 2.71 (m, 1H), 1.79 - 1.65 (m, 2H), 1.47 - 1.31 (m, 2H).
Example 1-115: Synthesis of (2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)naphthalen-l-yl)pyrazolo[l,5- a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 89)
Step 1: To a stirred solution of 2-(5-(2-((4-methoxypiperidin-l-yl) sulfonyl) naphthalen-l-yl) pyrazolo[l,5- a] pyridin-3-yl) acetic acid (100 mg, 0.209 mmol, 1.0 equiv) in DMF (1 mL). were added KHCO3 (2 equiv) and chloromethyl pivalate (1.5 equiv) and the resulting solution was stirred at RT for 2 h. After completion, the reaction was quenched with water and extracted with ethyl acetate. The combined organic fractions were washed with water, brine, dried over Na2SO4 and concentrated. Crude mixture was purified by preparative HPLC to give pure (2-(5-(2-((4-methoxypiperidin-l-yl) sulfonyl) naphthalen-l-yl) pyrazolo[l,5- a] pyridin-3-yl) acetoxy) methyl pivalate (14 mg, 11% yield).
LCMS (ESI+) m/z 594.4 [M+H]+
XH NMR (400 MHz, DMSO) δ 8.71 (dd, J = 7.1, 0.9 Hz, 1H), 8.28 - 8.21 (m, 1H), 8.17 - 8.10 (m, 1H), 8.01 (d, J = 8.8 Hz, 1H), 7.99 (s, 1H), 7.73 (ddd, J = 8.1, 6.8, 1.2 Hz, 1H), 7.60 - 7.54 (m, 2H), 7.44 (dd, J = 8.6, 1.1 Hz, 1H), 6.82 (dd, J = 7.1, 1.9 Hz, 1H), 5.68 (s, 2H), 3.96 - 3.82 (m, 2H), 3.28 - 3.20 (m, 1H), 3.16 (s, 3H), 3.15 - 3.09 (m, 1H), 3.09 - 3.01 (m, 1H), 2.82 (ddd, J = 12.6, 8.5, 3.7 Hz, 1H), 2.75 (ddd, J = 12.6, 8.5, 4.1 Hz, 1H), 1.78 - 1.68 (m, 2H), 1.40 (dqd, J = 12.5, 8.3, 3.8 Hz, 2H), 1.00 (s, 9H).
Example 1-116: Synthesis of 2-(5-(2-((4-acetamidopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-
3-yl)acetic acid (Compound 92)
Step 1: 2-(5-(2-((4-acetamidopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (31% yield) using N-(l-((2-bromophenyl)sulfonyl)piperidin-4-yl)acetamide (150 mg, 0.415 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (172 mg, 0.415 mmol, 1 equiv) as starting materials, Pdll8 (0.1 equiv) as catalyst and Cs2CO3 (1.5 equiv) as a base.
N-(l-((2-bromophenyl)sulfonyl)piperidin-4-yl)acetamide was prepared as described in US2015259357A1.
LCMS (ESI+) m/z 457.2 [M+H]+
TH NMR (400 MHz, DMSO-d6) 6 8.59 (d, J = 7.2 Hz, 1H), 8.09 (s, 1H), 8.02 - 7.97 (m, 1H), 7.91 (s, 1H), 7.79 - 7.71 (m, 1H), 7.71 - 7.62 (m, 1H), 7.60 - 7.54 (m, 1H), 7.50 - 7.43 (m, 1H), 6.82 (dd, J= 7.2, 2.0 Hz, 1H), 3.62 (s, 2H), 3.55 - 3.50 (m, 1H), 3.21 - 3.13 (m, 2H), 2.60 - 2.52 (m, 2H), 1.74 (s, 3H), 1.58 - 1.49 (m, 2H), 1.16 - 1.03 (m, 2H).
Example 1-117: Synthesis of (2-(5-(5-amino-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 93)
Step 1: To the stirred solution of 7-bromo-5-nitroquinolin-8-ol (56 g, 208.1 mmol, 1 equiv) in 1,4 dioxane (1120 mL), were added DIPEA (53.79 g, 416.2 mmol, 2 equiv) and (4-methoxyphenyl)methanethiol (96.29 g, 624.3 mmol, 3 equiv) under nitrogen atmosphere at RT. After that, Xantphos (60.2 g, 104.0 mmol, 0.5 equiv) was added, followed by tris(dibenzylideneacetone)dipalladium(0) (19.05 g, 20.8 mmol, 0.1 equiv). The suspension was purged with nitrogen for 20 min and the reaction mixture was stirred at 120°C for 20 hours. After completion, the reaction mixture was concentrated under reduced pressure, redissolved in DCM, filtered through Celite® and concentrated under reduced pressure again. Crude mixture was purified by flash chromatography to give pure 7-((4-methoxybenzyl)thio)-5-nitroquinolin-8-ol (36.5 g 51% yield).
7-bromo-5-nitroquinolin-8-ol was was prepared as described in W02016154051A1.
LCMS (ESI+) m/z 343.2 [M+H]+
NMR (400 MHz, CDCI3): 6 9.26 (dd, J = 8.8, 1.2 Hz, 1H), 8.88 (dd, J = 4.0, 1.2 Hz, 1H), 8.53 (s, 1H), 7.71 - 7.68 (m, 1H), 7.25 (d, J = 8.8 Hz, 2H), 6.79 (d, J = 8.8 Hz, 2H), 4.26 (s, 2H), 3.76 (s, 3H).
Step 2: To a stirred solution of 7-((4-methoxybenzyl)thio)-5-nitroquinolin-8-ol (36.5 g , 106.6 mmol, 1 equiv) in MeCN (730 mL), cooled in an ice-water bath, were added acetic acid (54.75 mL) and water (54.75 mL). l,3-dichloro-5,5-dimethyl-hydantoin (42 g, 213.2 mmol, 2 equiv) was added and the resulting mixture was stirred at O°C for 2 hours. After completion, the reaction mixture was added to pre-stirred solution of 4-methoxypiperidine (31.92 g, 277.18 mmol, 2.6 equiv) and trimethylamine (59.87 mL, 426.4 mmol, 4 equiv) in DCM (912.5 mL) and stirred at RT for 16 hours. The reaction mixture was quenched with water and extracted with DCM. The combined organic fraction was washed with water, dried over anhydrous Na2SO4, filtered and concentrated to afford crude product. 7-((4-methoxypiperidin-l-yl)sulfonyl)-5- nitroquinolin-8-ol (14 g, 36% yield) was obtained by precipitation of the product in DCM and diethyl ether mixture (1:1), filtration and drying under reduced pressure.
LCMS (ESI+) m/z 368.2 [M+H]+
NMR (400 MHz, CDCI3): 6 9.40 (dd, J = 8.8, 1.2 Hz, 1H), 9.01 (dd, J = 4.4, 1.6 Hz, 1H), 9.00 (s, 1H), 7.90
- 7.87 (m, 1H), 3.48 - 3.36 (m, 5H), 3.34 (s, 3H), 2.01 - 1.87 (m, 2H), 1.80-1.74 (m, 2H).
Step 3: To a stirred solution of 7-((4-methoxypiperidin-l-yl)sulfonyl)-5-nitroquinolin-8-ol (20 g , 54.4 mmol, 1 equiv) in DCM (400 mL), cooled in an ice-water bath, was added triethylamine (11.3 mL, 81.7 mmol, 1.5 equiv) followed by dropwise addition of trifluoromethanesulfonic anhydride (18.4 g, 65.3 mmol, 1.2 equiv). The reaction mixture was stirred for 2 hours at RT and then quenched with water and extracted with DCM. The combined organic fraction was washed with washed with water, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. Crude mixture was purified by flash chromatography to give pure 7-((4-methoxypiperidin-l-yl)sulfonyl)-5-nitroquinolin-8-yl trifluoromethane sulfonate (10 g, 37% yield).
LCMS (ESI+) m/z 500.2 [M+H]+
TH-NMR (400 MHz, CDCI3): 6 9.20 (dd, J = 4.0, 1.6 Hz, 1H), 9.12 (dd, J = 8.8, 1.2 Hz, 1H), 8.88 (s, 1H), 7.92
- 7.88 (m, 1H), 3.40 - 3.38 (m, 3H), 3.31 - 3.26 (m, 5H), 1.90 - 1.81 (m, 4H).
Step 4: To the solution of 7-((4-methoxypiperidin-l-yl)sulfonyl)-5-nitroquinolin-8-yl trifluoromethanesulfonate (10 g, 20.0 mmol, 1 equiv) in an ice-cooled mixture of 1,2-dimethoxyethane (275 mL) and water (25 mL), were added (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- a]pyridin-3-yl)acetoxy)methyl pivalate (8.33 g, 20.0 mmol, 1 equiv) and potassium carbonate (5.5 g, 40.0 mmol, 2 equiv). The resulting mixture was bubbled with nitrogen for about 20 min and then Pd-118 (2.6 g, 4.0 mmol, 0.2 equiv) was added. Reaction mixture was degassed with nitrogen for another 20 minutes and stirred at RT for 16 h. After completion, the reaction was diluted with water and extracted with ethyl acetate. The combined organic fractions were dried over Na2SO4 and concentrated under reduced pressure. Crude mixture was purified by flash chromatography to give (2-(5-(7-((4-methoxypiperidin-l- yl)sulfonyl)-5-nitroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (6 g, 47% yield).
LCMS (ESI+) m/z 640.5 [M+H]+
TH-NMR (400 MHz, CDCI3): 6 9.07 - 9.02 (m, 3H), 8.53 (dd, J = 7.2, 0.8 Hz, 1H), 7.99 (s, 1H), 7.76 - 7.73 (m, 1H), 7.55 (d, J = 0.8 Hz, 1H), 6.74 (dd, J = 7.2, 2.0 Hz, 1H), 5.76 - 5.72 (m, 2H), 3.80 (s, 2H), 3.33 - 3.30 (m, 1H), 3.25 (s, 3H), 3.12 - 3.06 (m, 1H), 3.03 - 2.98 (m, 1H), 2.92 - 2.83 (m, 2H), 1.75 - 1.70 (m, 2H), 1.61 - 1.56 (m, 1H), 1.28 - 1.24 (m, 1H), 1.14 (s, 9H).
Step 5: To a stirred solution of (2-(5-(7-((4-methoxypiperidin-l-yl)sulfonyl)-5-nitroquinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (6 g, 9.38 mmol, 1 equiv) in THF (390 mL) was added sodium dithionite solution (8.16 g, 46.9 mmol) in water (240 mL). The reaction mixture was stirred at 55°C for 1 hour. After completion, the reaction was diluted with water and extracted with DCM and THF mixture (2:1), organic layers combined, dried over anhydrous MgSO4, filtered and concentrated under reduced pressure. Crude mixture was purified by flash chromatography to give (2-(5-(5-amino-7- ((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (3.7 g, 65% yield).
LCMS (ESI+) m/z 610.5 [M+H]+
TH-NMR (400 MHz, DMSO-d6): 6 8.78 (dd, J = 4.0, 1.6 Hz, 1H), 8.65 (dd, J = 8.8, 1.6 Hz, 1H), 8.54 (d, J = 7.2 Hz, 1H), 7.90 (s, 1H), 7.55 - 7.51 (m, 1H), 7.38 (s, 1H), 7.26 (s, 1H), 6.70 - 6.63 (m, 3H), 5.68 (s, 2H), 3.86 - 3.81 (m, 2H), 3.24 - 3.22 (m, 1H), 3.16 (s, 3H), 3.09 - 3.02 (m, 2H), 2.77 - 2.67 (m, 2H), 1.71 - 1.67 (m, 2H), 1.42 - 1.38 (m, 2H), 1.01 (s, 9H).
Example 1-118: Synthesis of 2-(5-(2-((4-methylpiperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 94) Step 1: 2-(5-(2-((4-methylpiperazin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (41% yield) using l-((2-bromophenyl)sulfonyl)-4-methylpiperazine (100 mg, 0.313 mmol, 1 equiv) and (2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (130.4 mg, 0.313 mmol, 1 equiv) as starting materials, Pd-118 (0.1 equiv) as catalyst and Cs2CO3 (2 equiv) as a base. l-((2-bromophenyl)sulfonyl)-4-methylpiperazine was prepared as described in J. Med. Chem. 2012, 55, 21, 9107
LCMS (ESI+) m/z 415.2 [M+H]+
NMR (401 MHz, DMSO) δ 8.59 (d, J = 7.2 Hz, 1H), 8.02 - 7.96 (m, 1H), 7.92 (s, 1H), 7.81 - 7.72 (m, 1H), 7.72 - 7.64 (m, 1H), 7.60 - 7.58 (m, 1H), 7.51 - 7.45 (m, 1H), 6.82 (dd, J= 7.2, 1.9 Hz, 1H), 3.64 - 3.60 (m, 2H), 2.84 - 2.77 (m, 4H), 2.13 - 2.07 (m, 4H), 2.04 (s, 3H).
Example 1-119: Synthesis of 2-(5-(2-((4-aminopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid (Compound 95)
Step 1: 2-(5-(2-((4-aminopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above using tert-butyl (l-((2-bromophenyl)sulfonyl)piperidin-4-yl)carbamate (200 mg, 0.48 mmol, 1 equiv) and (2-(5- (4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (198 mg, 0.48 mmol, 1 equiv) as starting materials, Pd-118 (0.1 equiv) as catalyst and Cs2CO3 (3 equiv) as a base. After completion, the volatiles were removed under reduced pressure and the resulting crude was forwarded directly into the next step. tert-butyl (l-((2-bromophenyl)sulfonyl)piperidin-4-yl)carbamate was prepared as described in
US2015259357A1.
LCMS (ESI+) m/z 629.3 [M+H]+ Step 2: 2-(5-(2-((4-((tert-butoxycarbonyl)amino)piperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above using 2-(5-(2-((4-aminopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (350 mg, 0.56 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 515.3 [M+H]+
Step 3: 2-(5-(2-((4-aminopiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (41% yield over 3 steps), using 2-(5-(2-((4-((tert-butoxycarbonyl)amino)piperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (300 mg, 0.58 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 415.2 [M+H]+
NMR (400 MHz, DMSO) δ 8.53 (d, J = 7.2 Hz, 1H), 8.10 (dd, J = 1.4, 7.8 Hz, 1H), 7.81 (s, 1H), 7.77 (td, J= 1.5 Hz, 7.7 Hz, 1H), 7.67 (td, J= 1.4 Hz, 7.7 Hz, 1H) 7.59 - 7.55 (m, 1H), 7.47 (dd, J= 1.3, 7.5 Hz, 1H), 6.81 (dd, J= 2.0, 7.2 Hz, 1H), 3.50 (s, 2H), 3.35 - 3.31 (m, 2H), 3.00 - 2.89 (m, 2H), 2.82 - 2.73 (m, 1H), 1.54 - 1.39 (m, 2H), 1.31 - 1.19 (m, 2H).
Example 1-120: Synthesis of (2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (Compound 1035) and 2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1034)
Step 1: To a stirred solution of tert-butyl 4-hydroxypiperidine-l-carboxylate [commercial] (1.0 g, 4.94 mmol, 1 equiv) in DMF (20 mL), cooled in an ice-water bath, was added NaH (1.2 g, 49.44 mmol, 10 equiv, 60% suspension in mineral oil) followed by ((oxybis(ethane-2,l-diyl))bis(oxy))bis(ethane-2,l-diyl) bis(4- methylbenzenesulfonate) [commercial] (2.48 g, 4.94 mmol, 1 equiv). The reaction mixture was stirred at RT for 16 h, quenched with cold NH4CI solution and extracted with ethyl acetate. Combined organic fractions were dried over Na2SO4 and evaporated. tert-butyl 4-(2-(2-(2-(2- (tosyloxy)ethoxy)ethoxy)ethoxy)ethoxy)piperidine-l-carboxylate (500 mg, 0.94 mmol, 19% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 532.2 [M+H]+
Step 2: 2-(2-(2-(2-(piperidin-4-yloxy)ethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate trifluoroacetate was synthesized was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above using tert-butyl 4-(2-(2-(2-(2- (tosyloxy)ethoxy)ethoxy)ethoxy)ethoxy)piperidine-l-carboxylate (500 mg, 0.94 mmol, 1 equiv) as a starting material. After completion, the volatiles were removed under reduced pressure. The residue was three times co-evaporated with toluene and then lyophilized to afford crude material, which was directly forwarded into the next step.
LCMS (ESI+) m/z 431.8 [M+H]+ Step 3: 2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (10% yield over 2 steps), using 8-bromoquinoline-7-sulfonyl chloride (220 mg, 0.72 mmol, 1 equiv) and 2-(2-(2-(2-(piperidin-4-yloxy)ethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate trifluoroacetate (464 mg, 1.07 mmol, 1.5 equiv) as starting materials.
TH NMR (400 MHz, DMSO-d6) 6 9.16 (dd, J = 4.1, 1.7 Hz, 1H), 8.57 (dd, J = 8.3, 1.8 Hz, 1H), 8.24 - 8.19 (m, 1H), 8.16 - 8.11 (m, 1H), 7.81 - 7.75 (m, 3H), 7.50 - 7.44 (m, 2H), 4.14 -4.05 (m, 2H), 3.58 - 3.39 (m, 16H), 3.27 - 3.20 (m, 1H), 3.03 - 2.90 (m, 2H), 2.40 (s, 3H), 1.87 - 1.77 (m, 2H), 1.53 - 1.43 (m, 2H).
Step 4: To a stirred solution of 5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(4-(piperazin-l- yl)phenyl)pyrimidine-2,4-diamine (76 mg, 0.157 mmol, 1.1 equiv) in DMF (1 mL), were added 2-(2-(2-(2- ((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate (100 mg, 0.143 mmol, 1 equiv) and K2CO3 (59 mg, 0.43 mmol, 3 equiv). The reaction was stirred at 80 °C for 4h and after completion, the resulting mixture was quenched with cold water, extracted with ethyl acetate and the volatiles were removed under reduced pressure. N2-(4-(4-(2- (2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)piperazin-l- yl)phenyl)-5-chloro-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (80 mg, 55% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 1017.4 [M+H]+
TH NMR (400 MHz, DMSO-d6) 6 9.48 (s, 1H), 9.35 - 9.26 (m, 1H), 9.17 - 8.13 (m, 1H), 8.70 - 8.60 (m, 1H), 8.58 - 8.52 (m, 1H), 8.25 - 8.17 (m, 2H), 8.15 - 8.10 (m, 1H), 7.95 (s, 1H), 7.86 - 7.70 (m, 3H), 7.49 - 7.32 (m, 2H), 6.95 - 6.78 (m, 2H), 3.54 - 3.39 (m, 22H), 3.18 - 3.01 (m, 6H), 2.59 - 2.54 (m, 2H), 1.88 - 1.76 (m, 2H), 1.53 - 1.43 (m, 2H), 1.25 - 1.21 (m, 6H).
Step 5: (2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate and 2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid were synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using N2-(4-(4-(2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)piperazin-l-yl)phenyl)-5-chloro-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (80 mg, 0.08 mmol, 1 equiv) and (2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (1.25 equiv) as starting materials, Cs2CO3 (2 equiv) as base and Pdll8 as catalyst (0.1 equiv). After completion the products were separated using preparative HPLC.
(2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1035) (4 mg, 4% yield)
LCMS (ESI+) m/z 1225.6, 1227.4 [M+H]+
NMR (400 MHz, DMSO-d6) 6 9.48 (s, 1H), 9.30 (s, 1H), 8.94 - 8.89 (m, 1H), 8.62 (d, J = 7.1 Hz, 1H), 8.58
- 8.51 (m, 2H), 8.32 - 8.20 (m, 2H), 8.11 (d, J = 8.8 Hz, 1H), 7.94 (s, 1H), 7.86 - 7.80 (m, 1H), 7.75 - 7.66 (m, 2H), 7.48 (s, 1H), 7.45 - 7.33 (m, 3H), 6.84 (d, J = 8.7 Hz, 2H), 6.78 (dd, J = 1.8, 7.0 Hz, 1H), 5.68 (s, 2H), 3.89 - 3.83 (m, 2H), 3.55 - 3.43 (m, 14H), 3.40 - 3.27 (m, 4H), 3.21 - 3.00 (m, 7H), 2.82 - 2.65 (m, 3H), 2.57 - 2.54 (m, 2H), 1.75 - 1.66 (m, 2H), 1.41 - 1.33 (m, 2H), 1.16 (d, J = 6.9 Hz, 6H), 0.99 (s, 9H).
2-(5-(7-((4-(2-(2-(2-(2-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)ethoxy)ethoxy)ethoxy)ethoxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1034) (6 mg, 6% yield)
LCMS (ESI+) m/z 1111.5, 1113.4 [M+H]+
TH NMR (400 MHz, DMSO-d6) 6 9.48 (s, 1H), 9.33 (s, 1H), 8.94 - 8.89 (m, 1H), 8.67 - 8.62 (m, 1H), 8.60 - 8.45 (m, 2H), 8.33 - 8.24 (m, 1H), 8.22 (s, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.91 (s, 1H), 7.83 (d, J = 8.1 Hz, 1H), 7.77 - 7.65 (m, 2H), 7.47 (s, 1H), 7.44 - 7.32 (m, 3H), 6.84 (d, J = 8.8 Hz, 2H), 6.71 (d, J = 7.1 Hz, 1H), 3.55
- 3.43 (m, 18H), 3.22 - 3.00 (m, 9H), 2.83 - 2.71 (m, 3H), 2.58 - 2.52 (m, 2H), 1.76 - 1.66 (m, 2H), 1.43 - 1.30 (m, 2H), 1.16 (d, J = 6.8 Hz, 6H).
Example 1-121: Synthesis of (2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)-3,6,9,12- tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1037) and 2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)-3,6,9,12- tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1036)
Step 1: To a stirred solution of tert-butyl 4-hydroxypiperidine-l-carboxylate [commercial] (264 mg, 1.83 mmol, 1 equiv) in DMF (20 mL), cooled in an ice-water bath, was added NaH (10 equiv, 60% suspension in mineral oil) followed by 3,6,9,12-tetraoxatetradecane-l,14-diyl bis(4-methylbenzenesulfonate) [commercial] (1 g, 1.83 mmol, 1 equiv). The reaction mixture was stirred at RT for 16 h, quenched with cold NH4CI solution and extracted with ethyl acetate. Combined organic fractions were dried over Na2SO4 and evaporated to afford crude tert-butyl 4-((14-(tosyloxy)-3,6,9,12-tetraoxatetradecyl)oxy)piperidine-l- carboxylate, which was directly forwarded into the next step.
LCMS (ESI+) m/z 576.4 [M+H]+
Step 2: 14-(piperidin-4-yloxy)-3,6,9,12-tetraoxatetradecyl 4-methylbenzenesulfonate trifluoroacetate was synthesized was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above using tert-butyl 4-((14-(tosyloxy)-3,6,9,12-tetraoxatetradecyl)oxy)piperidine-l- carboxylate (250 mg, 0.43 mmol, 1 equiv) as a starting material. After completion, the volatiles were removed under reduced pressure. The residue was three times co-evaporated with toluene and then lyophilized to afford crude material, which was directly forwarded into the next step.
LCMS (ESI+) m/z 476.1 [M+H]+ Step 3: 14-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl 4- methylbenzenesulfonate was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (9% yield over 3 steps), using 8-bromoquinoline-7-sulfonyl chloride (220 mg, 0.72 mmol, 1 equiv) and 14-(piperidin-4-yloxy)-3,6,9,12-tetraoxatetradecyl 4-methylbenzenesulfonate trifluoroacetate (464 mg, 0.97 mmol, 1.35 equiv) as starting materials.
LCMS (ESI+) m/z 745.1 [M+H]+
Step 4: To a stirred solution of 5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(4-(piperazin-l- yl)phenyl)pyrimidine-2,4-diamine (180 mg, 0.37 mmol, 1.1 equiv) in DMF (2 mL), were added 14-((l-((8- bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl 4-methylbenzenesulfonate (250 mg, 0.34 mmol, 1 equiv) and K2CO3 (141 mg, 1.02 mmol, 3 equiv). The reaction was stirred at 80 °C for 4h and after completion, the resulting mixture was quenched with cold water, extracted with ethyl acetate and the volatiles were removed under reduced pressure to afford crude N2-(4-(4-(14-((l-((8- bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12-tetraoxatetradecyl)piperazin-l-yl)phenyl)-5- chloro-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine, which was directly forwarded into the next step.
LCMS (ESI+) m/z 1061.3 [M+H]+
Step 5: (2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)-3,6,9,12-tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate and 2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)-3,6,9,12- tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid were synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above, using N2-(4-(4-(14-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)-3,6,9,12- tetraoxatetradecyl)piperazin-l-yl)phenyl)-5-chloro-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4- diamine (80 mg, 0.08 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5- a]pyridin-3-yl)acetoxy)methyl pivalate (1.2 equiv) as starting materials, Cs2CO3 (2 equiv) as base and Pdll8 as catalyst (0.1 equiv). After completion the products were separated using preparative HPLC. (2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)-3,6,9,12-tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1037) (1.5 mg, 0.5% yield)
LCMS (ESI+) m/z 1267.7, 1269.1 [M+H]+
NMR (400 MHz, DMSO-d6) 6 9.51 - 9.46 (m, 1H), 9.32 - 9.28 (m, 1H), 8.95 - 8.89 (m, 1H), 8.62 (d, J = 7.2 Hz, 1H), 8.59 - 8.52 (m, 1H), 8.28 (d, J = 8.8 Hz, 1H), 8.25 - 8.20 (m, 1H), 8.15 - 8.05 (m, 1H), 7.94 (s, 1H), 7.85 - 7.80 (m, 1H), 7.75 - 7.66 (m, 2H), 7.51 - 7.46 (m, 1H), 7.45 - 7.39 (m, 2H), 7.39 - 7.32 (m, 1H), 6.87 - 6.81 (m, 3H), 6.81 - 6.75 (m, 1H), 5.68 (s, 2H), 3.89 - 3.84 (m, 2H), 3.56 - 3.39 (m, 22H), 3.29 - 3.26 (m, 4H), 3.19 - 3.12 (m, 1H), 3.07 - 3.00 (m, 5H), 2.57 - 2.53 (m, 2H), 1.75 - 1.65 (m, 2H), 1.39 - 1.32 (m, 2H), 1.16 (d, J = 6.6 Hz, 6H), 1.00 (s, 9H).
2-(5-(7-((4-((14-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)-3,6,9,12-tetraoxatetradecyl)oxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1036) (7.2 mg, 2.6% yield)
LCMS (ESI+) m/z 1155.8, 1157.6 [M+H]+
XH NMR (400 MHz, DMSO-d6) 6 9.52 - 9.41 (m, 1H), 9.35 - 9.28 (m, 1H), 8.93 - 8.82 (m, 1H), 8.62 - 8.57 (m, 1H), 8.54 (d, J = 7.2 Hz, 1H), 8.49 (d, J = 8.5 Hz, 1H), 8.24 (d, J = 8.8 Hz, 1H), 8.17 (s, 1H), 8.07 (d, J = 8.8 Hz, 1H), 7.91 (s, 1H), 7.79 (d, J = 8.0 Hz, 1H), 7.73 - 7.57 (m, 2H), 7.42 (s, 1H), 7.40 - 7.30 (m, 3H), 6.82 (d, J = 8.6 Hz, 2H), 6.71 (d, J = 7.2 Hz, 1H), 3.62 - 3.58 (m, 3H) 3.55 - 3.29 (m, 23H), 3.19- 3.10 (m, 1H), 3.08 - 2.99 (m, 5H), 2.80 - 2.64 (m, 3H), 2.57 - 2.53 (m, 2H), 1.74 - 1.61 (m, 2H), 1.39 - 1.24 (m, 2H), 1.13 (d, J = 6.9 Hz, 6H).
Example 1-122: Synthesis of (S)-(2-(5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l- yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1039) and (S)-2- (5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)- 2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid (Compound 1038)
Step 1: l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-ol was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (81% yield) using 8-bromoquinoline-7-sulfonyl chloride (255 mg, 0.832 mmol, 1 equiv) and piperidin-4-ol (Cpd-1) (168 mg, 1.664 mmol, 2 equiv) as starting materials.
LCMS (ESI+) m/z 371.2 [M+H]+
Step 2: To a stirred solution of l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-ol (200 mg, 0.539 mmol, 1 equiv) in DMF (8 mL), cooled in an ice-water bath, was added NaH (128 mg, 3.232 mmol, 6 equiv, 60% suspension in mineral oil) followed by tert-butyl (2-(2-(2-(2-bromoethoxy)ethoxy)ethoxy)ethyl)carbamate (1.9 g, 5.387 mmol, 10 equiv). The reaction mixture was stirred at RT for 6 h, quenched with NH4CI and extracted with ethyl acetate. Combined organic fractions were dried over Na2SO4 and evaporated, tertbutyl (2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (190 mg, 0.294 mmol, 54% yield) was purified by flash column chromatography.
LCMS (ESI+) m/z 646.2 [M+H]+
Step 3: 2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethan-l- amine hydrochloride was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5 using tert-butyl (2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)carbamate (190 mg, 0.294 mmol) as starting material. After completion the volatiles were removed under reduced pressure and the crude product was used directly in the next step.
LCMS (ESI+) m/z 545.9 [M+H]+
Step 4: To a solution of (S)-2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetic acid (75 mg, 0.187 mmol, 1 equiv) in DMF (4 mL) were added HATU (106 mg, 0.281 mmol, 1.5 equiv) and DIPEA (0.1 mL, 0.561 mmol, 3 equiv) and the resulting mixture was stirred at RT for 15 min. 2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethan-l-amine hydrochloride (204 mg, 0.374 mmol, 2 equiv) in DMF (1 mL) was added and the reaction mixture was stirred at RT for 2 h. After completion, the reaction was diluted with water and extracted with ethyl acetate. The combined organic fractions were dried over Na2SO4 and evaporated. (S)-N-(2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4- yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide (120 mg, 0.129 mmol, 44% yield over two steps) was purified by flash column chromatography.
LCMS (ESI+) m/z 928.7, 930.3 [M+H]+
Step 5: (S)-(2-(5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate and (S)-2-(5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9- trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan- 14-yl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid were synthesized using the general procedure shown in Reaction Scheme 3 and Example Example Method 3, above using (S)-N-(2-(2-(2-(2-((l-((8-bromoquinolin-7-yl)sulfonyl)piperidin-4-yl)oxy)ethoxy)ethoxy)ethoxy)ethyl)-2-(4- (4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide (120 mg, 0.129 mmol, 1 equiv) and (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (1.3 equiv) as starting materials, Cs2CO3 (2 equiv) as base, Pdll8 as catalyst (0.1 equiv). (S)-(2-(5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1039) (13 mg, 9% yield)
LCMS (ESI+) m/z 1138.5, 1140.4 [M+H]+
NMR (400 MHz, DMSO-d6) 6 8.92 - 8.86 (m, 1H), 8.62 (d, J = 7.2 Hz, 1H), 8.56 (dd, J = 1.8, 8.4 Hz, 1H), 8.31 - 8.22 (m, 2H), 8.09 (d, J = 8.8 Hz, 1H), 7.94 (s, 1H), 7.70 (dd, J = 4.1, 8.4 Hz, 1H), 7.50 - 7.45 (m, 3H), 7.44 - 7.38 (m, 2H), 6.78 (dd, J = 1.8, 7.1 Hz, 1H), 5.68 (s, 2H), 4.50 (dd, J = 6.0, 8.1 Hz, 1H), 3.93 - 3.79 (m, 2H), 3.54 - 3.39 (m, 14H), 3.27 - 3.14 (m, 5H), 3.11 - 3.02 (m, 1H), 2.81 - 2.68 (m, 2H), 2.58 (s, 3H), 2.40 (s, 3H), 1.76 - 1.66 (m, 2H), 1.61 (s, 3H), 1.42 - 1.29 (m, 2H), 0.99 (s, 9H).
(S)-2-(5-(7-((4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin- 6-yl)-2-oxo-6,9,12-trioxa-3-azatetradecan-14-yl)oxy)piperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid (Compound 1038) (10 mg, 8% yield)
LCMS (ESI+) m/z 1024.7, 1026.6 [M+H]+
XH NMR (401 MHz, DMSO-d6) 6 8.94 - 8.88 (m, 1H), 8.60 (d, J = 7.2, 1H), 8.55 (d, J= 8.3 Hz, 1H), 8.32 - 8.22 (m, 2H), 8.11 (d, J = 8.8 Hz, 1H), 7.94 (s, 1H), 7.73 - 7.65 (m, 1H), 7.51 - 7.35 (m, 3H), 7.44 - 7.38 (m, 2H), 6.75 (d, J = 7.2 Hz, 1H), 4.50 (t, J = 7.1 Hz, 1H), 3.70 - 3.64 (m, 2H), 3.51 - 3.37 (m, 14H), 3.36 - 3.01 (m, 7H), 2.81 - 2.63 (m, 2H), 2.58 (s, 3H), 2.37 (s, 3H), 1.74 - 1.62 (m, 2H), 1.58 (s, 3H), 1.41 - 1.26 (m, 2H).
Example 1-123: Synthesis of (2-(5-(5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH-benzo[d]imidazol-
4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 97)
Step 1: To a stirred solution of 2-bromo-3-fluoro-6-nitroaniline (3.0 g, 12.82 mmol, 1 equiv) in THF (30 mL), were added TEA (3.5 mL, 25.6 mmol, 2 equiv), DMAP (0.156 g, 1.2 mmol, 0.1 equiv) and di-tert-butyl dicarbonate (4.41 mL, 19.23 mmol, 1.5 equiv) and the resulting solution was stirred at 80 °C for 16h. After completion, the reaction was quenched with water and extracted with with ethyl acetate. The combined organic fractions were dried over Na2SO4 and concentrated, tert-butyl (2-bromo-3-fluoro-6- nitrophenyl)(tert-butoxycarbonyl)carbamate (4.4 g, 79% yield) was purified by flash column chromatography.
NMR (400 MHz, DMSO-d6): 6 8.30 (dd, J = 9.3, 5.3 Hz, 1H), 7.76 (t, J = 8.5 Hz, 1H), 1.32 (s, 18H).
Step 2: To a stirred solution of tert-butyl (2-bromo-3-fluoro-6-nitrophenyl)(tert- butoxycarbonyl)carbamate (4.4 g, 10.14 mmol, 1 equiv) in DMF (20 mL), were added (4- methoxyphenyl)methanethiol (2.6 g, 15.21 mmol, 1.5 equiv) and K2CO3 (2.8g, 20.3 mmol) and reaction mixture was stirred at RT for 16 h. After completion, the reaction was quenched with ice cold water and extracted with ethyl acetate. The combined organic fractions were washed with ice cold water, dried over Na2SO4 and concentrated under reduced pressure, tert-butyl (2-bromo-3-((4-methoxybenzyl)thio)-6- nitrophenyl)(tert-butoxycarbonyl)carbamate (5.0 g, 87% yield) was purified by flash column chromatography.
NMR (400 MHz, DMSO-d6): 6 8.15 (d, J = 8.8 Hz, 1H), 7.65 (d, J = 9.0 Hz, 1H), 7.43 - 7.36 (m, 2H), 6.95 - 6.88 (m, 2H), 4.43 (s, 2H), 3.74 (s, 3H), 1.31 (s, 18H).
Step 3: To a stirred solution of tert-butyl (2-bromo-3-((4-methoxybenzyl)thio)-6-nitrophenyl)(tert- butoxycarbonyl)carbamate (500 mg, 1.09 mmol, 1 equiv) in ethanol (20 mL) and water (5 mL), were added NH4CI (150 mg, 2.72 mmol, 2.5 equiv), iron powder (150 mg, 2.72 mmol, 2.5 equiv) and the reaction mixture was stirred at 80°C for 6 h. After completion, the reaction mixture was filtered through celite and concentrated under reduced pressure to remove the excess solvent. The crude mixture was extracted with ethyl acetate, the organic layer was separated, dried over anhydrous Na2SO4 and concentrated under reduced pressure, tert-butyl (6-amino-2-bromo-3-((4-methoxybenzyl)thio)phenyl)carbamate (240 mg, 62% yield) was purified by flash column chromatography.
'H NMR (400 MHz, DMSO-d6): 6 8.25 (s, 1H), 7.23 - 7.14 (m, 2H), 7.06 - 7.00 (m, 1H), 6.87 - 6.80 (m, 2H), 6.65 - 6.58 (m, 1H), 5.16 (s, 2H), 3.98 (s, 2H), 3.72 (s, 3H), 1.39 (s, 9H).
Step 4: To an ice-cold solution of tert-butyl (6-amino-2-bromo-3-((4- methoxybenzyl)thio)phenyl)carbamate (1.0 g, 2.28 mmol, 1 equiv) in dioxane (3 mL) was added 4 M HCI in dioxane (6 mL) and the reaction mixture was allowed to stirred for 2 h under nitrogen. After consumption of the starting material the reaction mixture was evaporated under reduced pressure and the residue was triturated with ether to afford crude product which was forwarded directly for the next step.
LCMS (ESI+) m/z 339.2 [M+H]+
Step 5: To a stirred solution of 3-bromo-4-((4-methoxybenzyl)thio)benzene-l,2-diamine (450 mg, 1.32 mmol, 1 equiv) in THF (10 mL) were added triethyl orthoformate (0.532 ml, 3.2mmol) and p- Toluenesulfonic acid (23 mg, 0.13 mmol, 0.1 equiv) and the reaction mixture was stirred at 60°C for 24 h under nitrogen. After completion, the reaction mixture was evaporated under reduced pressure and the residue was triturated with diethyl ether and pentane to afford crude 4-bromo-5-((4- methoxybenzyl)thio)-lH-benzo[d]imidazole which was forwarded directly for the next step.
LCMS (ESI+) m/z 349.1 [M+H]+
Step 6: To a solution of 4-bromo-5-((4-methoxybenzyl)thio)-lH-benzo[d]imidazole (570 mg, 1.63 mmol, 1 equiv) in DMF (10 mL) was added sodium hydride (130 mg, 1.36 mmol, 0.83 equiv) and reaction was stirred at 0°C under nitrogen. After 30 min, methyl iodide (0.305mL, 4.896 mmol, 3 equiv) was added and the reaction mixture was allowed to stir at RT for 16 h under nitrogen. After completion, the mixture was quenched with water and extracted with ethyl acetate. The combined organic fractions were dried over anhydrous Na2SO4 and evaporated under reduced pressure. 4-bromo-5-((4-methoxybenzyl)thio)-l- methyl-lH-benzo[d]imidazole (300 mg, 0.825 mmol, 35% yield over three steps) was purified by flash column chromatography.
LCMS (ESI+) m/z 362.7 [M+H]+
NMR (400 MHz, DMSO-d6): 6 8.23 (s, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.34 (d, J = 8.4 Hz, 1H), 7.22 (d, J = 8.4 Hz, 2H), 6.81 (d, J = 8.5 Hz, 2H), 4.17 (s, 2H), 3.80 (s, 3H), 3.68 (s, 3H).
Step 7: To a stirred solution of 4-bromo-5-((4-methoxybenzyl)thio)-l-methyl-lH-benzo[d]imidazole (500 mg, 1.38 mmol, 1 equiv) in ACN (20 mL), were added AcOH (10 mL), water (10 mL) and the solution was cooled to 0°C. After 10 min, l,3-dichloro-5,5-dimethyl-hydantoin (540 mg, 2.76 mmol, 2 equiv) was added and the reaction was stirred at RT for 1 h. After completion, the reaction was quenched with water and extracted with DCM. The combined organic fractions were washed with brine, dried over Na2SO4 and evaporated to afford crude 4-bromo-l-methyl-lH-benzo[d]imidazole-5-sulfonyl chloride, which was forwarded directly for the next step.
Step 8: 4-bromo-5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH-benzo[d]imidazole was synthesized using the general procedure shown in Reaction Scheme 8 and Example Method 8, above (68% yield over two steps), using 4-bromo-l-methyl-lH-benzo[d]imidazole-5-sulfonyl chloride (500 mg, 1.62 mmol, 1 equiv) and 4-methoxypiperidine (1.5 equiv) as starting materials.
LCMS (ESI+) m/z 388.2, 390.2 [M+H] Step 9: (2-(5-(5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH-benzo[d]imidazol-4-yl)pyrazolo[l,5- a]pyridin-3-yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Example Method 3, above (40% yield), using 4-bromo-5-((4-methoxypiperidin-l- yl)sulfonyl)-l-methyl-lH-benzo[d]imidazole (130 mg, 0.335 mmol, 1 equiv) and (2-(5-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (3 equiv) as starting materials, Cs2CO3 (2.0 equiv) as base, Pdll8 as catalyst (0.1 equiv).
LCMS (ESI+) m/z 598.3 [M+H]+
TH NMR (400 MHz, DMSO) δ 8.63 (d, J = 7.1 Hz, 1H), 8.33 (s, 1H), 7.97 - 7.82 (m, 3H), 7.54 (s, 1H), 6.81 (dd, J = 1.9, 7.1 Hz, 1H), 5.69 (s, 2H), 3.92 (s, 3H), 3.88 (s, 2H), 3.21 - 3.15 (m, 1H), 3.13 (s, 3H), 3.02 - 2.93 (m, 2H), 2.73 - 2.62 (m, 2H), 1.70 - 1.59 (m, 2H), 1.38 - 1.17 (m, 2H), 1.02 (s, 9H).
Example 1-124: Synthesis of 2-(5-(5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH-benzo[d]imidazol-
4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 96)
Step 1: 2-(5-(5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH-benzo[d]imidazol-4-yl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (12% yield), using (2-(5-(5-((4-methoxypiperidin-l-yl)sulfonyl)-l-methyl-lH- benzo[d]imidazol-4-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (40 mg, 0.067 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 484.2 [M+H]+
TH NMR (400 MHz, DMSO) δ 12.87 - 11.82 (m, 1H), 8.62 (d, J = 7.1 Hz, 1H), 8.33 (s, 1H), 7.94 - 7.81 (m, 3H), 7.52 - 7.48 (m, 1H), 6.77 (dd, J = 1.9, 7.2 Hz, 1H), 3.89 (s, 3H), 3.68 - 3.64 (m, 2H), 3.21 - 3.16 (m, 1H), 3.13 (s, 3H), 3.01 - 2.96 (m, 2H), 2.72 - 2.59 (m, 2H), 1.65 - 1.60 (m, 2H), 1.36 - 1.26 (m, 2H). Example 1-125: Synthesis of 2-(5-{5-amino-7-[(4-methoxypiperidin-l-yl)sulfonyl]quinolin-8- yl}pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 91 ABX-555)
Step 1: 2-(5-{5-amino-7-[(4-methoxypiperidin-l-yl)sulfonyl]quinolin-8-yl}pyrazolo[l,5-a]pyridin-3- yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (74% yield), using (2-(5-(5-amino-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (25.0 mg, 0.041 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 496.15 [M+H]+
NMR (500 MHz, DMSO) δ 12.23 (s, 1H), 8.78 (dd, J = 4.1, 1.7 Hz, 1H), 8.65 (dd, J = 8.5, 1.7 Hz, 1H), 8.52 (dd, J = 7.2, 1.0 Hz, 1H), 7.89 (s, 1H), 7.53 (dd, J = 8.6, 4.1 Hz, 1H), 7.39 - 7.35 (m, 1H), 7.27 (s, 1H), 6.65 (dd, J = 7.1, 1.9 Hz, 1H), 6.62 - 6.60 (m, 2H), 3.71 - 3.59 (m, 2H), 3.26 - 3.19 (m, 1H), 3.16 (s, 3H), 3.15 - 3.00 (m, 2H), 2.75 (dddd, J = 20.6, 12.2, 8.5, 3.4 Hz, 2H), 1.74 - 1.66 (m, 2H), 1.37 (ddtd, J = 25.1, 12.3, 8.1, 3.7 Hz, 2H).
Example 1-126: Synthesis of (S)-(2-(5-(5-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)octanamido)-7-((4-methoxypiperidin-l- yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1027) Step 1: To a solution of (S)-8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamido)octanoic acid (40 mg, 0.074 mmol, 1 equiv) in DMF (4 mL) were added HATU (1.5 equiv) and DIPEA (3 equiv) and the resulting mixture was stirred at RT for 15 min. (2-(5-(5- amino-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (1.05 equiv) was added and the reaction mixture was stirred at RT for 20 h. After completion, the reaction mixture was concentrated under reduced pressure and purified by preparative HPLC to give pure (S)-(2-(5-(5-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin- 6-yl)acetamido)octanamido)-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (14 mg, 0.012 mmol, 17% yield).
(S)-8-(2-(4-(4-Chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6- yl)acetamido)octanoic acid was prepared as described in W02017197051A1.
LCMS (ESI+) m/z 1133.35 [M+H]+
NMR (500 MHz, DMSO) δ 10.30 (s, 1H), 8.91 (dd, J = 4.1, 1.7 Hz, 1H), 8.69 - 8.64 (m, 1H), 8.61 (dd, J = 7.1, 1.0 Hz, 1H), 8.44 (s, 1H), 8.16 (t, J = 5.7 Hz, 1H), 7.94 (s, 1H), 7.72 (dd, J = 8.6, 4.1 Hz, 1H), 7.51 - 7.46 (m, 3H), 7.46 - 7.39 (m, 2H), 6.77 (dd, J = 7.1, 1.9 Hz, 1H), 5.68 (s, 2H), 4.51 (dd, J = 8.1, 6.0 Hz, 1H), 3.92 - 3.80 (m, 2H), 3.27 - 3.18 (m, 2H), 3.16 (s, 3H), 3.20 - 3.05 (m, 4H), 2.86 - 2.73 (m, 2H), 2.59 (s, 3H), 2.61 - 2.52 (m, 3H), 2.41 - 2.39 (m, 3H), 1.73 - 1.68 (m, 4H), 1.63 - 1.61 (m, 3H), 1.47 (t, J = 6.8 Hz, 2H), 1.40 - 1.34 (m, 8H), 1.00 (s, 9H).
Example 1-127: Synthesis of (S)-2-(5-(5-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)octanamido)-7-((4-methoxypiperidin-l- yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1026)
Step 1: (S)-2-(5-(5-(8-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamido)octanamido)-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8- yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (40% yield), using (S)-(2-(5-(5-(8-(2-(4-(4-chlorophenyl)-2,3,9- trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamido)octanamido)-7-((4- methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (8.0 mg, 0.007 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1019.30 [M+H]+
TH NMR (500 MHz, DMSO) δ 12.33 (s, 1H), 10.30 (s, 1H), 8.91 (dd, J = 4.0, 1.7 Hz, 1H), 8.70 - 8.63 (m, 1H), 8.61 - 8.56 (m, 1H), 8.45 (s, 1H), 8.17 (t, J = 5.7 Hz, 1H), 7.93 (s, 1H), 7.72 (dd, J = 8.6, 4.0 Hz, 1H), 7.50 - 7.46 (m, 3H), 7.45 - 7.39 (m, 2H), 6.74 (dd, J = 7.1, 1.9 Hz, 1H), 4.51 (dd, J = 8.0, 6.1 Hz, 1H), 3.71 - 3.60 (m, 2H), 3.27 - 3.19 (m, 2H), 3.16 (s, 3H), 3.20 - 3.06 (m, 4H), 2.86 - 2.73 (m, 2H), 2.59 (s, 3H), 2.57 - 2.53 (m, 3H), 2.40 (s, 3H), 1.73 - 1.68 (m, 4H), 1.62 (s, 3H), 1.51 - 1.44 (m, 2H), 1.39 - 1.35 (m, 8H).
Example 1-128: Synthesis of (2-(5-(4-(6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-
2-yl)amino)phenyl)piperazin-l-yl)hexanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1029)
Step 1: To a stirred solution of 5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(4-(piperazin-l- yl)phenyl)pyrimidine-2,4-diamine (300 mg, 0.616 mmol, 1 equiv) in DMF (3 mL) was added K2CO3 (255 mg, 1.840 mmol, 3 equiv), followed by the addition of tert-butyl 6-bromohexanoate (232, 0.922 mmol, 1.5 equiv) at 0°C. Reaction mixture was stirred at 80°C for 8 h. After completion of the reaction it was quenched with ice water and extracted with ethyl acetate. Combined organic fractions were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. Crude mixture was purified by preparative HPLC to give tert-butyl 6-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)hexanoate (125 mg, 0.190 mmol, 30% yield).
LCMS (ESI+) m/z 657.3 [M+H]+
NMR (400 MHz, DMSO-d6) 5 9.48 (s, 1H), 9.30 (s, 1H), 8.67 - 8.63 (m, 1H), 8.23 (s, 1H), 7.87 - 7.80 (m, 1H), 7.77 - 7.69 (m, 1H), 7.43 (s, 1H), 7.43 - 7.32 (m, 2H), 6.86 (d, J = 9.1 Hz, 2H), 3.54 - 3.36 (m, 1H), 3.09 - 2.98 (m, 4H), 2.34 - 2.25 (m, 4H), 2.19 (t, J = 7.3 Hz, 2H), 1.56 - 1.41 (m, 4H), 1.40 (s, 9 H), 1.35 - 1.20 (m, 4H), 1.17 (d, J = 6.8 Hz, 6H).
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(4-(piperazin-l-yl)phenyl)pyrimidine-2,4-diamine was synthesized as in Example 1-099.
Step 2: 6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)arnino)pyrimidin-2-yl)arnino)phenyl)piperazin- l-yl)hexanoic acid was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above (100% yield), using tert-butyl 6-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)hexanoate (66 mg, 0.100 mmol) as starting material. After completion of the reaction the volatiles were removed under reduced pressure and the crude product was directly forwarded into the next step.
LCMS (ESI+) m/z 601.2 [M+H]+
TH NMR (500 MHz, DMSO) δ 10.41 (s, 1H), 9.61 (s, 1H), 9.54 (s, 1H), 8.57 (s, 1H), 7.86 (dd, J = 8.0, 1.6 Hz, 1H), 7.84 - 7.71 (m, 1H), 7.51 - 7.43 (m, 2H), 7.42 - 7.39 (m, 1H), 6.99 - 6.88 (m, 2H), 3.73 (d, J = 12.1 Hz, 2H), 3.58 - 3.53 (m, 3H), 3.45 (hept, J = 6.6 Hz, 1H), 3.17 - 3.09 (m, 4H), 3.08 - 3.01 (m, 2H), 2.25 (t, J = 7.3 Hz, 2H), 1.78 - 1.68 (m, 2H), 1.59 - 1.51 (m, 2H), 1.37 - 1.29 (m, 2H), 1.16 (d, J = 6.8 Hz, 6H).
Step 3: To a solution of 6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)hexanoic acid (70.0 mg, 0.110 mmol, 1 equiv) in DMF (1.1 mL) were added HATU (1.5 equiv) and DIPEA (3 equiv) and the resulting mixture was stirred at RT for 15 min. (2-(5-(5- amino-7-((4-methoxypiperidin-l-yl)sulfonyl)quinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (1.05 equiv) was added and the reaction mixture was stirred at RT for 20 h. After completion, the reaction mixture was concentrated under reduced pressure and purified by preparative HPLC to give (2- (5-(4-(6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)hexanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (75 mg, 0.064 mmol, 59% yield).
LCMS (ESI+) m/z 1141.35 [M+H]+
NMR (500 MHz, DMSO) δ 10.37 (s, 1H), 9.50 (s, 1H), 9.36 (s, 1H), 8.71 - 8.63 (m, 1H), 8.62 (dd, J = 7.1, 0.9 Hz, 1H), 8.31 (d, J = 2.2 Hz, 1H), 8.25 (s, 1H), 7.95 (s, 1H), 7.94 - 7.92 (m, 1H), 7.85 (dd, J = 8.0, 1.6 Hz, 1H), 7.78 - 7.71 (m, 1H), 7.55 - 7.52 (m, 1H), 7.52 - 7.44 (m, 2H), 7.41 (d, J = 8.3 Hz, 1H), 7.39 - 7.34 (m, 1H), 6.99 - 6.88 (m, 2H), 6.82 (dd, J = 7.1, 2.0 Hz, 1H), 5.71 (s, 2H), 3.89 (s, 2H), 3.84 - 3.50 (m, 2H), 3.45 (p, J = 6.8 Hz, 1H), 3.30 - 3.27 (m, 4H), 3.23 - 3.17 (m, 2H), 3.17 - 3.14 (m, 4H), 3.04 - 2.96 (m, 4H), 2.70 (ddd, J = 12.1, 8.2, 3.5 Hz, 2H), 2.42 (t, J = 7.2 Hz, 2H), 1.76 - 1.64 (m, 4H), 1.64 - 1.57 (m, 2H), 1.43 - 1.34 (m, 2H), 1.34 - 1.25 (m, 2H), 1.17 (d, J = 6.8 Hz, 6H), 1.05 (s, 9H).
Example 1-129: Synthesis of 2-(5-(4-(6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-
2-yl)amino)phenyl)piperazin-l-yl)hexanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1028)
Step 1: 2-(5-(4-(6-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)hexanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (62% yield), using (2-(5-(4-(6-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)hexanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (8.0 mg, 0.007 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1027.30 [M+H]+
XH NMR (500 MHz, DMSO) δ 12.45 (s, 1H), 10.34 (s, 1H), 9.48 (s, 1H), 9.29 (s, 1H), 8.65 (s, 1H), 8.58 (dd, J = 7.1, 0.9 Hz, 1H), 8.30 (d, J = 2.2 Hz, 1H), 8.25 - 8.20 (m, 1H), 7.94 (dd, J = 8.4, 2.2 Hz, 1H), 7.93 (s, 1H), 7.83 (dd, J = 8.0, 1.6 Hz, 1H), 7.77 - 7.70 (m, 1H), 7.55 - 7.52 (m, 1H), 7.45 - 7.41 (m, 2H), 7.40 (d, J = 8.4 Hz, 1H), 7.38 - 7.34 (m, 1H), 6.88 - 6.83 (m, 2H), 6.79 (dd, J = 7.2, 1.9 Hz, 1H), 3.70 (s, 2H), 3.44 (p, J = 6.7 Hz, 1H), 3.23 - 3.16 (m, 1H), 3.15 (s, 3H), 3.09 - 3.04 (m, 4H), 3.04 - 2.98 (m, 2H), 2.69 (ddd, J = 12.1, 8.2, 3.5 Hz, 2H), 2.54 - 2.51 (m, 4H), 2.38 (t, J = 7.3 Hz, 2H), 2.34 (t, J = 7.3 Hz, 2H), 1.71 - 1.63 (m, 2H), 1.63 - 1.57 (m, 2H), 1.55 - 1.48 (m, 2H), 1.40 - 1.33 (m, 2H), 1.33 - 1.26 (m, 2H), 1.17 (d, J = 6.8 Hz, 6H).
Example 1-130: Synthesis of (2-(5-(4-(ll-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)undecanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1032)
Step 1: To a solution of ll-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)undecanoic acid (65.0 mg, 0.097 mmol, 1 equiv) in DMF (1.0 mL) were added HATU (1.5 equiv) and DIPEA (3 equiv) and the resulting mixture was stirred at RT for 15 min. (2-(5- (4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (1.05 equiv) was added and the reaction mixture was stirred at RT for 20 h. After completion, the reaction mixture was concentrated under reduced pressure and purified by preparative HPLC to give (2- (5-(4-(ll-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin- l-yl)undecanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (29 mg, 0.024 mmol, 25% yield).
LCMS (ESI+) m/z 1211.45 [M+H]+
TH NMR (500 MHz, DMSO) δ 10.33 (s, 1H), 9.49 (s, 1H), 9.35 (s, 1H), 8.65 (s, 1H), 8.61 (dd, J = 7.1, 0.9 Hz, 1H), 8.29 (d, J = 2.3 Hz, 1H), 8.24 (s, 1H), 7.94 (s, 1H), 7.95 - 7.90 (m, 1H), 7.85 (dd, J = 8.0, 1.7 Hz, 1H), 7.77 - 7.70 (m, 1H), 7.53 (dd, J = 2.0, 1.0 Hz, 1H), 7.47 (d, J = 8.3 Hz, 2H), 7.40 (d, J = 8.4 Hz, 1H), 7.39 - 7.33 (m, 1H), 6.92 (d, J = 8.6 Hz, 2H), 6.83 (dd, J = 7.2, 2.0 Hz, 1H), 5.70 (s, 2H), 3.89 (s, 2H), 3.45 (p, J = 6.8 Hz, 1H), 3.24 - 3.17 (m, 1H), 3.15 (s, 3H), 3.03 - 2.96 (m, 8H), 2.69 (ddd, J = 12.1, 8.2, 3.5 Hz, 2H), 2.47 - 2.43 (m, 4H), 2.39 - 2.36 (m, 2H), 1.65 - 1.59 (m, 6H), 1.36 - 1.27 (m, 14H), 1.17 (d, J = 6.8 Hz, 6H), 1.04 (s, 9H). ll-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)undecanoic acid was synthesized as in Example 1-128.
Example 1-131: Synthesis of 2-(5-(4-(ll-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)undecanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 1030)
Step 1: 2-(5-(4-(ll-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)undecanamido)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (43% yield), using (2-(5-(4-(ll-(4-(4-((5- chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)undecanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (9.0 mg, 0.007 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1097.40 [M+H]+
TH NMR (500 MHz, DMSO) δ 12.55 (s, 1H), 10.32 (s, 1H), 9.48 (s, 1H), 9.28 (s, 1H), 8.65 (s, 1H), 8.58 (d, J = 7.1 Hz, 1H), 8.29 (d, J = 2.2 Hz, 1H), 8.25 - 8.21 (m, 1H), 7.95 - 7.92 (m, 1H), 7.93 (s, 1H), 7.83 (dd, J = 7.9, 1.6 Hz, 1H), 7.76 - 7.69 (m, 1H), 7.55 - 7.51 (m, 1H), 7.42 (d, J = 8.4 Hz, 2H), 7.39 (d, J = 8.4 Hz, 1H), 7.38 - 7.34 (m, 1H), 6.88 - 6.82 (m, 2H), 6.80 (dd, J = 7.1, 1.9 Hz, 1H), 3.69 (s, 2H), 3.44 (p, J = 6.9 Hz, 1H), 3.23 - 3.17 (m, 1H), 3.15 (s, 3H), 3.08 - 3.04 (m, 4H), 3.04 - 2.98 (m, 2H), 2.70 (ddd, J = 12.2, 8.2, 3.5 Hz, 2H), 2.39 - 2.33 (m, 2H), 2.33 - 1.T1 (m, 2H), 1.65 - 1.57 (m, 4H), 1.47 - 1.43 (m, 2H), 1.33 - 1.27 (m, 16H), 1.26 - 1.23 (m, 2H), 1.17 (d, J = 6.8 Hz, 6H). Example 1-132: Synthesis of (2-(5-(4-(ll-(4-(4-((5-chloro-4-((2-
(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)undecanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1033)
Step 1: To a solution of 8-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)octanoic acid (60 mg, 0.095 mmol, 1 equiv) in DMF (1 mL) were added HATU (1.5 equiv) and DIPEA (3 equiv) and the resulting mixture was stirred at RT for 15 min. (2-(5-(4- amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (1.05 equiv) was added and the reaction mixture was stirred at RT for 20 h. After completion, the reaction mixture was concentrated under reduced pressure and purified by preparative HPLC to give pure (2-(5-(4- (8-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)octanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (11 mg, 0.009 mmol, 10% yield).
LCMS (ESI+) m/z 1169.45 [M+H]+
NMR (500 MHz, DMSO) δ 10.33 (s, 1H), 9.48 (s, 1H), 9.29 (s, 1H), 8.65 (s, 1H), 8.60 (dd, J = 7.1, 0.9 Hz, 1H), 8.29 (d, J = 2.3 Hz, 1H), 8.23 (s, 1H), 7.97 - 7.92 (m, 1H), 7.93 (s, 1H), 7.83 (dd, J = 8.0, 1.7 Hz, 1H), 7.76 - 7.69 (m, 1H), 7.53 (dd, J = 2.0, 1.0 Hz, 1H), 7.42 (d, J = 8.5 Hz, 2H), 7.40 (d, J = 8.4 Hz, 1H), 7.40 - 7.33 (m, 1H), 6.89 - 6.83 (m, 2H), 6.83 (dd, J = 7.2, 1.9 Hz, 1H), 5.70 (s, 2H), 3.88 (s, 2H), 3.44 (p, J = 6.8 Hz, 1H), 3.24 - 3.16 (m, 1H), 3.15 (s, 3H), 3.07 (s, 4H), 3.05 - 2.97 (m, 2H), 2.70 (ddd, J = 12.2, 8.3, 3.5 Hz, 2H), 2.58 - 2.51 (m, 4H), 2.40 - 2.34 (m, 4H), 1.67 - 1.58 (m, 4H), 1.52 - 1.44 (m, 2H), 1.36 - 1.25 (m, 8H), 1.17 (d, J = 6.7 Hz, 6H), 1.04 (s, 9H).
8-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l- yl)octanoic acid was synthesized as in Example 1-128. Example 1-133: Synthesis of 2-(5-(4-(8-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin- 2-yl)amino)phenyl)piperazin-l-yl)octanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid (Compound 1031)
Step 1: 2-(5-(4-(8-(4-(4-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2- yl)amino)phenyl)piperazin-l-yl)octanamido)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5- a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (40% yield), (2-(5-(4-(ll-(4-(4-((5-chloro-4-((2- (isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)phenyl)piperazin-l-yl)undecanamido)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (14.0 mg, 0.012 mmol, 1 equiv) as starting material.
LCMS (ESI+) m/z 1055.35 [M+H]+
NMR (500 MHz, DMSO) δ 12.48 (s, 1H), 10.33 (s, 1H), 9.48 (s, 1H), 9.29 (s, 1H), 8.66 (s, 1H), 8.60 - 8.55 (m, 1H), 8.29 (d, J = 2.2 Hz, 1H), 8.23 - 8.22 (m, 1H), 7.95 - 7.93 (m, 1H), 7.93 (s, 1H), 7.83 (dd, J = 8.0, 1.7 Hz, 1H), 7.76 - 7.69 (m, 1H), 7.55 - 7.52 (m, 1H), 7.44 - 7.41 (m, 2H), 7.39 (d, J = 8.4 Hz, 1H), 7.38 - 7.33 (m, 1H), 6.88 - 6.83 (m, 2H), 6.80 (dd, J = 7.2, 2.0 Hz, 1H), 3.69 (s, 2H), 3.44 (p, J = 6.8 Hz, 1H), 3.24 - 3.16 (m, 1H), 3.15 (s, 3H), 3.08 - 3.04 (m, 4H), 3.04 - 2.98 (m, 2H), 2.70 (ddd, J = 12.2, 8.3, 3.5 Hz, 2H), 2.48 - 2.42 (m, 4H), 2.40 - 2.35 (m, 2H), 2.34 - 2.28 (m, 2H), 1.65 - 1.57 (m, 4H), 1.50 - 1.43 (m, 2H), 1.38 - 1.26 (m, 8H), 1.17 (d, J = 6.7 Hz, 6H).
Example 1-134: Synthesis of 2-(5-(2-chloro-6-cyanophenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 98) Step 1: Mixture of methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3- yl)acetate (30.0 mg, 0.095 mmol, 1 equiv), 2-bromo-3-chlorobenzonitrile (21.6 mg, 0.095 mmol, 1 equiv) and Pd(PPh3)4 (11.0 mg, 0.009 mmol, 0.1 equiv) was dissolved in 1,4-dioxane (1 mL) at RT, then 2 M aq. Na2CO3 (0.095 mL, 0.190 mmol, 2.000 eq) solution was added to the reaction mixture which was stirred at 90°C for 18 h. After that time, 1 M aq. LiOH solution (1.9 mL, 1.898 mmol, 20 equiv) was added to the reaction mixure which was stirred at rt for 3 h. Reaction mixture was then neutralized with 1 M aq. HCI and extracted with EtOAc. Combined organic fractions were washed with water and brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. Crude mixture was purified by preparative HPLC to afford pure 2-[5-(2-chloro-6-cyanophenyl)pyrazolo[l,5-a]pyridin-3-yl]acetic acid (3.0 mg, 0.010 mmol, 10 % yield).
LCMS (ESI+) m/z 312.05 [M+H]+
NMR (500 MHz, DMSO) δ 8.77 (d, J = 7.2 Hz, 1H), 8.03 - 7.95 (m, 3H), 7.83 - 7.80 (m, 1H), 7.66 (t, J = 8.0 Hz, 1H), 6.89 (dd, J = 7.2, 2.0 Hz, 1H), 3.69 (s, 2H).
Methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was prepared as described in Example 1-002.
Example 1-135: Synthesis of 2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid
(Compound 100)
Step 1: Methyl 2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (83% yield), using methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate (16.3 mg, 0.052 mmol, 1.1 equiv) and l-(2-bromophenyl)-N-methylmethanesulfonamide [commercial] (1 equiv) as starting materials, K2CO3 (3 equiv) as base, Pd(dppf)CI2 as catalyst (0.1 equiv). After completion of the reaction the solution was filtered, the volatiles were removed under reduced pressure and the resulting crude was used in the next step without purification. LCMS (ESI+) m/z 352.2 [M+H]+
Methyl 2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate was prepared as described in Example 1-002.
Step 2: 2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid was synthesized using the general procedure shown in Reaction Scheme 1 and Example Method 1, above (38% yield), using methyl 2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetate as starting material.
LCMS (ESI+) m/z 338.05 [M+H]+
TH NMR (500 MHz, DMSO) δ 9.10 (dd, J = 4.2, 1.7 Hz, 1H), 8.74 (dd, J = 7.2, 1.0 Hz, 1H), 8.54 (dd, J = 8.4, 1.7 Hz, 1H), 8.10 (d, J = 8.4 Hz, 1H), 7.99 (s, 1H), 7.85 (dd, J = 2.1, 1.0 Hz, 1H), 7.73 (d, J = 8.4 Hz, 1H), 7.71 (dd, J = 8.3, 4.2 Hz, 1H), 7.07 (dd, J = 7.2, 1.9 Hz, 1H), 3.76 (s, 2H).
Example 1-136: Synthesis of 2-(5-(4-(dimethylcarbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 101)
Step 1: 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5- a]pyridin-5-yl)benzoic acid (25.0 mg, 0.043 mmol, 1 equiv) and HATU (19.4 mg, 0.051 mmol, 1.2 equiv) were dissolved in DMF (0.5 mL). The mixture was stirred for 15 min at rt, then dimethylamine hydrochloride (3.8 mg, 0.047 mmol, 1.1 equiv) and DIPEA (0.022 mL, 0.128 mmol, 3.0 equiv) were added and the resulting mixture was stirred for 4 h at rt. After this time solvent was evaporated and the residue material was purified by preparative HPLC to give pure (2-(5-(4-(dimethylcarbamoyl)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (14.0 mg, 0.023 mmol, 53 % yield).
LCMS (ESI+) m/z 615.20 [M+H]+
3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-
5-yl)benzoic acid was prepared as described in Example 1-081. Step 2: (2-(5-(4-(dimethylcarbamoyl)-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin- 3-yl)acetoxy)methyl pivalate (14.0 mg, 0.023 mmol, 1.0 equiv) was dissolved in MeOH (1.1 mL) and water (0.2 mL). To the resulting mixture cesium carbonate (37.1 mg, 0.114 mmol, 5.0 equiv) was added and the reaction was stirred at 60°C for 1.5 h. After this time solvents were evaporated and the residue material was purified by preparative HPLC to afford pure 2-(5-(4-(dimethylcarbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (7.0 mg, 0.014 mmol, 61.3 % yield).
LCMS (ESI+) m/z 501.15 [M+H]+
TH NMR (500 MHz, DMSO) δ 8.65 - 8.60 (m, 1H), 7.97 - 7.93 (m, 2H), 7.78 (dd, J = 7.8, 1.8 Hz, 1H), 7.64 - 7.59 (m, 1H), 7.53 (d, J = 7.8 Hz, 1H), 6.85 (dd, J = 7.1, 2.0 Hz, 1H), 3.69 (s, 2H), 3.24 - 3.16 (m, 1H), 3.14 (s, 3H), 3.03 (s, 3H), 3.02 - 2.99 (m, 2H), 2.97 (s, 3H), 2.69 (ddd, J = 12.2, 8.4, 3.5 Hz, 2H), 1.65 - 1.56 (m, 2H), 1.34 - 1.23 (m, 2H).
Example 1-137: Synthesis of 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)-4-
(methylcarbamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 102)
Step 1: 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5- a]pyridin-5-yl)benzoic acid (17.0 mg, 0.029 mmol, 1 equiv) and HATU (13.2 mg, 0.035 mmol, 1.2 equiv) were dissolved in DMF (0.3 mL), then DIPEA (0.015 mL, 0.087 mmol, 3.0 equiv) was added and the mixture was stirred for 15 min at rt, then methylamine hydrochloride (2.1 mg, 0.032 mmol, 1.1 equiv) was added and the resulting mixture was stirred for 2 h at rt. After this time the solvent was evaporated and the residue material was purified by preparative HPLC to give pure (2-(5-(2-((4-methoxypiperidin-l- yl)sulfonyl)-4-(methylcarbamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (10.0 mg, 0.017 mmol, 58 % yield).
LCMS (ESI+) m/z 601.84 [M+H]+
3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-
5-yl)benzoic acid was prepared as described in Example 1-081. Step 2: (2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)-4-(methylcarbamoyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (10.0 mg, 0.017 mmol, 1.0 equiv) was dissolved in MeOH (0.5 mL) and water (0.1 mL). To the resulting mixture cesium carbonate (27.1 mg, 0.083 mmol, 5.0 equiv) was added and the reaction was stirred at 60°C for 20 h. After this time LCMS showed full conversion to the desired product. Solvents were evaporated and the residue material was purified by preparative HPLC (H2O + 0.1% FA, MeCN + 0.1% FA) to give pure 2-(5-(2-((4-methoxypiperidin-l-yl)sulfonyl)-4-
(methylcarbamoyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (8.0 mg, 0.016 mmol, 98.8 % yield).
LCMS (ESI+) m/z 487.10 [M+H]+
NMR (500 MHz, DMSO) δ 12.34 (s, 1H), 8.81 (q, J = 4.4 Hz, 1H), 8.64 (dd, J = 7.2, 1.0 Hz, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.16 (dd, J = 7.9, 1.8 Hz, 1H), 7.96 (s, 1H), 7.63 - 7.60 (m, 1H), 7.58 (d, J = 7.9 Hz, 1H), 6.85 (dd, J = 7.2, 1.9 Hz, 1H), 3.72 (s, 2H), 3.19 (tt, J = 7.4, 3.5 Hz, 1H), 3.14 (s, 3H), 3.04 - 2.97 (m, 2H), 2.84 (d, J = 4.6 Hz, 3H), 2.67 (ddd, J = 12.3, 8.4, 3.5 Hz, 2H), 1.65 - 1.56 (m, 2H), 1.33 - 1.22 (m, 2H).
Example 1-138: Synthesis of 2-(5-(4-acetamido-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (Compound 103)
Step 1: (2-(5-(4-acetamido-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 11 and Example Method 11, above (46% yield), using (2-(5-(4-amino-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (20.0 mg, 0.036 mmol, 1.0 equiv) and acetyl chloride (3.0 equiv) as starting materials.
LCMS (ESI+) m/z 601.15 [M+H]+
(2-(5-(4-amino-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate was prepared as described in Example 1-083. Step 2: ((2-(5-(4-acetamido-2-((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3- yl)acetoxy)methyl pivalate (10.0 mg, 0.017 mmol, 1.0 equiv) was dissolved in MeOH (0.8 mL) and water (0.2 mL). To the resulting mixture cesium carbonate (27.1 mg, 0.083 mmol, 5.0 equiv) was added and the reaction was stirred at 60°C for 1.5 h. After that time, solvents were evaporated and the residue material was purified by preparative HPLC to give pure 2-(5-(4-acetamido-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetic acid (7.9 mg, 0.016 mmol, 97 % yield).
LCMS (ESI+) m/z 487.10 [M+H]+
TH NMR (500 MHz, DMSO) δ 10.41 (s, 1H), 8.60 - 8.54 (m, 2H), 8.27 (d, J = 2.3 Hz, 1H), 7.94 - 7.88 (m, 2H), 7.55 - 7.51 (m, 1H), 7.39 (d, J = 8.2 Hz, 1H), 6.79 (dd, J = 7.2, 2.0 Hz, 1H), 3.64 (s, 2H), 3.24 - 3.16 (m, 1H), 3.15 (s, 3H), 3.06 - 2.98 (m, 2H), 2.70 (ddd, J = 12.2, 8.3, 3.6 Hz, 2H), 2.10 (s, 3H), 1.67 - 1.57 (m, 2H), 1.30 (dtd, J = 12.0, 8.0, 3.7 Hz, 2H).
Example 1-139: Synthesis of (2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 99):
Step 1: (2-(5-(7-chloroquinolin-8-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate was synthesized using the general procedure shown in Reaction Scheme 3 and Example Method 3, above (33% yield), using (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (15.0 mg, 0.059 mmol, 1.05 equiv) and 8-Bromo-7-chloroquinoline [commercial] (1 equiv) as starting materials, K2CO3 (3 equiv) as base, Pd(dppf)CI2 as catalyst (0.1 equiv). After completion of the reaction the solution was filtered, the volatiles were removed under reduced pressure and the resulting crude was used in the next step without purification.
LCMS (ESI+) m/z 452.1 [M+H]+
NMR (500 MHz, DMSO) δ 9.10 (dd, J = 4.2, 1.7 Hz, 1H), 8.76 (dd, J = 7.2, 1.0 Hz, 1H), 8.54 (dd, J = 8.3, 1.7 Hz, 1H), 8.11 (d, J = 8.5 Hz, 1H), 8.00 (s, 1H), 7.83 (dd, J = 2.0, 1.0 Hz, 1H), 7.76 - 7.68 (m, 2H), 7.11 (dd, J = 7.2, 2.0 Hz, 1H), 5.72 (s, 2H), 3.97 (s, 2H), 1.00 (s, 9H). (2-(5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate was prepared as described in Example 1-079.
Example 1-140: Synthesis of (S)-(2-(5-(4-(4-((4-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetyl)piperazin-l-yl)methyl)piperidine-l-carbonyl)-2-((4- methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (Compound 1042):
Step 1: 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-l,8,ll,12-tetraazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid [commercial] (65.0 mg, 0.162 mmol, 1.000 eq) and HATU (74.0 mg, 0.195 mmol, 1.200 eq) were dissolved in DMF (0.765 mL). The resulting mixture was stirred at rt for 15 min, then tert-butyl 4-[(piperazin-l-yl)methyl]piperidine-l-carboxylate [commercial] (50.5 mg, 0.178 mmol, 1.100 eq) and DIPEA (0.056 mL, 0.324 mmol, 2.000 eq) were added and the mixture was stirred at rt for 3 h. After this time, the solvent was evaporated, residue material was dissolved in DMSO and purified by reverse-phase flash chromatography to afford tert-butyl 4-[(4-{2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-l,8,ll,12-tetraazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]acetyl}piperazin-l-yl)methyl]piperidine-l-carboxylate (65.0 mg, 0.098 mmol, 60.2%) was obtained as yellow solid.
LCMS (ESI+) m/z 666.2 [M+H]+ 2H NMR (500 MHz, DMSO) δ 7.49 (d, J = 8.4 Hz, 2H), 7.47 - 7.41 (m, 2H), 4.57 (t, J = 6.7 Hz, 1H), 4.40 (s, 1H), 3.93 (s, 2H), 3.62 (s, 3H), 3.44 - 3.40 (m, 3H), 3.08 (s, 2H), 2.71 (d, J = 19.2 Hz, 2H), 2.60 (s, 3H), 2.55
- 2.51 (m, 2H), 2.44 - 2.40 (m, 3H), 2.30 (s, 1H), 2.16 (s, 1H), 1.70 (s, 2H), 1.63 (d, J = 1.0 Hz, 3H), 1.40 (s, 9H), 1.14 - 0.91 (m, 2H).
Step 2: tert-butyl (S)-4-((4-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetyl)piperazin-l-yl)methyl)piperidine-l-carboxylate was synthesized using the general procedure shown in Reaction Scheme 5 and Example Method 5, above, using tert-butyl 4-[(4-{2- [(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-l,8,ll,12-tetraazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetyl}piperazin-l-yl)methyl]piperidine-l-carboxylate (65 mg, 0.098 mmol) as starting material. After completion the volatiles were removed under reduced pressure and the crude product was used directly in the next step.
LCMS (ESI+) m/z 566.2 [M+H]+
Step 3: The solution of 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-
((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-5-yl)benzoic acid (21.5 mg, 0.037 mmol, 1.1 equiv) and HATU (1.2 equiv) in DMF (0.16 mL) was stirred at RT for 10 min. 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-l,8,ll,12-tetraazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]-l-{4- [(piperidin-4-yl)methyl]piperazin-l-yl}ethan-l-one hydrochloride (20.0 mg, 0.033 mmol, 1 equiv) and DIPEA (2 equiv) were added and the reaction was stirred at RT for 4 h, until full conversion was indicated by LCMS. (S)-(2-(5-(4-((10-(2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3- a][l,4]diazepin-6-yl)acetamido)decyl)carbamoyl)-2-((4-methoxypiperidin-l- yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate (10.0 mg, 0.008 mmol, 25% yield) was purified by preparative HPLC.
LCMS (ESI+) m/z 1135.2 [M+H]+
2H NMR (500 MHz, DMSO) δ 8.66 (dd, J = 7.3, 0.9 Hz, 1H), 7.97 (s, 1H), 7.92 (d, J = 1.7 Hz, 1H), 7.76 (dd, J = 7.7, 1.8 Hz, 1H), 7.62 (dd, J = 1.9, 0.9 Hz, 1H), 7.55 (d, J = 7.8 Hz, 1H), 7.52 - 7.47 (m, 2H), 7.47 - 7.40 (m, 2H), 6.90 (dd, J = 7.2, 1.9 Hz, 1H), 5.71 (s, 2H), 4.57 (t, J = 6.7 Hz, 1H), 3.90 (s, 2H), 3.67 - 3.64 (m, 2H), 3.64
- 3.56 (m, 1H), 3.50 - 3.44 (m, 2H), 3.43 - 3.35 (m, 1H), 3.36 (s, 4H), 3.28 (s, 4H), 3.15 (s, 3H), 3.02 - 2.96 (m, 2H), 2.74 - 2.67 (m, 2H), 2.59 (s, 3H), 2.54 (p, J = 1.9 Hz, 1H), 2.47 - 2.45 (m, 2H), 2.43 - 2.40 (m, 3H), 2.35 - 2.31 (m, 2H), 2.25 - 2.20 (m, 2H), 1.92 - 1.82 (m, 2H), 1.66 - 1.59 (m, 5H), 1.33 - 1.26 (m, 2H), 1.05
(s, 9H).
The preparation of 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-
((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-5-yl)benzoic acid was described in Example 1-081.
Example 1-141: Synthesis of (S)-(2-(5-(4-((l-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)-2-oxo-6,9,12,15-tetraoxa-3-azaheptadecan-17-yl)carbamoyl)-2- ((4-methoxypiperidin-l-yl)sulfonyl)phenyl)pyrazolo[l,5-a]pyridin-3-yl)acetoxy)methyl pivalate
(Compound 1043):
Step 1: To 4-[3-(2-{[(2,2-dimethylpropanoyl)oxy]methoxy}-2-oxoethyl)pyrazolo[l,5-a]pyridin-5-yl]-3-[(4- methoxypiperidin-l-yl)sulfonyl]benzoic acid (15.0 mg, 0.026 mmol, 1.000 eq) in DM F (3.0 mL) were added HATU (19.4 mg, 0.051 mmol, 2.000 eq) and DIPEA (0.044 mL, 0.255 mmol, 10.000 eq) at room temperature. Mixture was stirred for 10 min and then, (S)-N-(14-amino-3,6,9,12-tetraoxatetradecyl)-2-(4- (4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide trifluoroacetate (23.6 mg, 0.031 mmol, 1.200 eq) was added. Reaction was carried out at room temperature for 10 min and then, solvent was evaporated. {[2-(5-{4-[(14-{2-[(9S)-7-(4-chlorophenyl)- 4,5,13-trimethyl-3-thia-l,8,ll,12-tetraazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]acetamido}-3,6,9,12-tetraoxatetradecan-l-yl)carbamoyl]-2-[(4-methoxypiperidin-l- yl)sulfonyl]phenyl}pyrazolo[l,5-a]pyridin-3-yl)acetyl]oxy}methyl 2,2-dimethylpropanoate (18.0 mg, 0.015 mmol, 57.5%) was purified by preparative HPLC.
LCMS (ESI+) m/z 1188.3 [M+H]+
2H NMR (500 MHz, DMSO) δ 8.95 (t, J = 5.6 Hz, 1H), 8.68 (dd, J = 7.2, 0.9 Hz, 1H), 8.47 (d, J = 1.8 Hz, 1H), 8.28 (t, J = 5.6 Hz, 1H), 8.21 (dd, J = 8.0, 1.8 Hz, 1H), 7.99 (s, 1H), 7.63 (dd, J = 2.0, 1.0 Hz, 1H), 7.60 (d, J = 7.9 Hz, 1H), 7.52 - 7.48 (m, 2H), 7.46 - 7.42 (m, 2H), 6.88 (dd, J = 7.2, 2.0 Hz, 1H), 5.73 (s, 2H), 4.52 (dd, J = 8.1, 6.0 Hz, 1H), 3.92 (s, 2H), 3.61 - 3.58 (m, 2H), 3.58 - 3.55 (m, 4H), 3.55 - 3.50 (m, 8H), 3.50 - 3.45 (m, 4H), 3.31 - 3.18 (m, 5H), 3.16 (s, 3H), 3.04 - 2.97 (m, 2H), 2.72 - 2.65 (m, 2H), 2.61 (s, 3H), 2.42 (d, J = 0.9 Hz, 3H), 1.64 (s, 3H), 1.63 - 1.57 (m, 2H), 1.29 (dtd, J = 11.9, 7.8, 3.5 Hz, 2H), 1.06 (s, 9H).
The preparation of 3-((4-methoxypiperidin-l-yl)sulfonyl)-4-(3-(2-oxo-2-
((pivaloyloxy)methoxy)ethyl)pyrazolo[l,5-a]pyridin-5-yl)benzoic acid was described in Example 1-081.
(S)-N-(14-amino-3,6,9,12-tetraoxatetradecyl)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-yl)acetamide was prepared according to the procedure described in
US2017008904A1.
Example 2: Fluorescence polarization displacement assay (FP)
The interaction of KLHDC2 ligands of the present invention with the KLHDC2 protein was investigated by using a Fluorescence polarization displacement assay (FP). First, the protein solution was rapidly thawed and centrifuged at 14000 ref for 5 min at 4°C. The concentration of the supernatant was measured and the assay buffer containing 1 nM KLHDC2 in PBS pH 7.4, 2 mM DTT, 0.02% Tween-20 was prepared. Subsequently, compounds and DMSO were dispensed using Echo® 555 liquid handler, Labcyte Inc. to a black 384-well plate (Greiner, 784076). The final DMSO concentration was adjusted to 2% DMSO in a final assay volume of 10 pL. Generated dose-response curves were based on 16 concentration points from 1500 pM to 0.05 pM or from 300 pM to 0.009 pM in 2 technical replicates. Then, 1 nM final concentration of the peptide-based tracer modified with Cy5 fluorescent dye was dispensed by using Echo® 555 liquid handler, Labcyte Inc. into wells containing tested compounds or DMSO. 10 pl of assay buffer (containing 1 nM KLHDC2) was added using MultiFlo FX Microplate Dispenser to wells with compounds and a column containing only DMSO and the tracer (positive control). Then, 10 pl of PBS pH 7.4, 2 mM DTT, 0.02% Tween-20 buffer was added to wells with DMSO (blank) and with DMSO and tracer (negative control). The plates were shaken by VibroTurbulator for 60 sec at room temperature. After shaking the plates were centrifuged for 1 min at 1000 ref and incubated for 60 min in the dark at room temperature. The readout was performed with PHERAstar in a fluorescence polarization mode. Data were processed by using MARS software. The outliers from "Intensity-based on raw data" were removed from the columns containing blank, negative and positive controls. Results were uploaded and plotted by the dedicated software to determine the negative logarithm of the inhibitory constant (pKi value). The results are presented in Table
2. Example 3: Differential Scanning Fluorimetry (DSF)
The effect on stabilization of the KLHDC2 ligase by the KLHDC2 ligands of the present invention was investigated by using Differential Scanning Fluorimetry (DSF) assay. First, the KLHDC2 protein was thawed and centrifuged at 18000 ref at 4°C for 5 min. The protein concentration in the supernatant was measured and used to prepare a master mix containing 0.75 pM of KLHDC2 in the assay buffer PBS, pH 7.4; 1 mM DTT. The master mix solution was pipetted to a 4titude® PCR solid 384 well plate. The plate was spined down at 1000 ref for 10 sec at room temperature. Subsequently, SYPRO Orange dye and tested compounds were added to the plate by using Echo® 555 liquid handler, Labcyte Inc. The final DMSO concentration was adjusted to 2%. The plate was spined down at 1000 ref, 22°C for 1 min, shaken by VibroTurbulator for 1 min, level 10, spined down again, and incubated in the darkness at room temperature. The readout was done by using Viaa7 after 15 min from adding the compounds. The ATm and SSMD values were determined by the dedicated in-house script. The results are presented in Table 2.
Table 2 Biophysical parameters for selected compounds
Fluorescence polarization level description pKi:
7.0 < A
5.8 < B < 7.0
4.6 < C < 5.8
D < 4.6
Differential scanning fluorimetry level description DSF ATm (°C):
10 < a
5 < b < 10 c < 5
Example 4: Formation of ternary complexes by bifunctional compounds
HTRF ternary complex assay
The effect of the disclosed bifunctional compounds on the mediation of a ternary complex composed of [KLHDC2]-[compound of formula (ll)]-[Halo protein] or [KLHDC2]-[compound of formula (ll)]-[BRD4 protein] was investigated by using Homogeneous Time-Resolved Fluorescence assay (HTRF). Firstly, protein solutions were centrifuged at 18000 ref at 4°C for 5 min. The concentration of each protein was measured 3 times and the median of these measurements was used to prepare protein solutions. A mix of the proteins, acceptor, and the donor was prepared in PPI Europium detection buffer to obtain final concentrations of 100 nM KLHDC2, 100 nM Halo protein, 52.8 nM of Mab Anti-FLAG-d2 (acceptor), and 12 nM of Anti-6Xhis-Eu cryptate Gold (donor) or 100 nM KLHDC2, 50 nM BRD4 protein, 6.7 nM of Mab Anti-6xHis-d2 (acceptor), and 3 nM of Streptavidin-Eu cryptate (donor). 10 pl of the prepared mix was dispensed to selected wells on a 384-well low-volume plate (Greiner, 784075). Subsequently, tested compounds were added to the plate in the range of concentrations 0.0097-20 pM by using Echo® 555 liquid handler, Labcyte Inc. DMSO was backfilled to all wells, resulting in a final DMSO content of 0.5%. Wells containing only DMSO served as a background. The plate was sealed with a transparent film and it was shaken by VibroTurbulator for 30 sec at RT, level 3. Then, the plate was spun down (60 s, 1000 ref, RT) to make sure the liquids were at the bottom of the wells. The plate was incubated at RT and the readout was performed after 60 min by using a plate reader (Pherastar, BMG Labtech) in time-resolved fluorescence mode (filters: TR 337 665 620). The data were analyzed using GraphPad prism 9 and the dedicated script developed in-house. The results are presented in Table 3.
Table 3 Ternary complex formation assay for selected compounds assessed by HTRF method pEC5o level description:
6.0 < B < 7.0
C < 6.0
Max normalized signal-to-background HTRF ratio level description:
5 < a
2.5 < b < 5 c < 2.5
Example 5: Determination of the Cell Permeability and Target Engagement
NanoBRET - in cell CRBN Target Engagement Assay Commercially available, NanoBRET In-cell CRBN Target Engagement Assay (NanoBRET CRBN TE Assay), which uses NanoLuc CRBN HEK293 cells and NanoBRET In-cell CRBN Tracer was used (N2910, N2912, PROMEGA) to determine the cell membrane permeability of KLHDC2 ligands conjugated to CRBN binding moiety.
The assay was run in 96-well plate format (cat. 3600, Corning). NanoLuc CRBN HEK293 cells were treated with CRBN Tracer and compounds. Briefly, prepared was cell suspension (0.17 x 106cells/ml) in Opti-MEM without phenol red (Life Technologies). The cells were treated with CRBN Tracer (20x concentrated, 5 pl/well. For the live mode: 0.8 pM and for the lytic: 0.17 pM final concentration) or DMSO vehicle control (20x concentrated, 5 pl/well). Such prepared cells were seeded into the wells of 96-well plate on top of compounds (lOOx concentrated, 1 pl/well; 11-point 2-fold dilution) pre-plated on them with the help of Echo 555 acoustic dispenser (Labcyte). For the lytic mode assay, Digitonin (0.05 mg/ml, lOx concentrated, 10 pl/well) was added additionally. In the live mode assay, the plate was incubated at 37°C, 5% CO2 for 2 hours, and then at room temperature for 15 minutes to cool down. In the lytic mode assay, there was no incubation. The 3 x Complete NanoBRET reagent (50 pl/well) was prepared according to the manufacturer's recommendations, appropriately for live and lytic mode. The plate was incubated for 2 minutes at room temperature and the luminescence and fluorescence readings followed using CLARIOstar Multimode Plate Reader (donor emission RLU - 450 nm; acceptor emission RFU - 610 nm).
Table 4 NanoBRET plC50 values for tested bifunctional compounds in intact and lysed cells. plC5o level description:
6.0 < A
5.0 < B < 6.0
C < 5.0 Based on the evidence, presented herein KLHDC2 ligands are cell membrane permeable. Importantly, the activity for prodrug-modified ligands is 1-2 orders of magnitude higher than for a non-modified ligand indicating an enhanced cell membrane permeability for prodrugs.
Example 6: Intracellular Target Engagement (iTE) assay
The HEK293T-based cell line was used to overexpress HiBiT-KLHDC2 (full-length protein). Stable expression in pools of cells was achieved using the piggyBac transposon system and antibiotic selection (Puromycin, 2 pg/ml, 24h). Signal detection relies on the NanoBiT system that has established a sensor system by an in-frame fusion with an 11 amino acid tag ( Hi BiT) that has a high affinity towards the LgBiT. Those two comprise the scaffold of the NanoLuciferase (NLuc) and their complementation allows bioluminescent detection. A chemical entity tested as a potential KLHDC2 ligand, in the event of stabilizing the protein, causes a shift in its temperature of aggregation (Tagg). Briefly, stably transfected cells (pool of clones) in serum-free cell culture medium (DMEM GlutaMAX) were resuspended into 384-well PCR plates (2 x 103 cells/lOpl/well) (Bio-Rad, HSP3865) using the multichannel Finnpipette (Thermo Fisher Scientific). Tested compounds (32 pM, 25 nl) or DMSO vehicle control (0.25%) were subsequently added using Echo-Acoustic Liquid Handler (E550, Labcyte) and incubated for lh (or 2h - if a tested compound is a prodrug) at 37°C with 5% CO2. The plate was sealed and heated using a pre-heated temperature gradient thermal cycler (C1000 Touch with 384-well module, Bio-Rad) for 3.5 minutes and subsequently cooled to 25°C. PheraSTAR microplate reader and Nano-Gio® HiBiT Lytic Detection system (PROMEGA) were used for the signal read-out (RLU values).
KLHDC2 - Tagg - 51.5°C (DMSO sample, HEK293 HiBiT-KLHDC2 cells)
Table 5 Intracellular target engagement assay's results with temperatures of aggregation in intact cells.
AT - calculated as temperature differences to the DMSO control.
Thermal shift level description ATm (°C):
3.0 < a
1.5 < b < 3.0 c < 1.5
Based on this evidence, presented herein exemplary KLHDC2 ligands interact with the target in cellular experiments indicating the target engagement.
Example 7: HiBiT-BRD4 HEK293 degradation assay
Cell culture. HEK293 HiBiT-BRD4 cells (Promega, HEK293 LgBiT with the HiBiT on the N-term of BRD4) were cultured according to the manufacturer's recommendations. Briefly, cells were grown in DMEM high glucose with pyruvate (Gibco, cat. 11995), supplemented with 10% fetal bovine serum (FBS, cat. S181H- 500, Biowest), 1% Penicillin-Streptomycin (cat. L0022-100, Biowest) and 0.8 mg/ml G418 (Gibco, cat. 10- 131-035).
Degradation experiment with Luminescent read-out 2x103 cells were seeded into a well of the 384-well plate (cat. HSP3865, Bio-Rad) in a total volume of 30 pl. Compounds dilutions in DMSO were prepared and applied using the Echo 555 Liquid Handler (Labcyte). Final DMSO concentration was 0.1%, the dilution range for tested compounds was from 10 pM to 3 nM (5-fold diution) and for the dBET6 reference compound (MedChem, cat. HY-112588) from lpM to 0.3 nM (5-fold dilution). Cells were left for 4 h in an incubator (37°C, 5% CO2). Subsequently, plate was centrifuged briefly (100 ref, 1 min, room temperature) and 30 pl of the Nano-Gio HiBiT Lytic Assay (cat. N3050, Promega), prepared according to the manufacturer's recommendation, was added and mixed (300 rpm, 5 min, room temperature). The plate was briefly centrifuged (100 ref, 1 min, room temperature) and left for 10 minutes on the bench, equilibrating, protected from light. Measurement of the luminescent signal was performed using the PHERAstar Multimode Plate Reader (BMG Labtech). Tested samples were compared to the DMSO-treated control (100% BRD4 expression level).
Cell Viability Assay - CTG (Cell Titer Gio)
HiBiT-BRD4 HEK293 cells were seeded into a 384-well white, F-bottom plate (cat. 781080, Grainer Bio- One) in a final volume of 50 pl. DMSO-diluted compounds were added to the cells with the help of Echo 555 Liquid Handler (Labcyte). Final DMSO concentration: 0.25%. Cells were incubated (37°C, 5% CO2) with titrated compounds for 24h (highest concentration for compounds: 10 pM, and for the reference compound dBET6: 1 pM). Subsequently, CellTiter-Glo Luminescent Cell Viability Assay (cat. G7572, Promega) was added, according to the manufacturer's recommendations. Luminescent signal was detected using the CLARIOstar Multimode Plate Reader and the percentage of cell viability was calculated, normalizing to the DMSO control wells.
Table 6 Results of the HiBiT-BRD4 degradation assay. pDC5o level description:
7.0 < A
6.0 < B < 7.0
5.0 < C < 6.0
D < 5.0
Percentage of degraded BRD4 protein Dmax (%):
60% < a
30% < b < 60% c < 30%
The invention is further described herein with reference to the following clauses: wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl), -NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -SO(alkyl), and -P(O)(alkyl)2; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl or heteroaryl, each of which is independently unsubstituted or is substituted with at least one of -OH, -NH2, -NH(alkyl) or -N (alkyl )2; each R6 is independently selected from unsubstituted alkyl, haloalkyl, unsubstituted aryl, - C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CFbJqOIVIe, -(OCHzCFbJrNHR73, -C(O)alkyl and -S(O)2alkyl; wherein q is 1 or 2; r is 1, 2, 3, 4 or 5; and R7a is H or - C(O)alkyl;
R8 is selected from -COOR10, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -B(OH)2, - P(O)(OH)2 -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and ^(OjOCHjCHjSOjIVIe; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkyl)2, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(0)NMe2; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen. 2. The compound of clause 1, wherein X2 is N.
3. The compound of clause 1, wherein X3 is N.
4. The compound of clause 1, wherein X2 is CR3 and X3 is CR1.
5. The compound of any preceding clause, wherein R1 is H, halogen or methyl.
6. The compound of any preceding clause, wherein R3 is H, halogen, unsubstituted alkyl or -CN.
7. The compound of any preceding clause, wherein R1 is H.
8. The compound of clause 7, wherein R1 and R3 are each H.
9. The compound of clause 7, wherein R1 and R2 are each H.
10. The compound of any one of clauses 1-6, wherein R2 and R3 are each H.
11. The compound of any one of clauses 1-6, wherein
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
12. The compound of any one of clauses 1-8 and 11, wherein R2 is -B(OH)2, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
13. The compound of any one of clauses 1-8, 11 and 12, wherein R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
14. The compound of clause 13, wherein R2 is selected from: n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3. pound of clause 14, wherein R2 is selected from n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3. pound of clause 15, wherein R2 is selected from
wherein n is 1, 2 or 3; and wherein m is 0 or 1. The compound of clause 16, wherein R2 is
18. The compound of any preceding clause, wherein each R4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -
O(cycloalkyl), -O(heterocycloalkyl), -NH2, -N (a lkyl)z, -S(O)zalkyl, -S(O)zaryl, -S(O)2N(alkyl)2, - CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), -C(O)(heterocycloalkyl), and -C(O)OH; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
19. The compound of any preceding clause, wherein each R7 is independently selected from -Me, - OMe, -O(CH2)qOMe, -CH2OMe, -C(O)Me and -S(O)2Me.
20. The compound of any preceding clause, wherein each R4 is independently selected from -F, -Cl, - lBu, -Me, -CF3, -OH, -OMe, -OCF3, -CN, -NH2, -NMe2, -S(O)2Me, -S(O)2NMe2, -CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
21. The compound of any preceding clause, wherein each R6 is independently selected from haloalkyl, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
22. The compound of any preceding clause, wherein each R6 is independently selected from -CF3, -
C(O)piperidine, -
23. The compound of any preceding clause, wherein R2 is selected from:
24. The compound of any preceding clause, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C- S(O)2(heterocycloalkyl), C-S(O)2aryl, C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
25. The compound of any one of clauses 1-23, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then: ach R4 of — e ”R4) is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, -
NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), -
C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, -C(O)NH(alkyl), -SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
26. The compound of any one of clauses 1-25, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl);
27. The compound of any one of clauses 1-25, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH or -CH2C(O)OCH2OC(O)tBu;
X2 and X3 are both CH; and (i) each is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; or
28. The compound of any one of clauses 1-6 and 10-27, wherein R1 is methyl. 29. The compound of any preceding clause, wherein -
30. The compound of any preceding clause, wherein R8 is selected from -COOR9, -CHR9C(O)OR10, - CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, - CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2.
31. The compound of clause 30, wherein R8 is selected from -COOR9, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH2B(OH)2, -CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2.
32. The compound of clause 31, wherein R8 is selected from -COOH, -COOMe, -CH2C(O)OH, - CHMeC(O)OH, -CH2C(O)OMe, -CH2C(O)OEt, -CH^OjOCHzOCfOPBu, -CH2C(O)NH2, -CH2S(O)2NH2, - S(O)2NH2, -CH2-oxetane, -CH2(l,2,3-triazole), -CH2B(OH)2, -CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2.
33. The compound of clause 32, wherein R8 is selected from -CH2C(O)OCH2OC(O)tBu, -CH2C(O)OH and -CHMeC(O)OH.
34. The compound of clause 33, wherein R8 is selected from
35. The compound of clause 33, wherein R8 is -CH2C(O)OH. 36. The compound of any preceding clause, wherein the compound is selected from Compound ID Nos. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87 and 88.
37. The compound of clause 36, wherein the compound is selected from Compound ID Nos. 1, 2, 3,
4, 5, 6, 7, 8, 9, 10, 11, 12, 14, 15, 16, 17, 18, 19, 20, 21, 23, 24, 26, 28, 30, 31, 33, 37, 38, 39, 40, 41, 42,
43, 44, 45, 46, 47, 48, 49, 51, 52, 53, 54, 55, 56, 58, 59, 60, 61, 62, 63, 64, 65, 67, 68, 69, 70, 71, 72, 73,
75, 76, 77, 78, 79, 80, 81, 82, 84, 86 and 88.
38. The compound of any preceding clause, wherein:
R1 is H;
R3 is H or methyl;
R2 is aryl substituted with one or more R4 or is fused bicyclic heteroaryl substituted with one or more R6; and
R8 is -CH2C(O)OH or CHR9(heteroaryl).
39. The compound of clause 38, wherein: each R4 is independently selected from halogen, unsubstituted alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -NH2, -N(alkyl)2, -S(O)2(aryl), -S(O)2(heterocycloalkyl) and -C(O)OH; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R6 is -S(O)2(heterocycloalkyl); wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7
40. The compound of clause 39, wherein the compound is selected from Compound ID Nos. 1, 2, 3, 8, 10, 12, 13, 14, 15, 22, 24, 25, 27, 34, 37, 38, 41, 44, 45, 46, 47, 50, 59, 64, 72, 73, 75, 76, 77, 78, 79, 81, 82, 84 and 86.
41. The compound of clause 39, wherein R3 is H; each R4 is independently selected from -OH, -NH2, -O(alkyl) and -S(O)2(heterocycloalkyl); wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, and -
(OCH2CH2)rNHR7a; wherein q is 1 or 2, r is 4 and R7a is -C(O)Me.
42. The compound of clause 41, wherein the compound is Compound ID No. 2, 24, 37, 46, 47, 76, 77, 79, 82 or 86.
43. Use of a compound of any one of clauses 1-42 in a bifunctional protein degrader compound.
44. A bifunctional protein degrader compound comprising a compound of any one of clauses 1-42.
45. A bifunctional protein degrader compound comprising a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)27 -S(0)2alkyl, -S(0)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH20(alkyl), -CH2heterocycloalkyl, -CH2C(0)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from unsubstituted alkyl, haloalkyl, unsubstituted aryl, - C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl and R19; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CH2)q0Me, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19 and -OR19; wherein q is 1 or 2; r is 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -COOR9, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -B(OH)2, - P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and -C(O)OCH2CH2SO2l\/le; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alkylhcycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
46. A compound of formula (II)
[KLHDC2 ligase binding moiety] - linker - [Target protein binding moiety]
CD or a salt, solvate, hydrate or isomer thereof, wherein [Target protein binding moiety] is: wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl; or
(b) wherein indicates attachment to R18 of the linker; and k is an integer from 1-10; or
(c)
wherein
X4 and X5 are each independently N or CH;
X6 is N or CH;
R30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R40, or -C(O)R40;
R31 is H, -OMe, -heteroaryl, -heteroaryl-R40 or R40;
R32 is H, unsubstituted C1-C6 alkyl,
R33 is -N( C1-C6 alkyl)2, -NH(C1-C6 alkyl), -NH(aryl),
R34 is -Me or -C(O)R40; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; wherein
X7 is N or CH;
R35 is -heterocycloalkyl-R40, or R40;
R36 is H or -OMe; and R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from unsubstituted alkyl, haloalkyl, unsubstituted aryl, - C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl and R19; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19, -OR19 and -(OCH2CH2)rR19; wherein q is 1 or 2; r is 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -COOR9, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR9 2, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -B(OH)2, - P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)pNMe2, -C(O)O(CH2)pNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CMe2OH, and -C(O)OCH2CH2SO2Me; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkyl)2, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alkyl)2cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connected to R14 of the linker, wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and (c) when R1 and R3 are each H, then R2 is not halogen; and wherein [linker] has the following formula
R14.R15.R16.R17.R18 wherein
R14 is -Ci-6 alkyl, -C2.6 alkenyl, -C2.6alkynyl, C1-6 alkyl-N(C1-6 alkyl)-, -C(O)-, -SO2- or is absent
R15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, C1-6 alkyl-NH-, -C1-6 alkyl-N(C1-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
R16 is -Ci.6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH2-C(O)-, -CH2-C(O)-NH-, -CH2-C(O)O- or is absent R17 is -CH2(C2H4-O)y, (C2H4-O)X, (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
(a) when [Target protein binding moiety] is cycloalkyl, CH2-NH-C(O)-, heterocycloalkyl, or is absent; and (b) when [Target protein binding moiety] is
C(O)-NH-, heterocycloalkyl, heterocycloalkyl-NH, or is absent. The compound of any one of clauses 45-46, wherein X2 is N. 48. The compound of any one of clauses 45-46, wherein X3 is N.
49. The compound of any one of clauses 45-46, wherein X2 is CR3 and X3 is CR1.
50. The compound of any one of clauses 45-49, wherein R1 is H, halogen or methyl.
51. The compound of any one of clauses 45-50, wherein R3 is H, halogen, unsubstituted alkyl or -CN.
52. The compound of any one of clauses 45-51, wherein R1 is H.
53. The compound of clause 52, wherein R1 and R3 are each H.
54. The compound of clause 52, wherein R1 and R2 are each H.
55. The compound of any one of clauses 45-51, wherein R2 and R3 are each H.
56. The compound of any one of clauses 45-50, wherein
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
57. The compound of any one of clauses 45-53 and 56, wherein R2 is -B(OH)2, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6. 58. The compound of any one of clauses 45-53, 56 and 57, wherein R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
59. The compound of clause 58, wherein R2 is selected from wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3. 60. The compound of clause 59, wherein R2 is selected from n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
61. The compound of clause 60, wherein R2 is selected from
wherein n is 1, 2 or 3; and wherein m is 0 or 1. The compound of clause 61, wherein R2 is
63. The compound of any one of clauses 45-62, wherein each R4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), - O(cycloalkyl), -O(heterocycloalkyl), -NH2, -N (a lkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, - CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), -C(O)(heterocycloalkyl), -C(O)OH, R19 and -OR19; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
64. The compound of any one of clauses 45-63, wherein each R7 is independently selected from - Me, -OMe, -O(CH2)qOMe, -CH2OMe, -C(O)Me, -S(O)2Me, R19 and -OR19. 65. The compound of any one of clauses 45-64, wherein each R4 is independently selected from R19,
-OR19, -NHR19, -F, -Cl, -lBu, -Me, -CF3, -CN, -OH, -OMe, -OCF3, -NH2, -NMe2, -S(O)2Me, -S(O)2NMe2, -
CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
66. The compound of clause 65, wherein each R4 is independently selected from R19, -NHR19 -OR19,
67. The compound of any one of clauses 45-66, wherein each R6 is independently selected from haloalkyl, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl and R19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7. 68. The compound of any one of clauses 45-67, wherein each R6 is independently selected from -
CF3, -C(O)piperidine, -
69. The compound of any one of clauses 45-68, wherein R2 is selected from:
70. The compound of clause 69, wherein R2 is selected from:
71. The compound of any one of clauses 45-69, wherein: when
R8 is -CHR9C(O)OR10 , -CHR9C(O)NR92 or -CHR9(heterocycloalkyl), then: C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C- S(O)2(heterocycloalkyl), C-S(O)2aryl, C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
72. The compound of any one of clauses 45-69, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl), then: each R4 of - (~R4 /)n is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
73. The compound of any one of clauses 45-72, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH;
X2 and X3 are both CH; and
74. The compound of any one of clauses 45-72, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH or -CH^O^CH^CfOfBu;
X2 and X3 are both CH; and
(i) each is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; or
75. The compound of any one of clauses 45-51 and 71-74, wherein R1 is methyl.
76. The compound of any one of clauses 45-75, wherein -CHR9C(O)OR10 is
77. The compound of any one of clauses 45-73, wherein R8 is selected from -COOR9, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, - CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2.
78. The compound of clause 77, wherein R8 is selected from -COOR9, -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH2B(OH)2, -CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2. 79. The compound of clause 78, wherein R8 is selected from -COOH, -COOMe, -CH2C(O)OH, - CHMeC(O)OH, -CH2C(O)OMe, -CH2C(O)OEt, -CH2C(O)OCH2OC(O)tBu, -CH2C(O)NH2, -CH2S(O)2NH2, - S(O)2NH2, -CH2-oxetane, -CH2(l,2,3-triazole), -CH2B(OH)2, -CH2P(O)(OH)2, -B(OH)2 and -P(O)(OH)2.
80. The compound of clause 79, wherein R8 is selected from -CHjCfOjOCHjOCfOfBu, -CH2C(O)OH and -CHMeC(O)OH.
81. The compound of clause 80, wherein R8 is selected from
82. The compound of clause 80, wherein R8 is -CH2C(O)OH.
83. The compound of any one of clauses 45-82, wherein:
R1 and R3 are each H;
R2 is selected from aryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the fused aryl/heterocycloalkyl is unsubstituted or is substituted with one or more R6; and
R8 is -CHR9C(O)OR10.
84. The compound of clause 83, wherein R2 is aryl substituted with R4 and R8 is -CH2C(O)OR10
85. The compound of clause 83 or 84, wherein each R4 is S(O)2(heterocycloalkyl) substituted with R19 or OR19.
86. The compound of any one of clauses 46-85, wherein at least one of R14, R15, R16, R17 and R18 is present.
87. The compound of any one of clauses 46-86, wherein R14 is absent. 88. The compound of any one of clauses 46-86, wherein R14 is -C(O)-.
89. The compound of any one of clauses 46-88, wherein R15 is absent.
90. The compound of any one of clauses 46-88, wherein R15 is heterocycloalkyl.
91. The compound of any one of clauses 46-90, wherein R16 is absent.
92. The compound of any one of clauses 46-90, wherein R16 is -Ci.g alkyl.
93. The compound of any one of clauses 46-90, wherein R16 is -C(O)-.
94. The compound of any one of clauses 46-90, wherein R16 is -C(O)-NH-.
95. The compound of any one of clauses 46-94, wherein R17 is absent.
96. The compound of any one of clauses 46-94, wherein R17 is (Cjl- -Ojx.
97. The compound of any one of clauses 46-94 and 96, wherein x is 3-9.
98. The compound of clause 97, wherein x is 3, 5, 7 or 9.
99. The compound of any one of clauses 46-98, wherein R18 is absent.
100. The compound of any one of clauses 46-98, wherein R18 is -Ci-io alkyl or -Ci-io alkyl-NH-.
101. The compound of clause 100, wherein R18 is -Ci.g alkyl or -Ci.g alkyl-NH-; optionally wherein R18 is
-CH2CH2- or -CH2CH2-NH-.
102. The compound of any one of clauses 46-98, wherein R18 is heterocycloalkyl or heterocycloalkyl-
NH-. 103. The compound of any one of clauses 46-85, wherein R14, R15, R16, R17 and R18 are each absent.
104. The compound of any one of clauses 46-102, wherein [linker] is
_,1 wherein indicates attachment to [KLHDC2 ligase binding moiety] indicates attachment to [Target protein binding moiety]. The compound of any one of clauses 46-104, wherein the [Target protein binding moiety] is: wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl. The compound of any one of clauses 46-105, wherein R12 is Me. The compound of any one of clauses 46-105, wherein R12 is H. The compound of clause 105, wherein [Target protein binding moiety] is: The compound of clause 108, wherein [Target protein binding moiety] is: ses 46-109, wherein M is absent. ses 46-110, wherein L' is H. erein [Target protein binding moiety] is: ses 46-104, wherein [Target protein binding moiety] is: ses 46-104, wherein [Target protein binding moiety] is:
115. The compound of any one of clauses 46-104, wherein [Target protein binding moiety] is:
116. The compound of clause 115, wherein [Target protein binding moiety] is:
117. The compound of clause 116, wherein [Target protein binding moiety] is: 118. The compound of clause 116, wherein [Target protein binding moiety] is:
119. The compound of any one of clauses 115-118, wherein [linker] is
-,1 2 wherein indicates attachment to [KLHDC2 ligase binding moiety] indicates attachment to [Target protein binding moiety].
120. The compound of any one of clauses 46-119, wherein k is an integer from 3-8; optionally wherein k is 4, 6, or 8; further optionally wherein k is 6.
121. The compound of any one of clauses 46-120, wherein the compound is selected from Compound ID Nos. 1001, 1002, 1003, 1004, 1005, 1006, 1007, 1008, 1009, 1010, 1011, 1012, 1013, 1014, 1015, 1016, 1017, 1018, 1019, 1020, 1021, 1022, 1023, 1024 and 1025. 122. The compound of clause 121, wherein the compound is selected from Compound ID Nos. 1001, 1002 and 1003.
123. The compound of clause 121, wherein the compound is selected from Compound ID Nos. 1002, 1003, 1005, 1007, 1008, 1010, 1012, 1014, 1016, 1019, 1020 and 1022
124. The compound of clause 123, wherein the compound is selected from Compound ID Nos. 1003,
1008, 1010, 1012, 1014, 1016, 1020 and 1022,
125. The compound of clause 123, wherein the compound is selected from Compound ID Nos. 1005 and 1007.
126. The compound of clause 121, wherein the compound is selected from Compound ID Nos. 1008,
1009, 1010, 1011, 1012, 1013, 1014, 1015, 1016, 1017, 1018 and 1019.
127. The compound of clause 121, wherein the compound is selected from Compound ID Nos. 1024 and 1025.
128. A pharmaceutical composition comprising a compound of any one of clauses 1-127.
129. The compound of any one of clauses 1-127 or the pharmaceutical composition of clause 128 for use in medicine.

Claims

1. A compound of formula (la): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, -
NH(alkyl), -NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), and -P(O)(alkyl)2; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl or heteroaryl, each of which is independently unsubstituted or is substituted with at least one of -OH, -NH2, -NH(alkyl) or -N (alkyl )2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -0(CH2)q0Me, -(CFbJqOIVIe, -(OCHzCFbJrNHR73, -C(O)alkyl and -S(O)2alkyl; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or - C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2 -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and ^(OjOCHjCHjSOjIVIe; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkyl)2, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
2. The compound of claim 1, wherein X2 is N.
3. The compound of claim 1, wherein X3 is N.
4. The compound of claim 1, wherein X2 is CR3 and X3 is CR1.
5. The compound of any preceding claim, wherein R1 is H, halogen or methyl.
6. The compound of any preceding claim, wherein R3 is H, halogen, unsubstituted alkyl or -CN.
7. The compound of any preceding claim, wherein R1 is H.
8. The compound of claim 7, wherein R1 and R3 are each H.
9. The compound of claim 7, wherein R1 and R2 are each H.
10. The compound of any one of claims 1-6, wherein R2 and R3 are each H.
11. The compound of any one of claims 1-6, wherein
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
12. The compound of any one of claims 1-8 and 11, wherein R2 is -B(OH)2, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl or benzyl; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
13. The compound of any one of claims 1-8, 11 and 12, wherein R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6. n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
15. The compound of claim 14, wherein R2 is selected from wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
16. The compound of claim 15, wherein R2 is selected from
wherein n is 1, 2 or 3; and wherein m is 0 or 1.
17. The compound of claim 16, wherein R2 is
18. The compound of any preceding claim, wherein each R4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), - O(heterocycloalkyl), -NH2, -N(alkyl)z, -S(O)zalkyl, -S(O)zaryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), - S(O)2(heterocycloalkyl), -C(O)(heterocycloalkyl), and -C(O)OH; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
19. The compound of any preceding claim, wherein each R7 is independently selected from -Me, - OMe, -O(CH2)qOMe, -CH2OMe, -C(O)Me and -S(O)2Me.
20. The compound of any preceding claim, wherein each R4 is independently selected from -F, -Cl, - lBu, -Me, -CF3, -OH, -OMe, -OCF3, -CN, -NH2, -NMe2, -NHC(O)Me, -S(O)2Me, -S(O)2NMe2, -CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
21. The compound of any preceding claim, wherein each R6 is independently selected from haloalkyl, halogen, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl) and -C(O)alkyl; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
22. The compound of any preceding claim, wherein each R6 is independently selected from -CF3, -Cl,
-NH2, -C(O)piperidine, -
23. The compound of any preceding claim, wherein R2 is selected from:
24. The compound of any preceding claim, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C- S(O)2(heterocycloalkyl), C-S(O)2aryl, C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
25. The compound of any one of claims 1-23, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then: each R4 of — r ' R4) is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, -
NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, -C(O)NH(alkyl), -SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
26. The compound of any one of claims 1-25, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl);
27. The compound of any one of claims 1-25, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); n is 1 or 2; then:
R8 is -CH2C(O)OH or -CH^O^CH^CfOfBu;
X2 and X3 are both CH; and
(i) each is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; or
28. The compound of any one of claims 1-6 and 10-27, wherein R1 is methyl.
29. The compound of any preceding claim, wherein -
30. The compound of any preceding claim, wherein R8 is selected from -CHR9C(O)OR10, - CHR9C(O)NR9 2, -CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, - CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2.
31. The compound of claim 30, wherein R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, - CH2S(O)2NR9 2, -S(O)2NR9 2, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2.
32. The compound of claim 31, wherein R8 is -CHR9C(O)OR10.
33. The compound of claim 31, wherein R8 is selected from - -CH2C(O)OH, -CHMeC(O)OH, - CH2C(O)OMe, -CH2C(O)OEt, -CH^fOjOCH^CfOfBu, -CH2C(O)NH2, -CH2S(O)2NH2, -S(O)2NH2, -CH2- oxetane, -CH2(l,2,3-triazole), -CH2B(OH)2, -CH2P(O)(OH)2, COOH, -COOMe, -B(OH)2 and -P(O)(OH)2.
34. The compound of claim 32 or 33, wherein R8 is selected from -CH2C(O)OCH2OC(O)tBu, -
CH2C(O)OH and -CHMeC(O)OH. 35. The compound of claim 34, wherein R8 is selected from
36. The compound of claim 35, wherein R8 is -CH2C(O)OH.
37. The compound of any preceding claim, wherein the compound is selected from:
38. The compound of claim 37, wherein the compound is selected from:
R1 is H;
R3 is H or methyl;
R2 is aryl substituted with one or more R4 or is fused bicyclic heteroaryl substituted with one or more R6; and
R8 is -CH2C(O)OH, -CH(Me)C(O)OH, or CHR9(heteroaryl).
40. The compound of claim 39, wherein: each R4 is independently selected from halogen, unsubstituted alkyl, haloalkyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -NH2, -N(alkyl)z, -NHC(O)alkyl, -C(O)NHalkyl, -C(O)N(alkyl)2, - S(O)2N(alkyl)2, -S(0)2(aryl), -S(O)2(heterocycloalkyl) and -C(O)OH; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R6 is -S(O)2(heterocycloalkyl), alkyl or -NH2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7
1. The compound of claim 40, wherein the compound is selected from:
42. The compound of claim 40, wherein
R3 is H; each R4 is independently selected from -OH, -NHz, -O(alkyl), -NHC(O)alkyl, -C(O)NHalkyl, - S(O)z(heterocycloalkyl) and -C(O)OH; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from -O(alkyl), -O(CHz)qOI\/le, -(CHz)qOI\/le, and - (OCH2CHz)rNHR7a; wherein q is 1 or 2, r is 4 and R7a is -C(O)Me.
43. The compound of claim 42, wherein the compound is:
44. Use of a compound of any one of claims 1-43 in a bifunctional protein degrader compound.
45. A bifunctional protein degrader compound comprising a compound of any one of claims 1-43.
46. A bifunctional protein degrader compound comprising a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2;
R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)2, - NH(alkyl), -NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)z, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19 and -OR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR92, -S(O)2NR92, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, -C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CI\/le2OH, and -qojOCHjCHjSOjMe; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with 0P(0)(0H)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - 0C(0)0(CH2)mS(0)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - 0C(0)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alkyl)2cycloalkyl, N-(unsubstituted C1-C6alkyl)piperidinium cation, N- (unsubstituted C1-C6alkyl)morpholinium cation, and N-(unsubstituted C1-C6alkyl)imidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(O)NMe2;
R19 is a bond connecting the compound of formula (la') to a Target protein binding moiety or to a linker, wherein the linker is attached to a Target protein binding moiety, and wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen.
47. A compound of formula (II)
[KLHDC2 ligase binding moiety] - linker - [Target protein binding moiety] CD or a salt, solvate, hydrate or isomer thereof, wherein [Target protein binding moiety] is:
(a)
wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl;
(b) wherein indicates attachment to R18 of the linker; and k is an integer from 1-10;
(c)
wherein
X4 and X5 are each independently N or CH;
X6 is N or CH;
R30 is H, halogen, -OMe, -CN, unsubstituted C1-C6 alkyl, -CECH, R40, or -C(O)R40;
R31 is H, -OMe, -heteroaryl, -heteroaryl-R40 or R40;
R32 is H, unsubstituted C1-C6 alkyl, R33 is -N(C1-C6 alkyl)2, -NH(C1-C6 alkyl), -NH(aryl), or R40;
R34 is -Me or -C(O)R40; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; or wherein
X7 is N or CH;
R35 is -heterocycloalkyl-R40, or R40;
R36 is H or -OMe; and
R40 is a bond connected to R18 of the linker, wherein the [Target protein binding moiety] contains a single R40; wherein [KLHDC2 ligase binding moiety] is a compound of formula (la'): wherein:
X2 is N or CR3;
X3 is N or CR1; wherein when X2 is N, then X3 is CR1; and when X3 is N, then X2 is CR3;
R1 is H, halogen, haloalkyl, methyl, -OH or -NH2; R3 is H, halogen, haloalkyl, unsubstituted alkyl, -OH, -O(alkyl), -C(O)NH(alkyl), -N (alkyl)z, - NH(alkyl),- NH2 or -CN;
R2 is H, -B(OH)2, halogen, -CN, -NR5 2, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6; each R4 is independently selected from halogen, -CN, unsubstituted alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), -O(cycloalkyl), -O(heterocycloalkyl), -NH2, - N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2O(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -C(O)OH, - C(O)NH(alkyl), -C(O)N(alkyl)2, -SO(alkyl), -P(O)(alkyl)2, R19, -NHR19 and -OR19; wherein each aryl, cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; each R5 is independently selected from alkyl, aryl, heteroaryl and R19, wherein the alkyl, aryl and heteroaryl are independently unsubstituted or are substituted with at least one of -OH, -NH2, -NH(alkyl) or -N(alkyl)2; each R6 is independently selected from halogen, unsubstituted alkyl, haloalkyl, unsubstituted aryl, -NH2, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and -NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; and each R7 is independently selected from unsubstituted alkyl, -O(alkyl), -O(CH2)qOMe, -(CH2)qOMe, -(OCH2CH2)rNHR7a, -C(O)alkyl, -S(O)2alkyl, R19, -OR19 and -(OCH2CH2)rR19; wherein q is 1 or 2; r is 0, 1, 2, 3, 4 or 5; and R7a is H or -C(O)alkyl;
R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR9 2, - CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, -CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR10, - B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)pNMe2, -C(O)O(CH2)pNHMe, -C(O)OCH2CH(OH)CH2OH, - C(O)OCH2CH2CMe2OH, and -C(O)OCH2CH2SO2Me; wherein each Rp is independently -(CH2)mOC(O)(unsubstituted C1-C6alkyl), wherein each m is independently 1, 2, 3 or 4; each p is independently 2, 3, 4, 5, or 6; each R9 is independently H or unsubstituted alkyl; and each R10 is independently H, unsubstituted alkyl, or C1-C6alkyl substituted by one or more groups selected from -OC(O)(unsubstituted C1-C6alkyl), -OC(O)(C1-C6alkyl substituted with at least one R10a), - OC(O)cycloalkyl, -OC(O)heterocycloalkyl, -OC(O)aryl, -OC(O)heteroaryl, - OC(O)(CH2)mO(CH2)mO(unsubstituted C1-C6alkyl), -OC(O)(CH2)mO(CH2CH2O)m(unsubstituted C1-C6alkyl), - OC(O)O(unsubstituted C1-C6alkyl), -OC(O)O(C1-C6alkyl substituted with at least one R10a), - OC(O)Ocycloalkyl, -OC(O)O(CH2CH2O)m(unsubstituted C1-C6alkyl), -OC(O)O(CH2CH2O)m(C1-C6alkyl substituted with OP(O)(OH)2), -OC(O)O(CH2CH2S)m(unsubstituted C1-C6alkyl), - OC(O)O(CH2)mS(O)(unsubstituted C1-C6alkyl), -OC(O)O(CH2)mS(O)2(unsubstituted C1-C6alkyl), - OC(O)NH(C1-C6alkyl substituted with OMe), -OC(O)NMe(C1-C6alkyl substituted with OMe), -P(O)(OH)2, - OP(O)(OH)2, -C(O)heterocycloalkyl, -C(O)N(unsubstituted C1-C6alkylh, heteroaryl, heterocycloalkyl, - S(unsubstituted C1-C6alkyl), -S(O)(unsubstituted C1-C6alkyl), -N+(unsubstituted C1-C6alkyljs, - N+(unsubstituted C1-C6alky^cycloalkyl, N-(unsubstituted C1-C6alkyljpiperidinium cation, N- (unsubstituted C1-C6alkyljmorpholinium cation, and N-(unsubstituted C1-C6alkyljimidazolium cation; wherein each R10a is independently -OH, -O(unsubstituted C1-C6alkyl), -OBn, -NH2, -NMe2, - NHC(O)(unsubstituted C1-C6alkyl), -NHC(O)O(unsubstituted C1-C6alkyl), heterocycloalkyl, heteroaryl, aryl, -C(0)NMe2;
R19 is a bond connected to R14 of the linker, wherein formula (la') contains a single R19; and wherein:
(a) at least one of R1, R2 and R3 is not H;
(b) when R2 and R3 are each H, then R1 is halogen; and
(c) when R1 and R3 are each H, then R2 is not halogen; and wherein [linker] has the following formula
R14.R15.R16.R17.R18 wherein
R14 is -C1-6 alkyl, -C2-6 alkenyl, -C2-6alkynyl, Ci.g alkyl-N(Ci-6 alkyl)-, -C(O)-, -SO2- or is absent
R15 is cycloalkyl, heterocycloalkyl, aryl, heteroaryl, Ci.g alkyl-NH-, -Ci.g alkyl-N(Ci-6 alkyl)-, - cycloalkyl-NH-, -heterocycloalkyl-NH- or is absent
R16 is -C1-6 alkyl, -C(O)-, -C(O)-NH-, -C(O)O-, -CH2-C(O)-, -CH2-C(O)-NH-, -CH2-C(O)O- or is absent
R17 is -CH2(C2H4-O)y, (C2H4-O)X, (CaHg-Ojx, or is absent x is 1-10 y is 2-10; and wherein
(a) when [Target protein binding moiety] is cycloalkyl, CH2-NH-C(O)-, heterocycloalkyl, or is absent; and
(b) when [Target protein binding moiety] is
C(O)-NH-, heterocycloalkyl, heterocycloalkyl-NH, or is absent.
48. The compound of any one of claims 46-47, wherein X2 is N.
49. The compound of any one of claims 46-47, wherein X3 is N.
50. The compound of any one of claims 46-47, wherein X2 is CR3 and X3 is CR1.
51. The compound of any one of claims 46-50, wherein R1 is H, halogen or methyl.
52. The compound of any one of claims 46-51, wherein R3 is H, halogen, unsubstituted alkyl or -CN.
53. The compound of any one of claims 46-52, wherein R1 is H.
54. The compound of claim 53, wherein R1 and R3 are each H.
55. The compound of claim 53, wherein R1 and R2 are each H.
56. The compound of any one of claims 46-52, wherein R2 and R3 are each H.
57. The compound of any one of claims 46-51, wherein
R3 is H,
R1 is halogen or methyl, and
R2 is -B(OH)2, halogen, -CN, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
58. The compound of any one of claims 46-54 and 57, wherein R2 is -B(OH)2, -NR52, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the cycloalkyl, aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heterocycloalkyl, heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
59. The compound of any one of claims 46-5457 and 58, wherein R2 is selected from aryl, heteroaryl, fused bicyclic heteroaryl, fused aryl/heterocycloalkyl, benzyl or R19; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the heteroaryl, fused bicyclic heteroaryl, and fused aryl/heterocycloalkyl are unsubstituted or are substituted with one or more R6.
60. The compound of claim 59, wherein R2 is selected from
wherein n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
61. The compound of claim 60, wherein R2 is selected from
n is 0, 1, 2, 3, 4 or 5; m is 0, 1, 2, 3 or 4; and p is 0, 1, 2 or 3.
62. The compound of claim 61, wherein R2 is selected from wherein n is 1, 2 or 3; and wherein m is 0 or 1.
63. The compound of claim 62, wherein R2 is
64. The compound of any one of claims 46-63, wherein each R4 is independently selected from halogen, -CN, alkyl, haloalkyl, alkynyl, cycloalkyl, heterocycloalkyl, -OH, -O(alkyl), -O(haloalkyl), - O(cycloalkyl), -O(heterocycloalkyl), -NH2, -N (a lkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, - CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), -C(O)(heterocycloalkyl), -C(O)OH, R19 and -OR19; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
65. The compound of any one of claims 46-64, wherein each R7 is independently selected from -Me, -OMe, -O(CH2)qOMe, -CH2OMe, -C(O)Me, -S(O)2Me, R19 and -OR19.
66. The compound of any one of claims 46-65, wherein each R4 is independently selected from R19, - OR19, -NHR19, -F, -Cl, -lBu, -Me, -CF3, -CN, -OH, -OMe, -OCF3, -NH2, -NMe2, -S(O)2Me, -S(O)2NMe2, -
CH2S(O)2NHMe, cyclopropyl, -C(O)OH,
67. The compound of claim 66, wherein each R4 is independently selected from R19, -NHR19, -OR19,
68. The compound of any one of claims 46-67, wherein each R6 is independently selected from haloalkyl, -C(O)(heterocycloalkyl), -S(O)2(heterocycloalkyl), -C(O)alkyl, R19 and NHR19; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
69. The compound of any one of claims 46-68, wherein each R6 is independently selected from -CF3,
-C(O)piperidine, -
70. The compound of any one of claims 46-69, wherein R2 is selected from:
71. The compound of claim 70, wherein R2 is selected from:
72. The compound of any one of claims 46-70, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl), then: C4-R4 is selected from C-alkynyl, C-S(O)2alkyl, C-S(O)2N(alkyl)2, C-CH2S(O)2NH(alkyl), C- S(O)2(heterocycloalkyl), C-S(O)2aryl, C-CH2heterocycloalkyl, C-CH2C(O)NH(alkyl), C-NHC(O)(alkyl), C- SO(alkyl) and C-P(O)(alkyl)2; wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
73. The compound of any one of claims 46-70, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl), then: each R4 of - (~R4 /)n is independently selected from -CN, alkynyl, cycloalkyl, heterocycloalkyl, - NH2, -N(alkyl)2, -S(O)2alkyl, -S(O)2aryl, -S(O)2N(alkyl)2, -CH2S(O)2NH(alkyl), -S(O)2(heterocycloalkyl), - C(O)(heterocycloalkyl), -NHC(O)(alkyl), -CH2heterocycloalkyl, -CH2C(O)NH(alkyl), -COOH, -C(O)NH(alkyl), - SO(alkyl), and -P(O)(alkyl)2; wherein each cycloalkyl and heterocycloalkyl is independently unsubstituted or is substituted with at least one R7.
74. The compound of any one of claims 46-73, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR92 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH;
X2 and X3 are both CH; and
75. The compound of any one of claims 46-73, wherein: when
R8 is -CHR9C(O)OR10, -CHR9C(O)NR9 2 or -CHR9(heterocycloalkyl); then:
R8 is -CH2C(O)OH or -CH^O^CH^CfOfBu;
X2 and X3 are both CH; and is -S(O)2(heterocycloalkyl), wherein each heterocycloalkyl is independently unsubstituted or is substituted with at least one R7; or
76. The compound of any one of claims 46-52 and 72-75, wherein R1 is methyl.
11. The compound of any one of claims 46-76, wherein -CHR9C(O)OR10 is
78. The compound of any one of claims 46-74, wherein R8 is selected from -CHR9C(O)OR10, - CHR9C(O)NR9 2, -CHR9S(O)2NR9 2, -S(O)2NR92, -CHR9(heterocycloalkyl), -CHR9(heteroaryl), -CHR9B(OH)2, CHR9P(O)(OH)2, -CHR9P(O)(ORP)2, -COOR9, -B(OH)2, -P(O)(OH)2, -P(O)(ORP)2, -C(O)O(CH2)PNMe2, - C(O)O(CH2)PNHMe, -C(O)OCH2CH(OH)CH2OH, -C(O)OCH2CH2CI\/le2OH, and -C(O)OCH2CH2SO2l\/le; or wherein R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR9 2, -CH2S(O)2NR92, -S(O)2NR9 2, - CH2(heterocycloalkyl), -CH2(heteroaryl), -CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2.
79. The compound of claim 78, wherein R8 is selected from -CHR9C(O)OR10, -CHR9C(O)NR92, -
CH2S(O)2NR92, -S(O)2NR92, -CH2(4- or 5-membered heterocycloalkyl), -CH2(5-membered heteroaryl), - CH2B(OH)2, -CH2P(O)(OH)2, -COOR9, -B(OH)2 and -P(O)(OH)2.
80. The compound of claim 79, wherein R8 is -CHR9C(O)OR10,
81. The compound of claim 80, wherein R8 is selected from -CH2C(O)OH, -CHMeC(O)OH, - CH2C(O)OMe, -CH2C(O)OEt, -CH2C(O)OCH2OC(O)tBu, -CH2C(O)NH2, -CH2S(O)2NH2, -S(O)2NH2, -CH2- oxetane, -CH2(l,2,3-triazole), -CH2B(OH)2, -CH2P(O)(OH)2, -COOH, -COOMe, -B(OH)2 and -P(O)(OH)2.
82. The compound of claim 80 or 81, wherein R8 is selected from -CH2C(O)OCH2OC(O)lBu, -
CH2C(O)OH and -CHMeC(O)OH.
83. The compound of claim 82, wherein R8 is selected from
84. The compound of claim 82, wherein R8 is -CH2C(O)OH.
85. The compound of any one of claims 46-84, wherein:
R1 and R3 are each H;
R2 is selected from aryl, fused aryl/heterocycloalkyl and benzyl; wherein the aryl and benzyl are unsubstituted or are substituted with one or more R4; and wherein the fused aryl/heterocycloalkyl is unsubstituted or is substituted with one or more R6; and
R8 is -CHR9C(O)OR10.
86. The compound of claim 85, wherein R2 is aryl substituted with R4 and R8 is -CH2C(O)OR10
87. The compound of claim 85 or 86, wherein each R4 is S(O)2(heterocycloalkyl) substituted with R19 or OR19.
88. The compound of any one of claims 47-88, wherein at least one of R14, R15, R16, R17 and R18 is present.
89. The compound of any one of claims 47-88, wherein R14 is absent.
90. The compound of any one of claims 47-88, wherein R14 is -C(O)-.
91. The compound of any one of claims 47-90, wherein R15 is absent.
92. The compound of any one of claims 47-90, wherein R15 is heterocycloalkyl.
93. The compound of any one of claims 47-92, wherein R16 is absent.
94. The compound of any one of claims 47-92, wherein R16 is -Ci.g alkyl.
95. The compound of any one of claims 47-92, wherein R16 is -C(O)-.
96. The compound of any one of claims 47-92, wherein R16 is -C(O)-NH-.
97. The compound of any one of claims 47-96, wherein R17 is absent.
98. The compound of any one of claims 47-96, wherein R17 is (C2H4-O)x.
99. The compound of any one of claims 47-96and 98, wherein x is 3-9.
100. The compound of claim 99, wherein x is 3, 5, 7 or 9.
101. The compound of any one of claims 47-100, wherein R18 is absent.
102. The compound of any one of claims 47-100, wherein R18 is -Ci-io alkyl or -Ci-w alkyl-N H-.
103. The compound of claim 102, wherein R18 is -Ci.g alkyl or -Ci.g alkyl-N H-; optionally wherein R18 is -
CH2CH2- or -CH2CH2-NH-.
104. The compound of any one of claims 47-100, wherein R18 is heterocycloalkyl or heterocycloalkyl-
NH-.
105. The compound of any one of claims 47-87, wherein R14, R15, R16, R17 and R18 are each absent.
106. The compound of any one of claims 47-104, wherein [linker] is
rT' ein 1 rvT wher indicates attachment to [KLHDC2 ligase binding moiety] and 2 indicates attachment to [Target protein binding moiety].
107. The compound of any one of claims 47-106, wherein the [Target protein binding moiety] is: wherein
M is O, S or NH, or is absent; indicates attachment to R18 of the linker;
R11 is H, halogen, -OMe, an amino group, heterocycloalkyl, or unsubstituted C1-C6 alkyl;
R12 is H or Me; and
L' is H, alkyl, benzyl, acetyl or pivaloyl.
108. The compound of any one of claims 47-107, wherein R12 is Me.
109. The compound of any one of claims 47-107, wherein R12 is H.
110. The compound of claim 107, wherein [Target protein binding moiety] is:
111. The compound of claim 110, wherein [Target protein binding moiety] is:
112. The compound of any one of claims 47-111, wherein M is absent.
113. The compound of any one of claims 47-112, wherein L' is H.
114. The compound of claim 113, wherein [Target protein binding moiety] is:
115. The compound of any one of claims 47-106, wherein [Target protein binding moiety] is:
116. The compound of any one of claims 47-106, wherein [Target protein binding moiety] is:
117. The compound of any one of claims 47-106, wherein [Target protein binding moiety] is:
118. The compound of claim 117, wherein [Target protein binding moiety] is:
119. The compound of claim 118, wherein [Target protein binding moiety] is:
120. The compound of claim 118, wherein [Target protein binding moiety] is:
121. The compound of any one of claims 117-120, wherein [linker] is
~,1 2 wherein indicates attachment to [KLHDC2 ligase binding moiety] indicates attachment to [Target protein binding moiety].
122. The compound of any one of claims 47-121, wherein k is an integer from 3-8; optionally wherein k is 4, 6, or 8; further optionally wherein k is 6.
123. The compound of any one of claims 47-123, wherein the compound is selected from:
124. The compound of claim 123, wherein the compound is selected from:
125. The compound of claim 123, wherein the compound is selected from:
26. The compound of claim 125, wherein the compound is selected from:
27. The compound of claim 125, wherein the compound is selected from:
129. The compound of claim 123, wherein the compound is selected from
130. The compound of claim 123, wherein the compound is selected from
131. A pharmaceutical composition comprising a compound of any one of claims 1-130.
132. The compound of any one of claims 1-130 or the pharmaceutical composition of claim 131 for use in medicine.
PCT/EP2025/060749 2024-04-17 2025-04-17 Klhdc2 (kelch domain-containing protein 2) ligase ligands Pending WO2025219579A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PCT/PL2024/000014 WO2025221154A1 (en) 2024-04-17 2024-04-17 Klhdc2 (kelch domain-containing protein 2) ligase ligands
PLPCT/PL2024/000014 2024-04-17

Publications (1)

Publication Number Publication Date
WO2025219579A1 true WO2025219579A1 (en) 2025-10-23

Family

ID=91301990

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/PL2024/000014 Pending WO2025221154A1 (en) 2024-04-17 2024-04-17 Klhdc2 (kelch domain-containing protein 2) ligase ligands
PCT/EP2025/060749 Pending WO2025219579A1 (en) 2024-04-17 2025-04-17 Klhdc2 (kelch domain-containing protein 2) ligase ligands

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/PL2024/000014 Pending WO2025221154A1 (en) 2024-04-17 2024-04-17 Klhdc2 (kelch domain-containing protein 2) ligase ligands

Country Status (1)

Country Link
WO (2) WO2025221154A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140031559A1 (en) 2011-12-16 2014-01-30 State of Oregon, acting by and through the State Board of Higher Education on Behalf of University Tunable phenylacetylene hosts
US20150259357A1 (en) 2013-02-04 2015-09-17 Janssen Pharmaceutica Nv Flap modulators
US20170008904A1 (en) 2015-07-10 2017-01-12 Arvinas, Inc. Mdm2-based modulators of proteolysis and associated methods of use
WO2017011590A1 (en) 2015-07-13 2017-01-19 Arvinas, Inc. Alanine-based modulators of proteolysis and associated methods of use
WO2017197051A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Amine-linked c3-glutarimide degronimers for target protein degradation
WO2018217809A1 (en) 2017-05-23 2018-11-29 Abide Therapeutics, Inc. Pyrazole magl inhibitors
WO2019238816A1 (en) 2018-06-13 2019-12-19 University Of Dundee Bifunctional molecules for targeting uchl5
WO2021077010A1 (en) 2019-10-17 2021-04-22 Arvinas Operations, Inc. Bifunctional molecules containing an e3 ubiquitine ligase binding moiety linked to a bcl6 targeting moiety
WO2023192578A1 (en) 2022-03-31 2023-10-05 Kymera Therapeutics, Inc. Protein degraders and uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015012328A1 (en) * 2013-07-24 2015-01-29 武田薬品工業株式会社 Heterocyclic compound
US20230069104A1 (en) * 2019-06-28 2023-03-02 Kymera Therapeutics, Inc. Irak degraders and uses thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140031559A1 (en) 2011-12-16 2014-01-30 State of Oregon, acting by and through the State Board of Higher Education on Behalf of University Tunable phenylacetylene hosts
US20150259357A1 (en) 2013-02-04 2015-09-17 Janssen Pharmaceutica Nv Flap modulators
US20170008904A1 (en) 2015-07-10 2017-01-12 Arvinas, Inc. Mdm2-based modulators of proteolysis and associated methods of use
WO2017011590A1 (en) 2015-07-13 2017-01-19 Arvinas, Inc. Alanine-based modulators of proteolysis and associated methods of use
WO2017197051A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Amine-linked c3-glutarimide degronimers for target protein degradation
WO2018217809A1 (en) 2017-05-23 2018-11-29 Abide Therapeutics, Inc. Pyrazole magl inhibitors
WO2019238816A1 (en) 2018-06-13 2019-12-19 University Of Dundee Bifunctional molecules for targeting uchl5
WO2021077010A1 (en) 2019-10-17 2021-04-22 Arvinas Operations, Inc. Bifunctional molecules containing an e3 ubiquitine ligase binding moiety linked to a bcl6 targeting moiety
WO2023192578A1 (en) 2022-03-31 2023-10-05 Kymera Therapeutics, Inc. Protein degraders and uses thereof

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
CHEN, H ET AL., J. AM. CHEM. SOC, vol. 143, 2021, pages 15073
CHEN, H ET AL., J. AM. CHEM. SOC., vol. 143, 2021, pages 15073
FUJIHARA, T ET AL., ANGEW. CHEM. INT. ED, vol. 51, 2012, pages 11487
GAO, S ET AL., J. ORG. CHEM., vol. 83, 2018, pages 9250
HICKEY CH.M.DIGIANANTONIO K.M.ZIMMERMANN K.HARBIN A.QUINN C.PATEL A.GAREISS P.CHAPMAN A.TIBERI B.DOBRODZIEJ J.: "Co-opting the E3 ligase KLHDC2 for targeted protein degradation by small molecules", NAT. STRUCT. MOL. BIOL., vol. 31, 2023, pages 311 - 322, XP093256737, DOI: 10.1038/s41594-023-01146-w
KIM Y.SEO CJEON E.YOU I.HWANG K.KIM N.CHOI H.S.HINSHAW S.M.GRAY N.SSIM T.: "Targeted Kinase Degradation via the KLHDC2 Ubiquitin E3 Ligase", BIORXIV, 2022
KOPKA, I.E ET AL., BIOMOL
POIRSON J.DHILLON A.CHO H.LAM M.H.Y.ALERASOOL NLACOSTE J.MIZAN LTAIPALE M.: "Proteome-scale induced proximity screens reveal highly potent protein degraders and stabilizers", BIORXIV, 2022
RÖTH S.CARTON B.SATHYAMURTHI P.S.WATT M.MACARTNEY T.J.CHAN K.H.ISIDRO-LLOBET A.KONOPACKA A.QUEISSER M.ASAPKOTA G: "Screening of E3 Ligases Uncovers KLHDC2 as an Efficient Proximity-Induced Degrader of K-RAS, STK33, β-catenin and FoxP3", STK33, Β-CATENIN AND FOXP3
RUSNAC D.V.LIN H.C.CANZANI D.TIEN K.X.HINDS T.R.TSUE A.F.BUSH M.F.YEN H.C.SZHENG N.: "Recognition of the diglycine C-end degron by CRL2KLHDC2 ubiquitin ligase", MOL. CELL, vol. 72, no. 5, 2018, pages 813 - 822

Also Published As

Publication number Publication date
WO2025221154A1 (en) 2025-10-23

Similar Documents

Publication Publication Date Title
AU2008296974B2 (en) Soluble guanylate cyclase activators
JP4423043B2 (en) Rho-kinase inhibitor
KR102400920B1 (en) Inhibitors of lysine specific demethylase-1
US10662187B2 (en) Bruton&#39;s tyrosine kinase inhibitors
RU2734760C2 (en) Anti-infectious compounds
ES2821821T3 (en) Tetrahydroisoquinoline derivatives
US20050245520A1 (en) 2-Phenylpyridin-4-yl derivatives as alk5 inhibitors
AU2014315109A1 (en) Spirocyclic compounds as tryptophan hydroxylase inhibitors
KR101721739B1 (en) Kinase modulating compounds, compositions containing the same and use thereof
JP7299167B2 (en) Syk inhibitor and method of use thereof
CN111196804B (en) TGF-beta R1 inhibitors and uses thereof
CA2908098A1 (en) Mk2 inhibitors and uses thereof
FR3000492A1 (en) NOVEL AZAINDOLE DERIVATIVES AS MULTIKINASE INHIBITORS
US10550080B2 (en) Acyl sulfonamide NaV1.7 inhibitors
US20060058329A1 (en) Pyrazole inhibitors of the transforming growth factor
MX2010010018A (en) Novel hsp90 inhibitory carbazole derivatives, compositions containing same, and use thereof.
WO2012159557A1 (en) Substituted triazine phenylurea derivative and use thereof against tumors
ES2468221T3 (en) Benzimidazole derivatives and pharmaceutical compositions thereof
US20230227429A1 (en) Sulphonamide compounds
KR20250112837A (en) Modulator of TNF-α activity
TW201319063A (en) Medicinal agent comprising thiazolidine derivative or salt thereof as active ingredient
CN109641909A (en) The mechanism target and its treatment use of rapamycin signal pathway inhibitor
KR20190022728A (en) Mitochondrial inhibitors for the treatment of proliferative disorders
KR20240151737A (en) Bifunctional sulfonamide compounds
WO2025219579A1 (en) Klhdc2 (kelch domain-containing protein 2) ligase ligands

Legal Events

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

Ref document number: 25719437

Country of ref document: EP

Kind code of ref document: A1