Movatterモバイル変換


[0]ホーム

URL:


WO2012034095A1 - Compounds and compositions as trk inhibitors - Google Patents

Compounds and compositions as trk inhibitors
Download PDF

Info

Publication number
WO2012034095A1
WO2012034095A1PCT/US2011/051115US2011051115WWO2012034095A1WO 2012034095 A1WO2012034095 A1WO 2012034095A1US 2011051115 WUS2011051115 WUS 2011051115WWO 2012034095 A1WO2012034095 A1WO 2012034095A1
Authority
WO
WIPO (PCT)
Prior art keywords
fluorophenyl
pyrazolo
pyrimidine
pyrrolidin
carboxamide
Prior art date
Application number
PCT/US2011/051115
Other languages
French (fr)
Inventor
Jon Loren
Valentina Molteni
Yi Fan
Gregory Chopiuk
Pamela A. Albaugh
Brenton T. Flatt
Jeffrey M. Smith
Original Assignee
Irm Llc
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 Irm LlcfiledCriticalIrm Llc
Publication of WO2012034095A1publicationCriticalpatent/WO2012034095A1/en

Links

Classifications

Definitions

Landscapes

Abstract

The invention provides compounds of formula (I), pharmaceutical compositions comprising such compounds and methods of using such compounds to treat or prevent diseases or disorders associated with abnormal or deregulated TRK kinase activity. wherein: and the rest of the variables are as specified in the claims.

Description

COMPOUNDS AND COMPOSITIONS AS TRK INHIBITORS
FIELD OF THE INVENTION
[0001] The invention relates to protein kinase inhibitors, and methods of using such
compounds.
BACKGROUND OF THE INVENTION
[0002] Protein kinases (PK) are a large set of structurally related phosphoryl transferases having highly conserved structures and catalytic functions. Protein kinases are enzymatic components of the signal transduction pathways which catalyze the transfer of the terminal phosphate from ATP to the hydroxy group of tyrosine, serine and/or threonine residues of proteins, and are therefore categorized into families by the substrates they phosphorylate: Protein Tyrosine Kinases (PTK), and Protein Serine/Threonine Kinases.
[0003] Protein kinases play a critical role in the control of cell growth and differentiation and are responsible for the control of a wide variety of cellular signal transduction processes, wherein protein kinases are key mediators of cellular signals leading to the production of growth factors and cytokines. The overexpression or inappropriate expression of normal or mutant protein kinases plays a significant role in the development of many diseases and disorders including, central nervous system disorders such as Alzheimer's, inflammatory disorders such as arthritis, bone diseases such as osteoporosis, metabolic disorders such as diabetes, blood vessel proliferative disorders such as angiogenesis, autoimmune diseases such as rheumatoid arthritis, ocular diseases, cardiovascular disease, atherosclerosis, cancer, thrombosis, psoriasis, restenosis, schizophrenia, pain sensation, transplant rejection and infectious diseases such as viral, and fungal infections.
[0004] Examples of protein-tyro sine kinases include, but are not limited to, Irk, IGFR-1, Syk, Zap-70, Bmx, Btk, CHK (Csk homologous kinase), CSK (C-terminal Src Kinase), Itk-1, Src (c- Src, Lyn, Fyn, Lck, Hck, Yes, Blk, Fgr and Frk), Tec, Txk/Rlk, Abl, EGFR (EGFR-l/ErbB-1, ErbB-2/NEU/HER-2, ErbB-3 and ErbB-4), FAK, FGF1R (also FGFR1 or FGR-1), FGF2R (also FGR-2), MET (also Met-I or c-MET), PDGFR (a and β), Tie-1, Tie-2 (also Tek-1 or Tek), VEGFR1 (also FLT-1), VEGFR2 (also KDR), FLT-3, FLT-4, c-KIT, JAK1, JAK2, JAK3, TYK2, LOK, RET, TRKA, TRKB, TRKC, PYK2, ALK (Anaplastic Lymphoma Kinase), EPHA (1-8), EPHB (1-6), RON, Ros, Fes, Fer or EPHB4 (also EPHB4-1).
[0005] Examples of protein-serine/threonine kinases include, but are not limited to, Ark, ATM (1-3), CamK (1-IV), CamKK, Chkl and 2 (Checkpoint kinases), CKI, CK2, Erk, IKK-I (also IKK- ALPHA or CHUK), IKK-2 (also IKK-BETA), Ilk, Jnk (1-3), LimK (1 and 2), MLK3Raf (A, B and C), CDK (1-10), PKC (including all PKC subtypes), Plk (1-3), NIK, Pak (1-3), PDKl, PKR, RhoK, RIP, RIP-2, GSK3 (a and β), PKA, P38, Erk (1-3), PKB (including all PKB subtypes) (also AKT-1, AKT-2, AKT-3 or AKT3-1), IRAKI, FRK, SGK, TAK1 or Tpl-2 (also COT).
SUMMARY OF THE INVENTION
[0006] Provide herein are compounds and pharmaceutical compositions thereof, which are useful as inhibitors of Tropomyosin-Related Kinases (TRKA, TRKB and/or TRKC).
[0007] In one aspect, the present invention provides compounds having Formula (I), and the pharmaceutically acceptable salts, pharmaceutically acceptable solvates (e.g. hydrates), the N- oxide derivatives, prodrug derivatives, protected derivatives, individual stereoisomers and mixture of stereoisomers thereof:
Figure imgf000003_0001
Formula (I)
wherein
Figure imgf000003_0002
R1 is H, -C(0)NH2, -L^4, -L2R13, -C(0)OR5-, -C(0)NHOR5, -C(0)NHC(0)OR5-,
-C(0)OC(0)OR5-, -C(0)NHC(0)NR5R5, -C(0)NR5OR14, -C(0)NHNH2,
-NR5C(0)NR5R9,
Figure imgf000003_0003
; -S02NR5R5 or phenyl substituted with 1 to 3 groups independently selected from -NR5C(0)NR5R7, halo, d-C4alkyl, and -CN; R3 is H;
X is C(R5)2, O or NR5;
L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q-, -C(O)-, -C(0)NR50(CR6R6)q-, -C(0)0-,
-C(0)-C1-C8alkylene or -C(0)-C2-C8alkenylene ;
L2 is -NR5C(0)(CR6R6)q-; each L is independently selected from a CrCsalkylene optionally substituted with 1 to 3 substituents independently selected from C Cealkyl;
R4 is selected from H, R15, -N(R5)2, Q-Cealkyl, CrC6alkyl substituted with 1 to 4 hydroxyl groups, Ci-Cgalkyl substituted with 1 to 6 groups independently selected from halo, C - C4alkyl, and -R , phenyl, Cs-Cscycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms,
or R4 is selected from phenyl, C3-Cgcycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms each of which is substituted with 1 to 3 substituents independently selected from halo, Q-CeaLkyl, C Cealkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R8, -OR6, -C(0)R6, -C(0)OR6, -C(0)NR6R6, -C(0)L3R8, -S(0)2R6, and - S(0)2NR6R6;
each R5 is independently selected from H or C Cealkyl;
each R6 is independently selected from H, Q-Cealkyl, and Q-Cealkyl substituted with 1 to 4 hydroxyl groups;
R is phenyl or phenyl substituted with 1 to 3 groups independently selected from halo,
Ci-C4alkyl, Ci-C4haloalkyl and -R8;
R8 is selected from -OC(0)R6, -NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6,
-NHS(0)2R6, -S(0)2NR6R6, -C(0)OR6 and -OR6;
R9 is Ci-C4haloalkyl or -OR5;
each R10 is independently selected from halo, CrC6alkyl, CrCehaloalkyl, -C(=N)OR5, -CN, - (CR6R6)qCN, -NR5R5, -(CR5R5)NR5R5, -C(0)OR6, -C(0)NR5R5, -(CR6R6)qR6, -C(0)R14, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, -S(0)2NR6R6, -S(0)2R6, -OR6, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S and 5 membered heteroaryl containing 1 to 4 N heteroatoms;
each R11 is independently selected from halo, -OR5, -CN, deuterium, Q-CeaLkyl, hydroxyl substitutedCi-Cealkyl, C Cehalolkyl, and n is 0, 1 or 2;
or each R11 is deuterium, and n is 0, 1, 2, 3, 4, 5, 6 or 7;
R 12 is H, C C6alkyl, phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, Ci-C4alkyl, and -R ; R13 is halo, -OC(0)R12, -OR12, -CN, -NHC(0)OR12, -NHC(0)R12 or -NR5R5;
R14 is a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, or a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S substituted with 1 to 4 substituents independently selected from halo, -OR6, Q- C6alkyl and CrCehaloalkyl;
R15 is cyclohexyl having
Figure imgf000005_0001
or
Figure imgf000005_0002
h is optionally substituted with 1-3 substituents independently selected from halo, -CN, Ci-Cealk l, -R14, and -OR5;
R
Figure imgf000005_0003
or substituted with 1 to 3 substituents independently selected from R10,
or R20 is selected from
Figure imgf000005_0004
each of which is optionally substituted with 1 to 3 substituents independently selected from R10;
p is 1 or 2 and
q is 1, 2, 3, 4, 5 or 6.
[0008] In certain embodiments, such compounds of Formula (I):
R2 is or
Figure imgf000005_0005
R1 is H, -C(0)NH2, -LlR4, -L2R13, -C(0)OR5-, -C(0)NHOR5, -, -C(0)NHC(0)NR5R5, -
/OR6
C(0)NR5OR14, -C(0)NHNH2, -NR5C(0)NR5R9, % I"NH2 ; _S02NR5R5 or phenyl substituted with 1 to 3 groups independently selected from -NR5C(0)NR5R7, halo, Ci-C4alkyl, and -CN; R3 is H;
X is C(R5)2, O or NR5;
L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q-, -C(O)-, -C(0)NR50(CR6R6)q-, -C(0)0-,
-C(0)-C1-C8alkylene or -C(0)-C2-C8alkenylene ;
L2 is -NR5C(0)(CR6R6)q-;
each L is independently selected from a CrCgalkylene optionally substituted with 1 to 3 substituents independently selected from Q-Cealkyl;
R4 is selected from H, R15, -N(R5)2, CrC6alkyl, CrC6alkyl substituted with 1 to 4 hydroxyl groups, Q-Qalkyl substituted with 1 to 6 groups independently selected from halo, CrG^alkyl, and - R , phenyl, C3-C8cycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms,
or R4 is selected from phenyl, C3-C8cycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms each of which is substituted with 1 to 3 substituents independently selected from halo, C Cealkyl, C Cealkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R8, -OR6, - C(0)R6, -C(0)OR6, -C(0)NR6R6, -C(0)L3R8, -S(0)2R6, and -S(0)2NR6R6;
each R5 is independently selected from H or Q-Cealkyl;
each R6 is independently selected from H, C Cealkyl, and C Cealkyl substituted with 1 to 4
hydroxyl groups;
R is phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, C1-C4alkyl,
Q-Qhaloalkyl and -R8;
R8 is selected from -OC(0)R6, -NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6, -NHS(0)2R6,
-S(0)2NR6R6, -C(0)OR6 and -OR6;
R9 is Q-Qhaloalkyl or -OR5;
each R10 is independently selected from halo, CrC6alkyl, Q-Cehaloalkyl, -C(=N)OR5, -CN,
-(CR6R6)qCN, -NR5R5, -C(0)OR6, -C(0)NR5R5, -(CR6R6)qR6, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, -S(0)2NR6R6, -S(0)2R6, -OR6, 4-6 membered
heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S and 5 membered heteroaryl containing 1 to 4 N heteroatoms;
each R11 is independently selected from halo, -OR5, deuterium, Q-CeaLkyl, hydroxyl substitutedC
C6alkyl, CrCehalolkyl, and n is 0, 1 or 2;
or each R11 is deuterium, and n is 0, 1, 2, 3, 4, 5, 6 or 7;
R 12 is H, Ci-Cealkyl, phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, C1-C4alkyl, and -R8;
R13 is halo, -OC(0)R12, -OR12, -CN, -NHC(0)OR12, -NHC(0)R12 or -NR5R5;
R14 is a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, or a
4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S ssuubbssttiittuutteedd w with 1 to 4 substituents independently selected from halo, -OR6, C Cealkyl and Q- Cehaloalkyl;
is cyclohexyl having a
Figure imgf000007_0001
wherein each is optionally substituted with 1-3 substituents independently selected from halo, -Cealkyl, -R14, and -OR5;
Figure imgf000007_0002
3 substituents independently selected from R1U,
or R20 is selected from
Figure imgf000007_0003
, each of which is optionally
substituted with 1 to 3 substituents independently selected from R10;
p is 1 or 2 and q is 1, 2, 3, 4, 5 or 6.
[0009] In certain embodiments, such compounds of Formula (I) are compounds having the structure of Formula (I-a):
Figure imgf000008_0001
Formula (I-a).
[00010] In certain embodiments, such compounds of Formula (I) are compounds having the structure of Formula (II- a):
Figure imgf000008_0002
Formula (II- a).
[00011] In certain embodiments, such compounds of Formula (I) are compounds having the structure of Formula (III- a):
Figure imgf000008_0003
Formula (III- a).
[00012] In certain embodiments, such compounds of Formula (I-a) are compounds having the structure of Formula (I-b):
Figure imgf000008_0004
Formula (I-b)
wherein m is 1, 2 or 3.
[00013] In certain embodiments, such compounds of Formula (Il-a) are compounds having the structure of Formula (Il-b):
Formula (II-b)
wherein m is 1, 2 or 3.
[00014] In certain embodiments, such compounds of Formula (Ill-a) are compounds having the structure of Formula (Ill-b):
Figure imgf000009_0001
Formula (Ill-b)
wherein m is 1, 2 or 3.
[00015] In certain embodiments of compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), R1 is -C(0)NH2.
[00016] In other embodiments of compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), R1 is -LXR4. In certain embodiments of such compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q- or - C(0)NR50(CR6R6)q-. In other embodiments of such compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), L1 is -C(O)- or - C(0)0-. In still other embodiments of such compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), L1 is -C(0)-C
Cgalkylene or -C(0)-C2-Cgalkenylene.
[00017] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), R4 is phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl. In other embodiments of the aforementioned compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), R4 is phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, each of which is optionally substituted with 1 to 3 substituents independently selected from halo, Cr C6alkyl, C^Cealkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R8, -OR4, -C(0)R4, -C(0)OR4, - C(0)L3R6, -S(0)2R4, and -S(0)2NR4R4.
[00018] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (II-b) and Formula (Ill-b), R4 is tetrahydro-2H-pyranyl, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, morpholinyl, oxetanyl, tetrahydro-2H-thiopyranyl, azetidinyl, piperazinyl, pyridyl, pyrazolyl, benzthiazolyl or pyrrolyl. In other embodiments of the aforementioned compounds of Formula (I), Formula (I-a), Formula (II- a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R4 is tetrahydro-2H- pyranyl, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, morpholinyl, oxetanyl, tetrahydro-2H- thiopyranyl, azetidinyl, piperazinyl, pyridyl, pyrazolyl, benzthiazolyl or pyrrolyl, each of which is substituted with 1 to 3 substituents independently selected from halo, Q-Cealkyl, C Cealkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R14, -OR6, -C(0)R6, -C(0)OR6, -C(0)L3R6, - S(0)2R6, and -S(0)2NR6R6.
[00019] In certain embodiments of compounds of Formula (I), Formula (I-a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R1 is -C(0)NHOR5, -
Figure imgf000010_0001
[00020] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R14 is pyrrolidinyl, tetrahydro-2H-pyranyl, morpholinyl or piperidinyl, each of which is optionally substituted with 1 to 2 -OR6 groups.
[00021] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R4 is R15, - N(R5)2, methyl, ethyl, propyl, isopropyl, isobutene, t-butyl, or C Csalkyl substituted with 1 to 6 groups independently selected from halo, C1-C4alkyl, and -R .
[00022] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R15 is
cyclohexyl having a Qalkyl bridge,
Figure imgf000010_0002
wherein each is optionally substituted with 1-3 substituents independently selected from C Cealkyl.
[00023] In certain embodiments of compounds of Formula (I), Formula (I-a), Formula (Il-a),
Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R 1 is -L 2 R 13.
[00024] In certain embodiments of compounds of Formula (I), Formula (I-a), Formula (Il-a),
Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R1 is -Br, -CN and -S02NR5R5. [00025] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), each R10 is independently selected from F, CI, methyl, -CF3), -C(=N)OR5, -CN, -(CR6R6)qCN, -NR5R5, - C(0)OR6, -C(0)NR5R5, -(CR5R5)qR8, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, - S(0)2NR6R6, -S(0)2R6, -OR6, morpholinyl and tetrazolyl.
[00026] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), each R11 is independently selected from F, deuterium and methyl, and n is 1 or 2.
[00027] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), each R11 is independently selected from deuterium, and n is 1, 2, 3, 4, 5, 6 or 7.
[00028] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R is selected from -OC(0)R6, -NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6, -S(0)2NR6R6, - C(0)OR6 and -OR6, and wherein each R6 is independently selected from H, methyl, ethyl, propyl, isopropyl, t-butyl or a C Cealkyl substituted with 1 to 4 hydroxyl groups.
[00029] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), R5 is H, methyl or ethyl.
[00030] In certain embodiments of the aforementioned compounds of Formula (I), Formula (I- a), Formula (Il-a), Formula (Ill-a), Formula (I-b), Formula (Il-b) and Formula (Ill-b), q is 1, 2 or 3.
[00031] In certain embodiments compounds of Formula (I) are selected from ethyl 5-[(2R,4S)-
4- fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[2- (3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[(2R)-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[4,4-difluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[5-(3- fluorophenyl)-3-methyl-4,5-dihydro-lH-pyrazol-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[2-(3-fluorophenyl)-5-oxopyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl
5- [5-(3-fluorophenyl)-2,2,3,3,4,4-hexadeuteriumpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate; ethyl 5-[2-(3-fluorophenyl)-2,3, 3,4,4,5, 5-heptadeuteriumpyrrolidin-l-yl]pyrazolo[ 1,5- a]pyrimidine-3-carboxylate; ethyl 5-[2-(3-fluorophenyl)-2-deuteriumopyrrolidin- 1- yl]pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylate; ethyl 5- [2-(3-fluorophenyl)-3- azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[2-(3- fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; 5-[(2R)-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-2-(3- fluorophenyl)pyrrolidin-l-yl]-N,N-dimethylpyrazolo[l,5-a]pyrimidine-3-carboxamide; 5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-hydroxyethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2- sulfamoylethyl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 2-({5-[(2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}formamido)acetamide; N-(l,l-dioxo- thiolan-3-yl)-5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide; tert-butyl 4- { 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin- l-yl]pyrazolo[ 1 ,5- a]pyrimidine-3-amido}piperidine-l-carboxylate; tert-butyl N-[2-({5-[(2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}formamido)ethyl]carbamate; 5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(l-methanesulfonylpiperidin-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]-N-(2-methanesulfonamidoethyl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4- fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(piperidin-4-yl)pyrazolo[l,5-a]pyrimidine-3- carboxamide; N-(2-aminoethyl)-5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]-N-[l-(2-hydroxyacetyl)piperidin-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2S)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-ethylpyrazolo[l,5-a]pyrimidine-3-carboxamide; 5- [5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5- [(5R)-5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide 5-[(lR,2S,5S)-2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide; 5-[(lR,2R,5S)-2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[5-(3-fluorophenyl)-2,2-dideuteriumpyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[2-(3-fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide; 5-[(2R)-2-(3-fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; (2R)-2-(3-fluorophenyl)-l-{pyrazolo[l,5-a]pyrimidin-5-yl}pyrrolidine; 5-[(2R)-4,4-difluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]-N-ethoxypyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4- fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylic acid; 5-[(2R,4S)-
4- fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-[l-(hydroxymethyl)cyclopropyl]pyrazolo[l,5- a]pyrimidine-3-carboxamide; ethyl l-{5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidine-3-amido}cyclopropane-l-carboxylate; 5-[(2R,4S)-4-fhioro-2-(3- fluorophenyl)pyrrolidin-l-yl]-N-(3-methyloxetan-3-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-methoxyethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2- hydroxy-2-methylpropyl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; {5-[(2R)-4,4-difluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonylurea; l-({5-[(2R)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonyl)-3-(propan-2- yl)urea; l-{5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- amido} cyclopropane- 1-carboxylic acid; 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N- phenylpyrazolo[l,5-a]pyrimidine-3-carboxamide; [l-({5-[(2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonyl)azetidin-3-yl]methanol; (3R)-l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl}carbonyl)pyrrolidin-3-ol; l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonyl)piperidine; N-(l-carbamoylcyclopropyl)-5-[(2R,4S)-4- fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; (3S)-l-({5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl}carbonyl)pyrrolidin-3-ol; l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonyl)-3-methylazetidin-3-ol; 5-[(2R)-4,4-difluoro-2-(3- fluorophenyl)pyrrolidin-l-yl]-N-(2-hydroxyethyl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; ethyl 5-[(2R)-4-fluoro-2-(3-fluorophenyl)-4-deuteriumpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate; ethyl 5-[(2R,4S)-4-fluoro-2-(3-methyl-l,2,4-oxadiazol-5-yl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; 5-[(2R)-4-fluoro-2-(3-fluorophenyl)-4- deuteriumpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; methyl N-({5-[(2R)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3-yl}carbonyl)carbamate; 5- (4,4-difluoro-2-{5-fluoro-2-[(propan-2-yl)carbamoyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[2-(3-cyano-5-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-cyano-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide; {5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidin-3-yl}[(ethoxycarbonyl)oxy]methanone; (2R)-l-{3- bromopyrazolo[l,5-a]pyrimidin-5-yl}-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine; 5-[2-(2- cyanophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5 -[2- (2- chlorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-(2-{5-fluoro-2- [(propan-2-yl)carbamoyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-(2- {5-fluoro-2-[(morpholin-4-yl)carbonyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxamide; 5-{2-[2-(dimethylcarbamoyl)-5-fluorophenyl]pyrrolidin-l-yl}pyrazolo[l,5- a]pyrimidine-3-carboxamide; 5-{2-[5-fluoro-2-(trifluoromethyl)phenyl]pyrrolidin-l- yl }pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide; 5- { 2-[2-(trifluoromethyl)phenyl]pyrrolidin- 1- yl }pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide; 5- { 2-[3-(aminomethyl)-5-fluorophenyl]pyrrolidin-
1- yl}pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-(2-{3-fluoro-5-[(propan-2- ylamino)methyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[2-(5-cyano-
2- fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-2-(3- cyanophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-2-(3-cyano-5- fluorophenyl)-4-fluoropyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-(2- phenylpyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-2-(3-cyano-5- fluorophenyl)-4,4-difluoropyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; ethyl 5- [(2R)-2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate; ethyl 5-[(2R,4S)-2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate; 5-[(2R,4S)-2-(3-carbamoyl-5-fluorophenyl)-4-fluoropyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide, and 5-[(2R)-4,4-difluoro-2-{5-fluoro-2-[(propan-2- yl)carbamoyl]phenyl}pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide.
[00032] Another aspect provided herein are pharmaceutical compositions comprising a therapeutically effective amount of a compound of Formula (I) and a pharmaceutically acceptable carrier.
[00033] In certain embodiments of such pharmaceutical compositions, the pharmaceutical compositions are formulated for intravenous, oral administration, rectal administration inhalation, nasal administration, topical administration, ophthalmic administration or otic administration. In certain embodiments of such pharmaceutical compositions, the pharmaceutical composition is a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a solution, an emulsion, an ointment, eye drop or ear drop.
[00034] Another aspect provided herein are medicaments for treating a TRK mediated disease or condition in a patient wherein the medicament comprises a therapeutically effective amount of a compound of Formula (I).
[00035] Another aspect provided herein is the use of a compound of Formula (I) in the manufacture of a medicament for treating a TRK mediated disease or condition. In certain embodiments of such methods, the disease or condition is cancer, a proliferative diseases, a pain disorder, a dermatological disease, a metabolic disease, a muscle disease, a neurodegenerative disease, a neurological disease, an immunodeficiency disease, an immunologically-mediated disease, an autoimmune disease, an autoimmune mediated disease, a bone disease, an
inflammatory disease, fibrosis, an ophthalmic disease, an infectious disease, a viral disease, wound repair, a respiratory disease, a pulmonary disease, a renal disease, a kidney disease, a liver disease, a cardiovascular disease, a vascular disease, heart disease, cell death and hyperplasiaan inflammatory disease. In certain embodiments of such methods, the disease or condition is asthma, chronic obstructive pulmonary disease (COPD), adult respiratory distress syndrome (ARDS), ulcerative colitis, Crohns disease, bronchitis, dermatitis, allergic rhinitis, psoriasis, scleroderma, urticaria, rheumatoid arthritis, multiple sclerosis, lymphoma, metastasis, anaplastic large-cell lymphoma, osteosarcoma, fibrosarcoma, melanoma, breast cancer, renal cancer, brain cancer, prostate cancer, colorectal cancer, thyroid cancer, ovarian cancer, pancreatic cancer, neuronal cancer, neuroblastoma, lung cancer, uterine cancer, gastrointestinal cancer, HIV or lupus. In certain embodiments of such uses, the disease or condition is condition is papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis or asthma.
[00036] Another aspect provided herein is a method for inhibiting a TRK kinase comprising administering to a system or a subject in need thereof, a therapeutically effective amount of a compound of Formula (I), or pharmaceutically acceptable salts or pharmaceutical compositions thereof.
[00037] Another aspect provided herein is a method for treating a TRK kinase-mediated disease or condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound of Formula (I), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, wherein the disease or condition is selected from cancer, pain, cachexia, anorexia nervosa, a proliferative diseases, a pain disorder, a dermatological disease, a metabolic disease, a muscle disease, a neurodegenerative disease, a neurological disease, an immunodeficiency disease, an immunologically-mediated disease, an autoimmune disease, an autoimmune mediated disease, a bone disease, an inflammatory disease, fibrosis, an ophthalmic disease, an infectious disease, a viral disease, wound repair, a respiratory disease, a pulmonary disease, a renal disease, a kidney disease, a liver disease, a cardiovascular disease, a vascular disease, heart disease, cell death and hyperplasiaan inflammatory disease, and wherein the compound is a compound of Formula (I). In certain embodiments the disease is asthma, chronic obstructive pulmonary disease (COPD), adult respiratory distress syndrome (ARDS), ulcerative colitis, Crohns disease, bronchitis, dermatitis, allergic rhinitis, psoriasis, scleroderma, urticaria, rheumatoid arthritis, multiple sclerosis, lymphoma, metastasis, anaplastic large-cell lymphoma, osteosarcoma, fibrosarcoma, melanoma, breast cancer, renal cancer, brain cancer, prostate cancer, colorectal cancer, thyroid cancer, ovarian cancer, pancreatic cancer, neuronal cancer,
neuroblastoma, lung cancer, uterine cancer, gastrointestinal cancer, HIV, lupus, colon cancer or papillary thyroid carcinoma. In certain embodiments of such methods, the disease or condition is condition is papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis or asthma.
[00038] Another aspect provided herein are methods for treating a cell-proliferative condition, comprising administering to a system or subject in need of such treatment an effective amount of a compound of Formula (I), or pharmaceutically acceptable salts or pharmaceutical compositions thereof; wherein the cell-proliferative condition is lymphoma, metastasis, anaplastic large-cell lymphoma, osteosarcoma, fibrosarcoma, melanoma, breast cancer, renal cancer, brain cancer, prostate cancer, colorectal cancer, thyroid cancer, ovarian cancer, pancreatic cancer, neuronal cancer, neuroblastoma, lung cancer, uterine cancer or gastrointestinal cancer. In certain
embodiments of such methods, the cell-proliferative condition is anaplastic large-cell lymphoma, pancreatic cancer, ovarian cancer and lung cancer.
[00039] Another aspect provided herein are compounds for use in a method of medical treatment, wherein the method of medical treatment is for treating a disease selected from cancer, pain, cachexia, anorexia nervosa, a proliferative diseases, a pain disorder, a dermatological disease, a metabolic disease, a muscle disease, a neurodegenerative disease, a neurological disease, an immunodeficiency disease, an immunologically-mediated disease, an autoimmune disease, an autoimmune mediated disease, a bone disease, an inflammatory disease, fibrosis, an ophthalmic disease, an infectious disease, a viral disease, wound repair, a respiratory disease, a pulmonary disease, a renal disease, a kidney disease, a liver disease, a cardiovascular disease, a vascular disease, heart disease, cell death and hyperplasia, an inflammatory disease, and wherein the compound is a compound of Formula (I). In certain embodiments the disease is asthma, chronic obstructive pulmonary disease (COPD), adult respiratory distress syndrome (ARDS), ulcerative colitis, Crohns disease, bronchitis, dermatitis, allergic rhinitis, psoriasis, scleroderma, urticaria, rheumatoid arthritis, multiple sclerosis, lymphoma, metastasis, anaplastic large-cell lymphoma, osteosarcoma, fibrosarcoma, melanoma, breast cancer, renal cancer, brain cancer, prostate cancer, colorectal cancer, thyroid cancer, ovarian cancer, pancreatic cancer, neuronal cancer,
neuroblastoma, lung cancer, uterine cancer, gastrointestinal cancer, HIV, lupus, colon cancer or papillary thyroid carcinoma. In certain embodiments of such methods, the disease or condition is condition is papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis or asthma.
[00040] Another aspect provided herein is a method of treating anorexia nervosa, comprising administering to a system or subject in need of such treatment an effective amount of a compound of Formula (I), or pharmaceutically acceptable salts or pharmaceutical compositions thereof
DETAILED DESCRIPTION OF THE INVENTION
Definitions
[00041] The terms "alkenyl" and "alkene," as used herein, refers to a partially unsaturated branched or straight chain hydrocarbon having at least one carbon-carbon double bond. Atoms oriented about the double bond are in either the cis (Z) or trans (E) conformation. An alkenyl or alkene group can be optionally substituted. As used herein, the terms "C2-C3alkyenyl", "C2- C4alkyenyl", "C2-C5alkenyl", "C2-C6alkenyl", "C2-C7alkenyl", and "C2-C8alkenyl" refer to an alkenyl group containing at least 2, and at most 3, 4, 5, 6, 7 or 8 carbon atoms, respectively. Non- limiting examples of alkenyl groups, as used herein, include ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl and the like. As used herein, the terms "C2- C3alkene", "C2-C4alkene", "C2-C5alkene", "C2-C6alkene", "C2-C7alkene", and "C2-C8alkene" refer to an alkene group containing at least 2, and at most 3, 4, 5, 6, 7 or 8 carbon atoms, respectively. Non-limiting examples of alkene groups, as used herein, include ethene, propene, butene, pentene, hexene, heptene, octene, nonene, decene and the like.
[00042] The term "alkenylene," as used herein, refers to a partially unsaturated branched or straight chain divalent hydrocarbon radical derived from an alkenyl group. An alkenylene group can be optionally substituted. As used herein, the terms "C2-C3 alkenylene", "C2-C4alkenylene", "C2-C5alkenylene", "C2-C6alkenylene", "C2-C7alkenylene", and "C2-C8alkenylene" refer to an alkenylene group containing at least 2, and at most 3, 4, 5, 6, 7 or 8 carbon atoms respectively. Non-limiting examples of alkenylene groups as used herein include, ethenylene, propenylene, butenylene, pentenylene, hexenylene, heptenylene, octenylene, nonenylene, decenylene and the like.
[00043] The term "alkyl," as used herein, refers to a saturated branched or straight chain hydrocarbon. An alkyl group can be optionally substituted. As used herein, the terms "Ci-Csalkyl", "Ci-C4alkyl", "Ci-C5alkyl", "Ci-C6alkyl", "Ci-C7alkyl" and "Ci-C8alkyl" refer to an alkyl group containing at least 1, and at most 3, 4, 5, 6, 7 or 8 carbon atoms, respectively. Non-limiting examples of alkyl groups as used herein include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec -butyl, t-butyl, n-pentyl, isopentyl, hexyl, heptyl, octyl, nonyl, decyl and the like.
[00044] The term "alkylene," as used herein, refers to a saturated branched or straight chain divalent hydrocarbon radical derived from an alkyl group. An alkylene group can be optionally substituted. As used herein, the terms "CrQalkylene", "C1-C4alkylene", "Q-Csalkylene", "Q- Cealkylene", "C1-C7alkylene" and "CrCgalkylene" refer to an alkylene group containing at least 1, and at most 3, 4, 5, 6, 7 or 8 carbon atoms respectively. Non-limiting examples of alkylene groups as used herein include, methylene, ethylene, n-propylene, isopropylene, n-butylene, isobutylene, sec-butylene, t-butylene, n-pentylene, isopentylene, hexylene and the like.
[00045] The term "aryl," as used herein, refers to monocyclic or fused bicyclic ring systems having a total of six, ten or fourteen carbon atom ring members. An aryl group can be optionally substituted with one or more substituents. Non-limiting examples of aryl groups, as used herein, include phenyl and naphthyl.
[00046] The term "arylene," as used means a divalent radical derived from an aryl group. An arylene group can be optionally substituted.
[00047] The term "cyano," as used herein, refers to a -CN group.
[00048] The term "cycloalkyl," as used herein, refers to a saturated monocyclic ring system or a saturated fused bicyclic ring system. As used herein, the terms "C3-C5 cycloalkyl", "C3-C6 cycloalkyl" and "C3-C7 cycloalkyl" refer to a cycloalkyl group wherein the saturated ring system contains at least 3, and at most 5, 6 or 7 carbon atoms. A cycloalkyl group can be optionally substituted. Non-limiting examples of cycloalkyl groups, as used herein, include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
[00049] The term "halo," as used herein, refers to fluoro (-F), chloro (-C1), bromo (-Br), and iodo (-1).
[00050] The terms "haloalkyl" or "halo-substituted alkyl," as used herein, refers to an alkyl group as defined herein, substituted with one or more halogen groups, wherein the halogen groups are the same or different. A haloalkyl group can be optionally substituted. Non-limiting examples of such branched or straight chained haloalkyl groups, as used herein, include methyl, ethyl, propyl, isopropyl, isobutyl and n-butyl substituted with one or more halogen groups, wherein the halogen groups are the same or different. By way of example ony, trifluoromethyl,
pentafluoroethyl, and the like.
[00051] The term "heteroaryl," as used herein, refers to monocyclic or fused bicyclic ring systems having a total of 5, 6, 9 or 10 ring members, wherein at least one ring member is a heteroatom selected from nitrogen, oxygen and sulfur. A heteroaryl group can be optionally substituted with one or more substituents. Non-limiting examples of heteroaryl groups, as used herein, include benzofuranyl, benzofurazanyl, benzoxazolyl, benzopyranyl, benzthiazolyl, benzothienyl, benzazepinyl, benzimidazolyl, benzothiopyranyl, benzo[b]furyl, benzo[b]thienyl, cinnolinyl, furazanyl, furyl, imidazolyl, indolyl, indolizinyl, indazolyl, isoindolyl, isoquinolinyl, isoxazolyl, isothiazolyl, 1,8-naphthyridinyl, oxazolyl, oxaindolyl, oxadiazolyl, pyrazolyl, pyrrolyl, phthalazinyl, pteridinyl, purinyl, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, quinoxalinyl, quinolinyl, quinazolinyl, thiazolyl, thiadiazolyl, thienyl, triazinyl, triazolyl and tetrazolyl.
[00052] The term "heteroarylene," as used means a divalent radical derived from a heteroaryl group. A heteroarylene group can be optionally substituted.
[00053] The term "heterocycloalkyl," as used herein, refers to a cycloalkyl, as defined herein, wherein one or more of the ring carbons are replaced by a moiety selected from -0-, -N=, -NR-, - C(O)-, -S-, -S(O) - or -S(0)2-, wherein R is hydrogen, C1-C4alkyl or a nitrogen protecting group. A heterocycloalkyl group can be optionally substituted. Non-limiting examples of heterocycloalkyl groups, as used herein, include morpholino, pyrrolidinyl, pyrrolidinyl-2-one, piperazinyl, piperazinyl-2-one, piperidinyl, piperidinylone, 1,3-dioxolanyl, imidazolidinyl, pyrazolidinyl, 1,4- dioxanyl, 1,4-dithianyl, thiomorpholinyl, azepanyl, hexahydro-l,4-diazepinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, thioxanyl, azetidinyl, oxetanyl, thietanyl, oxepanyl, thiepanyl, dioxanyl, 1,3-dioxolanyl, dithianyl, dithiolanyl, imidazolidinyl, 3- azabicyclo[3.1.0]hexanyl, 2,5-diazabicyclo[2.2.1]heptane and 3-azabicyclo[4.1.0]heptanyl.
[00054] The term "heteroatom," as used herein, refers to an oxygen atom, sulfur atom, or nitrogen atom.
[00055] The term "hydroxyl," as used herein, refers to the group -OH.
[00056] The term "hydroxyalkyl," as used herein refers to an alkyl group as defined herein substituted with one or more hydroxyl group. Non-limiting examples of branched or straight chained "Ci-Ce hydroxyalkyl groups as used herein include methyl, ethyl, propyl, isopropyl, isobutyl and n-butyl groups substituted with one or more hydroxyl groups.
[00057] The term "optionally substituted," as used herein, means that the referenced group may or may not be substituted with one or more additional group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, heterocycloalkyl, hydroxyl, alkoxy, mercaptyl, cyano, halo, carbonyl, thiocarbonyl, isocyanato, thiocyanato, isothiocyanato, nitro, perhaloalkyl, perfluoroalkyl, and amino, including mono- and di-substituted amino groups, and the protected derivatives thereof. Non-limiting examples of optional substituents, where proper valency is maintained, include, halo, -CN, =0, -OR, -C(0)R, -C(0)OR, -OC(0)R, - OC(0)OR, -C(0)NHR, -C(0)NR2, -OC(0)NHR, -OC(0)NR2, -SR-, -S(0)R, -S(0)2R, -NHR, - N(R)2, -NHC(0)R, -NRC(0)R, -NHC(0)OR, -NRC(0)OR, S(0)2NHR, -S(0)2N(R)2, -NHS(0)2, -NRS(0)2, -NHS(0)2R, -NRS(0)2R, Ci-C8alkyl, Ci-C8alkoxy, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, halo-substituted CrCgalkyl, halo-substituted CrCgalkoxy, where each R is independently selected from H, halo, CrCgalkyl, CrCgalkoxy, aryl, heteroaryl, cycloalkyl, heterocycloalkyl, halo-substituted CrCgalkyl, and halo-substituted CrCgalkoxy. The placement and number of such substitutent groups is done in accordance with the well-understood valence limitations of each group, for example =0 is a suitable substituent for an alkyl group but not for an aryl group.
[00058] The term "solvate," as used herein, refers to a complex of variable stoichiometry formed by a solute (by way of example, a compound of Formula (I), or a salt thereof, as described herein) and a solvent. Non-limiting examples of a solvent are water, acetone, methanol, ethanol and acetic acid.
[00059] The term "acceptable" with respect to a formulation, composition or ingredient, as used herein, means having no persistent detrimental effect on the general health of the subject being treated.
[00060] The term "administration" or "administering" of the subject compound means providing a compound of the invention and prodrugs thereof to a subject in need of treatment.
[00061] The term "cancer," as used herein refers to an abnormal growth of cells which tend to proliferate in an uncontrolled way and, in some cases, to metastasize (spread). The types of cancer include, but is not limited to, solid tumors (such as those of the bladder, bowel, brain, breast, endometrium, heart, kidney, lung, lymphatic tissue (lymphoma), ovary, pancreas or other endocrine organ (thyroid), prostate, skin (melanoma) or hematological tumors (such as the leukemias). [00062] The term "carrier," as used herein, refers to chemical compounds or agents that facilitate the incorporation of a compound described herein into cells or tissues.
[00063] The terms "co-administration" or "combined administration" or the like as used herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
[00064] The term "dermatological disorder," as used herein refers to a skin disorder. Such dermatological disorders include, but are not limited to, proliferative or inflammatory disorders of the skin such as, atopic dermatitis, dermatitis, bullous disorders, collagenoses, contact dermatitis, eczema, Kawasaki Disease, rosacea, Sjogren-Larsso Syndrome, and urticaria.
[00065] The term "diluent" as used herein, refers to chemical compounds that are used to dilute a compound described herein prior to delivery. Diluents can also be used to stabilize compounds described herein.
[00066] The terms "effective amount" or "therapeutically effective amount," as used herein, refer to a sufficient amount of a compound described herein being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an "effective amount" for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate "effective" amount in any individual case may be determined using techniques, such as a dose escalation study.
[00067] The terms "enhance" or "enhancing," as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term "enhancing" refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An "enhancing-effective amount," as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.
[00068] The terms "fibrosis" or "fibrosing disorder," as used herein, refers to conditions that follow acute or chronic inflammation and are associated with the abnormal accumulation of cells and/or collagen and include but are not limited to fibrosis of individual organs or tissues such as the heart, kidney, joints, lung, or skin, and includes such disorders as idiopathic pulmonary fibrosis and cryptogenic fibrosing alveolitis.
[00069] The term "iatrogenic", as used herein, means a condition, disorder, or disease created or worsened by medical or surgical therapy.
[00070] The term "inflammatory disorders", as used herein, refers to those diseases or conditions that are characterized by one or more of the signs of pain (dolor, from the generation of noxious substances and the stimulation of nerves), heat (calor, from vasodilatation), redness (rubor, from vasodilatation and increased blood flow), swelling (tumor, from excessive inflow or restricted outflow of fluid), and loss of function (functio laesa, which may be partial or complete, temporary or permanent). Inflammation takes many forms and includes, but is not limited to, inflammation that is one or more of the following: acute, adhesive, atrophic, catarrhal, chronic, cirrhotic, diffuse, disseminated, exudative, fibrinous, fibrosing, focal, granulomatous, hyperplastic, hypertrophic, interstitial, metastatic, necrotic, obliterative, parenchymatous, plastic, productive, proliferous, pseudomembranous, purulent, sclerosing, seroplastic, serous, simple, specific, subacute, suppurative, toxic, traumatic, and/or ulcerative. Inflammatory disorders further include, without being limited to those affecting the blood vessels (polyarteritis, temporarl arteritis); joints (arthritis: crystalline, osteo-, psoriatic, reactive, rheumatoid, Reiter's); gastrointestinal tract (Disease,); skin (dermatitis); or multiple organs and tissues (systemic lupus erythematosus).
[00071] The terms "neurogenerative disease" or "nervous system disorder," as used herein, refers to conditions that alter the structure or function of the brain, spinal cord or peripheral nervous system, including but not limited to Alzheimer's Disease, cerebral edema, cerebral ischemia, multiple sclerosis, neuropathies, Parkinson's Disease, those found after blunt or surgical trauma (including post-surgical cognitive dysfunction and spinal cord or brain stem injury), as well as the neurological aspects of disorders such as degenerative disk disease and sciatica. The acronym "CNS" refers to disorders of the central nervous system (brain and spinal cord).
[00072] The term "pharmaceutically acceptable," as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compounds described herein. Such materials are administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
[00073] The term "pharmaceutically acceptable salt", as used herein, refers to a formulation of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compounds described herein.
[00074] The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound of Formula I and a coagent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound of Formula I and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of 3 or more active ingredients.
[00075] The term "pharmaceutical composition", as used herein, refers to a mixture of a compound described herein with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
[00076] The term "prodrug", as used herein, refers to an agent that is converted into the parent drug in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. Prodrugs are bioavailable by oral administration whereas the parent is not. Prodrugs improve solubility in pharmaceutical compositions over the parent drug. A non-limiting example of a prodrug of the compounds described herein is a compound described herein administered as an ester which is then metabolically hydrolyzed to a carboxylic acid, the active entity, once inside the cell. A further example of a prodrug is a short peptide bonded to an acid group where the peptide is metabolized to reveal the active moiety.
[00077] The term "respiratory disease," as used herein, refers to diseases affecting the organs that are involved in breathing, such as the nose, throat, larynx, trachea, bronchi, and lungs.
Respiratory diseases include, but are not limited to, asthma, adult respiratory distress syndrome and allergic (extrinsic) asthma, non-allergic (intrinsic) asthma, acute severe asthma, chronic asthma, clinical asthma, nocturnal asthma, allergen-induced asthma, aspirin- sensitive asthma, exercise-induced asthma, isocapnic hyperventilation, child-onset asthma, adult-onset asthma, cough- variant asthma, occupational asthma, steroid-resistant asthma, seasonal asthma, seasonal allergic rhinitis, perennial allergic rhinitis, chronic obstructive pulmonary disease, including chronic bronchitis or emphysema, pulmonary hypertension, interstitial lung fibrosis and/or airway inflammation and cystic fibrosis, and hypoxia.
[00078] The term "subject" or "patient", as used herein, encompasses mammals and non- mammals. Examples of mammals include, but are not limited to, humans, chimpanzees, apes monkeys, cattle, horses, sheep, goats, swine; rabbits, dogs, cats, rats, mice, guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like.
[00079] The term "therapeutically effective amount", as used herein, refers to any amount of a compound which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.
[00080] The terms "treat", "treating" or "treatment," as used herein, refers to methods of alleviating, abating or ameliorating a disease or condition symptoms, preventing additional symptoms, ameliorating or preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophylactic ally and/or therapeutically.
[00081] The terms "use" or "used," as used herein, are intended to include a compound of Formula (I) provided herein for use in the prophylactic and/or therapeutic treatment of one or more diseases provided herein, a method of use or a method of treatment comprising administering a compound of the Formula (I) to a person in need of such treatment in an effective amount for the prophylactic and/or therapeutic treatment of one or more diseases provided herein, the preparation or a method or preparation of a pharmaceutical formulation/preparation for use in the prophylactic and therapeutic treatment of one or more diseases provided herein, especially involving mixing a compound of the Formula (I) (as therapeutically active ingredient) with at least one pharmaceutically acceptable carrier material, including making it ready for use in such treatment (e.g. adding an instruction insert (e.g. package leaflet or the like), formulation, appropriate preparation, adaptation for specific uses, customizing and the like), and the use of a compound of the Formula (I) for such preparation, and/or all other prophylactic or therapeutic uses mentioned herein.
[00082] Other objects, features and advantages of the methods and compositions described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only.
Compounds
[00083] Provided herein are compounds, pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, which are inhibitrs of TRKA, TRKB and TRKC kinase activity. Also provided herein are compounds, pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, pharmaceutical combinations for the treatment diseases or conditions/disorders associated with TRKA, TRKB and TRKC kinase activity. Also provided herein are methods of treaing diseases or conditions/disorders associated with TRKA, TRKB and TRKC kinase activity, wherein the method includes administration of a therapeutically effective amount of a compound provided herein, pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, and pharmaceutical compositions containing such pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof.
[00084] In certain embodiments, such diseases and/or disorders include, but are not limited to, cancer, proliferative diseases, pain, dermatological diseases and/or disorders, metabolic diseases and/or disorders, muscle diseases and/or disorders, neurodegenerative diseases and/or disorders, neurological diseases and/or disorders, inflammatory diseases, fibrosis, infectious diseases, respiratory diseases and/or disorders, pulmonary diseases and/or disorders and hyperplasia.
[00085] Such cancer and proliferative diseases include, but are not limited to, hematopoietic disorders, hematopoietic malignancies, non-hematopoietic malignancies, benign or malignant tumors, tumors of the neck and head, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, neuronal cancer, neuroblastoma, bladder cancer, breast cancer, secretory breast carcinoma, stomach cancer, gastric tumors, ovarian cancer, uterine cancer, colon cancer, rectal cancer, colorectal adenoma, prostate cancer, renal cancer, brain cancer, endometrial cancer, pancreatic cancer, lung cancer, non- small cell lung cancer, human adenoid cystic carcinoma, vaginal cancer, thyroid cancer, papillary thyroid carcinoma, sarcoma, congenital fibrosarcoma, osteolytic sarcoma, osteosarcoma, fibrosarcoma, myeloma, tumor metastasis to bone, congenital mesoblastic nephroma, glioblastomas, melanoma, multiple myeloma, gastrointestinal cancer, gastrointestinal stromal tumors (GIST), mastocytosis, neuroblastoma, fibrotic cancers, tumor metastasis growth, epidermal hyperproliferation, psoriasis, metastasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, lymphomas, diffuse large B-cell lymphoma, B-cell lymphoma, mammary carcinoma, Wilm's tumor, Cowden syndrome, Lhermitte-Dudos disease and Bannayan-Zonana syndrome.
[00086] Such hematopoietic disorders include, but are not limited to, myeloproliferative disorders, thrombocythemia, essential thrombocytosis (ET), angiogenic myeloid metaplasia, myelofibrosis (MF), myelofibrosis with myeloid metaplasia (MMM), chronic idiopathic myelofibrosis (IMF), polycythemia vera (PV), the cytopenias, and pre-malignant myelodysplasia syndromes.
[00087] Such hematological malignancies include, but are not limited to, leukemias, myeloid leukemias, hairy cell leukemia, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, including, but are not limited to, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML with trilineage myelodysplasia (AML/TMDS), mixed lineage leukemia (MLL),
myelodysplasia syndromes (MDSs), myeloproliferative disorders (MPD), multiple myeloma, (MM), myeloid sarcoma and acute promyelocytic leukemia (APL).
[00088] Such pain disorders include, but are not limited to, cancer-related pain, skeletal pain caused by tumor metastasis, osteoarthritis, visceral pain, inflammatory pain and neurogenic pain.
[00089] Such dermatological diseases and/or disorders include, but are not limited to, inflammatory or allergic conditions of the skin, dermatitis, eczema, psoriasis, atopic dermatitis, seborrhoeic dermatitis (Dandruff, Cradle cap), diaper rash, urushiol-induced contact dermatitis, contact dermatitis, erythroderma, lichen simplex chronicus, prurigo nodularis, itch, pruritus ani, nummular dermatitis, dyshidrosis, pityriasis alba, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphigus, epidermolysis bullosa acquisita, peritoneal and sub dermal adhesion and photoaging of the skin.
[00090] Such metabolic diseases and/or disorders and eating disorder include, but are not limited to, obesity, diabetes and anoerexia.
[00091] Such muscle diseases and/or disorders include, but are not limited to, muscular atrophies (e.g. disuse), muscular dystrophies (e.g. Duchenne's muscle dystrophy, Becker's muscle dystrophy, Limb-Girdle muscle dystrophy), sarcopenia, cachexia, wasting and
Facioscapulohumeral dystrophy.
[00092] Such neurological diseases and/or disorders and neurodegenerative disorders include, but are not limited to, impaired neurological function and Alzheimer's disease.
[00093] Such inflammatory diseases and/or disorders include, but are not limited to, uveitis, atherosclerosis, atherogenesis, glomerulonephritis, Kawasaki disease, inflammatory responses, polymyositis, arthritis, neurological inflammation, chronic arthritis inflammation and
osteoarthritis.
[00094] Such fibrosis diseases and/or disorders include, but are not limited to, extracellular matrix accumulation and fibrosis, scleroderma, fibrosclerosis, radiation-induced fibrosis, kidney fibrosis, lung fibrosis and liver fibrosis, haemochromatosis, primary biliary cirrhosis, restenosis, retroperitoneal fibrosis, mesenteric fibrosis, endometriosis and keloids.
[00095] Such ophthalmic/ocular diseases and/or disorders include, but are not limited to, proliferative vitreoretinopathy, ocular scarring, corneal scarring, ocular disorders, corneal wounds, conjunctivitis, keratoconjunctivitis sicca, and vernal conjunctivitis.
[00096] Such infectious diseases and/or disorders include, but are not limited to, Chagas disease.
[00097] Such respiratory diseases and/or disorders and pulmonary disorders include, but are not limited to, asthma, bronchial asthma, allergic asthma, intrinsic (non-allergic) asthma, extrinsic (allergic) asthma, exercise-induced asthma, drug-induced asthma (including aspirin and NSAID- induced) and dust-induced asthma, chronic obstructive pulmonary disease (COPD); chronic obstructive airways disease (COAD), chronic obstructive lung disease (COLD), bronchitis, chronic bronchitis, acute bronchitis, dyspnea, arachidic bronchitis, catarrhal bronchitis, croupus bronchitis, phthinoid bronchitis, rhinitis, acute rhinitis, chronic rhinitis, rhinitis medicamentosa, vasomotor rhinitis, perennial and seasonal allergic rhinitis, rhinitis nervosa (hay fever), inflammatory or obstructive airways diseases, pulmonary hypertension, acute lung injury, adult/acute respiratory distress syndrome (ARDS), pulmonary fibrosis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary disease due to infectious or toxic agents, emphysema,
pneumoconiosis, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis, byssinosis, acute lung injury (ALI), hypereosinophilia, Loffler's syndrome, eosinophilic pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg-Strauss syndrome), eosinophilic granuloma, eosinophil-related disorders affecting the airways occasioned by drug-reaction, pulmonary hypertension, primary pulmonary hypertension (PPH), secondary pulmonary hypertension (SPH), familial PPH, sporadic PPH, precapillary pulmonary hypertension, pulmonary arterial hypertension (PAH), pulmonary artery hypertension, idiopathic pulmonary hypertension, thrombotic pulmonary arteriopathy (TPA), plexogenic pulmonary arteriopathy, functional classes I to Γ pulmonary hypertension, and pulmonary hypertension associated with, related to, or secondary to, left ventricular dysfunction, mitral valvular disease, constrictive pericarditis, aortic stenosis, cardiomyopathy, mediastinal fibrosis, anomalous pulmonary venous drainage, pulmonary venoocclusive disease, collagen vascular disease, congenital heart disease, HIV virus infection, drugs and toxins such as fenfluramines, hypoxemia, pulmonary venous hypertension, chronic obstructive pulmonary disease, interstitial lung disease, sleep-disordered breathing, alveolar hypoventilation disorder, chronic exposure to high altitude, neonatal lung disease, alveolar- capillary dysplasia, sickle cell disease, other coagulation disorder, chronic thromboemboli, connective tissue disease, lupus, schistosomiasis, sarcoidosis or pulmonary capillary
hemangiomatosis.
[00098] The aforementioned compounds and pharmaceutically acceptable salts, solvates, N- oxides, prodrugs and isomers thereof, are compounds having structures according to Formula (I):
Figure imgf000028_0001
Formula (I)
wherein R 1 , R2 and R 3 are as defined herein.
[00099] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having th ):
Figure imgf000028_0002
Formula (I- a)
wherein n, R 1 , R 11 , R 20 and R 3 are as defined herein.
[000100] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having the structure of Formula (II- a):
Figure imgf000028_0003
Formula (Il-a)
wherein n, X, R 1 , R 11 , 20 and R 3J are as defined herein.
[000101] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having the structure of Formula (Ill-a):
Figure imgf000029_0001
Formula (Ill-a)
wherein n, R 1 , R 11 , R 20 and R 3 are as defined herein.
[000102] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having ):
Figure imgf000029_0002
Formula (I-b)
wherein n, m, R1, R11, R10 and R3 are as defined herein.
[000103] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having the structure of Formula (Il-b):
Figure imgf000029_0003
Formula (Il-b)
wherein n, m, X, R1, R11, R10 and R3 are as defined herein.
[000104] In certain embodiments of the aforementioned compounds and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, having structures according to Formula (I) are compounds having the structure of Formula (Ill-b):
Figure imgf000030_0001
Formula (III-b)
wherein n, m, R^ R^ R^ and R3 are as defined herein.
[000105] The compounds, pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, and pharmaceutical compositions provided herein also includes all suitable isotopic variations of such compounds, and pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, and pharmaceutical compositions. An isotopic variation of a compound of the invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that may be incorporated into the compounds of the invention and pharmaceutically acceptable salts thereof include but are not limited to isotopes of hydrogen, carbon, nitrogen and oxygen such as 2 H, 3 H,UC,13C,14C,15N,170,180,35S,18F,36C1 and123I. Certain isotopic variations of the compounds of the invention and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as3H or14C is incorporated, are useful in drug and/or substrate tissue distribution studies. In particular examples,3H and14C isotopes may be used for their ease of preparation and detectability. In other examples, substitution with isotopes such as H may afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements. Isotopic variations of the compounds, and
pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, and pharmaceutical compositions provided herein are prepared by conventional procedures using appropriate isotopic variations of suitable reagents. Isotopic variations of the compounds have the potential to change a compound' s metabolic fate and/or create small changes in physical properties such as hydrophobicity, and the like. Isotopic variation have the potential to enhance efficacy and safety, enhance bioavailability and half-life, alter protein binding, change biodistribution, increase the proportion of active metabolites and/or decrease the formation of reactive or toxic metabolites.
Processes for Making Compounds of Formula (I)
[000106] General procedures for preparing compounds of Formula (I) are described in the Examples, infra. In the reactions described, reactive functional groups, for example hydroxyl, amino, imino, thio or carboxy groups, where these are desired in the final product, may be protected to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice (see e.g., T.W. Greene and P. G. M. Wuts in "Protective Groups in Organic Chemistry," John Wiley and Sons, 1991).
[000107] In certain embodiments, the compounds of Formula (I) described herein are prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound of Formula (I) with a pharmaceutically acceptable organic acid or inorganic acid. In other embodiments, a pharmaceutically acceptable base addition salt of compounds of Formula (I) described herein is prepared by reacting the free acid form of the compound of Formula (I) with a pharmaceutically acceptable organic base or inorganic base. Alternatively, the salt forms of the compounds of Formula (I) described herein are prepared using salts of the starting materials or intermediates. In certain embodiments, the compounds of Formula (I) described herein are in the form of other salts including, but not limited to, oxalates and trifluoroacetates. In certain embodiments, hemisalts of acids and bases are formed, for example, hemisulphate and
hemicalcium salts.
[000108] The pharmaceutically acceptable organic acid or inorganic acids used to form pharmaceutically acceptable acid addition salts of compounds of Formula (I) include, but are not limited to, hydrofluoric acid, hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, sulfuric acid, phosphoric acid, formic acid, acetic acid, trifluoroacetic acid, propionic acid, butyric acid, caprylic acid, dichloroacetic acid, hippuric acid, lactic acid, citric acid, tartaric acid, malic acid, gluconic acid, mandelic acid, maleic acid, succinic acid, adipic acid, aspartic acid, fumaric acid, glutamic acid, malonic acid, sebacic acid, salicylic acid, hexanoic acid, benzoic acid, p- chloro-benzoic acid, nicotinic acid, diphenylacetic acid, triphenylacetic acid, o-hydroxybenzoic acid, p-hydroxybenzoic acid, l-hydroxynaphthalene-2-carboxylic acid or 3-hydroxynaphthalene-2- carboxylic acid, sulfonic acids, methanesulfonic acid, benzenesulfonic acid, ethanesulfonic acid, ethane- 1,2-disulfonic acid, 2-hydroxy-ethanesulfonic acid, (+) camphor- 10-sulfonic acid, naphthalene-2- sulfonic acid, naphthalene- 1,5-disulfonic acid and p-toluenesulfonic acid.
Pharmaceutically acceptable solvates are generally hydrates.
[000109] Such pharmaceutically acceptable acid addition salts of compounds of Formula (I) include, but are not limited to, a hydrobromide, hydrochloride, sulfate, nitrate, succinate, maleate, formate, acetate, adipate, besylatye, bicarbonate/carbonate, propionate, fumarate, citrate, tartrate, lactate, benzoate, salicylate, glutamate, aspartate, p-toluenesulfonate, benzenesulfonate, methanesulfonate, ethanesulfonate, naphthalenesulfonate (e.g. 2-naphthalenesulfonate), hexanoate salt, bisulphate/sulphate, borate, camsylate, cyclamate, edisylate, esylate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen
phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, tannate, tosylate, trifluoroacetate and xinofoate salts.
[000110] Such pharmaceutically acceptable base addition salt of a compound of Formula (I) include, but are not limited to, aluminium, ammonium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine, ethanolamines, benzylamines, pyridine, benethamine, diethanolamine, 4-(2- hydroxy-ethyl)morpholine,l-(2-hydroxyethyl)pyrrolidine, N-methyl glutamine, piperazine, triethanol-amine and zinc salts.
[000111] In certain embodiments, the free acid or free base forms of the compounds of Formula (I) described herein are prepared from the corresponding base addition salt or acid addition salt from, respectively. For example a compound Formula (I) in an acid addition salt form is converted to the corresponding free base by treating with a suitable base (by way of example only, an ammonium hydroxide solution, a sodium hydroxide, and the like). For example, a compound of Formula (I) in a base addition salt form is converted to the corresponding free acid by treating with a suitable acid (by way of example only, hydrochloric acid).
[000112] In certain embodiments, the compounds of Formula (I) described herein in unoxidized form are prepared from N-oxides of compounds Formula (I) by treating with a reducing agent (by way of example only, sulfur, sulfur dioxide, triphenyl phosphine, lithium borohydride, sodium borohydride, phosphorus trichloride, tribromide, or the like) in a suitable inert organic solvent (by way of example only, acetonitrile, ethanol, aqueous dioxane, or the like) at 0 to 80°C.
[000113] In certain embodiments, prodrug derivatives of compounds Formula (I) described herein are prepared using methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et al., (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs are prepared by reacting a non-derivatized compound of Formula (I) with a suitable carbamylating agent (by way of example only, 1,1- acyloxyalkylcarbanochloridate, para-nitrophenyl carbonate, or the like). [000114] In certain embodiments, the compounds of Formula (I) described herein are prepared as protected derivatives using methods known to those of ordinary skill in the art. A detailed description of the techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, "Protecting Groups in Organic Chemistry," 3 rd edition, John Wiley and Sons, Inc., 1999.
[000115] In certain embodiments, the compounds of Formula (I) described herein are prepared or formed, as solvates (e.g., hydrates). In certain embodiments, hydrates of compounds of Formula (I) are prepared by recrystallization from an aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.
[000116] In certain embodiments, the compounds of Formula (I) provided herein are prepared as a racemic mixture. In certain embodiments, the compounds of Formula (I) described herein are prepared as their individual stereoisomers. In certain embodiments, the compounds of Formula (I) provided herein are prepared as a racemic mixture and their individual stereoisomers are aobtained using chiral chromatography, including , but not limited to, chiral liquid chromatogtaphy. In other embodiments, the compounds of Formula (I) described herein are prepared as their individual stereoisomers by reacting a racemic mixture of the compound with an optically active resolving agent to form a pair of diastereoisomeric compounds, separating the diastereomers and recovering the optically pure enantiomers. In certain embodiments, resolution of enantiomers is carried out using covalent diastereomeric derivatives of the compounds of Formula (I), or by using dissociable complexes (e.g., crystalline diastereomeric salts). Diastereomers have distinct physical properties (e.g., melting points, boiling points, solubility, reactivity, etc.) and are readily separated by taking advantage of these dissimilarities. In certain embodiments, the diastereomers are separated by chromatography, or by separation/resolution techniques based upon differences in solubility. The optically pure enantiomer is then recovered, along with the resolving agent, by any practical means that would not result in racemization. A more detailed description of the techniques applicable to the resolution of stereoisomers of compounds from their racemic mixture can be found in Jean Jacques, Andre Collet, Samuel H. Wilen, "Enantiomers, Racemates and Resolutions," John Wiley And Sons, Inc., 1981.
[000117] Compounds of Formula (I) are provided in substantially pure form. In certain embodiments compounds of Formula (I) are at least 60% pure. In certain embodiments compounds of Formula (I) are at least 75% pure. In certain embodiments compounds of Formula (I) are at 85% pure. In certain embodiments compounds of Formula (I) are at least 98% pure (% are on a weight for weight basis).
[000118] Compounds of Formula (I) are made by processes described herein and as illustrated in the Examples. Non-limiting examples of synthetic schemes used to make compounds of Formula
(I) described herein are illustrated in reaction schemes (I)-(V), wherein n, m, p, C[, X, L 1 , L2 , L 3 , R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15 and R20 are as defined herein.
Reaction Scheme (I)
Figure imgf000034_0001
Figure imgf000034_0002
Reaction Scheme (III)
Figure imgf000036_0001
 Reaction Scheme (IV)
Figure imgf000037_0001
Reaction Scheme (V)
Figure imgf000038_0001
Figure imgf000038_0002
Pharmacology and Utility
[000120] Protein kinases (PTK) play a central role in the regulation of a wide variety of cellular processes and maintaining control over cellular function. Protein kinases catalyze and regulate the process of phosphorylation, whereby the kinases covalently attach phosphate groups to proteins or lipid targets in response to a variety of extracellular signals. Examples of such stimuli include hormones, neurotransmitters, growth and differentiation factors, cell cycle events, environmental stresses and nutritional stresses. An extracellular stimulus may affect one or more cellular responses related to cell growth, migration, differentiation, secretion of hormones, activation of transcription factors, muscle contraction, glucose metabolism, control of protein synthesis, and regulation of the cell cycle.
[000121] Many diseases are associated with abnormal cellular responses triggered by protein kinase-mediated events. These diseases include, but are not limited to, autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, respiratory diseases, allergies and asthma, Alzheimer's disease, and hormone-related diseases.
[000122] Examples of protein-tyro sine kinases include, but are not limited to,
(a) tyrosine kinases such as Irk, IGFR-1, Zap-70, Bmx, Btk, CHK (Csk homologous kinase), CSK (C-terminal Src Kinase), Itk-1, Src (c-Src, Lyn, Fyn, Lck, Syk, Hck, Yes, Blk, Fgr and Frk), Tec, Txk/Rlk, Abl, EGFR (EGFR-l/ErbB-1, ErbB- 2/NEU/HER-2, ErbB-3 and ErbB-4), FAK, FGF1R (also FGFR1 or FGR-1), FGF2R (also FGR-2), MET (also Met-I or c-MET), PDGFR (a and β), Tie-1, Tie-2 (also Tek-1 or Tek), VEGFR1 (also FLT-1), c-FMS, VEGFR2 (also KDR), FLT-3, FLT-4, c-KIT, JAK1, JAK2, JAK3, TYK2, LOK, RET,
Ros,TRKA, TRKB, TRKC, PYK2, ALK (Anaplastic Lymphoma Kinase), EPHA (1-8), EPHB (1-6), RON, Fes, Fer or EPHB4 (also EPHB4-1), and
(b) and serine/threonine kinases such as Aurora, c-RAF, SGK, MAP kinases (e.g., MKK4, MKK6, etc.), SAPK2a, SAPK2P, Ark, ATM (1-3), CamK (1-IV), CamKK, Chkl and 2 (Checkpoint kinases), CKI, CK2, Erk, IKK-I (also IKK- ALPHA or CHUK), IKK-2 (also IKK-BETA), Ilk, Jnk (1-3), LimK (1 and 2), MLK3Raf (A, B and C), CDK (1-10), PKC (including all PKC subtypes), Plk (1- 3), NIK, Pak (1-3), PDK1, PKR, RhoK, RIP, RIP-2, GSK3 (a and β), PKA, P38, Erk (1-3), PKB (including all PKB subtypes) (also AKT-1, AKT-2, AKT-3 or AKT3-1), IRAKI, FRK, SGK, TAK1 and Tpl-2 (also COT).
[000123] Phosphorylation modulates or regulates a variety of cellular processes such as proliferation, growth, differentiation, metabolism, apoptosis, motility, transcription, translation and other signaling processes. Aberrant or excessive PTK activity has been observed in many disease states including, but not limited to, benign and malignant proliferative disorders, diseases resulting from inappropriate activation of the immune system and diseases resulting from inappropriate activation of the nervous systems. Specific diseases and disease conditions include, but are not limited to, autoimmune disorders, allograft rejection, graft vs. host disease, diabetic retinopathy, choroidal neovascularization due to age-related macular degeneration, psoriasis, arthritis, osteoarthritis, rheumatoid arthritis, synovial pannus invasion in arthritis, multiple sclerosis, myasthenia gravis, obesity, diabetes mellitus, diabetic angiopathy, retinopathy of prematurity, infantile hemangiomas, non-small cell lung, bladder and head and neck cancers, prostate cancer, breast cancer, ovarian cancer, gastric and pancreatic cancer, psoriasis, fibrosis, rheumatoid arthritis, atherosclerosis, restenosis, auto-immune disease, allergy, respiratory diseases, asthma, transplantation rejection, inflammation, thrombosis, retinal vessel proliferation, inflammatory bowel disease, Crohn's disease, ulcerative colitis, bone diseases, transplant or bone marrow transplant rejection, lupus, chronic pancreatitis, cachexia, septic shock, fibroproliferative and differentiative skin diseases or disorders, central nervous system diseases, neurodegenerative diseases, disorders or conditions related to nerve damage and axon degeneration subsequent to a brain or spinal cord injury, acute or chronic cancer, ocular diseases, viral infections, heart disease, lung or pulmonary diseases or kidney or renal diseases and bronchitis.
[000124] Tyrosine kinases can be broadly classified as receptor-type (having extracellular, transmembrane and intracellular domains) or the non-receptor type (being wholly intracellular) protein tyrosine kinases. Inappropriate or uncontrolled activation of many of these kinase (aberrant protein tyrosine kinase activity), for example by over-expression or mutation, results in
uncontrolled cell growth. Many of the protein tyrosine kinases, whether a receptor or non-receptor tyrosine kinase have been found to be involved in cellular signaling pathways involved in numerous pathogenic conditions, including, but not limited to, immunomodulation, inflammation, or proliferative disorders such as cancer.
[000125] Compounds of the invention are inhibitors TrkA, TrkB and TrkC, and as such the compounds and pharmaceutical compositions provided herein are useful for treating diseases or disorders in which such kinases contribute to the pathology and/or symptomology of a disease or disorder associated with such kinases. Such diseases or disorders include, but are not limited to, cancer, pain, cachexia, anorexia nervosa, a proliferative diseases, a pain disorder, a dermatological disease, a metabolic disease, a muscle disease, neurodegenerative diseases and/or disorders, neurological diseases and/or disorders, an inflammatory disease, fibrosis, an infectious disease, a respiratory disease, a pulmonary disease and hyperplasiaan inflammatory disease, and wherein the compound is a compound of Formula (I). In certain embodiments the disease is asthma, chronic obstructive pulmonary disease (COPD), adult respiratory distress syndrome (ARDS), bronchitis, dermatitis, allergic rhinitis, psoriasis, scleroderma, lymphoma, metastasis, anaplastic large-cell lymphoma, osteosarcoma, fibrosarcoma, melanoma, breast cancer, renal cancer, brain cancer, prostate cancer, colorectal cancer, thyroid cancer, ovarian cancer, pancreatic cancer, neuronal cancer, neuroblastoma, lung cancer, uterine cancer, gastrointestinal cancer, colon cancer or papillary thyroid carcinoma.
Receptor Tyrosine Kinases (RTKs).
[000126] The Receptor Tyrosine Kinases (RTKs) comprise a large family of transmembrane receptors with diverse biological activities. A number of distinct RTK subfamilies have been identified including, but not limited to, ALK receptor family, EGF receptor family, the Insulin receptor family, the PDGF receptor family, the FGF receptor family, the VEGF receptor family, the HGF receptor family, the Trk receptor family, the EPH receptor family, the AXL receptor family, the LTK receptor family, the TIE receptor family, the ROR receptor family, the DDR receptor family, the RET receptor family, the KLG receptor family, the RYK receptor family and the MuSK receptor family.
[000127] Receptor tyrosine kinases have been shown to be not only key regulators of normal cellular processes but also to have a critical role in the development and progression of many types of cancer. The receptor tyrosine kinase (RTK) family includes receptors that are crucial for the growth and differentiation of a variety of cell types. The intrinsic function of RTK mediated signal transduction is initiated by extracellular interaction with a specific growth factor (ligand), typically followed by receptor dimerization, stimulation of the intrinsic protein tyrosine kinase activity and receptor trans-phosphorylation. Binding sites are thereby created for intracellular signal transduction molecules and lead to the formation of complexes with a spectrum of cytoplasmic signaling molecules that facilitate the appropriate cellular response such as, by way of example only, cell division, differentiation, metabolic effects, and changes in the extracellular microenvironment.
Tropomyosin-Receptor-Kinase (Trk) Family
[000128] The Trk family receptor tyrosine kinases (NTRK genes), TrkA (NTRK1), TrkB
(NTRK2), and TrkC (NTRK3), are the signaling receptors that mediate the biological actions of the peptide hormones of the neurotrophin family. Trk receptors are membrane-bound receptors that, through several signal cascades, control neuronal growth and survival, and differentiation, migration and metastasis of tumor cells. The neurotrophin family of growth factors includes nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and two neurotrophins (NT), NT- 3, and NT-4. Neurotrophins are critical to the functioning of the nervous system, and the activation of Trk receptors by neurotrophin binding leads to activation of signal cascades resulting in promoting survival and other functional regulation of cells. Each type of neurotrophin has a different binding affinity toward its corresponding Trk receptor, and upon neurotrophin binding, the Trk receptors phosphorylates themselves and members of the MAPK pathway. The differences in the signaling initiated by these distinct types of receptors are important for generating diverse biological responses.
[000129] The Trk family kinase receptors promote tumorigenesis and are able to control tumor cell growth and survival as well as differentiation, migration and metastasis. The Trk receptors are implicated in the development and progression of cancer, through upregulation of either, the receptor, their ligand (NGF, BDNF, NT-3, and NT-4), or both. In many cases high Trk expression is associated with aggressive tumor behavior, poor prognosis and metastasis. Thus, diseases and disorders related to Trk receptors result from 1) expression of a Trk receptor(s) in cells which normally do not express such a receptor(s); 2) expression of a Trk receptor(s) by cells which normally do not express such a receptor(s); 3) increased expression of Trk receptor(s) leading to unwanted cell proliferation; 4) increased expression of Trk receptor(s) leading to adhesion independent cell survival; 5) mutations leading to constitutive activation of Trk receptor(s); 6) over stimulation of Trk receptor(s) due to abnormally high amount of, or mutations in, Trk receptor(s), and/or 7) abnormally high amount of Trk receptor(s) activity due to abnormally high amount of, or mutations in, Trk receptor(s).
[000130] Genetic abnormalities, i.e. point mutations and chromosomal rearrangements involving both the genes expressing TrkB and TrkC have been found in a variety of cancer types. In a kinome-wide approach to identify point mutants in tyrosine kinases, mutations in the genes expressing TrkB and TrkC were found in cell lines and primary samples from patients with colorectal cancer. In addition, chromosomal translocations involving the genes expressing TrkA and TrkB have been found in several different types of tumors. Gene rearrangements involving the genes expressing TrkA and a set of different fusion partners (TPM3, TPR, TFG) are a hallmark of a subset of papillary thyroid cancers. Moreover, secretary breast cancer, infant fibrosarcoma and congenital mesoblastic nephroma have been shown to be associated with a chromosomal rearrangement t(12;15) generating a ETV6-NTRK3 fusion gene that was shown to have constitutive kinase activity and transforming potential in several different cell lines including fibroblasts, hematopoietic cells and breast epithelial cells.
[000131] TrkA has the highest affinity to the binding nerve growth factor (NGF). NGF is important in both local and nuclear actions, regulating growth cones, motility, and expression of genes encoding the biosynthesis enzymes for neurotransmitters. Nocireceptive sensory neurons express mostly TrkA and not TrkB or TrkC.
[000132] TrkB serves as a receptor for both BDNF and NT-4, and is expressed in
neuroendocrine-type cells in the small intestine and the colon, in the alpha cells of the pancreas, in the monocytes and macrophages of the lymph nodes and of the spleen, and in the granular layers of the epidermis. TrkB is also expressed in cancerous prostate cells but not in normal cells. The binding of BDNF to TrkB receptor causes activation of intercellular cascades which regulate neuronal development and plasticity, long-term potentiation, and apoptosis. BDNF promotes the proliferation, differentiation and growth and survival of normal neural components such as retinal cells and glial cells. In addition, TrkB activation is a potent and specific suppressor of anchorage independent cell death (anoikis), which is apoptosis induced by loss of attachment of a cell to its matrix. By way of example, activation of the Phosphatidylinositol-3-kinase/Protein Kinase B signaling axis by TrkB promotes the survival of non-transformed epithelial cells in 3-dimensional cultures and induces tumor formation and metastasis of those cells in immunocompromised mice. Anchorage independent cell survival is a metastatic process allowing tumor cells to migrate through the systemic circulation and grow at distant organs. Agonism of TrkB results in the failure of induced cell death by cancer treatments. Thus, TrkB modulation is a target for treatment of benign and malignant proliferative diseases, especially tumor diseases.
[000133] Diseases and disorders related to the TrkB receptor include, but are not limited to, cancers, such as, by way of example only, neuroblastoma progression, Wilm's tumor progression, breast cancer, pancreatic cancer, colon cancer, prostate cancer, and lung cancer. The TrkB receptor has been shown to be associated with Alzheimer's disease. [000134] Additional research has discovered mutations in TrkB in humans that result in a partial loss of enzymatic activity of the receptor. This genetic legion results in an increase in apetite and obesity (hyperphagic obesity). Similar results have been obtained in mouse models, thus strengthening the hypothesis that lowering TrkB activity could serve to modulate feeding behavior, and would be useful in the treatment of disorders such as anorexia.
[000135] Other non-oncology indications for a Trk inhibitor include atopic dermatitis and psoriasis.
[000136] TrkC is activated by binding with NT-3 and is expressed by proprioceptive sensory neurons. The axons of these proprioceptive sensory neurons are much thicker than those of nocireceptive sensory neurons, which express TrkA. Signalling through TrkC leads to cell differentiation and development of proprioceptive neurons that sense body position. Mutations in this gene expressing TrkC is associated with medulloblastomas, secretory breast carcinomas and other cancers. In addition, high expression of TrkC is a hallmark of melanoma, especially in cases with brain metastasis.
[000137] Trk family members, especially NTRK1 and NTRK2, play a role in pancreatic cancer wherein: i) high expression of various members of the Trk family and their cognate ligands have been shown in tissue samples from patients with pancreatic cancer; ii) NTRK2 overexpression has been linked to a malignant, highly metastatic phenotype of pancreatic cancer; iii) high expression of NTRK1/NGF, has been correlated with enhanced proliferation, invasive behavior and pain in PC patients; and iv) nerve growth factor has been shown to increase the invasive potential of pancreatic cancer cell lines. Overexpression of TrkA in pancreatic cancer might be caused by methylation of negative regulatory AP-1 sites in the promoter region of TrkA.
[000138] Gene rearrangements involving NTRK1 are a hallmark of a subset of papillary thyroid cancers. Thyroid- specific TRK oncogenes are generated by rearrangements of the NTRK1 gene with three different activating genes, namely TPR, TPM3, and TFG.
[000139] Several loss of function mutations in thr TrkA are responsible for congenital insensitivity to pain with anhidrosis (CIPA), a disorder characterized by a lack of pain sensation and anhidrosis. More recently, an antagonistic TrkA antibody has been shown to be efficacious in inflammatory and neupathic pain animal models. In addition, TrkA and NGF have been implicated in eliciting cancer related pain. It was shown that NGF secreted by tumor cell and tumor invading macrophages secret NGF which directly stimulates TrkA located on peripheral pain fibers. Using various tumor models in both mouse and rats it was demonstrated that neutralizing NGF with a monoclonal antibody inhibits cancer related pain to a degree similar or superior to the highest tolerated dose of morphine. Therefore, a selective inhibitor of TrkA can be used in the treatment of pain associated with cancer.
[000140] High expression of Trk's are found in Wilm's tumor, prostate carcinoma and pancreatic cancers. High expression of TrkC is a hallmark of carcinoma. In neuroblastoma, high TRKB expression is correlated with an aggressive untreatable tumors and resistance to standard cytotoxic therapies. In mouse models of cancer metastasis, the NTRK2 gene (TrkB protein) can induce metastasis and removal of the gene reverses this metastatic potential. The bulk of evidence suggests that inhibition of Trk enzymes would block the growth and spread of various cancers where Trk is involved. Furthermore, activating mutations in Trk's are present in 7 % of cancers.
[000141] Certain compounds, pharmaceutical compositions and pharmaceutical combination provided herein are inhibitors of Trk receptor tyrosine kinases (TrkA, TrkB, and TrkC), thus such compounds, pharmaceutical compositions and pharmaceutical combination are useful for the treatment of diseases and/or disorders that respond to inhibition of Trk receptor tyrosine kinases (TrkA, TrkB, and TrkC). In certain embodiments, such compounds, pharmaceutical compositions and pharmaceutical combination are useful in the treatment of cancer by inhibiting the
development and/or progression of the cancer. In certain embodiments, such compounds, pharmaceutical compositions and pharmaceutical combination are useful in the treatment of diseases and/or disorders including, but are not limited to, neuroblastoma, Wilm's tumor, breast cancer, pancreatic cancer, colon cancer, prostate cancer, lung cancer, melanoma, anoerexia, atopic dermatitis, psoriasis and Alzheimer's disease.
[000142] In accordance with the foregoing, the present invention further provides a method for preventing or treating any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount (See, "Administration and Pharmaceutical Compositions ", infra) of a compound of Formula I or a pharmaceutically acceptable salt thereof. For any of the above uses, the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.
Routes of Administration and Pharmaceutical Compositions
[000143] For the therapeutic uses of compounds of Formula (I), or pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, described herein, such compounds are administered in therapeutically effective amounts either alone or as part of a pharmaceutical composition. Accordingly, provided herein are pharmaceutical compositions, which comprise at least one compound of Formulas (I) described herein, pharmaceutically acceptable salts and/or solvates thereof, and one or more pharmaceutically acceptable carriers, diluents, or excipients. In addition, such compounds and compositions are administered singly or in combination with one or more additional therapeutic agents. The routes of administration of compounds of Formula (I) and pharmaceutical compositions include, but are not limited to, oral administration, intravitreal administration, rectal administration, parenteral, intravenous administration, intraperitoneal administration, intramuscular administration, inhalation, transmucosal administration, pulmonary administration, intestinal administration, subcutaneous administration, intramedullary
administration, intrathecal administration, direct intraventricular, intranasal administration, topical administration, ophthalmic administration or otic administration.
[000144] In certain embodiments, compounds of Formula (I) or pharmaceutical compositions described herein are administered locally, while in other embodiments compounds of Formula (I) or pharmaceutical composite described herein are administered systemically. Local administration includes, but is not limited to, injection into an organ, optionally in a depot or sustained release formulation. Systemic administration includes, but is not limited to, oral administration or intravenous administration. In other embodiments, compounds of Formula (I) or pharmaceutical compositions described herein are administered in a targeted drug delivery system, such as, by way of example only, in a liposome coated with organ-specific antibody. The liposome is targeted to and taken up selectively by the organ. In other embodiments, compounds of Formula (I) or pharmaceutical compositions described herein are administered in the form of a rapid release formulation, while in other embodiments, compounds of Formula (I) or pharmaceutical compositions described herein are administered in the form of an extended release formulation . In other embodiments, compounds of Formula (I) or pharmaceutical compositions described herein are administered in the form of an intermediate release formulation.
[000145] The therapeutically effective amount will vary depending on, among others, the disease indicated, the severity of the disease, the age and relative health of the subject, the potency of the compound administered, the route of administration and the treatment desired. In certain embodiments, satisfactory results are indicated to be obtained at daily dosages daily dosage of a compound of Formula (I) from about 0.03 to 2.5mg/kg per body weight. In certain embodiments, the daily dosage of a compound of Formula (I), administered orally, is in the range from 0.05 micrograms per kilogram body weight ^g/kg) to 100 micrograms per kilogram body weight ^g/kg). In certain embodiments, the daily dosage of a compound of Formula (I), administered topically, is in the range from 0.05 micrograms per kilogram body weight ^g/kg) to 100 micrograms per kilogram body weight ^g/kg). In other embodiments, the daily dosage of a compound of Formula (I), administered parenterally, is in the range from 0.05 micrograms per kilogram body weight ^g/kg) to 100 milligrams per kilogram body weight (mg/kg). In certain embodiments, the daily dosage of a compound of Formula (I), administered intrermuscularlly, is in the range from 0.05 micrograms per kilogram body weight ^g/kg) to 100 micrograms per kilogram body weight ^g/kg). An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5mg to about lOOmg of a compound of Formula (I), conveniently administered, e.g. in divided doses up to four times a day or in controlled release form. In certain embodiment, unit dosage forms for oral administration comprise from about 1 to 50 mg of a compound of Formula (I).
[000146] Other aspects provided herein are processes for the preparation of pharmaceutical composition which comprise at least one compound of Formula (I) described herein. In certain embodiments, such processes include admixing a compound of Formula (I) described herein with one or more pharmaceutically acceptable carriers, diluents or excipients. In certain embodiments, the pharmaceutical compositions comprise a compound of Formula (I) in free form or in a pharmaceutically acceptable salt or solvate form. In certain embodiments, the pharmaceutical compositions comprising a compound of Formula (I) in free form or in a pharmaceutically acceptable salt or solvate form, in association with at least one pharmaceutically acceptable carrier, diluent or excipient are manufactured by mixing, dissolving, granulating dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes and/or coating methods. In other embodiments, such compositions are optionally contain excipients, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In other embodiments, such compositions are sterilized.
Oral Dosage Forms
[000147] In certain embodiments, the pharmaceutical compositions containing at least one compound of Formula (I) are administered orally as discrete dosage forms, wherein such dosage forms include, but are not limited to, capsules, gelatin capsules, caplets, tablets, chewable tablets, powders, pills, dragees, granules, liquids, gels, syrups, flavored syrups, elixirs, slurries, solutions or suspensions in aqueous or non-aqueous liquids, edible foams or whips, and oil-in-water liquid emulsions or water-in-oil liquid emulsions. The capsules, gelatin capsules, caplets, tablets, chewable tablets, powders or granules, used for the oral administration of at least one compound of Formula (I) are prepared by admixing at least one compound of Formula (I) (active ingredient) together with at least one excipient using conventional pharmaceutical compounding techniques. Non-limiting examples of excipients used in oral dosage forms described herein include, but are not limited to, binders, fillers, disintegrants, lubricants, absorbents, colorants, flavors,
preservatives and sweeteners.
[000148] Non-limiting examples of such binders include, but are not limited to, corn starch, potato starch, starch paste, pre- gelatinized starch, or other starches, sugars, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, tragacanth, guar gum, cellulose and its derivatives (by way of example only, ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethylcellulose, methyl cellulose,
hydroxypropyl methylcellulose and microcrystalline cellulose), magnesium aluminum silicate, polyvinyl pyrrolidone and combinations thereof.
[000149] Non-limiting examples of such fillers include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre- gelatinized starch, and mixtures thereof. In certain embodiments, the binder or filler in pharmaceutical compositions provided herein are present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
[000150] Non-limiting examples of such disintegrants include, but are not limited to, agar-agar, alginic acid, sodium alginate, calcium carbonate, sodium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre- gelatinized starch, other starches, clays, other algins, other celluloses, gums, and combinations thereof. In certain embodiments, the amount of disintegrant used in the pharmaceutical compositions provided herein is from about 0.5 to about 15 weight percent of disintegrant, while in other embodiments the amount is from about 1 to about 5 weight percent of disintegrant.
[000151] Non-limiting examples of such lubricants include, but are not limited to, sodium stearate, calcium stearate, magnesium stearate, stearic acid, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, sodium lauryl sulfate, talc, hydrogenated vegetable oil (by way of example only, peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, sodium oleate, ethyl oleate, ethyl laureate, agar, silica, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.) and combinations thereof. In certain embodiments, the amount of lubricants used in the pharmaceutical compositions provided herein is in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms.
[000152] Non-limiting examples of such diluents include, but are not limited to, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, glycine or combinations thereof.
[000153] In certain embodiments, tablets and capsules are prepared by uniformly admixing at least one compound of Formula (I) (active ingredients) with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary. In certain embodiments, tablets are prepared by compression. In other embodiments, tablets are prepared by molding.
[000154] In certain embodiments, at least one compound of Formula (I) is orally administered as a controlled release dosage form. Such dosage forms are used to provide slow or controlled-release of one or more compounds of Formula (I). Controlled release is obtained using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof. In certain embodiments, controlled-release dosage forms are used to extend activity of the compound of Formula (I), reduce dosage frequency, and increase patient compliance.
[000155] Administration of compound of Formula (I) as oral fluids such as solution, syrups and elixirs are prepared in unit dosage forms such that a given quantity of solution, syrups or elixirs contains a predetermined amount of a compound of Formula (I). Syrups are prepared by dissolving the compound in a suitably flavored aqueous solution, while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions are formulated by dispersing the compound in a nontoxic vehicle. Non-limiting examples of excipients used in as oral fluids for oral administration include, but are not limited to, solubilizers, emulsifiers, flavoring agents, preservatives, and coloring agents. Non-limiting examples of solubilizers and emulsifiers include, but are not limited to, water, glycols, oils, alcohols, ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers. Non-limiting examples of preservatives include, but are not limited to, sodium benzoate. Non-limiting examples of flavoring agents include, but are not limited to, peppermint oil or natural sweeteners or saccharin or other artificial sweeteners. Parenteral Dosage Forms
[000156] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered parenterally by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial.
[000157] Such parenteral dosage forms are administered in the form of sterile or sterilizable injectable solutions, suspensions, dry and/or lyophylized products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection (reconstitutable powders) and emulsions. Vehicles used in such dosage forms include, but are not limited to, Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, physiological saline buffer, Ringer's Injection solution, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection solution; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
[000158] In certain embodiments, a compound of Formula (I) or composition containing one or more compounds of Formula (I) is parenteral administration by bolus injection. In other embodiments, a compound of Formula (I) or composition containing one or more compounds of Formula (I) is parenteral administration by continuous infusion. Formulations for injection are presented in unit dosage form, by way of example only, in ampoules or formulations for injection are presented in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Transdermal Administration
[000159] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered transdermally. Such transdermal dosage forms include "reservoir type" or "matrix type" patches, which are applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of a compound of Formula (I). By way of example only, such transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. In other embodiments, matrix transdermal formulations are used. In certain embodiments transdermal administration is used to provide continuous, while in other embodiments transdermal administration is used to provide discontinuous infusion of a compound of Formula (I) in controlled amounts.
[000160] In certain embodiments, the rate of absorption is slowed by using rate-controlling membranes or by trapping the compound within a polymer matrix or gel. In certain embodiments, transdermal delivery is via a transdermal patch.
[000161] Formulations for transdermal delivery of a compound of Formula (I) include an effective amount of a compound of Formula (I), a carrier and an optional diluent. A carrier includes, but is not limited to, absorbable pharmacologically acceptable solvents to assist passage through the skin of the host, such as water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and combinations thereof.
[000162] In certain embodiments, such transdermal delivery systems include penetration enhancers to assist in delivering one or more compound of Formula (I) to the tissue. Such penetration enhancers include, but are not limited to, acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide;
dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
[000163] In other embodiments, the pH of such a transdermal pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, is adjusted to improve delivery of one or more compounds of Formula (I). In other embodiments, the polarity of a solvent carrier, the ionic strength, or tonicity are adjusted to improve delivery. In other embodiments, compounds such as stearates are added to advantageously alter the
hydrophilicity or lipophilicity of one or more compound of Formula (I) so as to improve delivery. In certain embodiments, such stearates serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery- enhancing or penetration-enhancing agent. In other embodiments, different salts, hydrates or solvates of the compound of Formula (I) are used to further adjust the properties of the resulting composition.
[000164] In other embodiments, transdermal delivery of the compound of Formula (I) is accomplished by means of iontophoretic patches and the like
Topical Dosage Forms
[000165] In certain embodiments at least one compound of Formula (I) is administered by topical application of pharmaceutical composition containing at least one compound of Formula (I) in the form of lotions, gels, ointments solutions, emulsions, suspensions or creams. Suitable formulations for topical application to the skin are aqueous solutions, ointments, creams or gels, while formulations for ophthalmic administration are aqueous solutions. Such formulations optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.
[000166] Such topical formulations include at least one carrier, and optionally at least one diluent. Such carriers and diluents include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and combinations thereof.
[000167] In certain embodiments, such topical formulations include penetration enhancers to assist in delivering one or more compound of Formula (I) to the tissue. Such penetration enhancers include, but are not limited to, acetone; various alcohols such as ethanol, oleyl, and
tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
Pulmonary Administration
[000168] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered by inhalation. Dosage forms for inhaled administration are formulated as aerosols or dry powders. Aerosol formulations for inhalation administration comprise a solution or fine suspension of at least one compound of Formula (I) in a
pharmaceutically acceptable aqueous or non-aqueous solvent. In addition, such pharmaceutical compositions optionally comprise a powder base such as lactose, glucose, trehalose, mannitol or starch, and optionally a performance modifier such as L-leucine or another amino acid, and/or metals salts of stearic acid such as magnesium or calcium stearate.
[000169] In certain embodiments, compound of Formula (I) are be administered directly to the lung by inhalation using a Metered Dose Inhaler ("MDI"), which utilizes canisters that contain a suitable low boiling propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas, or a Dry Powder Inhaler (DPI) device which uses a burst of gas to create a cloud of dry powder inside a container, which is then be inhaled by the patient. In certain embodiments, capsules and cartridges of gelatin for use in an inhaler or insufflator are formulated containing a powder mixture of a compound of Formula (I) and a powder base such as lactose or starch. In certain embodiments, compound of Formula (I) are delivered to the lung using a liquid spray device, wherein such devices use extremely small nozzle holes to aerosolize liquid drug formulations that can then be directly inhaled into the lung. In other embodiments, compound of Formula (I) are delivered to the lung using a nebulizer device, wherein a nebulizers creates an aerosols of liquid drug formulations by using ultrasonic energy to form fine particles that can be readily inhaled. In other embodiments, compound of Formula (I) are delivered to the lung using an electrohydrodynamic ("EHD") aerosol device wherein such EHD aerosol devices use electrical energy to aerosolize liquid drug solutions or suspensions.
[000170] In certain embodiments, the pharmaceutical composition containing at least one compound of Formula (I), or pharmaceutically acceptable salts and solvates thereof, described herein, also contain one or more absorption enhancers. In certain embodiments, such absorption enhancers include, but are not limited to, sodium glycocholate, sodium caprate, N-lauryl-β-ϋ- maltopyranoside, EDTA, and mixed micelles.
[000171] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered nasally. The dosage forms for nasal administration are formulated as aerosols, solutions, drops, gels or dry powders.
Rectal Administration
[000172] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered rectally in the form of suppositories, enemas, retention enemas ointment, creams rectal foams or rectal gels. In certain embodiments such suppositories are prepared from fatty emulsions or suspensions, cocoa butter or other glycerides.
Depot Administration
[000173] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are formulated as a depot preparation. Such long acting formulations are administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. In certain embodiments, such formulations include polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[000174] In certain embodiments injectable depot forms are made by forming microencapsulated matrices of the compound of Formula (I) in biodegradable polymers. The rate of compound of Formula (I) release is controlled by varying the ratio of compound of Formula (I) to polymer and the nature of the particular polymer employed. In other embodiments, depot injectable
formulations are prepared by entrapping the compound of Formula (I) in liposomes or microemulsions.
Ophthalmic Administration
[000175] In certain embodiments, a compound of Formula (I) or pharmaceutical composition described herein are ophthalmically administered to the eye. Administration to the eye generally results in direct contact of the agents with the cornea, through which at least a portion of the administered agents pass. In certain embodiments, such compounds of Formula (I) or
pharmaceutical compositions have an effective residence time in the eye of about 2 to about 24 hours. In certain embodiments, such compounds of Formula (I) or pharmaceutical compositions have an effective residence time in the eye of about 4 to about 24 hours. In certain embodiments, such compounds of Formula (I) or pharmaceutical compositions have an effective residence time in the eye of about 6 to about 24 hours.
[000176] Ophthalmic administration, as used herein, includes, but is not limited to, topical administration, intraocular injection, subretinal injection, intravitreal injection, periocular administration, subconjuctival injections, retrobulbar injections, intracameral injections (including into the anterior or vitreous chamber), sub-Tenon's injections or implants, ophthalmic solutions, ophthalmic suspensions, ophthalmic ointments, ocular implants and ocular inserts, intraocular solutions, use of iontophoresis, incorporation in surgical irrigating solutions, and packs (by way of example only, a saturated cotton pledget inserted in the fornix). In certain embodiments, the compounds of Formula (I) or pharmaceutical composition described herein are formulated as an ophthalmic composition and are administered topically to the eye. Such topically administered ophthalmic compositions include, but are not limited to, solutions, suspensions, gels or ointments.
[000177] In certain embodiments the pharmaceutical compositions, comprising at least one compound of Formula (I) described herein, used for ophthalmic administration take the form of a liquid where the compositions are present in solution, in suspension or both. In some
embodiments, a liquid composition includes a gel formulation. In other embodiments, the liquid composition is aqueous. In other embodiments, such liquid compositions take the form of an ointment. In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered ophthamically as eye drops formulated as aqueous solutions that optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. A desired dosage is administered via a known number of drops into the eye. By way of example only, for a drop volume of 25 μΐ, administration of 1-6 drops delivers 25-150 μΐ of the
composition. In certain embodiments, the aqueous compositions contain from about 0.01% to about 50% weight/volume of a compound of Formula (I). In other embodiments, the aqueous compositions contain from about 0.1% to about 20% weight/volume of a compound of Formula (I). In still other embodiments, the aqueous compositions contain from about 0.2% to about 10% weight/volume of a compound of Formula (I). In certain embodiments, the aqueous compositions contain from about 0.5% to about 5%, weight/volume of a compound of Formula (I).
[000178] In certain embodiments the aqueous compositions have an ophthalmically acceptable pH and osmolality. In certain embodiments the aqueous compositions include one or more ophthalmically acceptable pH adjusting agents or buffering agents, including acids such as acetic, boric, citric, lactic, phosphoric and hydrochloric acids; bases such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate and tris- hydroxymethylaminomethane; and buffers such as citrate/dextrose, sodium bicarbonate and ammonium chloride. Such acids, bases and buffers are included in an amount required to maintain pH of the composition in an ophthalmically acceptable range.
[000179] In certain embodiments the compositions also include also include one or more ophthalmically acceptable salts in an amount required to bring osmolality of the composition into an ophthalmically acceptable range. Such salts include those having sodium, potassium or ammonium cations and chloride, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate or bisulfite anions; suitable salts include sodium chloride, potassium chloride, sodium thiosulfate, sodium bisulfite and ammonium sulfate.
[000180] In certain embodiments the aqueous compositions also contain one or more polymers as suspending agents. Such polymers include, but are not limited to, water-soluble polymers such as cellulosic polymers described herein, (for example only, hydroxypropyl methylcellulose), and water-insoluble polymers described herein (for example only, cross-linked carboxyl-containing polymers). In certain embodiments, the aqueous compositions also include an ophthalmically acceptable mucoadhesive polymer, selected for example from carboxymethylcellulose, carbomer (acrylic acid polymer), poly(methylmethacrylate), polyacrylamide, polycarbophil, acrylic acid/butyl acrylate copolymer, sodium alginate and dextran.
[000181] In certain embodiments the compositions also include ophthalmically acceptable solubilizing agents to aid in the solubility of a compound of Formula (I). The term "solubilizing agent" generally includes agents that result in formation of a micellar solution or a true solution of the agent. In certain embodiments, ophthalmically acceptable nonionic surfactants including, but not limited to, polysorbate 80 are used as solubilizing agents. In other embodiments, ophthalmically acceptable glycols including, but not limited to, polyglycols, polyethylene glycol 400, and glycol ethers are used as solubilizing agents.
[000182] In certain embodiments the compositions also include one or more ophthalmically acceptable surfactants to enhance physical stability or for other purposes. Such nonionic surfactants include, but are not limited to, polyoxyethylene fatty acid glycerides and vegetable oils (by way of example only, polyoxyethylene (60) hydrogenated castor oil) and polyoxyethylene alkylethers and alkylphenyl ethers (by way of example only, octoxynol 10 and octoxynol 40).
[000183] In certain embodiments the compositions also include one or more ophthalmically acceptable preservatives to inhibit microbial activity. Such preservatives include, but are not limited to mercury-containing substances such as merfen and thiomersal; stabilized chlorine dioxide; and quaternary ammonium compounds such as benzalkonium chloride,
cetyltrimethylammonium bromide and cetylpyridinium chloride.
[000184] In certain embodiments the compositions also include one or more antioxidants to enhance chemical stability where required. Such antioxidants include, but are not limited to, ascorbic acid and sodium metabisulfite.
[000185] In certain embodiments, the aqueous compositions provided herein are packaged in single-dose non-reclosable containers, while in other embodiments the aqueous compositions provided herein are packaged in multiple-dose reclosable containers wherein a preservative is included in the composition.
Otic Administration
[000186] In certain embodiments pharmaceutical compositions containing at least one compound of Formula (I) are administered otically as ear drops. Such formulations are aqueous solutions that optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives. Combination Therapies
[000187] In certain embodiments, a compound of Formula (I) provided herein, or a
pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing at least one compound of Formula (I), is administered alone (without an additional therapeutic agent) for the treatment of one or more of the disease and/or disorders associated TrkA, TrkB, and TrkC kinase activity.
[000188] In other embodiments, a compound of Formula (I) provided herein, or a
pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing at least one compound of Formula (I) provided herein, is administered in combination with one or more additional therapeutic agents, for the treatment of one or more of the disease and/or disorders associated with TrkA, TrkB, and TrkC kinase activity.
[000189] In other embodiments, a compound of Formula (I) provided herein, or a
pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing at least one compound of Formula (I) provided herein, is formulated in combination with one or more additional therapeutic agents and administered for the treatment of one or more of the disease and/or disorders associated with TrkA, TrkB, and TrkC kinase activity.
[000190] In another embodiment, a compound of Formula (I) provided herein, or a
pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing at least one compound of Formula (I) provided herein, is administered sequentially with one or more additional therapeutic agents, for the treatment of one or more of the disease and/or disorders associated with TrkA, TrkB, and TrkC kinase activity.
[000191] In other embodiments, the combination treatments provided herein include
administration of a compound of Formula (I) provided herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing a compound of Formula (I) prior to administration of one or more additional therapeutic agents, for the treatment of one or more of the disease and/or disorders associated with with TrkA, TrkB, and TrkC kinase activity.
[000192] In other embodiments, the combination treatments provided herein include
administration of a compound of Formula (I) provided herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing a compound of Formula (I), subsequent to administration of one or more additional therapeutic agents, for the treatment of one or more of the disease and/or disorders associated with TrkA, TrkB, and TrkC kinase activity.
[000193] In other embodiments, the combination treatments provided herein include
administration of a compound of Formula (I) provided herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition containing a compound of Formula (I), concurrently with administration of one or more additional therapeutic agents, for the treatment of one or more of the disease and/or disorders associated with TrkA, TrkB, and TrkC kinase activity.
[000194] In certain embodiments of the combination therapies described herein, the compounds of Formula (I) provided herein, or a pharmaceutically acceptable salts or solvates thereof, and the additional therapeutics agent(s) act additively. In certain embodiments of the combination therapies described herein, the compounds of Formula (I) provided herein, or a pharmaceutically acceptable salts or solvates thereof, and the additional therapeutics agent(s) act synergistically. [000195] In other embodiments, a compound of Formula (I) provided herein, or a
pharmaceutically acceptable salts or solvates thereof, or a pharmaceutical composition containing a compound of Formula (I), is administered to a patient who has not previously undergone or is not currently undergoing treatment with another therapeutic agent.
[000196] The additional therapeutic agents used in combination with at least one compound of Formula (I) provided herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to chemo therapeutic agents, anti-inflammatory agents, bronchodilatory agents, antihistamine agents, decongestant agents, anti-tussive agents, antiproliferative agents, cytostatic agents, cytotoxic agents,
inhibitors of polyamine biosynthesis, inhibitors of a protein kinase, inhibitors of a serine/threonine protein kinase, inhibitors of protein kinase C, inhibitors of a tyrosine protein kinase, inhibitors of EGF receptor tyrosine kinase, (e.g. Iressa®, inhibitors of VEGF receptor tyrosine kinase, (e.g.
PTK787 or Avastin®), inhibitors of PDGF receptor tyrosine kinase, (e.g. STI571 (Glivec®)), a cytokine, a negative growth regulator, such as TGF-B or IFN-B, an aromatase inhibitor (e.g. letrozole (Femara®) or anastrozole), an inhibitor of the interaction of an SH2 domain with a phosphorylated protein, antiestrogens, bisphosphonates ( e.g. AREDIA® or ZOMETA®) and monoclonal antibodies (e.g. against HER2, such as trastuzumab).
[000197] The anti-inflammatory agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, non-steroidal anti-inflammatory drugs such as salicylic acid, acetylsalicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, acetaminophen, indomethacin, sulindac, etodolac, mefenamic acid, meclofenamate sodium, tolmetin, ketorolac, dichlofenac, ibuprofen, naproxen, naproxen sodium, fenoprofen, ketoprofen, flurbinprofen, oxaprozin, piroxicam, meloxicam, ampiroxicam, droxicam, pivoxicam, tenoxicam, nabumetome,
phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, apazone and nimesulide, leukotriene antagonists including, but not limited to, zileuton, aurothioglucose, gold sodium thiomalate and auranofin, steroids including, but not limited to, alclometasone diproprionate, amcinonide, beclomethasone dipropionate, betametasone, betamethasone benzoate, betamethasone
diproprionate, betamethasone sodium phosphate, betamethasone valerate, budesonide, ciclesonide, clobetasol proprionate, clocortolone pivalate, hydrocortisone, hydrocortisone derivatives, desonide, desoximatasone, dexamethasone, flunisolide, flucoxinolide, flurandrenolide, fluticasone propionate, glucocorticosteroids, halcinocide, medrysone, methylprednisolone, methprednisolone acetate, methylprednisolone sodium succinate, mometasone furoate, paramethasone acetate, prednisolone, prednisolone acetate, prednisolone sodium phosphate, prednisolone tebuatate, prednisone, triamcinolone, triamcinolone acetonide, triamcinolone diacetate, and triamcinolone hexacetonide and other anti-inflammatory agents including, but not limited to, methotrexate, colchicine, allopurinol, probenecid, thalidomide or a derivative thereof, 5 -amino salicylic acid, retinoid, dithranol or calcipotriol, sulfinpyrazone and benzbromarone.
[000198] Other anti-inflammatory agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, steroids described in WO 02/88167, WO 02/12266, WO 02/100879, WO 02/00679 (especially those of Examples 3, 11, 14, 17, 19, 26, 34, 37, 39, 51, 60, 67, 72, 73, 90, 99 and 101), WO 03/35668, WO 03/48181, WO 03/62259, WO 03/64445, WO 03/72592, WO 04/39827 and WO 04/66920; non-steroidal glucocorticoid receptor agonists, such as those described in DE 10261874, WO 00/00531, WO 02/10143, WO 03/82280, WO 03/82787, WO 03/86294, WO 03/104195, WO 03/101932, WO 04/05229, WO 04/18429, WO 04/19935, WO 04/26248 and WO 05/05452; LTB4 antagonists such as BIIL 284, CP-195543, DPC11870, LTB4 ethanolamide, LY 293111, LY 255283, CGS025019C, CP-195543, ONO-4057, SB 209247, SC- 53228 and those described in US 5451700 and WO 04/108720; LTD4 antagonists such as montelukast, pranlukast, zafirlukast, accolate, SR2640, Wy-48,252, ICI 198615, MK-571, LY- 171883, Ro 24-5913 and L-648051; dopamine receptor agonists such as cabergoline,
bromocriptine, ropinirole and 4-hydroxy-7-[2-[[2-[[3-(2-phenylethoxy)- propyl]sulfonyl]ethyl]amino]ethyl]-2(3H)-benzothiazolone and pharmaceutically acceptable salts thereof (the hydrochloride being Viozan® - AstraZeneca); PDE4 inhibitors such as cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden),V-11294A (Napp), BAY19-8004 (Bayer), SCH-351591 (Schering-Plough), Arofylline (Almirall Prodesfarma), PD189659 / PD168787 (Parke-Davis), AWD-12-281 (Asta Medica), CDC-801 (Celgene), SelCID(TM) CC-10004 (Celgene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (Kyowa Hakko Kogyo), GRC 3886 (Oglemilast, Glenmark), WO 92/19594, WO 93/19749, WO 93/19750, WO 93/19751, WO 99/16766, WO 01/13953, WO 03/104204, WO 03/104205, WO 04/000814, WO 04/000839 and WO 04/005258 (Merck), WO 04018450, WO 04/018451, WO 04/018457, WO 04/018465, WO 04/018431, WO 04/018449, WO 04/018450, WO 04/018451, WO 04/018457, WO 04/018465, WO 04/019944, WO 04/019945, WO 04/045607, WO 04/037805, WO 04/063197, WO
04/103998, WO 04/111044, WO 05012252, WO 05012253, WO 05/013995, WO 05/030212, WO 05/030725, WO 05/087744, WO 05/087745, WO 05/087749 and WO 05/090345 as well as those described in WO 98/18796 and WO 03/39544; A2a agonists such as those described in EP 409595A2, EP 1052264, EP 1241176, WO 94/17090, WO 96/02543, WO 96/02553, WO
98/28319, WO 99/24449, WO 99/24450, WO 99/24451, WO 99/38877, WO 99/41267, WO 99/67263, WO 99/67264, WO 99/67265, WO 99/67266, WO 00/23457, WO 00/77018, WO 00/78774, WO 01/23399, WO 01/27130, WO 01/27131, WO 01/60835, WO 01/94368, WO 02/00676, WO 02/22630, WO 02/96462, WO 03/086408, WO 04/039762, WO 04/039766, WO 04/045618 and WO 04/046083; and A2b antagonists such as those described in WO 02/42298 and WO 03/042214.
[000199] The bronchodilatory agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, beta-2 adrenoceptor agonists, anticholinergic agents, antimuscarinic agents, ipratropium bromide, oxitropium bromide, tiotropium salts, glycopyrrolate, CHF 4226 (Chiesi), SVT-40776, albuterol (salbutamol), metaproterenol, terbutaline, salmeterol, fenoterol, procaterol, formoterol, carmoterol, and GSK159797 and pharmaceutically acceptable salts thereof.
[000200] Other bronchodilatory agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, compounds (in free or salt or solvate form) of formula I of WO 0075114, preferably compounds of the Examples thereof, compounds (in free or salt or solvate form) of formula I of WO 04/16601 or of formula I of WO 04/087142, compounds, such as those described in EP 147719, EP 1440966, EP 1460064, EP 1477167, EP 1574501, JP 05025045, JP
2005187357, US 2002/0055651, US 2004/0242622, US 2004/0229904, US 2005/0133417, US 2005/5159448, US 2005/5159448, US 2005/171147, US 2005/182091, US 2005/182092, US 2005/209227, US 2005/256115, US 2005/277632, US 2005/272769, US 2005/239778, US 2005/215542, US 2005/215590, US 2006/19991, US 2006/58530, WO 93/18007, WO 99/64035, WO 01/42193, WO 01/83462, WO 02/66422, WO 02/ 70490, WO 02/76933, WO 03/24439, WO 03/42160, WO 03/42164, WO 03/72539, WO 03/91204, WO 03/99764, WO 04/16578, WO 04/22547, WO 04/32921, WO 04/33412, WO 04/37768, WO 04/37773, WO 04/37807, WO 04/39762, WO 04/39766, WO 04/45618 WO 04/46083 , WO 04/80964, WO 04/087142, WO 04/89892, WO 04/108675, WO 04/108676, WO 05/33121, WO 05/40103, WO 05/44787, WO 05/58867, WO 05/65650, WO 05/66140, WO 05/70908, WO 05/74924, WO 05/77361, WO 05/90288, WO 05/92860, WO 05/92887, WO 05/90287, WO 05/95328, WO 05/102350, WO 06/56471, WO 06/74897, WO 06/8173, EP 424021, US 3714357, US 5171744, US 2005/171147, US 2005/182091, WO 01/04118, WO 02/00652, WO 02/51841, WO 02/53564, WO 03/00840, WO 03/33495, WO 03/53966, WO 03/87094, WO 04/18422, WO 04/05285, WO 04/96800, WO 05/77361 and WO 06/48225.
[000201] Dual anti-inflammatory and bronchodilatory agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, dual beta-2 adrenoceptor agonist /muscarinic antagonists such as those disclosed in US 2004/0167167, US 2004/0242622, US 2005/182092, US
2005/256114, US 2006/35933, WO 04/74246, WO 04/74812, WO 04/89892 and WO 06/23475.
[000202] The antihistamine drug substances agents used in combination with at least one compound of Formula (I) described herein, or a pharmaceutically acceptable salt or solvate thereof, include, but are not limited to, cetirizine hydrochloride, levocetirizine, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, dimetinden, ebastine, epinastine, levocabastine, mizolastine and tefenadine as well as those disclosed in WO 03/099807, WO 04/026841 and JP 2004107299.
[000203] In certain embodiments, the additional thereapeutic agent(s) used in the combination therapies described herein include, but are not limited to, non-selective cyclo-oxygenase COX- l/COX-2 inhibitors (by way of example only, piroxicam, diclofenac, propionic acids such as naproxen, flubiprofen, fenoprofen, ketoprofen and ibuprofen, fenamates such as mefenamic acid, indomethacin, sulindac, azapropazone, pyrazolones such as phenylbutazone, salicylates such as aspirin), COX-2 inhibitors (by way of example only, meloxicam, celecoxib, rofecoxib, valdecoxib, lumarocoxib, parecoxib and etoricoxib); glucocorticosteroids; methotrexate, lefunomide;
hydroxychloroquine, d-penicillamine, auranofin or other parenteral or oral gold preparations.
[000204] Chemotherapeutic agents or other anti-proliferative agents used in combination with the compounds provided herein to treat proliferative diseases and cancer include, but are not limited to, surgery, radiotherapy (γ-radiation, neutron beam radiotherapy, electron beam
radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other chemotherapeutic drugs, including, but not limited to, anthracyclines, alkyl sulfonates, aziridines, ethylenimines, methylmelamines, nitrogen mustards, nitrosoureas, folic acid analogs, dihydrofolate reductase inhibitor, purine analogs, pyrimidine analogs, podophyllotoxins, platinum- containing agents, interferons, interleukins, alkylating agents (mechlorethamine, chlorambucil, Cyclophosphamide, Melphalan, Ifosfamide), antimetabolites (Methotrexate, gemcitabine or capecitabine), purine antagonists and pyrimidine antagonists (6-Mercaptopurine, 5-Fluorouracil, Cytarabile, Gemcitabine), spindle poisons/ microtubule active agents (Vinblastine, Vincristine, Vinorelbine, Paclitaxel, epothilone), topoisomerase I inhibitors, topoisomerase II inhibitors, podophyllotoxins (Etoposide, Irinotecan, Topotecan), antibiotics (Doxorubicin, Bleomycin, Mitomycin), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carboplatin), enzymes (Asparaginase), and hormones (Tamoxifen, Leuprolide, Flutamide, and Megestrol), GLEEVEC™, adriamycin, dexamethasone, cyclophosphamide, busulfan, improsulfan, piposulfan, benzodepa, carboquone, meturedepa, uredepa, altretamine, triethylenemelamine,
triethylenephosphoramide, triethylenethiophosphoramide, trimethylolomelamine, chlorambucil, chlornaphazine, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard, carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine, dacarbazine, mannomustine, mitobronitol, mitolactol, pipobroman, aclacinomycins, actinomycin F(l), anthramycin, azaserine, bleomycin, cactinomycin, carubicin, carzinophilin, chromomycin, dactinomycin, daunorubicin, daunomycin, 6-diazo-5-oxo-l-norleucine, doxorubicin, epirubicin, mitomycin C, mycophenolic acid, nogalamycin, olivomycin, peplomycin, plicamycin,
porfiromycin, puromycin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin, denopterin, methotrexate, pteropterin, trimetrexate, fludarabine, 6-mercaptopurine, thiamiprine, thioguanine, ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, fluorouracil, tegafur, L-asparaginase, pulmozyme, aceglatone, aldophosphamide glycoside, aminolevulinic acid, amsacrine, bestrabucil, bisantrene, carboplatin, cisplatin, defofamide, demecolcine, diaziquone, elfornithine, elliptinium acetate, etoglucid, etoposide, flutamide, gallium nitrate, hydroxyurea, interferon-alpha, interferon-beta, interferon-gamma, interleukin-2, lentinan, lonidamine, methotrexate, mitoguazone, mitoxantrone, mopidamol, nitracrine, pentostatin, phenamet, pirarubicin, podophyllinic acid, 2-ethylhydrazide, procarbazine, razoxane, sizofiran, spirogermanium, paclitaxel, tamoxifen, teniposide, tenuazonic acid, triaziquone or combinations thereof.
[000205] Other agents used in combination with the compounds provided herein include, but are not limited to: treatments for asthma such as albuterol and SINGULAIR™; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; agents for treating blood disorders such as corticosteroids and anti-leukemic agents. Treatment of Diseases Mediated by Kinase Activity
[000206] Compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and combination therapies provided herein are inhibitors of TrkA, TrkB, and TrkC kinase activity, and are useful in the treatment and/or prevention of diseases and/or disorders in which aberrant, abnormal or deregulated activity of TrkA, TrkB, and TrkC kinase contributes to the pathology and/or symptomology of such diseases and/or disorders. Such diseases and/or disorders mediated by TrkA, TrkB, and TrkC kinases are provided herein.
[000207] In certain embodiments, such diseases and/or disorders associated with TrkA, TrkB, and TrkC, kinases include, but are not limited to, cancer, proliferative diseases, pain,
dermatological diseases and/or disorders, metabolic diseases and/or disorders, muscle diseases and/or disorders, neurodegenerative diseases and/or disorders, neurological diseases and/or disorders, inflammatory diseases, fibrosis, infectious diseases, respiratory diseases and/or disorders, pulmonary diseases and/or disorders and hyperplasia.
[000208] Such cancer and proliferative diseases include, but are not limited to, hematopoietic disorders, hematopoietic malignancies, non-hematopoietic malignancies, benign or malignant tumors, tumors of the neck and head, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, neuronal cancer, neuroblastoma, bladder cancer, breast cancer, secretory breast carcinoma, stomach cancer, gastric tumors, ovarian cancer, uterine cancer, colon cancer, rectal cancer, colorectal adenoma, prostate cancer, renal cancer, brain cancer, endometrial cancer, pancreatic cancer, lung cancer, non- small cell lung cancer, human adenoid cystic carcinoma, vaginal cancer, thyroid cancer, papillary thyroid carcinoma, sarcoma, congenital fibrosarcoma, osteolytic sarcoma, osteosarcoma, fibrosarcoma, myeloma, tumor metastasis to bone, congenital mesoblastic nephroma, glioblastomas, melanoma, multiple myeloma, gastrointestinal cancer, gastrointestinal stromal tumors (GIST), mastocytosis, neuroblastoma, fibrotic cancers, tumor metastasis growth, epidermal hyperproliferation, psoriasis, metastasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, lymphomas, diffuse large B-cell lymphoma, B-cell lymphoma, mammary carcinoma, Wilm's tumor, Cowden syndrome, Lhermitte-Dudos disease and Bannayan-Zonana syndrome.
[000209] Such hematopoietic disorders include, but are not limited to, myeloproliferative disorders, thrombocythemia, essential thrombocytosis (ET), angiogenic myeloid metaplasia, myelofibrosis (MF), myelofibrosis with myeloid metaplasia (MMM), chronic idiopathic myelofibrosis (IMF), polycythemia vera (PV), the cytopenias, and pre-malignant myelodysplastic syndromes.
[000210] Such hematological malignancies include, but are not limited to, leukemias, myeloid leukemias, hairy cell leukemia, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, including, but are not limited to, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML with trilineage myelodysplasia (AML/TMDS), mixed lineage leukemia (MLL),
myelodysplastic syndromes (MDSs), myeloproliferative disorders (MPD), multiple myeloma, (MM), myeloid sarcoma and acute promyelocytic leukemia (APL).
[000211] Such pain disorders include, but are not limited to, cancer-related pain, skeletal pain caused by tumor metastasis, osteoarthritis, visceral pain, inflammatory pain and neurogenic pain.
[000212] Such dermatological diseases and/or disorders include, but are not limited to, inflammatory or allergic conditions of the skin, dermatitis, eczema, psoriasis, atopic dermatitis, seborrhoeic dermatitis (Dandruff, Cradle cap), diaper rash, urushiol-induced contact dermatitis, contact dermatitis, erythroderma, lichen simplex chronicus, prurigo nodularis, itch, pruritus ani, nummular dermatitis, dyshidrosis, pityriasis alba, alopecia areata, erythema multiforma, dermatitis herpetiformis, scleroderma, vitiligo, hypersensitivity angiitis, urticaria, bullous pemphigoid, lupus erythematosus, pemphigus, epidermolysis bullosa acquisita, peritoneal and sub dermal adhesion and photoaging of the skin.
[000213] Such metabolic diseases and/or disorders and eating disorder include, but are not limited to, obesity, diabetes and anoerexia.
[000214] Such muscle diseases and/or disorders include, but are not limited to, muscular atrophies (e.g. disuse), muscular dystrophies (e.g. Duchenne's muscle dystrophy, Becker's muscle dystrophy, Limb-Girdle muscle dystrophy), sarcopenia, cachexia, wasting and
Facioscapulohumeral dystrophy.
[000215] Such neurological diseases and/or disorders and neurodegenerative disorders include, but are not limited to, impaired neurological function and Alzheimer's disease. [000216] Such inflammatory diseases and/or disorders include, but are not limited to, uveitis, atherosclerosis, atherogenesis, glomerulonephritis, Kawasaki disease, inflammatory responses, polymyositis, arthritis, neurological inflammation, chronic arthritis inflammation and
osteoarthritis.
[000217] Such fibrosis diseases and/or disorders include, but are not limited to, extracellular matrix accumulation and fibrosis, scleroderma, fibrosclerosis, radiation-induced fibrosis, kidney fibrosis, lung fibrosis and liver fibrosis, haemochromatosis, primary biliary cirrhosis, restenosis, retroperitoneal fibrosis, mesenteric fibrosis, endometriosis and keloids.
[000218] Such infectious diseases and/or disorders include, but are not limited to, Chagas disease.
[000219] Such respiratory diseases and/or disorders and pulmonary disorders include, but are not limited to, asthma, bronchial asthma, allergic asthma, intrinsic (non-allergic) asthma, extrinsic (allergic) asthma, exercise-induced asthma, drug-induced asthma (including aspirin and NSAID- induced) and dust-induced asthma, chronic obstructive pulmonary disease (COPD); chronic obstructive airways disease (COAD), chronic obstructive lung disease (COLD), bronchitis, chronic bronchitis, acute bronchitis, dyspnea, arachidic bronchitis, catarrhal bronchitis, croupus bronchitis, phthinoid bronchitis, rhinitis, acute rhinitis, chronic rhinitis, rhinitis medicamentosa, vasomotor rhinitis, perennial and seasonal allergic rhinitis, rhinitis nervosa (hay fever), inflammatory or obstructive airways diseases, pulmonary hypertension, acute lung injury, adult/acute respiratory distress syndrome (ARDS), pulmonary fibrosis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, pulmonary disease due to infectious or toxic agents, emphysema, pneumoconiosis, aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis, byssinosis, acute lung injury (ALI), hypereosinophilia, Loffler's syndrome, eosinophilic pneumonia, parasitic (in particular metazoan) infestation (including tropical eosinophilia), bronchopulmonary aspergillosis, polyarteritis nodosa (including Churg- Strauss syndrome), eosinophilic granuloma, eosinophil-related disorders affecting the airways occasioned by drug-reaction, pulmonary hypertension, primary pulmonary hypertension (PPH), secondary pulmonary hypertension (SPH), familial PPH, sporadic PPH, precapillary pulmonary hypertension, pulmonary arterial hypertension (PAH), pulmonary artery hypertension, idiopathic pulmonary hypertension, thrombotic pulmonary arteriopathy (TPA), plexogenic pulmonary arteriopathy, functional classes I to IV pulmonary hypertension, and pulmonary hypertension associated with, related to, or secondary to, left ventricular dysfunction, mitral valvular disease, constrictive pericarditis, aortic stenosis, cardiomyopathy, mediastinal fibrosis, anomalous pulmonary venous drainage, pulmonary venoocclusive disease, collagen vascular disease, congenital heart disease, HIV virus infection, drugs and toxins such as fenfluramines, hypoxemia, pulmonary venous hypertension, chronic obstructive pulmonary disease, interstitial lung disease, sleep-disordered breathing, alveolar hypoventilation disorder, chronic exposure to high altitude, neonatal lung disease, alveolar-capillary dysplasia, sickle cell disease, other coagulation disorder, chronic thromboemboli, connective tissue disease, lupus, schistosomiasis, sarcoidosis or pulmonary capillary hemangiomatosis.
[000220] In certain embodiments, the compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and/or combinations provided herein are used in the treatment and/or prevention of respiratory diseases and/or disorders including, but not limited to, asthma, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, exercise-induced asthma, drug-induced asthma (including aspirin and NSAID-induced) and dust-induced asthma, chronic obstructive pulmonary disease (COPD);
bronchitis, acute and chronic rhinitis including rhinitis medicamentosa, and vasomotor rhinitis; perennial and seasonal allergic rhinitis including rhinitis nervosa (hay fever).
[000221] In certain embodiments, the compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and/or combinations provided herein are used in the treatment and/or prevention of dermatological disorders including, but not limited to, psoriasis, dermatitis, eczema, atopic dermatitis, contact dermatitis, urushiol-induced contact dermatitis, eczematous dermatoses, and delayed-type hypersensitivity reactions; phyto- and photodermatitis; seborrhoeic dermatitis, dermatitis herpetiformis, lichen simplex chronicus, lichen planus, lichen sclerosus et atrophica, discoid lupus erythematosus, diaper rash, erythroderma, prurigo nodularis, itch, pruritus ani, nummular dermatitis, dyshidrosis and pityriasis alba.
[000222] In certain embodiments, the compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and/or combinations provided herein are used in the treatment and/or prevention of cancer including, but not limited to, hematopoietic disorders, hematopoietic malignancies, non-hematopoietic malignancies, benign or malignant tumors, tumors of the neck and head, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, neuronal cancer, neuroblastoma, bladder cancer, breast cancer, secretory breast carcinoma, stomach cancer, gastric tumors, ovarian cancer, uterine cancer, colon cancer, rectal cancer, colorectal adenoma, prostate cancer, renal cancer, brain cancer, endometrial cancer, pancreatic cancer, lung cancer, non-small cell lung cancer, human adenoid cystic carcinoma, vaginal cancer, thyroid cancer, papillary thyroid carcinoma, sarcoma, congenital fibrosarcoma, osteolytic sarcoma, osteosarcoma, fibrosarcoma, myeloma, tumor metastasis to bone, congenital mesoblastic nephroma, glioblastomas, melanoma, multiple myeloma, gastrointestinal cancer, gastrointestinal stromal tumors (GIST), mastocytosis, neuroblastoma, fibrotic cancers, tumor metastasis growth, epidermal hyperproliferation, psoriasis, metastasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, lymphomas, diffuse large B-cell lymphoma, B-cell lymphoma, mammary carcinoma, Wilm's tumor, Cowden syndrome, Lhermitte- Dudos disease and Bannayan-Zonana syndrome.
[000223] In certain embodiments, the compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and/or combinations provided herein are used in the treatment and/or prevention of cancer including, but not limited to, hematopoietic disorders, hematopoietic malignancies, non-hematopoietic malignancies, benign or malignant tumors, tumors of the neck and head, brain cancer, kidney cancer, liver cancer, adrenal gland cancer, neuronal cancer, neuroblastoma, bladder cancer, breast cancer, secretory breast carcinoma, stomach cancer, gastric tumors, ovarian cancer, uterine cancer, colon cancer, rectal cancer, colorectal adenoma, prostate cancer, renal cancer, brain cancer, endometrial cancer, pancreatic cancer, lung cancer, non-small cell lung cancer, human adenoid cystic carcinoma, vaginal cancer, thyroid cancer, papillary thyroid carcinoma, sarcoma, congenital fibrosarcoma, osteolytic sarcoma, osteosarcoma, fibrosarcoma, myeloma, tumor metastasis to bone, congenital mesoblastic nephroma, glioblastomas, melanoma, multiple myeloma, gastrointestinal cancer, gastrointestinal stromal tumors (GIST), mastocytosis, neuroblastoma, fibrotic cancers, tumor metastasis growth, epidermal hyperproliferation, psoriasis, metastasis, prostate hyperplasia, neoplasia, neoplasia of epithelial character, lymphomas, diffuse large B-cell lymphoma, B-cell lymphoma, mammary carcinoma, Wilm's tumor, Cowden syndrome, Lhermitte- Dudos disease and Bannayan-Zonana syndrome. Such hematopoietic disorders include, but are not limited to, myeloproliferative disorders, thrombocythemia, essential thrombocytosis (ET), angiogenic myeloid metaplasia, myelofibrosis (MF), myelofibrosis with myeloid metaplasia (MMM), chronic idiopathic myelofibrosis (IMF), polycythemia vera (PV), the cytopenias, and pre- malignant myelodysplastic syndromes. Such hematological malignancies include, but are not limited to, leukemias, myeloid leukemias, hairy cell leukemia, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, including, but are not limited to, acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML with trilineage myelodysplasia (AML/TMDS), mixed lineage leukemia (MLL), myelodysplastic syndromes (MDSs), myeloproliferative disorders (MPD), multiple myeloma, (MM), myeloid sarcoma and acute promyelocytic leukemia (APL).
[000224] Compounds of Formula (I), pharmaceutically acceptable salts, solvates, N-oxides, prodrugs and isomers thereof, pharmaceutical compositions, and combination therapies provided herein are used in methods for inhibiting TrkA, TrkB, and TrkC kinase activity in a subject (human or other mammal) for the treatment and/or prevention of diseases and/or disorders associated with or mediated by TrkA, TrkB, and TrkC kinase activity. In certain embodiments, such methods include administering to a subject an effective amount of a compound of Formula (I), or a pharmaceutical composition containing a compound of Formula (I).
[000225] In certain embodiments, the methods for the treatment of a subject suffering from a disease and/or disorder associated with TrkA, TrkB, and TrkC kinase activity include
administering to the subject an effective amount of a compound of Formula (I), or a
pharmaceutically acceptable salt, solvate thereof, either alone or as part of a pharmaceutical composition as described herein.
[000226] In certain embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof, is used in the preparation of a medicament for the treatment of a disease or disorder associated with TrkA, TrkB, and TrkC kinase activity.
[000227] In accordance with the foregoing, provided herein are methods for preventing, treating and or ameliorating the condition of any of the diseases or disorders described above in a subject in need of such treatment, which method comprises administering to said subject a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. For any of the methods and uses provided herein, the required dosage will vary depending on the mode of administration, the particular condition to be treated and the effect desired.
Examples
[000228] The following examples are offered to illustrate, but not to limit, synthetic methods of compounds of Formula (I). Synthesis of intermediates
nthesis of ethyl 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-10)
Figure imgf000069_0001
1-9 1-10
[000229] A solution of ethyl 3-amino-lH-pyrazole-4-carboxylate (1.28 g, 8.3 mmol) and ethyl propiolate (0.89 g, 9.1 mmol) in EtOH/AcOH, 5: 1 (20 mL) was heated under microwave conditions at 140 °C for 20 minutes. The crude reaction mixture was reduced to dryness and recrystallized from EtOH to afford ethyl 5-hydroxypyrazolo[l,5-a]pyrimidine-3-carboxylate (1-9) as a white crystalline solid. Alternatively, the crude reaction mixture could be purified by column chromatography on silica with hexanes/EtOAc gradient as eluant. 1H NMR (400MHz, DMSO-dg) δ 11.8 (s, 1 H), 8.58 (d, J = 7.6 Hz, 1 H), 8.14 (s, 1 H), 6.15 (d, J = 7.6 Hz, 1 H), 4.28 (q, J = 7.2 Hz, 2 H), 1.30 (t, J = 6.8 Hz, 3 H). MS m/z 208.1 (M+l)+.
[000230] Ethyl 5-hydroxypyrazolo[l,5-a]pyrimidine-3-carboxylate (1-9) (0.43 g, 2.0 mmol) was treated with POCl3 (3 mL) and heated to 100 °C for 1 hour then cooled to room temperature. The reaction was evaporated to dryness, quenched with cold aqueous NaHC03 and extracted with DCM (3 x 25 mL). The combined organic extracts were washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. Crude product was purified by column chromatography on silica gel with hexanes/EtOAc gradient as eluant to afford ethyl 5-chloropyrazolo[l,5- a]pyrimidine-3-carboxylate (1-10). 1H NMR (400MHz, DMSO-d6) δ 8.61 (d, J = 1.2 Hz, 1 H),
8.54 (s, 1 H), 6.97 (d, J = 7.2 Hz, 1 H), 4.40 (q, J = 7.2 Hz, 2 H), 1.40 (t, J = 6.8 Hz, 3 H). MS m/z 226.1 (M+l)+.
-(3-fluorophenyl)pyrrolidine (1-12)
Figure imgf000069_0002
I-ll 1-12
[000231] 3-Chloropropan- 1 -amine hydrochloride (43 g, 331 mmol) was added to a suspension of 3-fluorobenzaldehyde (41 g, 331 mmol) and NaHC03 (39 g, 364 mmol) in water (100 mL). The slurry was stirred overnight. The resulting solution was extracted with EtOAc (3 x 150 mL) and the organic layer was dried with sodium sulfate. The EtOAc was removed to yield 3-chloro-N-(3- fluorobenzylidene)propan-l -amine (I-ll) as a light yellow oil. MS m/z 200.1 (M+l)+. [000232] To a solution of lithium granules (5.5 g, 800 mmol) and 4,4'-di-tert-butylbiphenyl (5.7 g, 21 mmol) in THF (50 mL) at -78 °C was added 3-chloro-N-(3-fluorobenzylidene)propan-l- amine (1-11) (53 g, 266 mmol). The solution was stirred at -78 °C for 2 hours then quenched with water. The solids were removed by filtration and the filtrate was reduced to dryness. The resulting residue was reconstituted in EtOAc and washed with 1 N HC1 (3 x 25 mL). The combined aqueous extracts were washed with EtOAc. The aqueous layer was then neutralized with 1 N NaOH and extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried with sodium sulfate, filtered and reduced to dryness to yield 2-(3-fluorophenyl)pyrrolidine (1-12) as a light yellow oil. MS m/z 165.2 (M+l)+. Note: In some instances the hydrochloride salt of (R)-(3-fluorophenyl)pyrrolidine (I-12A) was purchased from NetChem Company, New
Brunswick, NJ, USA.
Synthesis of ( 2R,4S)-4-fluoro-2-( 3-fluorophenyl)pyrrolidine (1-18)
Figure imgf000070_0001
[000233] To a solution of (R)-4-hydroxypyrrolidin-2-one (5.0 g, 49.5 mmol) in DMF (25 mL) at 0 °C was added TBDMSCl (7.8 g, 52 mmol) and imidazole (5.1 g, 74.3 mmol). The reaction was warmed to room temperature and stirred for 3 hours. The mixture was poured into water and the resulting precipitate was filtered and dried under vacuum overnight to yield (R)-tert-butyl 4-((tert- butyldimethylsilyl)oxy)-2-oxopyrrolidine-l-carboxylate. MS m/z 238.1 (M+23)+. To a solution of this precipitate (10.9 g, 50.7 mmol) in CH3CN (100 mL) at 0 °C under N2 was added TEA (8.5 mL, 61 mmol), DMAP (3.1 g, 25.45 mmol) and di-tert-butyl dicarbonate (14.4 g, 66.2 mmol). The mixture was warmed to room temperature and stirred overnight. The mixture was poured into water and extracted with EtOAc. The organic layer was separated, washed with 1 N HC1, 1 N NaOH and brine, dried over sodium sulfate, filtered and concentrated to yield (R)-tert-butyl 4- (tert-butyldimethylsilyloxy)-2-oxopyrrolidine-l-carboxylate (1-13). MS m/z 338.1 (M+23)+.
[000234] To a solution of (2R)-2-(tert-butyldimethylsilyloxy)-4-(3-fluorophenyl)-4- hydroxybutylcarbamate (1-13) (13.6 g, 43.2 mmol) in THF (100 mL) at 0 °C under N2 was added (3-fluorophenyl)magnesium bromide (52 mL of 1 M solution in THF, 51.84 mmol) over 1 hour. The reaction mixture was stirred at 0 °C for 1 hour. Methanol (80 mL) was added to the mixture followed by NaBH4 (2.45 g, 64.8 mmol) at 0 °C. The mixture was stirred at 0 °C for 1 hour then poured into 10% aqueous NH4C1. The mixture was extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield tert-butyl (2R)-2- (tert-butyldimethylsilyloxy)-4-(3-fluorophenyl)-4-hydroxybutylcarbamate (1-14). MS m/z 436.1 (M+23)+.
[000235] To a solution of tert-butyl (2R)-2-(tert-butyldimethylsilyloxy)-4-(3-fluorophenyl)-4- hydroxybutylcarbamate (1-14) (15.8 g, 38.2 mmol) in DCM (120 mL) at -60 °C under N2 was added TEA (16 mL, 114.6 mmol) and MsCl (3.3 mL, 42.0 mmol). The resulting mixture was stirred at -60 °C for 1 hour. The reaction was poured into water, washed with brine, dried over sodium sulfate, filtered and concentrated to yield (4R)-tert-butyl 4-(tert-butyldimethylsilyloxy)-2- (3-fluorophenyl)pyrrolidine-l-carboxylate (1-15). MS m/z 418.1 (M+23)+. The crude product was used directly in the next step.
[000236] To a solution of (4R)-tert-butyl 4-(tert-butyldimethylsilyloxy)-2-(3- fluorophenyl)pyrrolidine-l-carboxylate (1-15) (18.1 g, 38.2 mmol) in THF (76 mL) at room temperature was added TBAF (50 mL of a 1.0 M solution in THF, 49.7 mmol). The mixture was stirred at room temperature for 2 hours then poured into water. The mixture was extracted with EtOAc, washed with water and brine, dried over sodium sulfate, filtered and concentrated. The mixture was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield (4R)-tert-butyl 2-(3-fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (1-16). MS m/z 304.1 (M+23)+. NOTE: In some instinces the two diasteromers were separated by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield (2R,4R)-tert-butyl 2- (3-fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (I-16A) and (2S,4R)-tert-butyl 2-(3- fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (I-16B). However, better resolution is achieved in the subsequent step. [000237] To a solution of (4R)-tert-butyl 2-(3-fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (1-16) (2.7 g, 9.6 mmol) in DCM (25 mL) in a plastic bottle at -78 °C was added DAST (2.5 mL, 19.2 mmol). The mixture was stirred at -78 °C for 2 hours and then was warmed slowly to room temperature overnight. The mixture was added drop wise to aqueous NaHC03 at 0 °C and was extracted with DCM. The organic layers were combined, washed with brine, dried over sodium sulfate, filtered and concentrated. The two diasteromers were separated by column
chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield (2R,4S)-tert-butyl 4- fluoro-2-(3-fluorophenyl)pyrrolidine-l -carboxylate (I-17A) (first eluting compound) and (2S,4S)- tert-butyl 4-fluoro-2-(3-fluorophenyl)pyrrolidine-l -carboxylate (I-17B). MS m/z 284.1 (M+l)+.
[000238] To a solution of (2R, 4S)-tert-butyl 4-fluoro-2-(3-fluorophenyl)pyrrolidine-l- carboxylate (I-17A) (890 mg, 3.14 mmol) in DCM (5 mL) at room temperature was added TFA (5 mL). The mixture was stirred at room temperature for 2 hours. All the solvent was removed under reduced pressure. The crude was extracted with EtOAc, washed with aqueous NaHC03 and brine, dried over sodium sulfate, filtered and concentrated to yield (2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidine (1-18).1H NMR (400MHz, CDC13) δ 7.31 - 7.25 (m, 1 H), 7.15 - 7.10 (m, 2 H), 6.96 - 6.90 (m, 1 H), 5.29 (d, J = 53.6 Hz 1 H), 4.50 (dd, J = 9.6, 6.4 Hz, 1 H), 3.43 - 3.33 (m, 1 H), 3.31 - 3.28 (m, 1 H), 2.51 (ddd, J = 21.2, 14.0, 6.4 Hz, 1 H), 2.23 (bs, 1 H), 1.79 (dddd, J = 39.6, 14.4, 10.0, 4.4 Hz, 1 H). MS m/z 184.1 (M+l)+.
-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine (1-21 )
Figure imgf000072_0001
[000239] To a solution of (2R,4R)-tert-butyl 2-(3-fluorophenyl)-4-hydroxypyrrolidine-l- carboxylate (I-16A) (1.4 g, 5.0 mmol) and trichloroisocyanuric acid (1.2 g, 5.0 mmol) in DCM (70 mL) at -10 °C was added 2,2,6,6-tetramethylpiperidine-l-oxyl (TEMPO) (0.08 g, 0.5 mmol). The mixture was stirred at -10 °C for 15 minutes, then to room temperature over 1 hour and subsequently poured into cold aqueous NaHC03 containing ice while stirring. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and reduced to dryness to yield (R)-tert-butyl 2-(3-fluorophenyl)-4-oxopyrrolidine-l-carboxylate (1-19) as a light yellow oil. 1H NMR (400MHz, CD3OD) δ 7.40 (q, J = 7.6 Hz, 1 H), 7.01 (d, J = 1.6 Hz, 1 H), 7.03 - 6.99 (m, 2 H), 5.34 (bs, 1 H), 4.07 - 3.91 (m, 2 H), 3.30 (dd, J = 18.8, 10.0 Hz, 1 H), 2.50 (dd, J = 18.8, 3.2 Hz, 1 H), 1.31 (bs, 9H). MS m/z 224.1 (M-56)+.
[000240] To a solution of (R)-tert-butyl 2-(3-fluorophenyl)-4-oxopyrrolidine-l-carboxylate (I- 19) (1.3 g, 4.8 mmol) in DCM (15 mL) in a plastic bottle at -78 °C was added DAST (1.9 mL, 14.4 mmol) drop wise. The resulting orange homogeneous solution was stirred at -78 °C for 30 minutes. The solution was then warmed to room temperature and continued to stir for an additional 2 hours. The resulting solution was poured into stirring ice water (100 mL) and agitated for 15 minutes then extracted with DCM (3 x 50 mL). The combined organic extracts were dried over sodium sulfate, filtered and reduced to dryness. The crude product was purified by flash column chromatography on silica with hexanes/EtOAc gradient as eluant to yield (R)-tert-butyl 4,4-difluoro-2-(3-fluorophenyl)pyrrolidine-l-carboxylate (1-20) as a light yellow oil. 1H NMR (400MHz, CD3OD) δ 7.39 (q, J = 1.6 Hz, 1 H), 7.11 (d, J = 7.6 Hz, 1 H), 7.04 - 6.99 (m, 2 H), 5.03 (bs, 1 H), 4.02 - 3.89 (m, 2 H), 2.99 - 2.86 (m, 1 H), 2.39 - 2.28 (m, 1 H), 1.20 (bs, 9 H). MS m/z 246.1 (M-56)+.
[000241] To a solution of (R)-tert-butyl-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine-l- carboxylate (1-20) (0.9 g, 3.0 mmol) in DCM (20 mL) was added TFA (2 mL, 27 mmol) and stirred at room temperature for 2 hours. The reaction was cooled to 0 °C and aqueous NaHC03 was added while rapidly stirring. The organic phase was separated and washed with aqueous NaHC03, dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield (R)-4,4- difluoro-2-(3-fluorophenyl)pyrrolidine (1-21) as a light yellow oil. 1H NMR (400MHz, CD3OD) δ 7.40 (q, J = 8.0 Hz, 1 H), 7.23 (d, J = 8.8 Hz, 1 H), 7.20 (d, J = 10.0 Hz, 1 H), 7.05 (dt, J = 8.8, 2.4 Hz, 1 H), 4.41 (dd, J = 10.4, 7.2 Hz, 1 H), 3.48 (q, J = 12.0 Hz, 1 H), 3.30 (q, J = 15.2 Hz, 1 H), 2.74 - 2.63 (m, 1 H), 2.29 - 2.14 (m, 1 H). MS m/z 202.1 (M+l)+. nthesis of (2R)-4-fluoro-2-( 3-fluorophenyl)-4-methylpyrrolidine-4-d (1-24 )
Figure imgf000073_0001
1-19 1-22 1-23 1-24
[000242] To a solution of (R)-tert-butyl 2-(3-fluorophenyl)-4-oxopyrrolidine-l-carboxylate (I- 19) (0.2 g, 0.72 mmol) in THF (5 mL) was added sodium borodeuteride (0.12 g, 2.9 mmol) in a single portion. After stirring for 12 h, methanol (5 mL) was added and stirring was continued for 30 minutes. The reaction mixture was reduced to dryness and the residue was triturated with DCM (30 mL). The organic layer was filtered and reduced to dryness to yield (2R)-tert-butyl 2-(3- fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate-4-d (1-22). MS m/z 227.2 (M-56)+.
[000243] To a solution of (2R)-tert-butyl 2-(3-fluorophenyl)-4-hydroxypyrrolidine-l- carboxylate-4-d (1-22) (0.18 g, 0.63 mmol) in DCM (8 mL) at -78 °C was added DAST (0.2 g, 1.27 mmol). The reaction mixture was warmed to room temperature and stirred for 1 hour.
Saturated aqueous NaHC03 (10 mL) was added and stirred for 20 minutes. The organic layer was separated and washed with aqueous NaHC03 (3 x 50 mL), dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield (2R)-tert-butyl 4-fluoro-2-(3-fluorophenyl)pyrrolidine-l- carboxylate -4-d (1-23). MS m/z 228.1 (M-56)+.
[000244] To a solution of (2R)-tert-butyl 4-fluoro-2-(3-fluorophenyl)pyrrolidine-l-carboxylate - 4-d (1-23) (0.1 g, 0.35 mmol) in DCM (10 mL) was added TFA (200 μΐ,, 2.6 mmol). The reaction mixture was stirred for 1 hour at 65 °C and quenched with aqueous NaHC03 (10 mL). The mixture was extracted with DCM, washed with brine, dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield (2R)-4-fluoro-2-(3-fluorophenyl)-4-methylpyrrolidine-4-d (1-24). MS m/z 185.1 (M+l)+.
-(3-fluorophenyl)pyrrolidine-2-d (1-27)
Figure imgf000074_0001
[000245] To a solution of tert-butyl 2-oxopyrrolidine-l-carboxylate (2 g, 10.8 mmol) in THF (30 mL) at 0 °C under argon was slowly added (3-fluorophenyl)magnesium bromide (14.5 mL of a 1 M solution in THF, 14.5 mmol). The resulting solution was warmed to room temperature and stirred for 3 hours. The reaction mixture was quenched with MeOH (50 mL) and stirred for 1 hour. The mixture was reduced to dryness and the resulting residue was reconstituted in EtOAc (150 mL). This solution was washed with brine (3 x 100 mL) and aqueous NaHC03 (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness yielding the crude tert-butyl 2-(3-fluorophenyl)-2-hydroxypyrrolidine-l-carboxylate (1-25). MS m/z 226.1 (M-56)+.
[000246] To a solution of tert-butyl 2-(3-fluorophenyl)-2-hydroxypyrrolidine-l-carboxylate (I- 25) (1.8 g, 3.5 mmol) in MeOH (30 mL) was added concentrated HC1 (3 mL, 34.9 mmol). The resulting solution was stirred for 2 hours at 90 °C and then neutralized with aqueous NaHC03. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 5-(3- fluorophenyl)-3,4-dihydro-2H-pyrrole (1-26). MS m/z 164.1 (M+l)+.
[000247] To a solution of 5-(3-fluorophenyl)-3,4-dihydro-2H-pyrrole (1-26) (1.0 g, 6.1 mmol) in ethanol (30 mL) was added sodium borodeuteride (0.77 g, 18.3 mmol). The reaction mixture was stirred at room temperature for 72 hours then quenched with aqueous NaHC03 (50 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 2-(3-fluorophenyl)pyrrolidine-2-d (1-27). MS m/z 167.1 (M+l)+.
Synthesis of2-(3-fluorophenyl)pyrrolidine-5,5-d2 (1-29)
Figure imgf000075_0001
[000248] To (3-fluorophenyl)magnesium bromide in (100 mL of a 1 M solution in THF, 100 mmol) at -78 °C under argon was added a solution of pyrrolidine-2,5-dione (5 g, 50.5 mmol) in DCM (20 mL) over a period of 10 minutes. The resulting solution was slowly warmed to room temperature and stirred overnight. Sodium cyanoborohydride (3.8 g, 60.2 mmol) was then added to the reaction mixture and stirred for 1 hour. The pH was adjusted to 3-4 by the addition of HC1 (20 mL of a 6 M aqueous solution, 120 mmol) drop wise. After 1 hour the mixture was neutralized with NaOH (4.8 g, 120 mmol) in ethanol (50 mL) and water (10 mL) and extracted with DCM (3 x 100 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 5-(3-fluorophenyl)pyrrolidin-2-one (1-28). MS m/z 180.1 (M+l)+.
[000249] To a solution of 5-(3-fluorophenyl)pyrrolidin-2-one (1-28) (1.0 g, 5.6 mmol) in THF (10 mL) was added lithium aluminum deuteride (28 mL of a 1 M solution in THF) and stirred for 3 hours at room temperature. The reaction mixture was quenched with MeOH (50 mL) and stirred for 1 hour. The solvents were removed and the resulting residue was reconstituted in EtOAc (60 mL) and washed with a aqueous NaHC03. The organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 2-(3-fluorophenyl)pyrrolidine-5,5-d2 (1-29) as a colorless oil. MS m/z 168.1 (M+l)+.
Figure imgf000076_0001
[000250] To a solution of 2-pyrrolidine-3,3,4,4,5,5-d6 (0.5 g, 5.5 mmol) and N,N- dimethylpyridin-4-amine (70 mg, 0.6 mmol), in ACN (50 mL) at -5 °C was slowly added di-tert- butyl dicarbonate (2.4 g, 11 mmol) so not to exceed 0 °C. The reaction mixture was stirred at room temperature for 3 hours then poured into water (200 mL). The resulting mixture was extracted with EtOAc (3 x 75 mL). The combined organic extracts were washed with brine (3 x 50 mL), 1 M HCl (1 x 50 mL) and aqueous NaHC03 (3 x 50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield tert-butyl-2- oxopyrrolidine-l-carboxylate-3,3,4,4,5,5-d6 (1-30). MS m/z 136.1 (M-56)+.
[000251] To a solution of tert-butyl-2-oxopyrrolidine-l-carboxylate-3,3,4,4,5,5-d6 (1-30) (1.0 g, 5.2 mmol) in THF (30 mL) at 0 °C under argon gas was slowly added (3-fluorophenyl)magnesium bromide (10.5 mL of a 1 M solution in THF, 10.5 mmol). The resulting solution was warmed to room temperature and stirred for 3 hours. The reaction mixture was diluted with MeOH (50 mL) and stirred for 1 hour. The solvents were removed and the resulting residue was reconstituted in EtOAc (150 mL). This solution was washed with brine (3 x 100 mL) and aqueous NaHC03 (3 x 100 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield tert-butyl 2-(3-fluorophenyl)-2-hydroxypyrrolidine-l-carboxylate-3,3,4,4,5,5-d6 (1-31). MS m/z 232.1 (M-56)+.
[000252] To a solution of tert-butyl 2-(3-fluorophenyl)-2-hydroxypyrrolidine-l-carboxylate- 3,3,4,4,5,5-d6 (1-31) (1.0 g, 3.5 mmol) in MeOH (30 mL) was added concentrated HCl (2 mL, 23.3 mmol). The resulting solution was stirred for 2 hours at 90 °C then neutralized with aqueous NaHC03. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 5-(3- fluorophenyl)-3,4-dihydro-2H-pyrrole-2,2,3,3,4,4-d6 (1-32). MS m/z 170.1 (M+l)+.
Synthesis of 2 -(3 -fluorophenyl )pyrrolidine-3, 3,4,4, 5,5-d6 (1-33)
Figure imgf000077_0001
[000253] To a solution of 5-(3-fluorophenyl)-3,4-dihydro-2H-pyrrole-2,2,3,3,4,4-d6 (1-32) (0.4 g, 2.4 mmol) in ethanol (10 mL) was added sodium borohydride (0.4 g, 10.5 mmol). The reaction mixture was stirred at room temperature for 3 hours then quenched with aqueous NaHC03 (30 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 2-(3-fluorophenyl)pyrrolidine- 3,3,4,4,5,5-d6 (1-33). MS m/z 172.1 (M+l)+.
Synthesis of -( 3 -fluorophenyl )pyrrolidine-2, 3, 3,4, 4,5,5-d7 ( 1-34 )
Figure imgf000077_0002
[000254] To a solution of 5-(3-fluorophenyl)-3,4-dihydro-2H-pyrrole-2,2,3,3,4,4-d6 (1-32) (0.4 g, 2.4 mmol) and ethanol (10 mL) was added sodium borodeuteride (0.4 g, 9.5 mmol). The reaction mixture was stirred at room temperature overnight then quenched with aqueous NaHC03 (30 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 2-(3-fluorophenyl)pyrrolidine- 2,3,3,4,4,5,5-d7 (1-34). MS m/z 173.2 (M+l)+.
Synthesis of 5-(3-fluorophenyl)pyrrolidin-2-one (1-35)
Figure imgf000077_0003
1-35
[000255] To a solution of l-fluoro-3-iodobenzene (2.0 mL, 17.5 mmol) in anhydrous Et20 (10 mL) was added magnesium (0.48 g, 20 mmol) at room temperature and with cooling as
magnesium was consumed. Upon complete consumption of magnesium, a cold solution of succinimide (0.5 g, 5 mmol) in DCM (25 mL) was added and stirred at room temperature for 12 hours. Subsequently, solid NaCNBH3 (0.38 g, 6.0 mmol) was added. The reaction was acidified to pH 3-4 with 6 M HC1, stirred for 30 minutes and neutralized with 3 M NaOH. The organic layer was separated and the aqueous layers extracted with DCM (3 x 10 mL). The combined organic extracts were washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with
hexanes/EtOAc gradient as eluant to yield 5-(3-fluorophenyl)pyrrolidin-2-one (1-35). 1H NMR (400MHz, DMSO-6) δ 8.13 (s, 1 H), 7.44 - 7.38 (m, 1 H), 7.16 - 7.08 (m, 3 H), 4.69 (t, J = 6.8 Hz, 1 H), 2.48 - 2.43 (m, 1 H), 2.24 (d, J = 8.0 Hz, 1 H), 2.22 (dd, J = 8.0, 2.1 Hz, 1 H), 1.80 - 1.70 (m, 1 H). MS m/z 180.1 (M+l)+.
Synthesis of 5-(3-fluorophenyl)-2,2-dimethylpyrrolidine (1-38)
Figure imgf000078_0001
1-36 TEA, THF 1-37 1-38
[000256] To a solution of 2-methylbut-3-yn-2-amine (1.4 g, 16.6 mmol) in DCM (50 mL) and TEA (2.5 mL, 18.3 mmol) at 0 °C was added benzyl chloroformate (2.4 mL, 16.6 mmol). The reaction was stirred at room temperature for 12 hours and partitioned with water. The organic layer was separated, washed with aqueous NaHC03 and brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with hexanes/EtOAc gradient as eluant to yield benzyl (2-methylbut-3-yn-2-yl)carbamate (1-36) as a clear oil. 1H NMR (400MHz, DMSO-6) δ 7.55 (bs, 1 H), 7.40 - 7.30 (m, 5 H), 5.01 (s, 2 H), 3.01 (s, 1 H), 1.46 (s, 6 H). MS m/z 218.1 (M+l)+.
[000257] To a degassed anhydrous solution of Cul (21 mg, 0.1 mmol) and Pd2(PPh)2 (40 mg, 57 μιτιοΐ) in THF (10 mL) was added TEA (0.4 mL, 2.8 mmol), 3-fluorobenzoyl chloride (0.34 mL, 2.8 mmol) and benzyl (2-methylbut-3-yn-2-yl)carbamate (1-36) (0.61 g, 2.8 mmol). The reaction was stirred at room temperature for 12 hours and partitioned with Et20/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude benzyl (5-(3-fluorophenyl)-2-methyl-5-oxopent-3-yn-2-yl)carbamate (1-37) was used without further purification. 1H NMR (400MHz, DMSO-6) δ 7.98 - 7.94 (m, 2 H), 7.84 (d, J = 9.6 Hz, 1 H), 7.64 - 7.60 (m, 2 H), 7.40 - 7.30 (m, 5 H), 5.10 (s, 2 H), 1.62 (s, 6 H). MS m/z 340.1 (M+l)+.
[000258] To a solution of benzyl (5-(3-fluorophenyl)-2-methyl-5-oxopent-3-yn-2-yl)carbamate (1-37) (0.28 g, 0.82 mmol) in MeOH (20 mL) was added 10% PdOH-C (30 mg). The reaction was degassed under vacuum and backfilled with hydrogen. Stirring was continued under a hydrogen atmosphere for 2 hours. Subsequently, the reaction was filtered through a pad of Celite and reduced to dryness to yield 5-(3-fluorophenyl)-2,2-dimethylpyrrolidine (1-38) as a amber oil. MS mJz 194.2 (M+l)+.
Synthesis of 5-(3-fluorophenyl)-3-methyl-4,5-dihydro-lH-pyrazole (1-40)
Figure imgf000079_0001
1-39 1-40
[000259] 3-Fluorobenzaldehyde (0.50 mL, 5.0 mmol) and 1-
(triphenylphosphoranylidene)propan-2-one (1.72 g, 5.4 mmol) were combined and heated under neat conditions at 100 °C for 1 hour. Upon cooling the reaction was purified by column chromatography on silica with hexanes/EtOAc gradient as eluant to yield (E)-4-(3- fluorophenyl)but-3-en-2-one (1-39) as a clear oil. 1H NMR (400MHz, DMSO- d6) δ 7.44 (d, J = 16.4 Hz, 1 H), 7.32 - 7.33 (m, 1 H), 7.29 (bd, J = 8.0 Hz, 2 H), 7.22 (td, J = 10.0, 1.6 Hz, 1 H), 7.08 (tdd, J = 8.4, 2.8, 1.2 Hz, 1 H), 6.68 (d, J = 16.4 Hz, 1 H), 2.37 (s, 3 H).
[000260] To a solution of 5-(3-fluorophenyl)-3-methyl-4,5-dihydro-lH-pyrazole (1-39) (0.26 g, 1.6 mmol) in EtOH (6 mL) was slowly added hydrazine (64 μί, 2.0 mmol). The reaction was heated at reflux for 3 hours. Upon cooling the reaction was reduced to dryness to yield 5-(3- fluorophenyl)-3-methyl-4,5-dihydro-lH-pyrazole (1-40) which was used without further purification. MS m/z 179.1 (M+l)+.
-(3-fluorophenyl)-3-azabicyclo[ 3.1.0] hexane
Figure imgf000079_0002
[000261] To a solution of (3-fluorophenyl)magnesium bromide (16 mL of a 1 M solution in THF) at -78 °C was added 3-azabicyclo[3.1.0]hexane-2,4-dione (0.9 g, 8.1 mmol). The reaction mixture was warmed to room temperature while stirring overnight. Sodium cyanoborohydride (0.6 g, 9.7 mmol) was then added followed by a slow addition of concentrated HCl (2.6 mL, 30.0 mmol). After the solution was stirred for 1 hour at room temperature aqueous NaOH (50 mL, 0.12 mol) was added. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column
chromatography on silica gel with EtOAc/hexanes gradient to yield 4-(3-fluorophenyl)-3- azabicyclo[3.1.0]hexan-2-one (1-41). MS m/z 192.1 (M+l)+.
[000262] To a solution of 4-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-2-one (1-41) (0.8 g, 4.2 mmol) in THF (10 mL) at -78 °C was added a lithium aluminum hydride (20 mL of a 1 M solution in THF). After stirring for 2 hours the solution was slowly warmed to room temperature and stirred for an additional 2 hours. Methanol (20 mL) was added drop wise to the resulting mixture, stirred for 20 minutes, and reduced in volume on roto-vap to remove organic solvents. The resulting residue was reconstituted in EtOAc and washed with brine (3 x 50mL). The organic layer was dried over anhydrous sodium sulfate, filtered and reduced to dryness to yield 2-(3- fluorophenyl)-3-azabicyclo[3.1.0]hexane (1-42). MS m/z 178.1 (M+l)+.
Synthesis of(2R,4R)-l-(3-(ethoxycarbonyl)pyrazolo[l,5-a]pyrimidin-5-yl)-4-hydroxypyrrolidine-2- carboxylic acid
Figure imgf000080_0001
1-10 1-53
[000263] A solution of (2R,4R)-4-hydroxypyrrolidine-2-carboxylic acid (0.15 g, 1.1 mmol), ethyl 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-10) (0.25 g, 1.1 mmol) and potassium fluoride (0.19 g, 3.3 mmol) in DMSO (3 mL) was heated at 80 °C for 3h. Upon cooling the reaction was partitioned with water and EtOAc. The organic layer was washed with brine, dried with magnesium sulfate, filtered and reduced to dryness to yield (2R,4R)-l-(3- (ethoxycarbonyl)pyrazolo[ 1 ,5-a]pyrimidin-5-yl)-4-hydroxypyrrolidine-2-carboxylic acid (1-53) as a clear amorphous solid. The crude material was used without further purification. MS m/z 321.1 (M+l)+.
Synthesis of 5-chloropyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide (1-56)
Figure imgf000080_0002
1-9 1-55
1-56
[000264] A solution of methyl 5-oxo-4,5-dihydropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-9) (0.75g, 3.6 mmol), lithium hydroxide (0.467 g, 18 mmol) and water (20 mL) were stirred at ambient temperature for 3 hours. HPLC indicated complete reaction. Acetic acid (5 mL) was added and the precipitated solid was collected by vacuum filtration to give 5-oxo-4,5- dihydropyrazolo[l,5-fl]pyrimidine-3-carboxylic acid (1-55). 1H NMR (400MHz,6-DMSO) δ 12.5 (bs, 1 H), 11.3 (bs, 1 H), 8.57 (d, 1 H), 8.08 (d, 1 H), 6.15, (d, 1 H). MS m/z 180.1 (M+l)+.
[000265] A mixture of 5-hydroxypyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-55) (1.47 g, 8.2 mmol) and phosphorous oxychloride (7.6 mL, 82 mmol) was heated at a gentle reflux for 2 hours. After cooling to ambient temperature, the resulting mixture was diluted with 1,2-dichloroethane (20 mL) and concentrated under reduced pressure to dryness, twice. The resulting material was diluted with THF (10 mL, anhydrous) and added slowly to a well-stirred mixture of 28% ammonium hydroxide (4 mL), ammonium chloride (0.44 g, 8.2 mmol) and THF (15 mL) at 5 °C. The reaction mixture was stirred at ambient temperature for 30 minutes and then concentrated under reduced pressure. The resulting material was purified by column chromatography on silica gel with ethyl acetate/hexanes gradient as eluent to afford 5-chloropyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-56) as a colorless crystalline solid.1H NMR (400MHz,6-DMSO) δ 9.33 (d, J = 7.2 Hz, 1H), 8.60 (s, 1H), 7.54 (bs, 1H), 7.36 (d, J = 7.2 Hz, 1H), 7.19 (bs, 1H). MS m/z 197.1 (M+l)+.
Synthesis of(R)-3-(pyrrolidin-2-yl)benzonitrile hydrochloride (1-58)
Figure imgf000081_0001
Pd(OAc)2,
t-Bu3P - HBF4 1-57 1-58
[000266] Tert-butyl pyrrolidine- 1-carboxylate (0.5 g, 2.9 mmol), (-) sparteine (0.82 g, 3.5 mmol), and MTBE were added to a flask under nitrogen and cooled to - 78 °C. S-BuLi (1.4 M in cyclohexane, 2.5 mL, 2.3 mmol) was added dropwise and the mixture maintained at - 78 °C for 3 hours. ZnCl2 (1.0 M in Et20, 2.3 mL , 2.3 mmol) was added dropwise and stirred at - 78 °C for 30 minutes, then warmed to ambient temperature and stirred for 1 hour. 3-Bromobenzonitrile (0.58 g, 3.2 mmol), Pd(OAc)2 (0.034 g, 0.15 mmol), and tri-tert-butylphosphine tetrafluoroborate (0.049 g , 0.17 mmol) were added to the reaction and stirred overnight at ambient temperature. Concentrated NH4OH (0.4 mL) was added to the reaction and stirred for 30 minutes. The reaction was filtered through celite and the organics were washed with 1 N HCI and water. The organics were dried over Na2S04, filtered and purified by silica gel chromatography with an EtOAc/hexane gradient to afford (R)-tert-butyl 2-(3-cyanophenyl)pyrrolidine-l-carboxylate (1-57). 1H NMR (400MHz, CDCI3) δ 7.52 (m, 1 H), 7.47 (m, 1 H), 7.42 (m, 2 H), 4.86, (d, J = 69 Hz, 1 H) 3.69 - 3.51 (m, 2 H), 2.35 (m, 1 H), 1.90 (m, 2 H), 1.79 (m, 1 H), 1.46 (bs, 4 H), 1.18 (bs, 5 H). MS m/z 217.1 (M- 55)+.
[000267] (R)-tert-butyl 2-(3-cyanophenyl)pyrrolidine-l-carboxylate (1-57), (0.20 g, 0.73 mmol), DCM (1 mL), and HC1 (2 M in Et20, 4.5 mL, 2.9 mmol) were combined to give a clear solution which was stirred overnight at ambient temperature. The reaction was concentrated to give (R)-3- (pyrrolidin-2-yl)benzonitrile hydrochloride (1-58). MS m/z 173.1 (M+l)+.
Synthesis of 3-fluoro-5-(pyrrolidin-2-yl)benzonitrile (1-60)
Figure imgf000082_0001
1-59 1-60
[000268] To a solution of 3-bromo-5-fluorobenzonitrile (0.40g, 2.0 mmol) in THF (5 mL) at -78 °C was added isopropylmagnesium chloride (1.05 mL of a 2.0 M solution in THF, 2.1 mmol). The reaction was stirred at -20 °C for 30 minutes then cooled to -78 °C. The reaction mixture was added to a -78 °C solution of tert-butyl 2-oxopyrrolidine-l-carboxylate (0.33 mL, 2.0 mmol) in THF (5 mL). The reaction was stirred to ambient temperature over lhour, quenched with saturated aqueous ammonium chloride and diluted with ethyl acetate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford tert-butyl 2-(3-cyano-5- fluorophenyl)-2-hydroxypyrrolidine-l-carboxylate (1-59) as a clear oil that crystallizes on standing. MS m/z 251.1 (M-55)+.
[000269] To a solution of tert-butyl 2-(3-cyano-5-fluorophenyl)-2-hydroxypyrrolidine-l- carboxylate (1-59) (0.22g,0.7 mmol) in DCM (10 mL) was added TFA (5 mL). The reaction was stirred at ambient temperature until complete by LCMS then reduced to dryness. The crude mixture was dissolved in EtOH:AcOH (10: 1, 4 mL) and was added NaCNBH3 (O. l lg, 1.4 mmol). The reaction was stirred for lhour then partitioned with ethyl acetate and saturated aqueous sodium bicarbonate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford 3-fluoro-5-(pyrrolidin-2-yl)benzonitrile (1-60) as a clear oil. MS m/z 191.1 (M+l)+. Synthesis of2-(l-( 3-carbamoylpyrazolo[ 1,5 -a ]pyrimidin-5-yl )pyrrolidin-2 -yl ) -4-fluorobenzoic acid (1-64)
Figure imgf000083_0001
1-63 1-56 1-64
[000270] n-BuLi (2.5 M in hexane, 1.8 mL, 4.6 mmol) was added dropwise to a solution of 2- bromo-4-fluorobenzoic acid (0.50 g, 2.3 mmol) in THF (7 mL) under N2 at - 78 °C and stirred for 30 minutes. To the resulting mixture was added dropwise a solution of tert-butyl 2- oxopyrrolidine-l-carboxylate (0.45 g, 2.4 mmol) in THF (2 mL) at - 78 °C and stirred for 30 minutes. The reaction was quenched with saturated aqueous NH4C1 (2 mL) and warmed to ambient temperature. The mixture was concentrated, taken up in EtOAc, dried over Na2S04 and filtered. The crude product was purified by silica gel chromatography with a
EtOAc/ AcOH/hexane gradient to afford 2-(l-(tert-butoxycarbonyl)-2-hydroxypyrrolidin-2-yl)-4- fluorobenzoic acid (1-62). 1H NMR (400MHz, CDC13) δ 7.93 (m, 1 H), 7.25 (m, 1 H), 7.18 (m, 1 H), 4.77 (m, 1 H), 3.25 (m, 2 H), 2.33 (m, 2 H), 1.74 (m, 2H), 1.44 (bs, 9 H). MS m/z 348.1 (M+23)+.
[000271] To a solution of 2-(l-(tert-butoxycarbonyl)-2-hydroxypyrrolidin-2-yl)-4-fluorobenzoic acid (1-62) (0.105 g, 0.32 mmol) in DCM (1 mL) was added TFA (1 mL) and the reaction was stirred at ambient temperature for 1 hour. The reaction was concentrated and taken up in EtOH (1 mL) and was followed by 3 sequential iterations of addition of sodium cyanoborohydride (0.030 g, 0.48 mmol) and stirring for 1 hour. The reaction was quenched with 2M Na2C03 (1.5 mL) and concentrated to afford 4-fluoro-2-(pyrrolidin-2-yl)benzoic acid (1-63). The crude product was used without purification. MS m/z 210.1 (M+l)+.
[000272] 4-Fluoro-2-(pyrrolidin-2-yl)benzoic acid (1-63) (0.067 g, 0.32 mmol), 5- chloropyrazolo[l,5-a]pyrimidine-3-carboxamide (1-56) (0.078 g, 0.32 mmol), 2M Na2C03(l mL) and ACN (2 mL) were combined and heated at 85 °C overnight. The resulting solution was adjusted to pH 5 with 1 N HC1 and concentrated. The crude solid was trituated six times with a 20% MeOH/DCM solution and the combined trituants dried over Na2S04, filtered, and concentrated to afford 2-(l-(3-carbamoylpyrazolo[l,5-a]pyrimidin-5-yl)pyrrolidin-2-yl)-4- fluorobenzoic acid (1-64) which was carried on without further purification. MS m/z 370.1 (M+l)+.
Synthesis of2-(5-fluoro-2-(trifluoromethyl)phenyl)pyrrolidine (1-66)
Figure imgf000084_0001
1-65 1-66
[000273] To a solution of 2-bromo-4-fluoro-l-(trifluoromethyl)benzene (0.28 mL, 2.0 mmol) in THF (5 mL) at -78 °C was added wBuLi (0.72 mL of a 2.8 M solution in Hexane, 2.0 mmol). The reaction was stirred at -78 °C for 15 minutes then added to a solution of tert-butyl 2- oxopyrrolidine-l-carboxylate (0.33 mL, 2.0 mmol) in THF (5 mL) at -78 °C. The reaction was stirred to ambient temperature over lhour, quenched with saturated aqueous ammonium chloride and diluted with ethyl acetate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford tert-butyl 2-(5-fluoro-2-(trifluoromethyl)phenyl)-2- hydroxypyrrolidine-l-carboxylate (1-65) as a clear oil. MS m/z 294.1 (M-55)+.
[000274] To a solution of tert-butyl 2-(5-fluoro-2-(trifluoromethyl)phenyl)-2- hydroxypyrrolidine-l-carboxylate (1-65) (0.26g,0.7 mmol) in DCM (10 mL) was added TFA (5 mL). The reaction was stirred at ambient temperature until complete by LCMS then reduced to dryness. The crude mixture was dissolved in EtOH:AcOH (10: 1, 4 mL) and was added NaCNB¾ (0.1 lg, 1.4 mmol). The reaction was stirred for lh then partitioned with ethyl acetate and saturated aqueous sodium bicarbonate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford 2-(5-fluoro-2-(trifluoromethyl)phenyl)pyrrolidine (1-66) as a clear oil. MS m/z 234.1 (M+l)+.
Synthesis of2-(2-(trifluoromethyl)phenyl)pyrroUdine (1-68)
Figure imgf000085_0001
1-67 1-68
[000275] To a solution of l-bromo-2-(trifluoromethyl)benzene (0.27 mL, 2.0 mmol) in THF (5 mL) at -78 °C was added wBuLi (0.72 mL of a 2.8 M solution in Hexane, 2.0 mmol). The reaction was stirred at -78 °C for 15 minutes then added to a solution of tert-butyl 2-oxopyrrolidine-l- carboxylate (0.33 mL, 2.0 mmol) in THF (5 mL) at -78 °C. The reaction was stirred to ambient temperature over lhour, quenched with saturated aqueous ammonium chloride and diluted with ethyl acetate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford tert-butyl 2-hydroxy-2- (2- (trifluoromethyl)phenyl)pyrrolidine-l -carboxylate (1-67) as a clear oil. MS m/z 276.1 (M-55)+.
[000276] To a solution of tert-butyl 2-hydroxy-2-(2-(trifluoromethyl)phenyl)pyrrolidine-l- carboxylate (1-67) (0.44g,1.6 mmol) in DCM (20 mL) was added TFA (10 mL). The reaction was stirred at ambient temperature until complete by LCMS then reduced to dryness. The crude mixture was dissolved in EtOH:AcOH (10: 1, 8 mL) and was added NaCNBH3 (0.17g, 2.7 mmol). The reaction was stirred for lhour then partitioned with ethyl acetate and saturated aqueous sodium bicarbonate. The organic layer was separated, dried over magnesium sulfate, filtered and reduced to dryness to afford 2-(2-(trifluoromethyl)phenyl)pyrrolidine (1-68) as a clear oil. MS m/z 216.1 (M+l)+.
Synthesis of3-fluoro-5-((2R,4S)-4-fluoropyrrolidin-2-yl)benzonitrile (1-74)
Figure imgf000086_0001
[000277] To a solution of 3-bromo-5-fluorobenzonitrile (5.2 g, 0.26 mmol) in THF (26 mL) at - 78 °C was slowly added isopropylmagnesium chloride (13.6 mL of a 2.0 M solution in diethyl ether, 0.27 mmol). The mixture was warmed and stirred at -30 °C for 1 hour then cooled to -78 °C. The resulting aryl Grignard was added to a solution of (R)-tert-butyl 4-(tert- butyldimethylsilyloxy)-2-oxopyrrolidine-l-carboxylate (1-13) (7.8 g, 25 mmol) in THF (25 mL) at -78 °C. The reaction mixture was stirred at 0 °C for 30 minutes. Methanol (20 mL) was added to the reaction mixture followed by NaBH4 (1.4 g, 37 mmol). The mixture was stirred at room temperature for 12 hours then poured into 10% aqueous NFLCl. The mixture was extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield tert-butyl ((2R)-2-((tert-butyldimethylsilyl)oxy)-4-(3-cyano-5-fluorophenyl)-4- hydroxybutyl)carbamate (1-69). 1H NMR (400MHz, CDC13) δ 7.47 (s, 1 H), 7.38 - 7.33 (m, 1 H), 7.25 - 7.20 (m, 1 H), 4.95 (d, J = 92 Hz, 1 H), 4.81 (bs, 1 H), 4.12 - 4.05 (m, 1 H), 3.76 (bs, 1 H), 3.45 - 3.35 (m, 1 H), 3.23 - 3.12 (m, 1 H), 1.87 - 1.71 (m, 2 H), 1.45 (s, 9 H), 0.91 (s, 9 H), 0.12 (s, 6 H). MS m/z 461.2 (M+23)+.
[000278] To a solution of tert-butyl ((2R)-2-((tert-butyldimethylsilyl)oxy)-4-(3-cyano-5- fluorophenyl)-4-hydroxybutyl)carbamate (1-69) (5.3 g, 12.1 mmol) in DCM (50 mL) at -60 °C was added TEA (5.1 mL, 36.3 mmol) and MsCl (0.98 mL, 12.2 mmol). The resulting mixture was stirred at -60 °C for 2 hours. The reaction was poured into water, diluted with DCM and washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield (3R)-4- ((tert-butoxycarbonyl)amino)-3-((tert-butyldimethylsilyl)oxy)-l-(3-cyano-5-fluorophenyl)butyl methanesulfonate (1-70). MS m/z 539.1 (M+23)+. Compound decomposes on standing. Used immediately in next reaction without delay.
[000279] To a solution of (3R)-4-((tert-butoxycarbonyl)amino)-3-((tert-butyldimethylsilyl)oxy)- l-(3-cyano-5-fluorophenyl)butyl methanesulfonate (1-70) (5.3 g, 10.3 mmol) in DMF (30 mL) at 0 °C was added NaH (0.43 mg of a 60% mineral oil dispersion, 10.8 mmol). The reaction was stirred at room temperature for 12 hours. The reaction was quenched with water and extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield (4R)-tert-butyl 4-((tert-butyldimethylsilyl)oxy)-2-(3-cyano-5- fluorophenyl)pyrrolidine-l-carboxylate (1-71). 1H NMR (400MHz, CDC13) δ 7.38 (s, 1 H), 7.33 - 7.20 (m, 1 H), 7.20 - 7.12 (m, 1 H), 4.90 (dd, J = 7.6, 56.2 Hz, 1 H), 4.45 - 4.39 (m, 1 H), 3.78 - 3.65 (m, 1 H), 3.52 (dd, J = 11.2, 36.8 Hz, 1 H), 2.56 - 2.41 ( m, 1 H), 1.89 (t, J = 13.2 Hz, 1 H), 1.47 (s, 4 H), 1.23 (s, 5 H), 0.75 (d, J = 1.6 Hz, 9 H), 0.03 (s, 3 H), -0.08 (d, J = 10.8 Hz, 3 H). MS m/z 443.2 (M+23)+.
[000280] To a solution of (4R)-tert-butyl 4-((tert-butyldimethylsilyl)oxy)-2-(3-cyano-5- fluorophenyl)pyrrolidine-l-carboxylate (1-71) (2.7 g, 6.4 mmol) in THF (25 mL) at room temperature was added TBAF (7.1 mL of a 1.0 M solution in THF, 7.0 mmol). The mixture was stirred at room temperature for 1 hour then concentrated. The mixture was purified by column chromatography on silica gel with EtOAc/hexanes gradient as eluant to yield the desired isomer (2R,4R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (I-72A) (1.0 g, 51% yield) and (2S,4R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (I-72B). 1H NMR (400MHz, CDC13) δ 7.41 (s, 1 H), 7.35 -7.26 (m, 1 H), 7.24-7.16 (m, 1 H), 4.89 (d, / = 48.8 Hz, 1 H), 4.56 - 4.50 (m, 1 H), 3.85 - 3.70 (m, 1 H), 3.66 - 3.55 (m, 1 H), 2.59 (bs, 1 H), 2.00 - 1.90 (m, 1 H), 1.45 (bs, 3 H), 1.22 (bs, 6 H). MS m/z 304.1 (M+23)+.
[000281] To a solution of (2R,4R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-hydroxypyrrolidine- 1-carboxylate (I-72A) (0.7 g, 2.3 mmol) in DCM (4 mL) in a plastic bottle at -78 °C was added DAST (0.6 mL, 4.6 mmol). The reaction was stirred at -78 °C for 2 hours and then loaded directly onto silica gel and purified by column chromatography with EtOAc/hexanes gradient as eluant to yield (2R,4S)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidine-l-carboxylate (1-73). 1H NMR (400MHz, CDC13) δ 7.35 (s, 1 H), 7.30 -7.16 (m, 2 H), 5.25 (d, / = 52.0 Hz, 1 H), 5.06 - 4.86 (m, 1 H), 4.20 - 3.98 (m, 1 H), 3.72 (dd, J = 11.6, 38.8 Hz, 1 H), 2.80 - 2.66 (m, 1 H), 2.06 - 1.78 (m, 1 H), 1.45 (bs, 3 H), 1.19 (bs, 6 H). MS m z 331.1 (M+23)+.
[000282] To a solution of (2R,4S)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidine-l- carboxylate (1-73) (0.6 g, 1.95 mmol) in DCM (2 mL) at room temperature was added TFA (2 mL). The mixture was stirred at room temperature for 2 hours. All the solvents were removed under reduced pressure. The crude was diluted with EtOAc, washed with aqueous NaHC03 and brine, dried over sodium sulfate, filtered and concentrated to yield 3-fluoro-5-((2R,4S)-4- fluoropyrrolidin-2-yl)benzonitrile (1-74). 1H NMR (400MHz, CDC13) δ 7.52 (s, 1 H), 7.43 - 7.37 (m, 1 H), 7.24 - 7.20 (m, 1 H), 5.28 (dt, J = 3.6, 53.6 Hz, 1 H), 4.57 (dd, J = 6.4, 9.6 Hz, 1 H), 3.44 - 3.18 (m, 2 H), 2.63 - 2.50 (m, 1 H), 1.72 (dddd, J = 4.4, 10.0, 14.4, 39.6 Hz, 1 H). MS m/z 209.1 (M+l)+.
Synthesis of (R)-3-(4,4-difluoropyrrolidin-2-yl)-5-fluorobenzonitrile (1-77)
Figure imgf000088_0001
[000283] To a solution of (2R,4R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-hydroxypyrrolidine- 1-carboxylate (I-72A) (0.52 g, 1.7 mmol) and trichloroisocyanuric acid (0.4 g, 1.7 mmol) in DCM (20 mL) at -10 °C was added 2,2,6,6-tetramethylpiperidine-l-oxyl (TEMPO) (27 mg, 0.17 mmol). The mixture was stirred at -10 °C for 15 minutes, then to room temperature over 1 hour and subsequently poured into cold aqueous NaHC03 containing ice while stirring. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and reduced to dryness to yield (R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-oxopyrrolidine-l-carboxylate (1-75). 1H NMR (400MHz, CDC13) δ 7.27 - 7.22 (m, 2 H), 7.12 (td, J = 2.0, 8.8 Hz, 1 H), 5.33 (bs, 1 H), 4.06 (d, J = 19.6 Hz, 1 H), 3.86 (d, J = 19.6 Hz, 1 H), 3.15 (dd, J = 10.0, 18.8 Hz, 1 H), 2.47 (dd, J = 3.2, 18.8 Hz, 1 H), 1.35 (bs, 9 H). MS m/z 249.1 (M-56)+.
[000284] To a solution of (R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4-oxopyrrolidine-l- carboxylate (1-75) (0.54 g, 1.8 mmol) in DCM (15 mL) in a plastic bottle at -78 °C was added DAST (1.9 mL, 14.4 mmol). The resulting orange homogeneous solution was stirred at -78 °C for 30 minutes then room temperature for 12 hours. The reaction was diluted with DCM and washed with water, brine, dried over sodium sulfate, filtered and reduced to dryness. The crude product was purified by flash column chromatography on silica with hexanes/EtOAc gradient as eluant to yield (R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidine-l-carboxylate (1-76) as a clear oil that crystallizes on standing. 1H NMR (400MHz, CDC13) δ 7.33 (s, 1 H), 7.30-7.26 (m, 1 H), 7.23 - 7.17 (m, 1 H), 5.15 - 4.90 (m, 1 H), 4.02 - 3.84 (m, 2 H), 2.94 - 2.78 (m, 1 H), 2.36 - 2.22 (m, 1 H), 1.15 - 1.10 (m, 9 H). MS m/z 249.1 (M-56)+.
[000285] To a solution of (R)-tert-butyl 2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidine-l- carboxylate (1-76) (0.45 g, 1.4 mmol) in DCM (3 mL) was added TFA (3 mL) and stirred at room temperature for 1 hour. All the solvents were removed under reduced pressure. The crude was diluted with EtOAc, washed with aqueous NaHC03 and brine, dried over sodium sulfate, filtered and concentrated to yield (R)-3-(4,4-difluoropyrrolidin-2-yl)-5-fluorobenzonitrile (1-77) as a light yellow oil. 1H NMR (400MHz, CDC13) δ 7.52 (s, 1 H), 7.42 (d, J = 9.2 Hz, 1 H), 7.29 - 7.26 (m, 1 H), 4.46 (t, J = 8.8 Hz, 1 H), 3.52 - 3.30 (m, 2 H), 2.74 - 2.60 (m, 1 H), 2.20 - 2.04 (m, 1 H), 1.90 (bs, 1 H). MS m/z 227.1 (M+l)+.
Synthesis of(3S,5R)-5-(3-fluorophenyl)pyrrolidine-3-carbonitrile 2,2,2-trifluoroacetate (1-81 )
Figure imgf000089_0001
[000286] To a solution of (2R,4R)-tert-butyl 2-(3-fluorophenyl)-4-hydroxypyrrolidine-l- carboxylate (I-16A) (253 mg, 0.9 mmol) in DCM (anhyd, 6 mL) at 0 °C was added triethylamine (0.25 mL, 2.0 eq) followed by dropwise addition of methanesulfonyl chloride (95 μί, 1.4 eq). After stirring 2 hours at ambient temperature the reaction mixture was concentrated under reduced pressure and the resulting solid was extracted with Et20 (2 x 20 mL). The combined extracts were washed sequentially with 1% citric acid and brine, dried over magnesium sulfate, filtered and concentrated under reduced pressure to afford (2R,4R)-tert-butyl 2-(3-fluorophenyl)-4- ((methylsulfonyl)oxy)pyrrolidine-l-carboxylate (1-79) as a colorless oil. MS m/z 382.1 (M+23)+.
[000287] A mixture of (2R,4R)-tert-butyl 2-(3-fluorophenyl)-4-
((methylsulfonyl)oxy)pyrrolidine-l-carboxylate (1-79) (165 mg, 0.46 mmol), potassium cyanide (36 mg, 0.55 mmol) and DMSO (anhyd, 2 mL) was heated at 90 °C for 3 hours. After cooling, the reaction mixture was partitioned in Et20 and water. The organic layer was washed with brine, dried over magnesium sulfate, filtered and concentrated to dryness. The residue was purified by silica chromatography, eluting with EtOAc/Hex (0-50% gradient) to yield (2R,4S)-tert-butyl 4- cyano-2-(3-fluorophenyl)-pyrrolidine-l-carboxylate (1-80) as a colorless oil. 1H NMR (400 MHz, CDC13) δ 7.35 - 7.30 (m, 1H), 7.02 - 6.93 (m, 2H), 6.87 (d, J = 9.6 Hz, 1H), 5.16rl (bs, 0.4H), 5.00r2 (bs, 0.6H), 4.00 - 3.73 (m, 2H), 3.16 (bs, 1H), 2.67 (bs, 1H), 2.25 - 2.21 (m, 1H), 1.49 (bs, 3H), 1.23 (bs, 6H). MS m/z 313.1 (M+23)+.
[000288] To a solution of (2R,4S)-tert-butyl 4-cyano-2-(3-fluorophenyl)-pyrrolidine-l- carboxylate (1-80) (72 mg, 0.25 mmol) in DCM (0.5 mL) was added TFA (0.5 mL) with stirring. After 1 hour the volatile organics were removed under reduced pressure. The residue was triturated with Et20 and the resulting solids collected and dried under vacuum to give (3S,5R)-5- (3-fluorophenyl)pyrrolidine-3-carbonitrile 2,2,2-trifluoroacetate (1-81), which was used without purification. MS m/z 191.1 (M+l)+.
Synthesis of4-fluoro-3-(pyrrolidin-2-yl)benzonitrile (1-83)
Figure imgf000090_0001
1-82 1-83
[000289] To a solution of 3-bromo-4-fluorobenzonitrile (0.40 g, 2.0 mmol) in THF (anhyd, 3 mL) at -78 °C was added dropwise isopropylmagnesium chloride (1.05 mL of a 2.0M solution in THF, 2.0 mmol). The resulting solution was warmed to -30 °C with stirring. After 1 hour at -30 °C, the reaction mixture was added slowly to a solution of tert-butyl 2-oxopyrrolidine-l- carboxylate (0.31 μί, 1.8 mmol) in THF (1 mL) at -78. The resulting mixture was allowed to warm slowly to 0 °C. After 1 hour at 0 °C, the reaction was quenched by the addition of saturated aqueous ammonium chloride and stirred 20 minutes. The mixture was extracted with EtOAc and the combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to dryness. The residue was purified by silica chromatography with EtO Ac/Hex (10-30% gradient) as eluent to yield tert-butyl 2-(5-cyano-2-fluorophenyl)-2-hydroxypyrrolidine-l- carboxylate (1-82) as a clear oil that partially solidified on standing. MS m/z 251.1 (M-55)+.
[000290] To a solution of tert-butyl 2-(5-cyano-2-fluorophenyl)-2-hydroxypyrrolidine-l- carboxylate (1-82) (0.25 g, 0.82 mmol) in MeOH (4 mL) was added HC1 (2.05 mL of a 2N solution in Et20, 4.1 mmol). After stirring overnight, the reaction mixture was concentrated under a stream of nitrogen to afford white solids, which was diluted with MeOH (5 mL) and charged with sodium borohydride (51 mg, 1.6 eq) cautiously. After stirring at ambient temperature for 2 hours, the mixture was neutralized to pH 8-9 with saturated aqueous NaHC03, concentrated to minimum volume under reduced pressure and extracted with EtOAc (2x 25 mL). The combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to yield 4- fluoro-3-(pyrrolidin-2-yl)benzonitrile (1-83) as a colorless film, which was used without purification in the next step. MS m/z 191.1 (M+l)+.
Synthesis of Synthesis of(R)-2-(4,4-difluoropyrrolidin-2-yl)-4-fluoro-N-isopropylbenzamide (1-91 )
Figure imgf000091_0001
1. HCI, MeOH
2. A¾0, TEA, DCM
Figure imgf000091_0002
[000291] To a solution of 2-bromo-5-fluorobenzaldehyde (2.44 g, 12.0 mmol) and (S)-(-)-2- methyl-2-propane-sulfinamide (1.45 g, 12.0 mmol) in THF (30 mL, anhyd) was added titanium tetraethoxide (2.8 mL, 13.2 mmol, tech) at ambient temperature. After stirring 16 hours, the reaction mixture was charged with brine (25 mL) and EtOAc (40 mL). After stirring 10 minutes, the mixture was filtered through a pad of Celite™. Combined filtrates were washed with water and brine, dried over sodium sulfate and concentrated. The residue was purified by silica chromatography with EtOAc/Hex (0 - 25%) as eluent to give (lS',E)-N-(2-bromo-5- fluorobenzylidene)-2-methylpropane-2-sulfinamide (1-84) as a white solid. 1H NMR (400 MHz, CDC13) δ 8.94 (d, J = 2.2 Hz, 1 H), 7.77 (dd, J = 3.1, 9.1 Hz, 1 H), 7.64 (dd, J = 5.0, 8.8 Hz, 1 H), 7.12 (ddd, J = 3.1, 7.5, 8.8 Hz, 1 H), 1.30 (s, 9 H).
Method A
[000292] To a solution of (lS',E)-N-(2-bromo-5-fluorobenzylidene)-2-methylpropane-2- sulfinamide (1-81) (612 mg, 2.0 mmol) in DCM (6 mL) at -78 °C was added dropwise a solution of allylmagnesium chloride (2.0M in THF, 1.5 mL, 3.0 mmol). After stirring 5 hours at -78 °C, the reaction mixture was quenched by addition of saturated aqueous NH4CI (1 mL) and then allowed to warm to ambient temperature. The mixture was extracted with EtOAc (2x 25 mL) and the combined extracts were washed with brine, dried over sodium sulfate and concentrated. The residue was purified by silica chromatography with EtOAc/Hex (25-50 %) as eluent to afford a mixture of (S)-N-((R)-l-(2-bromo-5-fluorophenyl)but-3-en-l-yl)-2-methylpropane-2-sulfinamide (I-85A) and (S)-N-((S)-l-(2-bromo-5-fluorophenyl)but-3-en-l-yl)-2-methylpropane-2-sulfinamide (I-85B) as a clear waxy solid. The latter has been assigned tentatively as the major product of the mixture after analysis of NMR and chiral HPLC data.19F NMR (376 MHz, CDC13) δ -113.32, - 113.87. MS m/z 348.0, 350.0 (M+l)+.
Method B
[000293] To a stirred suspension of (S,E)-N-(2-bromo-5-fluorobenzylidene)-2-methylpropane-2- sulfinamide (1-81) (153 mg, 0.50 mmol) and indium powder (230 mg, 2.0 mmol) in saturated NaBr (10 mL, aq) was added allyl bromide (169 μί, 2.0 mmol). After stirring 36 hours, the reaction mixture was charged with saturates aqueous NaHC03 and stirred 30 minutes. The mixture was extracted with EtOAc and combined extracts were washed with brine, dried over sodium sulfate and concentrated. The residue was purified by silica chromatography with
EtOAc/Hex (5-35 %) as eluent to yield the (S)-N-((R)- l-(2-bromo-5-fluorophenyl)but-3-en- l-yl)- 2-methylpropane-2-sulfinamide (I-85A) as a clear oil. 1H NMR (400 MHz, CDC13) δ 7.52 (dd, J = 5.3, 8.8 Hz, 1 H), 7.14 (dd, J = 3.1, 9.8 Hz, 1 H), 6.88 (ddd, J = 3.1, 7.7, 8.7 Hz, 1 H), 5.77 (dddd, J = 6.1, 8.2, 10.9, 16.6 Hz, 1 H), 5.28 - 5.16 (m, 2 H), 4.98 - 4.89 (m, 1 H), 3.74 (s, 1 H), 2.76 - 2.63 (m, 1 H), 2.39 (dt, J = 8.3, 14.2 Hz, 1 H), 1.24 (s, 9 H).19F NMR (376 MHz, CDC13) δ - 113.87. MS m/z 348.0, 350.0 (M+l)+.
[000294] To a solution of I-85A (1.95 g, 5.6 mmol) in MeOH (15 mL) at 0 °C was added 4N solution of HC1 in MeOH (7.5 mL, 30 mmol). After stirring 45 minutes at ambient temperature, the reaction mixture was chilled to 0 °C and slowly made basic with 15 % NaOH. The resulting mixture was concentrated to a minimum volume on a rotary evaporator and then partitioned in EtOAc and saturated aqueous NaHC03. The combined EtOAc extracts were washed with brine, dried over sodium sulfate and concentrated to yield the corresponding homoallylamine (1.4 g). MS m/z 244.0, 246.0 (M+l)+. The amine was dissolved in DCM (20 mL), chilled to 0 °C and charged with triethylamine (0.98 mL, 7.0 mmol) and then acetic anhydride (0.57 mL, 6.05 mmol). After stirring 2 hours, the reaction mixture was washed successively with saturated aqueous NaHC03, water, IN HC1 and brine, then dried over sodium sulfate and concentrated to dryness to afford (R)-N-(l-(2-bromo-5-fluorophenyl)but-3-en-l-yl)acetamide (1-86) as a white solid. MS m/z 286.0, 288.0 (M+l)+.
[000295] To a solution of (R)-N-(l-(2-bromo-5-fluorophenyl)but-3-en-l-yl)acetamide (1-86) (1.50 g, 5.24 mmol) in THF (12 mL) was added water (3 mL) followed by iodine (4.0 g, 15.7 mmol). After stirring 6 hours, the reaction mixture was poured into a mixture of saturated aqueous NaHC03 (30 mL) and saturated aqueous Na2S203 (25 mL), extracted with EtOAc (2x 40 mL), washed successively with saturated aqueous Na2S203, water and brine, dried over sodium sulfate and concentrated to dryness to give (5R)-5-(2-bromo-5-fluorophenyl)pyrrolidin-3-yl acetate (1-87) as a pale amber oil, which was used without purification in the next step. MS m/z 302.0, 304.0 (M+l)+.
[000296] To a mixture of (5R)-5-(2-bromo-5-fluorophenyl)pyrrolidin-3-yl acetate (1-87) (1.59 g, 5.24 mmol), dioxane (15 mL) and water (15 mL) was added a solution of di-tert-butyl dicarbonate (1M in THF, 6.6 mL, 6.6 mmol). Added IN NaOH until pH 9 had been achieved. After 3 hours, the reaction mixture was partitioned into EtO Ac/water and the combined extracts were washed with brine, dried over sodium sulfate and concentrated to dryness to afford the corresponding carbamate. MS m/z 424.0, 426.0 (M+23)+.
[000297] The above carbamate was dissolved in MeOH (15 mL) and chilled to 0 °C, and then charged dropwise with IN NaOH (3.93 mL, 0.75 eq). After stirring 2 hours at ambient temperature, the reaction had not progressed to completion, as determined by LCMS analysis of an aliquot. Added another portion of IN NaOH (0.75 eq). After hydrolysis was complete (approx. 1 hour), the reaction mixture was partitioned into EtO Ac/brine. The combined EtOAc extracts were washed with brine, dried over sodium sulfate, and concentrated. The residue was purified by silica chromatography with EtOAc/Hex (25-50 %) as eluent, to yield (2R)-tert-butyl 2-(2-bromo-5- fluorophenyl)-4-hydroxypyrrolidine-l-carboxylate (1-88) as a white foam. MS m/z 382.0, 384.0 (M+23)+.
[000298] To a solution of (2R)-tert-butyl 2-(2-bromo-5-fluorophenyl)-4-hydroxypyrrolidine-l- carboxylate (1-88) (1.38 g, 3.83 mmol) in DCM (30 mL) at -10 °C was added trichloroisocyanuric acid (0.89 g, 3.8 mmol) followed by 2,2,6,6-tetramethylpiperidino-l-oxy (TEMPO) (60 mg, 0.38 mmol). After stirring 25 minutes at ambient temperature, the reaction mixture was poured into saturated aqueous NaHC03 and ice (2 g), then extracted with DCM. The combined extracts were washed with water (2x) and brine (2x), dried over sodium sulfate and concentrated to dryness to give the corresponding ketone as a pale yellow oil, which was used without purification in the next step. MS m/z 380.0, 382.0 (M+23)+.
[000299] The above ketone was dissolved in DCM (8 mL), at -78 °C and then charged with diethylamino sulfur trifluoride (DAST) (1.0 mL, 2 eq). After stirring 12 hours at ambient temperature, the reaction mixture was added to cold water and extracted with DCM. The combined extracts were washed with water and brine, dried over sodium sulfate and concentrated to dryness to afford (R)-tert-butyl 2-(2-bromo-5-fluorophenyl)-4,4-difluoro-pyrrolidine-l- carboxylate (1-89) as a pale yellow, crystalline solid. 1H NMR (400 MHz, CDC13) δ 7.52 (dd, J = 5.2, 8.7 Hz, 1 H), 7.06 - 6.85 (m, 2 H), 5.38 - 5.26 (m, 1 H), 4.16 - 4.01 (m, 1 H), 3.96 (q, J = 12.4 Hz, 1 H), 3.06 - 2.88 (m, 1 H), 2.32 - 2.22 (m, 1 H), 1.49 (bs, 3 H), 1.23 (bs, 6 H). MS m/z 324.0, 326.0 (M-55)+.
[000300] To a nitrogen-flushed microwave vial was added (R)-tert-butyl 2-(2-bromo-5- fluorophenyl)-4,4-difluoro-pyrrolidine-l-carboxylate (1-89) (304 mg, 0.80 mmol), molybdenum hexacarbonyl (211 mg, 0.80 mmol), iraws-di^-acetatobis[2-di-o-tolylphosphino)benzyl]- dipalladium(II) (Palladacycle) (30 mg, 4 mol ), THF (1.6 mL), l,8-diazabicyclo-[5.4.0]undec-7- ene (DBU) (239
Figure imgf000094_0001
1.6 mmol) and isopropylamine (171 2.0 mmol). The vial was capped and then heated at 150 °C for 30 minutes in a Biotage microwave reactor. The cooled reaction mixture was diluted with EtOAc and filtered through Celite. The combined filtrates were washed successively with saturated aqueous NH4C1, water, 2% citric acid and brine, then dried over sodium sulfate and concentrated. The residue was purified by silica chromatography with
EtOAc/Hex (5-30%) as eluent, to yield (R)-tert-butyl 4,4-difluoro-2-(5-fluoro-2- (isopropylcarbamoyl)phenyl)pyrrolidine-l-carboxylate (1-90) as an off-white foam. 1H NMR (400 MHz, CDC13) 5 7.44 (bs, 1 H), 7.11 (bs, 1 H), 7.00 (td, J = 2.5, 8.2 Hz, 1 H), 5.31 (t, J = 7.7 Hz, 1 H), 4.23 (bs, 1 H), 4.09 - 3.87 (m, 2 H), 2.91 (bs, 1 H), 2.41 (bs, 1 H), 1.45 (bs, 6 H), 1.26 (d, J = 6.5 Hz, 6 H), 1.19 (bs, 3 H). MS m/z 331.1 (M-55)+. [000301] To a solution of (R)-tert-butyl 4,4-difluoro-2-(5-fluoro-2-
(isopropylcarbamoyl)phenyl)pyrrolidine-l-carboxylate (1-90) (203 mg, 0.525 mmol) in DCM (0.4 mL) was added TFA (1 mL). After stirring for 2 hours, the mixture was concentrated to dryness, diluted with DCM, washed with saturated aqueous NaHC03 and brine, dried over sodium sulfate and concentrated to dryness to give (R)-2-(4,4-difluoropyrrolidin-2-yl)-4-fluoro-N- isopropylbenzamide (1-91) as a pale brown solid. MS m/z 287.1 (M+H)+. 1-91 was also isolated as a racemic mixture using un-resolved I-2A and I-2B from Method A in the above sequence.
Synthesis of Exemplary Compounds
Synthesis of ethyl 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[ l,5-a]pyrimidine-
3-carboxylate (X-14)
Figure imgf000095_0001
[000302] To a solution of ethyl 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-10) (0.49 g, 2.1 mmol) and (2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidine (1-18) (0.40 g, 2.1 mmol) in DMSO (8 mL) was added potassium fluoride (0.38 g, 6.6 mmol). The mixture was heated to 85 °C for 8 hours then cooled to room temperature and partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield ethyl 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylate (X-14). MS m/z 373.1 (M+l)+.
Synthesis of ethyl 5-(2-(3-fluorophenyl)-5-oxopyrrolidin-l-yl)pyrazolofl,5-a]pyrimidine-3- rboxylate (X-19)
Figure imgf000095_0002
1-35 1-10 X-19
[000303] To a degassed solution of 5-(3-fluorophenyl)pyrrolidin-2-one (1-35) (44 mg, 0.2 mmol), Cul (3 mg, 16 μιηοΐ) and potassium carbonate (61 mg, 0.4 mmol) in 1,4-dioxane (0.5 mL) was added ethyl 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-10) (50 mg, 0.2 mmol) and N,N'-dimethylcyclohexane-l,2-diamine (4 μί, 22 μιηοΐ). The reaction was heated at 110 °C for 12 hours and partitioned with DCM/H20 upon cooling. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by preparative LCMS to yield 5-(2-(3-fluorophenyl)-5-oxopyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylate (X-19). 1H NMR (400MHz, DMSO-6) δ 9.11 (d, J = 8.0 Hz, 1 H), 8.44 (s, 1 H), 8.18 (d, J = 7.6 Hz, 1 H), 7.35 - 7.19 (m, 3 H), 7.03 (bt, J = 8.8 Hz, 1 H), 5.84 (d, J = 6.4 Hz, 1 H), 4.23 (q, J = 6.0 Hz, 2 H), 2.26 - 2.88 (m, 1 H), 2.71 - 2.64 (m, 1 H), 2.00 - 1.95 (m, 1 H), 1.26 (t, J = 6.8 Hz, 3 H). MS m/z 369.1 (M+l)+.
Synthesis o R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazoloil,5-a]pyrimidine-3-carboxamide (X-
Figure imgf000096_0001
X-16 1-44 X-25
[000304] To a solution of (R)-ethyl 5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylate (X-16) (2.6 g, 7.3 mmol) in THF/MeOH/H20 (3/2/1, 200 mL) was added 3 M LiOH (12 mL). The reaction was stirred at 50 °C for 8 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HC1 and the aqueous layer was decanted away from the resulting amorphous solid. The solid was washed with water and dried under vacuum to yield (R)-5-(2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-44). MS m/z 327.1 (M+l)+.
Method A:
[000305] To a solution of (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-44) (0.50 g, 1.5 mmol) in NMP (5 mL) was added DIEA (0.8 mL, 4.6 mmol) and HATU (0.58 g, 1.5 mmol). The reaction was stirred at room temperature for 30 minutes then a mixture of NH4C1 (0.25 g, 4.7 mmol) and NH4OH (0.1 mL) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X- 25) as a white solid. 1H NMR (400MHz, CD3OD, mixture of rotamers at rt) δ 8.44ri (d, J = 8.0 Hz, 0.6 H), 8.25r2 (d, J = 8.0 Hz, 0.4 H), 8.12r2 (s, 0.4 H), 8.10rl (s, 0.6 H), 7.29 - 7.21rl>r2 (m, 1 H), 7.0 - 6.57r 2 (m, 3 H), 6.58rl (d, J = 7.6 Hz, 0.6 H), 6.04r2 (d, J = 7.6 Hz, 0.4 H), 5.19rl (bd, J = 8.0 Hz, 0.6 H), 5.09r2 (bd, J = 8.0 Hz, 0.4 H), 4.00 - 3.88rl,r2 (m, 1 H), 3.78 - 3.64r 2 (m, 1 H), 2.51 - 2.38r 2 (m, 1 H), 2.11 - 1.84rl>r2 (m, 3 H). MS m/z 326.1 (M+l)+.
Method B:
[000306] To a solution of (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-44) (30 mg, 0.10 mmol) in DCM (2 mL) was added DMF (2 drops) and oxalyl chloride (12 μί, 0.15 mmol). The reaction was stirred at room temperature for 6 hours (until complete by LCMS) then reduced to dryness. The residue was redissolved in DCM (4 mL) and anhydrous NH3 was bubbled through the reaction while stirring at 0 °C for 15 minutes. The reaction was filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield (R)-5-(2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-25) as a white solid. 1H NMR (400MHz, CD3OD, mixture of rotamers at rt) δ 8.44ri (d, J = 8.0 Hz, 0.6 H), 8.25r2 (d, J = 8.0 Hz, 0.4 H), 8.12r2 (s, 0.4 H), 8.10rl (s, 0.6 H), 7.29 - 7.21ri,r2 (m, 1 H), 7.0 - 6.57r 2 (m, 3 H), 6.58rl (d, J = 1.6 Hz, 0.6 H), 6.04r2 (d, J = 1.6 Hz, 0.4 H), 5.19rl (bd, J = 8.0 Hz, 0.6 H), 5.09r2 (bd, J = 8.0 Hz, 0.4 H), 4.00 - 3.88r 2 (m, 1 H), 3.78 - 3.64rl>r2 (m, 1 H), 2.51 - 2.38r 2 (m, 1 H), 2.11 - 1.84rl>r2 (m, 3 H). MS m/z 326.1 (M+l)+.
Method C:
[000307] To a solution of (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-44) (65 mg, 0.2 mmol) in 1,2-dichloroethane (2 mL) was added POCl3 (55 μί, 0.6 mmol) and heated to 90 °C for 30 minutes. The reaction was reduced to dryness. The residue was redissolved in DCM (4 mL) and anhydrous NH3 was bubbled through the reaction while stirring at 0 °C for 15 minutes. The reaction was filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X- 25) as a white solid. 1H NMR (400MHz, CD3OD, mixture of rotamers at rt) δ 8.44rl (d, J = 8.0 Hz, 0.6 H), 8.25r2 (d, J = 8.0 Hz, 0.4 H), 8.12r2 (s, 0.4 H), 8.10rl (s, 0.6 H), 7.29 - 7.21ri,r2 (m, 1 H), 7.0 - 6.57rl>r2 (m, 3 H), 6.58ri (d, J = 1.6 Hz, 0.6 H), 6.04r2 (d, J = 1.6 Hz, 0.4 H), 5.19rl (bd, J = 8.0 Hz, 0.6 H), 5.09r2 (bd, J = 8.0 Hz, 0.4 H), 4.00 - 3.88rl,r2 (m, 1 H), 3.78 - 3.64r 2 (m, 1 H), 2.51 - 2.38rl,r2 (m, 1 H), 2.11 - 1.84r 2 (m, 3 H). MS m/z 326.1 (M+l)+.
Synthesis of 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolofl,5-a]pyrimidine-3- -28)
Figure imgf000098_0001
[000308] To a solution of ethyl 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin- 1- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) (1.4 g, 3.7 mmol) in THF/MeOH/H20 (3/2/1,
120 mL) was added 3 M LiOH (10 mL). The reaction was stirred at 50 °C for 8 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HC1 and reduced to dryness. The crude solid was triturated with copious amounts of DCM and filtered. The filtrated was dried over sodium sulfate, filtered and reduced to dryness to yield 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (X-53). MS m/z (M+l)+.
[000309] To a solution of 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (X-53) (0.1 g, 0.3 mmol) in NMP (1.5 mL) was added DIEA (0.1 mL, 0.6 mmol) and HATU (0.1 g, 0.3 mmol). The reaction was stirred at room temperature for 30 minutes then a mixture of NH4C1 (87 mg, 1.5 mmol) and NH4OH (0.1 mL) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness.
The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-28) as a white solid. 1H NMR (400MHz, DMSO- d6, at 70 °C) δ
8.67 (bd, J = 5.6 Hz, 1 H), 8.09 (s, 1 H), 7.37 (q, J = 7.2 Hz, 1 H), 7.19 (d, J = 7.6 Hz, 1 H), 7.16
(bs, 1 H), 7.05 (bt, J = 8.0 Hz, 1 H), 6.83 (bs, 1 H), 5.50 (dd, J = 52.0, 2.0Hz, 1 H), 5.30 (t, J = 8.0
Hz, 1 H), 5.27 (bs, 1 H), 4.19 (bs, 1 H), 2.91 - 2.81 (m, 1 H), 2.27 - 2.10 (m, 1 H). MS m/z 344.1
(M+l)+.
Synthesis of5-((2R S)A-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)-N-(piperidin-4-yl)pyrazolofl,5- a]pyrimidine-3-carboxamide (X-37)
Figure imgf000099_0001
[000310] To a solution of tert-butyl 4-(5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamido)piperidine-l-carboxylate (X-33) (76 mg, 0.14 mmol) in DCM (2 mL) was added TFA (1 mL) and stirred at room temperature for 12 hours. The reaction was subsequently reduced to dryness and neutralized with silica-C03 using DCM/MeOH (2/1) as eluant to yield 5-((2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)-N-(piperidin-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-37) as an amorphous solid. MS m/z 427'.1 (M+l)+.
Synthesis of(R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazoloil,5-a]pyrimidine-3- carboxamide (X-40)
Metho
Figure imgf000099_0002
[000311] (R)-Ethyl 5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine- 3-carboxylate (1-45) was prepared in an identical manner as ethyl 5-((2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) starting from (R)- 4,4-difluoro-2-(3-fluorophenyl)pyrrolidine (1-21).
[000312] To a solution of (R)-ethyl 5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (1-45) (0.48 g, 1.2 mmol) in THF/MeOH/H20 (3/2/1, 20 mL) was added 3 M LiOH (4 mL). The reaction was stirred at 60 °C for 12 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HC1 and reduced to dryness. The crude solid was triturated with copious amounts of DCM and filtered. The filtrated was dried over sodium sulfate, filtered and reduced to dryness to yield (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-46). MS m/z 363.1 (M+l)+.
[000313] To a solution of (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (1-46) (0.44 g, 1.2 mmol) in NMP (10 mL) was added DIEA (0.5 mL, 2.4 mmol) and HATU (0.47 g, 1.2 mmol). The reaction was stirred at room temperature for 30 minutes then a mixture of NH4C1 (0.36 g, 6.0 mmol) and NH4OH (1 mL) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-40) as a white solid. 1H NMR (400MHz, CD3OD) δ 8.55 (bs, 1 H), 8.22 (s, 1 H), 7.39 (q, J = 8.0 Hz, 1 H), 7.16 (d, J = 8.0 Hz, 1 H), 7.10 (bd J = 10 Hz, 1 H), 7.02 (t, J = 8.4 Hz, 1 H), 6.52 (bs, 1 H), 5.53 - 5.50 (m, 1 H), 4.43 (q, J = 12.0 Hz, 1 H), 4.31 (bs, 1 H), 3.22 - 3.09 (m, 1 H), 2.52 (dq, J = 13.2, 5.2 Hz, 1 H). MS m/z 362.1 (M+l)+.
Method B
Figure imgf000100_0001
1-21 1-56 X-40
[000314] To a solution of (R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine (1-21) (25 mg, 0.12 mmol) in DMSO (0.5 mL) was added KF (15 mg, 0.26 mmol) and 5-chloropyrazolo[l,5- a]pyrimidine-3-carboxamide (1-56) (25 mg,0.12 mmol). The reaction was heated at 85 °C for 2 hours then cooled to 0 °C. The frozen reaction mixture was treated with water. The resulting white precipitate was filtered, washed with water and dried under vacuum. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield (R)- 5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X- 40) as a white solid. 1H NMR (400MHz, CD3OD) δ 8.55 (bs, 1 H), 8.22 (s, 1 H), 7.39 (q, J = 8.0 Hz, 1 H), 7.16 (d, J = 8.0 Hz, 1 H), 7.10 (bd J = 10 Hz, 1 H), 7.02 (t, J = 8.4 Hz, 1 H), 6.52 (bs, 1 H), 5.53 - 5.50 (m, 1 H), 4.43 (q, J = 12.0 Hz, 1 H), 4.31 (bs, 1 H), 3.22 - 3.09 (m, 1 H), 2.52 (dq, J = 13.2, 5.2 Hz, 1 H). MS m/z 362.1 (M+l)+.
Synthesis of (S)-5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazoloil,5-a]pyrimidine-3- carboxamide (X-45)
Figure imgf000101_0001
1-47 1-48 X-45
[000315] Ethyl 5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylate (1-47) was prepared in an identical manner as ethyl 5-((2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) starting from 5-(3-fluorophenyl)-2,2-dimethylpyrrolidine (1-38).
[000316] To a solution of ethyl 5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylate (1-47) (90 mg, 0.2 mmol) in THF/MeOH/H20 (3/2/1, 6 mL) was added 3 M LiOH (0.4 mL). The reaction was stirred at 50 °C for 6 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HCl and the resulting precipitate was filtered, washed with water and dried to yield 5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-48). MS m/z 355.1 (M+l)+.
[000317] To a solution of 5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (1-48) (60 mg, 0.17 mmol) in NMP (1.5 mL) was added DIEA (89 μί, 0.51 mmol) and HATU (65 mg, 0.17 mmol). The reaction was stirred at room temperature for 30 minutes then a mixture of NH4CI (27 mg, 0.51 mmol) and NH4OH (10 μί) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield 5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-44) as an amorphous solid. Chiral resolution was performed on LCMS using a 21x250mm ChiralPak AD-H column with 80g/min 70/30 C02/MeOH at 35 °C over 6 minutes to yield (S)-5-(5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-45). 1H NMR (400MHz, DMSO-6) δ 8.59 (bs, 1 H), 8.13 (bs, 1 H), 7.45 - 7.32 (m, 3 H), 7.11 - 7.08 (m, 4 H), 5.27 - 5.26 (m, 1 H), 1.89 -1.78 (m, 6 H), 1.62 - 1.57 (m, 3 H). MS m/z 354.1 (M+l)+.
Synthesis of'5-((lR,2R,5S)-2-(3-fluorophenyl)-3-azabicycloi3.1.0]hexan-3-yl)pyrazolol 1,5- a]pyrimidine-3-carboxamide (X-47)
Figure imgf000102_0001
X-23 1-49 X-47
[000318] Ethyl 5-(2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl)pyrazolo[l,5-a]pyrimidine- 3-carboxylate (X-23) was prepared in an identical manner as ethyl 5-((2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin- l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) starting from
2- (3-fluorophenyl)-3-azabicyclo[3.1.0]hexane (1-42).
[000319] To a solution of ethyl 5-(2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-23) (130 mg, 0.36 mmol) in THF/MeOH/H20 (3/2/1, 6 mL) was added 3 M LiOH (2.0 mL). The reaction mixture was stirred at 60 °C for 16 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HC1 and the resulting precipitate was filtered, washed with water and dried to yield 5-(2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-49). MS m/z 339.1 (M+l)+.
[000320] To a solution of 5-(2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (1-49) (100 mg, 0.30 mmol) in NMP (2.0 mL) was added triethylamine (120 μί, 0.89 mmol) and HATU (170 mg, 0.45 mmol). The reaction was stirred at room temperature for 30 minutes then NH4C1 (1.0 mL of a 2 M solution in NH4OH) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and reduced to dryness. The crude product was purified by preparative LCMS to yield 5-((lR,2R,5S)-2-(3-fluorophenyl)-
3- azabicyclo[3.1.0]hexan-3-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide. Chiral resolution was performed on LCMS using a 20x250mm ChiralCel OD column with 18mL/min, 8/1/1
Hexane/EtOH/MeOH+0.1 DEA over 22 minutes to yield 5-((lR,2R,5S)-2-(3-fluorophenyl)-3- azabicyclo[3.1.0]hexan-3-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-47) and its
diastereomer (X-46). 1H NMR (400MHz, CD3OD) δ 8.51 (bs, 1 H), 8.16 (s, 1 H), 7.38 (q, J = 7.6 Hz, 1 H), 7.07 (d, J = 7.6 Hz, 1 H), 6.99 (q, J = 7.6 Hz, 2 H), 6.62 (bs, 1 H), 5.35 (d, J = 5.2 Hz, 1 H), 4.10 (d, J = 10.4 Hz, 1 H), 3.97 (bs, 1 H), 2.19 (bs, 1 H), 2.02 - 1.96 (m, 1 H), 0.73 (q, J = 8.0 Hz, 1 H), 0.54 (q, J = 4.4 Hz, 1 H). MS m/z 338.1 (M+l)+. Synthesis of (R)-5-(2-<'3 ' -fluorophenyl)piperidin-l -yDpyrazolol 1,5-a] [pyrimidine-3-carboxamide (X-
50
Figure imgf000103_0001
X"24 1-50 X.50
[000321] Ethyl 5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X- 24) was prepared in an identical manner as ethyl 5-((2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) starting from commercially available 2-(3-fhiorophenyl)piperidine.
[000322] To a solution of ethyl 5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylate (X-24) (100 mg, 0.27 mmol) in THF/MeOH/H20 (3/2/1, 12 mL) was added 3 M
LiOH (1.8 mL). The reaction mixture was stirred at 60 °C for 18 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HCl and the resulting precipitate was filtered, washed with water and dried to yield 5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (I- 50). MS m z 341.1 (M+l)+.
[000323] To a solution of 5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-50) (80mg, 0.24 mmol) in NMP (2.5 mL) was added DIEA (150 μί, 0.86 mmol) and HATU (130 mg, 0.35 mmol). The reaction was stirred at room temperature for 30 minutes then NH4C1 (1.0 mL of a 2 M solution in NH4OH) was added in one portion and stirred for 1 hour. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and reduced to dryness. The crude product was purified by preparative LCMS to yield 5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-49). Chiral resolution was performed on LCMS using a
4.6x250mm ChiralCel OD-H column with lmL/min, 8/1/1 hexane/EtOH/MeOH + 0.1% DEA over 18 minutes to yield (R)-5-(2-(3-fluorophenyl)piperidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxamide (X-50). 1H NMR (400MHz, CD3OD @ 319K) δ 8.49 (d, J = 8 Hz, 1 H), 8.22 (s, 1 H), 7.39 (q, J = 8 Hz, 1 H), 7.10 (d, J = 7.6 Hz, 1 H), 7.03 - 6.95 (m, 2 H), 6.78 (d, J = 8 Hz, 1 H), 5.80 (t, J = 4.0 Hz, 1 H), 4.42 (d, J = 13.2 Hz, 1 H), 3.41 - 3.29 (m, 1 H), 2.41 - 2.35 (m, 1 H), 2.16 - 2.07 (m, 1 H), 1.90 - 1.80 (m, 1 H), 1.79 - 1.69 (m, 2 H), 1.66 - 1.55 (m, 1 H). MS m/z 340.2 (M+l)+.
Synthesis of (R)-5-(2-('3 -fluoropheny rrolidin-l -yDpyrazolol 1,5-a] 'pyrimidine (X-51 )
Figure imgf000104_0001
X-51
[000324] (R)-5-(2-(3-Fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine (X-51) was isolated as a byproduct in the synthesis of (R)-5-(2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-25) using Method C. Purified by preparative thin layer chromatography. 1H NMR (400MHz, DMSO-6) δ 8.56 (bs, 1 H), 7.81 (s, 1 H), 7.39 - 7.34 (m, 1 H), 7.07 - 7.04 (m, 1 H), 5.99 (bs, 1 H), 5.24 (bs, 1 H), 3.94 - 3.89 (m, 1 H), 3.63 (bs, 1 H), 2.43 - 2.40 (m, 1 H), 1.99 - 1.85 (m, 3 H). MS m/z 283.1 (M+l)+.
Synthesis of(R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrroUdin-l-yl)-N-ethoxypyrazolo[l,5- -3-carboxamide (X-52 )
Figure imgf000104_0002
[000325] (R)-Ethyl 5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine- 3-carboxylate (1-51) was prepared in an identical manner as ethyl 5-((2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-14) starting from (R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine (1-21) instead of (2R,4S)-4-fluoro-2-(3- fluorophenyl)pyrrolidine (1-18).
[000326] To a solution of (R)-ethyl 5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (1-51) (130 mg, 0.33 mmol) in THF/MeOH/H20 (3/2/1, 12 mL) was added 3 M LiOH (2.0 mL). The reaction mixture was stirred at 60 °C for 16 hours then cooled to room temperature and reduced in volume on roto-vap to remove the organic solvents. The crude mixture was neutralized with HCl and the resulting precipitate was filtered, washed with water and dried to yield (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-52). MS m/z 363.1 (M+l)+. [000327] To a solution of (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (1-52) (70 mg, 0.19 mmol) in NMP (2.0 mL) was added
triethylamine (100 μί, 0.72 mmol) and HATU (115 mg, 0.30 mmol). The reaction was stirred at room temperature for 30 minutes then O-ethylhydroxylamine hydrochloride (0.2 g, 2.1 mmol) was added in one portion and stirred for 2 hours. The reaction was partitioned with EtOAc/H20. The organic layer was separated, washed with brine, dried over anhydrous sodium sulfate, filtered and reduced to dryness. The crude product was purified by preparative LCMS to yield (R)-5-(4,4- difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)-N-ethoxypyrazolo[l,5-a]pyrimidine-3-carboxamide (X-52). 1H NMR (400MHz, CD3OD) δ 8.49 (bs, 1 H), 8.13 (s, 1 H), 7.32 (q, J = 8.0 Hz, 1 H), 7.09 (d, J = 7.6 Hz, 1 H), 7.04 (d, J = 10.0 Hz, 1 H), 6.95 (dt, J = 8.4, 2.0 Hz, 1 H), 6.54 (bs, 1 H), 5.46 (bs, 1 H), 4.39 (q, J = 11.6 Hz, 1 H), 4.21 (bs, 1 H), 3.83 (bs, 1 H), 3.66 (bs, 1 H), 3.83 (bs, 1 H), 3.65 (bs, 1 H), 3.09 - 2.99 (m, 1 H), 2.46 (dq, J = 12.0, 4.4 Hz, 1 H), 1.18 (bs, 3 H). MS m/z 406.1 (M+l)+.
Synthesis of (R)-N-carbamoyl-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[ 1,5- -3-carboxamide (X-59)
Figure imgf000105_0001
X-40 X-59
[000328] To a solution of (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-40) (62 mg, 0.17 mmol) in 1,2-dichloromethane (2 mL) was added oxalyl chloride (15 μί, 0.17 mmol). The reaction was heated to 85 °C for 4 hours then cooled to room temperature. Upon cooling NH4C1 (30 mg, 0.52 mmol) and NH4OH (0.2 mL) were added and the reaction was stirred at room temperature for 1 hour. The reaction was reduced to dryness and purified by column chromatography with DCM/MeOH gradient as eluant to yield (R)-N-carbamoyl-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxamide (X-59) as a white solid. MS m/z 405.1 (M+l)+.
Synthesis of ethyl 5-((2R,4S)-4-fluoro-2-(3-methyl-l,2,4-ox diazol-5-yl)pyrrolidin-l-yl)pyrazolofl,5- a I yrimidine -3 - carboxylate (X-71 )
Figure imgf000106_0001
1-53 1-54 X-71
[000329] To a solution of (2R,4R)-l-(3-(ethoxycarbonyl)pyrazolo[l,5-a]pyrimidin-5-yl)-4- hydroxypyrrolidine-2-carboxylic acid (1-53) (54 mg, 0.17 mmol) in NMP (1.5 mL) was added HATU (65 mg, 0.17 mmol) and DIEA (60 μί, 0.34 mmol). The reaction was stirred at room temperature for 15 minutes then N'-hydroxyacetimidamide (13 mg, 0.18 mmol) was added. The reaction was stirred from room temperature to 85 °C for 12 hours. Upon cooling the reaciton was filtered and purified by preparative LCMS to yield ethyl 5-((2R,4R)-4-hydroxy-2-(3-methyl- 1,2,4- oxadiazol-5-yl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (1-54) as a white solid. 1H NMR (400MHz, CD3OD) δ 8.54 (d, J = 7.6 Hz, 1 H), 8.22 (s, 1 H), 6.92 (d, J = 6.8 Hz, 1 H), 5.74 (bs, 1 H), 4.66 (bs, 1 H), 4.25 (q, J = 6.0 Hz, 2 H), 3.93 (bs, 1 H), 3.81 (bs, 1 H), 2.73 - 2.66 (m, 1 H), 2.38 (bs, 1 H), 2.31 (s, 1 H), 1.31 (t, J = 6.0 Hz, 3 H). MS m/z 361.1 (M+l)+.
[000330] To a solution of ethyl 5-((2R,4R)-4-hydroxy-2-(3-methyl-l,2,4-oxadiazol-5- yl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (1-54) (37 mg, 0.1 mmol) in DCM (2 mL) at -78 °C was added DAST (27 μί, 0.2 mmol). The reaction was stirred to room temperature over 12 hours then partitioned with water and EtOAc. The organic layer was washed with brine, dried with magnesium sulfate, filtered and reduced to dryness. The crude product was purified by column chromatography on silica gel with DCM/MeOH gradient as eluant to yield ethyl 5- ((2R,4S)-4-fluoro-2-(3-methyl- 1 ,2,4-oxadiazol-5-yl)pyrrolidin- 1 -yl)pyrazolo[ 1 ,5-a]pyrimidine-3- carboxylate (X-71) as a clear glassy solid. 1H NMR (400MHz, CD3OD) δ 8.62 (d, J = 7.6 Hz, 1 H), 8.23 (s, 1 H), 6.71 (d, J = 8.0 Hz, 1 H), 5.67 (t, J = 8.0 Hz, 1 H), 5.55 (d J = 52 Hz, 1 H), 4.30 (q, J = 6.8 Hz, 2 H), 4.17 - 4.10 (m, 2 H), 2.80 - 2.88 (m, 1 H), 2.48 - 2.38 (m, 1 H), 2.36 (s, 3 H), 1.37 (t, J = 7.2 Hz, 3 H). MS m/z 361.1 (M+l)+.
Synthesis of 5-(2-(3-cyano-5-fluorophenyl)pyrrolidin-l-yl)pyrazolofl,5-a]pyrimidine-3-carboxamide
(X-75)
Figure imgf000107_0001
[000331] To a solution of 3-fluoro-5-(pyrrolidin-2-yl)benzonitrile (1-60) (0.20 g, 1.05 mmol) and DIEA (0.55 mL, 3.15 mmol) in EtOH (6 mL) was added 5-chloropyrazolo[l,5-a]pyrimidine-3- carboxylic acid (1-56) (0.12 g, 1.05 mmol) and heated to 80 °C for 6 hours. The mixture was concentrated and redissolved in DCM. The organics were washed with water (2x), dried over Na2S04, and filtered. The crude product was purified by silica gel chromatography with a MeOH/ DCM gradient to afford 5-(2-(3-cyano-5-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine- 3-carboxamide (X-75). MS m/z 351.1 (M+l)+.
Synthesis of (ethyl carbonic) (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrroUdin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carbox lic anhydride (X-77)
Figure imgf000107_0002
[000332] To a solution of (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxylic acid (1-46) (0.093 g, 0.21 mmol) and TEA (0.032 mL, 0.23 mmol) in THF (1 mL) at -5 °C was added ethyl carbonochloridate (0.022 mL, 0.23 mmol). The reaction was maintained at -5 °C for 45 minutes and concentrated to dryness. The resulting material was partitioned between EtOAc and saturated aqueous NaHC03. The organic layer was washed with brine, separated and reduced to dryness. The crude material was purified by silica gel
chromatography with an EtOAc/hexane gradient to afford (ethyl carbonic) (R)-5-(4,4-difluoro-2- (3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic anhydride (X-77). 1H NMR (400MHz, CDC13) δ 8.34 (s, 1 H), 8.23 (bd, J = 7.5 Hz, 1 H), 7.40 - 7.33 (m, 1 H), 7.09 - 7.07 (m, 1 H), 7.04 - 6.97 (m, 2 H), 6.05 (bs, 1 H), 5.26 (bs, 1 H), 4.49 (bs, 1 H), 4.38 (q, J = 7.2 Hz, 2 H), 4.38 - 4.28 (m, 1 H), 3.13 - 3.03 (m, 1 H), 2.56 (qd, J = 13.3, 5.2 Hz, 1 H), 1.41 (t, J = 7.1 Hz, 3 H). MS m/z 435.1 (M+l)+. Synthesis of (R)-3-bromo-5-(4A-difluoro-2-(3-fluorophenyl)pyrroUdin-l-yl)pyrazolo[ 5- -78)
Figure imgf000108_0001
1-61 X-78
[000333] (R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine (1-21) (0.10 g, 0.65 mmol), 5- chloropyrazolo[l,5-a]pyrimidine (0.13 g, 0.65 mmol), DIEA (0.17 mL, 0.98 mmol) and EtOH (2 mL) were combined and heated for two iterations at 140 °C for 30 minutes in the microwave. The reaction was concentrated and the resulting residue was partitioned with EtOAc and saturated aqueous NaHC03. The aqueous layer was extracted with EtOAc. The combined organics were dried over Na2S04, filtered and purified by silica gel chromatography with an EtOAc/hexane gradient to afford (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine (1-61). 1H NMR (400MHz, CDC13) δ 8.20 (d, J = 8.4 Hz, 1 H), 7.88 (d, J = 2.2 Hz, 1 H), 7.33 (m, 1 H), 7.04 (d, J = 7.8 Hz, 1 H), 6.98 (m, 2 H), 6.17 (m, 1 H), 5.87 (d, J = 7.7 Hz, 1 H), 5.20 (dd, J = 9.9 Hz, 5.0 Hz, 1 H), 4.35 (q, J = 12.8 Hz, 1 H), 4.23 (q, J = 12.3 Hz, 1 H), 3.08 - 2.95 (m, 1 H), 2.50 (qd, J = 12.7 Hz, 5.0 Hz, 1 H). MS m/z 319.1 (M+l)+.
[000334] To a solution of (R)-5-(4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine (1-61) (0.025 g, 0.079 mmol) in ACN (1 mL) at 0 °C was added NBS (0.015 g, 0.087 mmol). The reaction was maintained at 0 °C for 2 hours and concentrated. The crude material was purified by silica gel chromatography with an EtOAc/hexane gradient afforded (R)-3-bromo-5- (4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine (X-78). 1H NMR (400MHz, CDC13) δ 8.14 (d, J = 7.7 Hz, 1 H), 7.86 (s, 1 H), 7.37 - 7.32 (m, 1 H), 7.07 (d, J = 7.7 Hz, 1 H), 7.02 - 6.98 (m, 2 H), 5.90 (d, J = 7.7 Hz, 1 H), 5.24 - 5.20 (m, 1 H), 4.43 (q, J = 12.7, 1 H), 4.30 (q, J = 12.7, 1 H), 3.10 - 2.97 (m, 1 H), 2.53 (qd, J = 13.0, 5.4 Hz, 1 H). MS m/z 397.0, 399.0 (M+l)+.
Synthesis of 5-(2-(5-fluoro-2-(isopropylcarbamoyl)phenyl)pyrrolidin-l-yl)pyrazolol 1,5- a]pyrimidine-3-carboxamide (X-82 )
Figure imgf000109_0001
[000335] To a solution of 2-(l-(3-carbamoylpyrazolo[l,5-a]pyrimidin-5-yl)pyrrolidin-2-yl)-4- fluorobenzoic acid (1-64) (0.024 g, 0.065 mmol) in THF (1 mL) was added DIEA (0.17 mL, 0.98 mmol), HATU (.012 g, 0.33 mmol) and isopropyl amine (0.019 g, 0.33 mmol). The resulting mixture was stirred at ambient temperature overnight and concentrated. The crude material was purified by preperative LCMS afforded 5-(2-(5-fluoro-2-(isopropylcarbamoyl)phenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-82).1H NMR (400MHz, CD3OD, 319K) δ 8.51 - 8.33 (m, 1 H), 8.17 (bs, 1 H), 7.48 - 7.43 (m, 1 H), 7.04 (bs, 1 H), 6.90 (dd, J = 10.1, 2.6, 1 H), 6.66 (bs, 1 H), 5.80 (bs, 1 H), 4.23 - 4.17 (m, 1 H), 4.04 (bs, 1 H), 3.78 (bs, 1 H), 2.60 - 2.51 (m, 1 H), 2.20 - 2.04 (m, 3 H), 1.32 - 1.25 (m, 6H). MS m/z 411.2 (M+l)+.
Synthesis of 5-(2-(3-(aminomethyl)-5-fluorophenyl)pyrrolidin-l-yl)pyrazolofl,5-a]pyrimidine-3- -87)
Figure imgf000109_0002
[000336] To a solution of 5-(2-(3-cyano-5-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (X-75) (0.010 g, 0.8 mmol) in MeOH (1 mL) and DCM (1 mL) was added Raney Ni (0.5 mL, slurry in H20). The reaction was placed under H2 (1 atm) and stirred overnight. The mixture was filtered through celite and purified by preparative LCMS to afford 5- (2-(3-(aminomethyl)-5-fluorophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-87). MS m/z 355.2(M+1)+.
Synthesis of 5-(2-(3-fluoro-5-((isopropylamino)methyl)phenyl)pyrrolidin-l-yl)pyrazolofl,5- a lpyrimidine-3 -carboxamide (X-88 )
Figure imgf000110_0001
X-87 X-88
[000337] To a supsension of 5-(2-(3-(aminomethyl)-5-fluorophenyl)pyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-87) (0.020 g, 0.056 mmol) in EtOH (1 mL) and AcOH (0.1 mL) was added acetone (0.04 mL, 0.56 mmol) and sodium cyanoborohydride (0.018 g, 0.28 mmol). The resulting mixture was stirred for 2 hours at ambient temperature and
concentrated. The crude product was purification by silica gel chromatography with a
MeOH/DCM gradient followed by preperative LCMS afforded 5-(2-(3-fluoro-5- ((isopropylamino)methyl)phenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X- 88). MS m/z 397.2(M+1)+.
Synthesis of 5-((2R,4S)-2-(3-cyano-5-fluorophenyl)-4-fluoropyrroUdin-l-yl)pyrazolol 1,5- -3-carboxamide (X-91 )
Figure imgf000110_0002
1-74 1-56 X_91
[000338] To a solution of 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxamide (1-56) (20 mg, 0.1 mmol) and 3-fluoro-5-((2R,4S)-4-fluoropyrrolidin-2-yl)benzonitrile (1-74) (20 mg, 0.1 mmol) in DMSO (1 mL) was added potassium fluoride (30 mg, 0.5 mmol). The reaction was heated to 120 °C overnight then cooled to room temperature. The crude was diluted with EtOAc, washed with H20 and brine, dried over magnesium sulfate, filtered, concentrated and purified by flash column chromatography on silica gel with DCM/MeOH gradient as eluant to yield 5-((2R,4S)-2-(3-cyano- 5-fluorophenyl)-4-fluoropyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-91). 1H NMR (400MHz, CD3OD) δ 8.62 (bs, 1 H), 8.29 (s, 1 H), 7.69 (s, 1 H), 7.60 - 7.50 (m, 2 H), 6.75 (bs, 1 H), 5.56 (d, J = 52.4 Hz, 1 H), 5.53 - 5.46 (m, 1 H), 4.45 - 4.25 (m, 2 H), 3.12 - 2.95 (m, 1 H), 2.30 (dt, J = 40.8, 10.4 Hz, 1 H). MS m/z 369.1 (M+l)+. Synthesis of(R)-ethyl 5-(2-('3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidin- 1 -yDpyrazolol 1 ,5- a ]pyrimidine-3-carboxylate (X-94 )
Figure imgf000111_0001
[000339] To a solution of ethyl 5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (1-10) (100 mg, 0.44 mmol) and (R)-3-(4,4-difluoropyrrolidin-2-yl)-5-fluorobenzonitrile (1-77) (100 mg, 0.44 mmol) in DMSO (2 mL) was added potassium fluoride (129 mg, 2.2 mmol). The reaction was heated to 120 °C overnight then cooled to room temperature. The crude was diluted with EtOAc, washed with H20 and brine, dried over magnesium sulfate, filtered, concentrated and purified by flash column chromatography on silica gel with hexanes/EtOAc gradient as eluant to yield (R)- ethyl 5-(2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3- carboxylate (X-94). 1H NMR (400MHz, CDC13) δ 8.33 (d, J = 8.0 Hz, 1 H), 8.31 (s, 1 H), 7.50 (s, 1 H), 7.35 (d, J = 8.4 Hz, 1 H), 7.28 (d, J = 7.2 Hz, 1 H), 6.06 (bs, 1 H), 5.49 (bs, 1 H), 4.34 (q, J = 7.2 Hz, 1 H), 4.32 - 4.20 (m, 2 H), 3.14 - 2.96 (m, 1 H), 2.62 - 2.48 (m, 1 H), 1.36 (t, J = 7.2 Hz, 1 H). MS m z 416.1 (M+l)+.
Synthesis of5-((2R,4S)-2-(3-carbamoyl-5-fluorophenyl)-4-fluoropyrrolidin-l-yl)pyrazoloi 1,5- a rimidine-3-carboxamide (X-96 )
Figure imgf000111_0002
X-95 1-78 X-96
[000340] To a suspensions of ethyl 5-((2R,4S)-2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylate (X-95) (50 mg, 0.126 mmol) in EtOH (1 mL) was added 6 N KOH (0.3 mL, 1.5 mmol). The reaction was heated to reflux overnight then cooled to room temperature. The mixture was concentrated and purified by flash column chromatography on silica gel with DCM/MeOH/0.1% AcOH gradient as eluant to yield 5-((2R,4S)-2-(3-carboxy-5- fluorophenyl)-4-fluoropyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-78). MS m/z 389. 1 (M+l)+. [000341] To a solution of 5-((2R,4S)-2-(3-carboxy-5-fluorophenyl)-4-fluoropyrrolidin-l- yl)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (1-78) (53 mg, 0.136 mmol) in DMF (3 mL) was added DIEA (0.12 mL, 0.68 mmol) and HATU (156 mg, 0.41 mmol). The reaction was stirred for 30 minutes at room temperature then a solution of NH4C1 (29 mg, 0.54 mmol) in NH4OH (0.25 mL) was added. The reaction was stirred at room temperature for 3 hours. The mixture was diluted with EtOAc, washed with brine, dried over magnesium sulfate, filtered and reduced to dryness. The crude product was purified by flash column chromatography on silica with
DCM/MeOH gradient as eluant to yield 5-((2R,4S)-2-(3-carbamoyl-5-fluorophenyl)-4- fluoropyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-96). 1H NMR (400MHz, CD3OD) δ 8.50 (bs, 1 H), 8.18 (s, 1 H), 7.68 (s, 1 H), 7.49 (d, J = 8.8 Hz, 1 H), 7.30 (d, J = 8.8 Hz, 1 H), 6.67 (bs, 1 H), 5.47 (d, J = 52.8 Hz, 1 H), 5.37 (t, J = 8.0 Hz, 1 H), 4.35 - 4.13 (m, 2 H), 3.02 - 2.86 (m, 1 H), 2.34 - 2.12 (m, 1 H). MS mJz 387.1 (M+l)+.
Synthesis of5-((2R,4S)-4-cyano-2-(3-fluorophenyl)pyrrolidin-l-yl)pyrazoloil,5-a]pyrimidine-3- -76)
Figure imgf000112_0001
1-81 1-56 X-76
[000342] To a microwave vial were combined (3S,5R)-5-(3-fluorophenyl)pyrrolidine-3- carbonitrile 2,2,2-trifluoroacetate (1-81) (38 mg, 0.13 mmol), 5-chloropyrazolo[l,5-a]pyrimidine- 3-carboxamide (1-56) (24 mg, 0.12 mmol), potassium fluoride (21 mg, 3 eq) and DMSO (anhyd, 0.5 mL) followed by DIEA (26 μί, 0.15 mmol). The vial was sealed and irradiated in a Biotage microwave reactor at 120 °C for 30 minutes. The cooled reaction mixture was partitioned in EtOAc and water. Combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to dryness. The residue was purified by silica chromatography with
MeOH/DCM (0-3% gradient) as eluent to yield 5-((2R,4S)-4-cyano-2-(3-fluorophenyl)pyrrolidin- l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (1-76) as a white solid. 1H NMR (400MHz, CDCI3) δ 8.42 (s, 1 H), 8.31 (bs, 1 H), 7.42 - 7.36 (m, 1 H), 7.07 - 7.04 (m, 1 H), 6.99 (d, J = 7.7 Hz, 1 H), 6.89 (d, J = 9.3 Hz, 1 H), 6.05 (bs, 1 H), 5.53 (bs, 1 H), 5.28 (bs, 1 H), 4.37 - 4.33 (m, 1 H), 4.17 (bs, 1 H), 3.44 - 3.36 (m, 1 H), 2.95 - 2.87 (m, 1 H), 2.52 - 2.46 (m, 1 H). MS m z 351.1 (M+l)+.
I l l Synthesis of 5-( -(2-cyanophenyl)pyrrolidin-l -yl)pyrazoloi 1 ,5-a]pyrimidine-3-carboxamide (X-79)
Figure imgf000113_0001
X-79
[000343] To a microwave vial added 5-(2-(2-bromophenyl)pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide (1-92, prepared in a similar manner to the synthesis of X-75 starting from 2-(2-bromophenyl)pyrrolidine) (93 mg, 0.24 mmol), zinc cyanide (17 mg, 0.14 mmol), zinc dust (4.7 mg, 0.07 mmol), tris(dibenzylideneacetone)dipalladium(0) (11 mg, 5 mol ), and 1,1'- bis(diphenyl-phosphino)ferrocene (DPPF) (13 mg, 10 mol ). After sealing the vial, it was evacuated and back-filled with argon (3x) and then charged with N,N-dimethylacetamide (0.6 mL). The vial was irradiated in a Biotage microwave reactor at 120 °C for 25 minutes. The cooled reaction mixture was partitioned in EtOAc and water and the combined extracts were washed with water (4 x 25 mL) and brine, then dried over sodium sulfate, filtered and concentrated to dryness. The residue was purified by silica chromatography with MeOH/DCM (0-3% gradient) as eluent, then triturated with Et20 and dried under vacuum to yield 5-(2-(2- cyanophenyl)pyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (X-79) as a white solid. 1H NMR (400 MHz, MeOD) δ 8.54 (d, J = 7.7 Hz, 1 H), 8.12 (s, 1 H), 7.75 (d, J = 7.7 Hz, 1 H), 7.62 - 7.58 (m, 1 H), 7.42 - 7.39 (m, 2 H), 6.67 (d, J = 7.7 Hz, 1 H), 5.51 - 5.48 (m, 1 H), 4.07 - 4.02 (m, 1 H), 3.87 - 3.81 (m, 1 H), 2.71 - 2.62 (m, 1 H), 2.28 - 2.16 (m, 2 H), 2.05 - 1.99 (m, 1 H). MS m/z 333.1 (M+l)+.
[000344] By repeating the procedures described in the above examples, using appropriate starting materials, the following compounds of Formula I, as identified in Table 1, were obtained.
Table 1
Figure imgf000113_0002
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Assays
Preparation of compound dilutions
[000345] Test compounds were dissolved in DMSO (10 mM) and transferred into 1.4mL flat bottom or V-shaped Matrix tubes carrying a unique 2D matrix chip by individual compound hubs. The numbers of these chips were distinctively linked to the individual compound identification numbers. The stock solutions were stored at -20°C if not used immediately. For the test procedure the vials were defrosted and identified by a scanner whereby a working sheet is generated that guides the subsequent working steps.
[000346] Compound dilutions were made in 384 well plates. This format enabled the assay of maximally 28 individual test compounds at 11 concentrations (single points) including 2 reference compounds. The dilution protocol included the production of pre-dilution plates, master plates and assay plates:
Compound plates: 30 μΐ^ of individual compound (10 mM) DMSO solution including reference compound were transferred into columns 1 and 13 of a 384 well plate. 20 μΐ^ of DMSO were added to the rest of the wells and the compounds were serially diluted (1:3) by transferring 10 μΐ^ from a well in column 1 or 13 to the next well in column 2 or 14 respectively and successively with the help of a
Minitrack robot.
Assay plates: Identical assay plates were then prepared by adding 50 nL each of compound dilutions of the compound plates into 384-well "assay plates". In the following the compounds were mixed with 50 μΐ^ of assays components (cells or enzyme) and tested for their inhibitory activity.
[000347] Compounds of Formula (I) were assayed to measure their capacity to inhibit TrkA, TrkB, and/or TrkC protein kinases. Other compounds of Formula (I) were assayed to measure their capacity to inhibit a Ba/F3 kinase panel, including but not limited to Ros, KDR, FMS, c-FMS, FLT3, c-Kit, JAK2, PDGFR, Lck, TrkA, TrkB, TrkC, IGF-1R and ALK protein kinases.
Ba/F3 cell proliferation assay panel
[000348] Compounds were tested for their ability to inhibit the proliferation of wt Ba/F3 cells and Ba/F3 cells transformed with constitutively expressed luciferase reporter and BCR-ABL or Tel-FMS or other Tel fusion kinases (EGFR, JAK2, ALK, BMX,FGFR3, FGFR4,FGR2, FLT1, FLT3, IGF1R,INSR,KDR, KIT ,LCK, LYN, MEK, MET, PDGFRcc, PDGFR , RET, RON, ROS, SRC, SYK, TIE and TYRO) or BRafV600E. Parental Ba/F3 cells were maintained in media containing recombinant mouse IL3 and the kinase transformed Ba/F3 cells were maintained in media without IL-3. 7.5 nl of compounds were spotted to each well of 1536-well assay plates by Liquid handling System Echo 555 (Labcyte). 700 cells were then plated into each well of the assay plates in 7 uL culture media per well and compounds were tested at 0.17 nM to 10 uM in 3-fold serial dilutions. The cells were then incubated for 48 hours at 37 °C. 3 uL of Bright-Glo®
(Promega) was added to each well and the plates were read using ViewLux (PerkinElmer).
Inhibition of cellular TrkA, TrkB and TrkC dependent proliferation
[000349] Compounds of Formula (I) were assayed to measure their capacity to selectively inhibit cell proliferation of Ba/F3 cells expressing activated TrkA, TrkB or TrkC through fusion to the dimerization domain of Tel (ETV6) transcription factor as well as Ba/F3 cells co-expressing full length rTrkA and mNGF compared with parental BaF3 cells.
[000350] The cell line used is the luciferase expressing Ba/F3 murine hematopoietic progenitor cell line transformed with human Tel-TrkA, Tel-TrkB or Tel-TrkC cDNAs (Ba/F3 EN A/B/C). These cells maintained in RPMI/10 fetal bovine serum (RPMI/FCS) supplemented with penicillin 50 mg/mL, streptomycin 50 mg/mL and L-glutamine 200 mM. Untransformed Ba/F3 cells were similarly maintained with the addition 5ng/ml of murine recombinant IL3. 50 μΐ of a Ba/F3 or Ba/F3 EN A/B/C cell suspension were plated in Greiner 384 well microplates (white ) at a density of 2000 cells per well. 50nl of serially diluted test compound (10-0.0001 mM in DMSO solution) is added to each well. The cells were incubated for 48 hours at 37 °C, 5% C02. 25μ1 of Bright glo® (Promega) luciferase substrate is added to each well. The emited luminiscence is quantified using ViewLux (PerkinElmer). IC50 values were calculated by linear regression analysis of the percentage inhibition of each compound at 11 concentrations.
Certain Assay Results
[000351] Various compounds of Formula (I) in free form or in pharmaceutically acceptable salt form, exhibit pharmacological properties, for example, as indicated by the in vitro and in vivo tests described in this application. The IC50 value in those experiments is given as that concentration of the test compound in question that results in a cell count that is 50 % lower than that obtained using the control without inhibitor. In certain examples compounds of Formula (I) have IC50 values from 0.1 nM to 200 μΜ. In some examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 100 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 50 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 25 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 20 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 15 μΜ In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 10 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.0001 μΜ to 5 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 2 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 1 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 8 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 6 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 4 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 2 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 1 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 08 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 06 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 04 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 02 μΜ. In other examples, compounds of Formula ( ) have IC50 values from 0 0001 μΜ to 0 01 μΜ.
[000352] In some examples, compounds of Formula (I) have IC50 values from 0.01 μΜ to 5 μΜ. In other examples, compounds of Formula (I) have IC50 values from 0.01 μΜ to 1 μΜ. In yet other examples, compounds of Formula (I) have IC50 values of less than 1 nM. In certain embodiments, compounds of Formula (I) exhibit a percentage inhibition of greater than 50%, or in other embodiments compounds of Formula (I) exhibit a percentage inhibition greater than about 70%.
[000353] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes.

Claims

WE CLAIM:
1. A compound of Formula (I):
Figure imgf000137_0001
Formula (I)
wherein:
Figure imgf000137_0002
R1 is H, -C(0)NH2, -LlR4, -L2R13, -C(0)OR5-, -C(0)NHOR5, -C(0)NHC(0)OR5-,
-C(0)OC(0)OR5- -C(0)NHC(0)NR5R5, -C(0)NR5OR14, -C(0)NHNH2, -
NR5C(0)NR5R9,
Figure imgf000137_0003
; -S02NR5R5 or phenyl substituted with 1 to 3 groups independently selected from -NR5C(0)NR5R7, halo, Ci-C4alkyl, and -CN; R3 is H;
X is C(R5)2, O or NR5;
L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q-, -C(O)-, -C(0)NR50(CR6R6)q-, -C(0)0-,
-C(0)-C1-C8alkylene or -C(0)-C2-C8alkenylene ;
L2 is -NR5C(0)(CR6R6)q-;
each L is independently selected from a CrCgalkylene optionally substituted with 1 to 3 substituents independently selected from C Cealkyl;
R4 is selected from H, R15, -N(R5)2, CrC6alkyl, CrC6alkyl substituted with 1 to 4 hydroxyl groups, Ci-Cgalkyl substituted with 1 to 6 groups independently selected from halo, C - C4alkyl, and -R , phenyl, C3-Cgcycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms,
or R4 is selected from phenyl, C3-Cgcycloalkyl, a 4-6 membered heterocycloalkyl
containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms each of which is substituted with 1 to 3 substituents independently selected from halo, C Cealkyl, C C6alkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R8, -OR6, -C(0)R6, -C(0)OR6, -C(0)NR6R6, - C(0)L3R8, -S(0)2R6, and -S(0)2NR6R6;
each R5 is independently selected from H or C Cealkyl;
each R6 is independently selected from H, Q-CeaLkyl, and C^Cealkyl substituted with 1 to 4 hydroxyl groups;
R is phenyl or phenyl substituted with 1 to 3 groups independently selected from halo,
Ci-C4alkyl, Ci-C4haloalkyl and -R8;
R8 is selected from -OC(0)R6, -NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6,
-NHS(0)2R6, -S(0)2NR6R6, -C(0)OR6 and -OR6;
R9 is CrC^aloalkyl or -OR5;
each R10 is independently selected from halo, C Cealkyl, CrCehaloalkyl, -C(=N)OR5, -CN, -(CR6R6)qCN, -NR5R5, -(CR5R5)NR5R5, -C(0)OR6, -C(0)NR5R5, -(CR6R6)qR6, -C(0)R14, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, -S(0)2NR6R6, -S(0)2R6, -OR6, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S and 5 membered heteroaryl containing 1 to 4 N heteroatoms;
each R11 is independently selected from halo, -OR5, -CN, deuterium, C Cealkyl, hydroxyl substitutedQ-Cealkyl, Q-Cehalolkyl, and n is 0, 1 or 2;
or each R11 is deuterium, and n is 0, 1, 2, 3, 4, 5, 6 or 7;
R 12 is H, C C6alkyl, phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, CrC4alkyl, and -R ;
R13 is halo, -OC(0)R12, -OR12, -CN, -NHC(0)OR12, -NHC(0)R12 or -NR5R5;
R14 is a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, or a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from
N, O and S substituted with 1 to 4 substituents independently selected from halo, -OR6,
Ci-C6alkyl and Ci-C6haloalkyl; R15 is cyclohexyl having a C Cealkyl bridge, T o ,
Figure imgf000139_0001
or
Figure imgf000139_0002
erein each is optionally substituted with 1-3 substituents independently selected from halo, -CN, C Cealkyl, -R14, and -OR5;
R
Figure imgf000139_0003
of which is
Figure imgf000139_0004
su sttute wt to su sttuents n epen enty seecte rom ;
p is 1 or 2 and
q is 1, 2, 3, 4, 5 or 6,
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1 wherein
Figure imgf000139_0005
R1 is H, -C(0)NH2, -LXR4, -L2R13, -C(0)OR5-, -C(0)NHOR5, -, -C(0)NHC(0)NR5R5,
/OR6
C(0)NR5OR14, -C(0)NHNH2, -NR5C(0)NR5R9, % I"NH2; _S02NR5R5 or phenyl substituted with 1 to 3 groups independently selected from -NR 5 C(0)NR 5 R 7 , halo, Cr C4alkyl, and -CN;
R3 is H;
X is C(R5)2, O or NR5;
L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q-, -C(O)-, -C(0)NR50(CR6R6)q-, -C(0)0-,
C(0)-C1-C8alkylene or -C(0)-C2-C8alkenylene ;
L2 is -NR5C(0)(CR6R6)q-;
each L is independently selected from a CrCgalkylene optionally substituted with 1 to 3 substituents independently selected from C C6alkyl;
R4 is selected from H, R15, -N(R5)2, Q-Cealkyl, CrCealkyl substituted with 1 to 4 hydroxyl groups, Ci-Cgalkyl substituted with 1 to 6 groups independently selected from halo, C - C4alkyl, and -R , phenyl, C3-Cgcycloalkyl, a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms,
or R4 is selected from phenyl, Cs-Cscycloalkyl, a 4-6 membered heterocycloalkyl
containing 1 to 2 heteroatoms selected from N, O and S, and a 5, 6, 9 or 10 membered heteroaryl containing 1 to 3 N heteroatoms each of which is substituted with 1 to 3 substituents independently selected from halo, C C6alkyl, C C6alkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R8, -OR6, -C(0)R6, -C(0)OR6, -C(0)NR6R6, - C(0)L3R8, -S(0)2R6, and -S(0)2NR6R6;
each R5 is independently selected from H or C C6alkyl;
each R6 is independently selected from H, C C6alkyl, and C C6alkyl substituted with 1 to 4 hydroxyl groups;
R is phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, C -
C4alkyl, Ci-C4haloalkyl and -R8;
R8 is selected from -OC(0)R6, -NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6,
-NHS(0)2R6, -S(0)2NR6R6, -C(0)OR6 and -OR6;
R9 is CrC^aloalkyl or -OR5;
each R10 is independently selected from halo, C C6alkyl, CrCehaloalkyl, -C(=N)OR5, - CN, -(CR6R6)qCN, -NR5R5, -C(0)OR6, -C(0)NR5R5, -(CR6R6)qR6, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, -S(0)2NR6R6, -S(0)2R6, -OR6, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S and 5 membered heteroaryl containing 1 to 4 N heteroatoms;
each R11 is independently selected from halo, -OR5, deuterium, Q-Cealkyl, hydroxyl
substitutedQ-Cealkyl, C Cehalolkyl, and n is 0, 1 or 2;
or each R11 is deuterium, and n is 0, 1, 2, 3, 4, 5, 6 or 7;
R 12 is H, C C6alkyl, phenyl or phenyl substituted with 1 to 3 groups independently selected from halo, C1-C4alkyl, and -R ;
R13 is halo, -OC(0)R12, -OR12, -CN, -NHC(0)OR12, -NHC(0)R12 or -NR5R5;
R14 is a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, or a 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from
N, O and S substituted with 1 to 4 substituents independently selected from halo, -OR6,
Ci-C6alkyl and Ci-C6haloalkyl;
R is cyclohexyl having a C C6alkyl
Figure imgf000141_0001
or
Figure imgf000141_0002
wherein each is optionally substituted with 1-3 substituents independently selected from halo, -CN, CrCealk l, -R14, and -OR5;
R
Figure imgf000141_0003
of which is substituted with 1 to 3 substituents independently selected from R10,
or R is selected from
Figure imgf000141_0004
, each of which is optionally substituted with 1 to 3 substituents independently selected from R10;
p is 1 or 2 and
q is 1, 2, 3, 4, 5 or 6,
a pharmaceutically acceptable salt thereof. The compound of claim 1, wherein the compound of Formula (I) is a compound having the structure of Formula (Ta):
Figure imgf000142_0001
Formula (Ta).
The compound of claim 1, wherein the compound of Formula (I) is a compound having the structure of Formula (I a):
Figure imgf000142_0002
Formula (I a).
The compound of claim 1, wherein the compound of Formula (I) is a compound having the structure of Formula (III- a):
Figure imgf000142_0003
Formula (III- a).
The compound of claim 3, wherein the compound of Formula (I-a) is a compound having the structure of Formula (Tb):
Figure imgf000142_0004
Formula (I-b)
wherein:
m is 1, 2 or 3.
The compound of claim 4, wherein the compound of Formula (II-a) is a compound having the structure of Formula (II-b):
Figure imgf000143_0001
Formula (II-b)
wherein:
m is 1, 2 or 3.
8. The compound of claim 5, wherein the compound of Formula (Ill-a) is a compound having the structure of Formula (Ill-b):
Figure imgf000143_0002
Formula (Ill-b)
wherein:
m is 1, 2 or 3.
9. The compound of any one of claims 1 to 8, wherein R1 is -C(0)NH2.
10. The compound of any one of claims 1 to 8, wherein R1 is -LXR4.
11. The compound of claim 10, wherein L1 is -C(0)NR5-, -C(0)NR5(CR6R6)q- or - C(0)NR50(CR6R6)q-.
12. The compound of claim 10, wherein L1 is -C(O)- or -C(0)0-.
13. The compound of claim 10, wherein L1 is -C(0)-C1-Cgalkylene or -C(0)-C2-Cgalkenylene.
14. The compound of any one of claims 1-8 or 10-13, wherein R4 is phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl.
15. The compound of any one of claims 1-8 or 10-13, wherein R4 is phenyl, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl, each of which is substituted with 1 to 3 substituents independently selected from halo, C Cealkyl, C C6alkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R14, -OR6, -C(0)R6, -C(0)OR6, -C(0)L3R6, -S(0)2R6, and -S(0)2NR6R6.
16. The compound of any one of claims 1-8 or 10-13, wherein R4 is tetrahydro-2H-pyranyl, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, morpholinyl, oxetanyl, tetrahydro-2H- thiopyranyl, azetidinyl, piperazinyl, pyridyl, pyrazolyl, benzthiazolyl or pyrrolyl.
17. The compound of any one of claims 1-8 or 10-13, wherein R4 is tetrahydro-2H-pyranyl, tetrahydrofuranyl, piperidinyl, pyrrolidinyl, morpholinyl, oxetanyl, tetrahydro-2H- thiopyranyl, azetidinyl, piperazinyl, pyridyl, pyrazolyl, benzthiazolyl or pyrrolyl, each of which is substituted with 1 to 3 substituents independently selected from halo, Q-Cealkyl, Q-Cealkyl substituted with 1 to 4 hydroxyl groups, 4-6 membered heterocycloalkyl containing 1 to 2 heteroatoms selected from N, O and S, -CN, -R14, -OR6, -C(0)R6, -
C(0)OR6, -C(0)L3R6, -S(0)2R6, and -S(0)2NR6R6.
18. The compound of any one of claims 1 to 8, wherein R1 is -C(0)NHOR5, -C(0)NR5OR14,
-C(0)NHNH2, -NR5C(0)NR5R9 or
Figure imgf000144_0001
.
19. The compound of any one of claims 1-8 or 16-18, wherein R14 is pyrrolidinyl, tetrahydro-2H- pyranyl, morpholinyl or piperidinyl, each of which is optionally substituted with 1 to 2 groups.
The compound of any one of claims 1-8 or 10-13, wherein R4 is R15, -N(R5)2, methyl, ethyl, propyl, isopropyl, isobutene, t-butyl, or CrCgalkyl substituted with 1 to 6 groups
independently selected from halo, C1-C4alkyl, and -R .
21.
Figure imgf000144_0002
substituents independently selected from C Cealkyl.
22. The compound of any one of claims 1 to 8, wherein R 1 is -L 2 R 13.
23. The compound of any one of claims 1 to 8, wherein R1 is -Br, -CN and -S02NR5R5.
24. The compound of any one of claims 1-23, wherein each R10 is independently selected from F, CI, methyl, -CF3), -C(=N)OR5, -CN, -(CR6R6)qCN, -NR5R5, -C(0)OR6, -C(0)NR5R5, - (CR5R5)qR8, - NR5C(0)NR5R5, - NR5S(0)2R6, -NR5S(0)2NR6R6, -S(0)2NR6R6, -S(0)2R6, - OR6, morpholinyl and tetrazolyl.
25. The compound of any one of claims 1-24, wherein each R is independently selected from F, deuterium and methyl, and n is 1 or 2.
26. The compound of any one of claims 1-25, wherein each R11 is independently selected from deuterium, and n is 1, 2, 3, 4, 5, 6 or 7.
27. The compound of any one of claims 1-26, wherein R8 is selected from -OC(0)R6, - NHC(0)OR6, -NR5R5, -C(0)N(R5R5), -S(0)2R6, -S(0)2NR6R6, -C(0)OR6 and -OR6, and wherein each R6 is independently selected from H, methyl, ethyl, propyl, isopropyl, t-butyl or a Ci-Cealkyl substituted with 1 to 4 hydroxyl groups.
28. The compound of any one of claims 1-27, wherein R5 is H, methyl or ethyl.
29. The compound of any one of claims 1-28, wherein q is 1, 2 or 3.
30. The compound of claim 1 selected from:
ethyl 5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate;
ethyl 5-[2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate;
ethyl 5-[(2R)-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate;
ethyl 5-[5-(3-fluorophenyl)-3-methyl-4,5-dihydro-lH-pyrazol-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate;
ethyl 5-[2-(3-fluorophenyl)-5-oxopyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate; ethyl 5-[5-(3-fluorophenyl)-2,2,3,3,4,4-hexadeuteriumpyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate;
ethyl 5-[2-(3-fluorophenyl)-2,3, 3,4,4,5, 5-heptadeuteriumpyrrolidin-l-yl]pyrazolo[ 1,5- a]pyrimidine-3-carboxylate;
ethyl 5-[2-(3-fluorophenyl)-2-deuteriumopyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate;
ethyl 5-[2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylate;
ethyl 5-[2-(3-fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxylate;
5-[(2R)-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-2-(3-fluorophenyl)pyrrolidin-l-yl]-N,N-dimethylpyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-hydroxyethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-sulfamoylethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
2-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl }formamido)acetamide;
N-(l,l-dioxo-thiolan-3-yl)-5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide;
tert-butyl 4- { 5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin- 1 -yl]pyrazolo[ 1 ,5- a]pyrimidine-3-amido Jpiperidine- 1 -carboxylate;
tert-butyl N-[2-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)p irolidin-l-yl]p razolo[l,5- a]pyrimidin-3-yl}formamido)ethyl]carbamate;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(l-methanesulfonylpiperidin-4- yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2- methanesulfonamidoethyl)pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(piperidin-4-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
N-(2-aminoethyl)-5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin- 1 -yl] -N- [ 1 -(2-hydroxyacetyl)piperidin-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[(2S)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide; 5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-ethylpyrazolo[l,5-a]pyrimidine- 3-carboxamide;
5-[5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[(5R)-5-(3-fluorophenyl)-2,2-dimethylpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[(lR,2S,5S)-2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5-a]pyrimidine- 3-carboxamide;
5-[(lR,2R,5S)-2-(3-fluorophenyl)-3-azabicyclo[3.1.0]hexan-3-yl]pyrazolo[l,5-a]pyrimidine- 3-carboxamide;
5-[5-(3-fluorophenyl)-2,2-dideuteriumpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-[2-(3-fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[(2R)-2-(3-fluorophenyl)piperidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
(2R)-2-(3-fluorophenyl)-l-{pyrazolo[l,5-a]pyrimidin-5-yl}pyrrolidine;
5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-ethoxypyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxylic acid;
5- [(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin- 1 -yl] -N- [ 1 -
(hydroxymethyl)cyclopropyl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
ethyl l-{5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-
3-amido Jcyclopropane- 1 -carboxylate;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(3-methyloxetan-3- yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-methoxyethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-hydroxy-2- methylpropyl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide;
{5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yljcarbonylurea; l-({5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl}carbonyl)-3-(propan-2-yl)urea;
l-{5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- amido} cyclopropane- 1-carboxylic acid;
5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-phenylpyrazolo[l,5-a]pyrimidine- 3-carboxamide;
[l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl}carbonyl)azetidin-3-yl]methanol;
(3R)-l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl }carbonyl)pyrrolidin-3-ol;
l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl }carbonyl)piperidine;
N-(l-carbamoylcyclopropyl)-5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l- yl]pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide;
(3S)-l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl }carbonyl)pyrrolidin-3-ol;
l-({5-[(2R,4S)-4-fluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl}carbonyl)-3-methylazetidin-3-ol;
5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]-N-(2-hydroxyethyl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
ethyl 5-[(2R)-4-fluoro-2-(3-fluorophenyl)-4-deuteriumpyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate;
ethyl 5- [(2R,4S)-4-fluoro-2-(3-methyl- 1 ,2,4-oxadiazol-5-yl)pyrrolidin- 1 -yl]pyrazolo[ 1 ,5- a]pyrimidine-3-carboxylate;
5-[(2R)-4-fluoro-2-(3-fluorophenyl)-4-deuteriumpyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine- 3-carboxamide;
methyl N-({5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidin-3-yl}carbonyl)carbamate;
5-(4,4-difluoro-2-{5-fluoro-2-[(propan-2-yl)carbamoyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-[2-(3-cyano-5-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R,4S)-4-cyano-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3- carboxamide;
{5-[(2R)-4,4-difluoro-2-(3-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidin-3- yl } [(ethoxycarbonyl)oxy]methanone;
(2R)-l-{3-bromopyrazolo[l,5-a]pyrimidin-5-yl}-4,4-difluoro-2-(3-fluorophenyl)pyrrolidine;
5-[2-(2-cyanophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[2-(2-chlorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-(2-{5-fluoro-2-[(propan-2-yl)carbamoyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-(2-{5-fluoro-2-[(morpholin-4-yl)carbonyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-{2-[2-(dimethylcarbamoyl)-5-fluorophenyl]pyrrolidin-l-yl}pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-{2-[5-fluoro-2-(trifluoromethyl)phenyl]pyrrolidin-l-yl}pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-{2-[2-(trifluoromethyl)phenyl]pyrrolidin-l-yl}pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-{2-[3-(aminomethyl)-5-fluorophenyl]pyrrolidin-l-yl}pyrazolo[l,5-a]pyrimidine-3- carboxamide;
5-(2-{3-fluoro-5-[(propan-2-ylamino)methyl]phenyl}pyrrolidin-l-yl)pyrazolo[l,5- a]pyrimidine-3-carboxamide;
5-[2-(5-cyano-2-fluorophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide; 5-[(2R)-2-(3-cyanophenyl)pyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[(2R,4S)-2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine- 3-carboxamide;
5-(2-phenylpyrrolidin-l-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide;
5-[(2R)-2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidin-l-yl]pyrazolo[l,5-a]pyrimidine-
3-carboxamide;
ethyl 5-[(2R)-2-(3-cyano-5-fluorophenyl)-4,4-difluoropyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate;
ethyl 5-[(2R,4S)-2-(3-cyano-5-fluorophenyl)-4-fluoropyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxylate; 5-[(2R,4S)-2-(3-carbamoyl-5-fluorophenyl)-4-fluoropyrrolidin-l-yl]pyrazolo[l,5- a]pyrimidine-3-carboxamide, and
5-[(2R)-4,4-difluoro-2-{5-fluoro-2-[(propan-2-yl)carbamoyl]phenyl}pyrrolidin-l- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide.
31. A pharmaceutical composition comprising a therapeutically effective amount of a compound of any one of claims 1-30 and a pharmaceutically acceptable carrier.
32. Use of a compound of any one of claims 1-30 in the manufacture of a medicament for treating a TRK kinase-mediated disease or condition, wherein the disease or condition is papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis or asthma.
33. A method for inhibiting a TRK kinase comprising administering to a system or a subject in need thereof, a therapeutically effective amount of a compound any one of claims 1-30, or pharmaceutically acceptable salts or pharmaceutical compositions thereof.
34. A method for treating a TRK kinase-mediated disease or condition, comprising
administering to a system or subject in need of such treatment an effective amount of a compound of any one of claims 1-30, or pharmaceutically acceptable salts or pharmaceutical compositions thereof, wherein the disease or condition is papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis or asthma.
35. A compound for use in a method of medical treatment, wherein the method of
medical treatment is for treating a disease selected from papillary thyroid carcinoma, pancreatic cancer, colon cancer, breast carcinoma, neuroblastoma, pain, cachexia, dermatitis and asthma, and wherein the compound is a compound of any one of claims 1-30.
PCT/US2011/0511152010-09-092011-09-09Compounds and compositions as trk inhibitorsWO2012034095A1 (en)

Applications Claiming Priority (2)

Application NumberPriority DateFiling DateTitle
US38139010P2010-09-092010-09-09
US61/381,3902010-09-09

Publications (1)

Publication NumberPublication Date
WO2012034095A1true WO2012034095A1 (en)2012-03-15

Family

ID=44654516

Family Applications (1)

Application NumberTitlePriority DateFiling Date
PCT/US2011/051115WO2012034095A1 (en)2010-09-092011-09-09Compounds and compositions as trk inhibitors

Country Status (1)

CountryLink
WO (1)WO2012034095A1 (en)

Cited By (65)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US8791123B2 (en)2009-07-092014-07-29Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US8933084B2 (en)2010-05-202015-01-13Array Biopharma Inc.Macrocyclic compounds as Trk kinase inhibitors
WO2015017533A1 (en)2013-07-302015-02-05Blueprint Medicines CorporationNtrk2 fusions
US9127013B2 (en)2008-10-222015-09-08Array Biopharma, Inc.Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US9227975B2 (en)2008-09-222016-01-05Array Biopharma, Inc.Method of treatment using substituted imidazo[1,2B]pyridazine compounds
WO2016097869A1 (en)2014-12-152016-06-23Cmg Pharmaceutical Co., Ltd.Fused ring heteroaryl compounds and their use as trk inhibitors
WO2017011776A1 (en)2015-07-162017-01-19Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JP2017507980A (en)*2014-03-172017-03-23レクシコン ファーマシューティカルズ インコーポレイテッド Inhibitor of adapter-related kinase 1, composition containing the same, and method of use thereof
WO2017075107A1 (en)2015-10-262017-05-04Nanda NishaPoint mutations in trk inhibitor-resistant cancer and methods relating to the same
WO2017087778A1 (en)2015-11-192017-05-26Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to ntrk
US9782414B2 (en)2014-11-162017-10-10Array Biopharma, Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
WO2017176751A1 (en)2016-04-042017-10-12Loxo Oncology, Inc.Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2017176744A1 (en)2016-04-042017-10-12Loxo Oncology, Inc.Methods of treating pediatric cancers
US9840510B1 (en)2016-04-062017-12-12Lysosomal Therapeutics Inc.Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US9868742B2 (en)2016-05-052018-01-16Lysosomal Therapeutics Inc.Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b] pyridazines, related compounds, and their use in the treatment of medical disorders
US9920061B2 (en)2016-04-062018-03-20Lysosomal Therapeutics Inc.Imidazo[1,5-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
WO2018071447A1 (en)2016-10-102018-04-19Andrews Steven WSubstituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
WO2018071454A1 (en)2016-10-102018-04-19Andrews Steven WSubstituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
CN108003161A (en)*2016-10-282018-05-08正大天晴药业集团股份有限公司Neurotrophic factor tyrosine kinase receptor inhibitor
US10017512B2 (en)2015-08-262018-07-10Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to NTRK
WO2018136663A1 (en)2017-01-182018-07-26Array Biopharma, Inc.Ret inhibitors
WO2018136661A1 (en)2017-01-182018-07-26Andrews Steven WSUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
US10045991B2 (en)2016-04-042018-08-14Loxo Oncology, Inc.Methods of treating pediatric cancers
WO2018170381A1 (en)2017-03-162018-09-20Andrews Steven WMacrocyclic compounds as ros1 kinase inhibitors
US10246466B2 (en)2014-01-242019-04-02Tp Therapeutics, Inc.Diaryl macrocycles as modulators of protein kinases
WO2019075114A1 (en)2017-10-102019-04-18Mark ReynoldsFormulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019075108A1 (en)2017-10-102019-04-18Metcalf Andrew TCrystalline forms
WO2019084285A1 (en)2017-10-262019-05-02Qian ZhaoFormulations of a macrocyclic trk kinase inhibitor
US10316044B2 (en)2015-07-022019-06-11Tp Therapeutics, Inc.Chiral diaryl macrocycles as modulators of protein kinases
KR20190067913A (en)*2016-10-282019-06-17치아타이 티안큉 파마수티컬 그룹 주식회사 Aminopyrazolopyrimidine compounds used as inhibitors of tyrosine kinase receptors which are neurotrophic factors
WO2019143994A1 (en)2018-01-182019-07-25Array Biopharma Inc.Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
WO2019143977A1 (en)2018-01-182019-07-25Array Biopharma Inc.Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
WO2019174598A1 (en)2018-03-142019-09-19Fochon Pharmaceuticals, Ltd.SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
CN110294761A (en)*2018-06-082019-10-01南京雷正医药科技有限公司Substituted pyrazolo [1,5-a] pyrimidine compound as Trk kinase inhibitor
WO2019191659A1 (en)2018-03-292019-10-03Loxo Oncology, Inc.Treatment of trk-associated cancers
WO2019233461A1 (en)*2018-06-082019-12-12江苏威凯尔医药科技有限公司Tropomyosin receptor kinase inhibitor, preparation method therefor and use thereof
CN110577532A (en)*2018-06-082019-12-17江苏威凯尔医药科技有限公司Tropomyosin receptor kinase inhibitor and preparation method and application thereof
WO2020028258A1 (en)2018-07-312020-02-06Loxo Oncology, Inc.Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
CN110804059A (en)*2019-09-302020-02-18郑州泰基鸿诺医药股份有限公司Carbamate compound, pharmaceutical composition and application thereof
US10570135B2 (en)2014-11-062020-02-25Lysosomal Therapeutics Inc.Substituted pyrazolo[1,5-A]pyrimidines and their use in the treatment of medical disorders
WO2020055672A1 (en)2018-09-102020-03-19Array Biopharma Inc.Fused heterocyclic compounds as ret kinase inhibitors
CN111138437A (en)*2019-12-042020-05-12杭州华东医药集团新药研究院有限公司Substituted pyrazolo [1,5-a ] pyrimidine amino acid derivatives and uses thereof
WO2020131627A1 (en)2018-12-192020-06-25Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
WO2020131674A1 (en)2018-12-192020-06-25Array Biopharma Inc.7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
US10751341B2 (en)2014-11-062020-08-25Lysosomal Therapeutics Inc.Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US10786508B2 (en)2014-11-062020-09-29Lysosomal Therapeutics Inc.Substituted imidazo[1,5-A]-pyrimidines and their use in the treatment of medical disorders
CN111718351A (en)*2019-03-192020-09-29华中师范大学 Oxygen-substituted pyrazolopyrimidine compound and pharmaceutical composition and application thereof
EP3722441A1 (en)2015-06-012020-10-14Loxo Oncology Inc.Method of diagnosing a cancer for a treatment with a trk inhibitor
CN111819179A (en)*2017-12-152020-10-23金字塔生物科技公司5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (1H-pyrazol-1-yl) pyrazolo [1,5-a ] pyrimidine derivatives and related compounds as TRK kinase inhibitors for the treatment of cancer
JP2021502413A (en)*2017-11-102021-01-28エンジェクス ファーマシューティカル インコーポレイテッド Macrocycles and their use as TRK kinase inhibitors
WO2021047584A1 (en)*2019-09-112021-03-18Fochon Pharmaceuticals, Ltd.SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
US11091486B2 (en)2016-10-262021-08-17Array Biopharma, IncProcess for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
CN113278022A (en)*2020-07-072021-08-20南京纳丁菲医药科技有限公司Pyrazolopyrimidine compounds, pharmaceutical compositions and uses thereof
US11124516B2 (en)2016-04-062021-09-21BIAL-BioTech Investments, Inc.Pyrrolo[1,2-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
WO2021228248A1 (en)2020-05-152021-11-18石药集团中奇制药技术(石家庄)有限公司Fused aza-heterocyclic amide compound and use thereof
US11214571B2 (en)2016-05-182022-01-04Array Biopharma Inc.Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US11345698B2 (en)2016-05-052022-05-31Bial—R&D Investments, S.A.Substituted imidazo[1,2-a]pyridines, substituted imidazo[1,2-a]pyrazines, related compounds, and their use in the treatment of medical disorders
RU2781618C2 (en)*2018-03-142022-10-14Фочон Байосайенсис, Лтд.Substituted (2-azabicyclo[3.1.0]hexane-2-yl)pyrazolo[1.5-a]pyrimidine and imidazo[1.2-b]pyridazine compounds as trk kinase inhibitors
US11524963B2 (en)2018-01-182022-12-13Array Biopharma Inc.Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
WO2022262671A1 (en)*2021-06-152022-12-22中国医药研究开发中心有限公司Macro heterocyclic compound and medical use thereof
CN116120323A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司Solid form aza-condensed ring amide compound and use thereof
CN116120322A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司Salt of aza-condensed ring amide compound, crystal form and use thereof
CN116115618A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司 a drug for treating tumors
EP2914622B1 (en)*2012-11-052023-06-07Foundation Medicine, Inc.Novel fusion molecules and uses thereof
US12187739B2 (en)2015-07-062025-01-07Turning Point Therapeutics, Inc.Diaryl macrocycle polymorph

Citations (186)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US3714357A (en)1968-07-151973-01-30Rech D Applic Scient Sogeras SPharmaceutical compositions comprising quinuclidinol derivatives
EP0147719A2 (en)1983-12-241985-07-10Tanabe Seiyaku Co., Ltd.Novel carbostyril derivative and process for preparing same
EP0409595A2 (en)1989-07-191991-01-23Glaxo Group LimitedProcess for the preparation of a carbocyclic nucleoside analogue
EP0424021A1 (en)1989-10-191991-04-24Pfizer LimitedAntimuscarinic bronchodilators
WO1992019594A1 (en)1991-05-021992-11-12Smithkline Beecham CorporationPyrrolidinones
JPH0525045B2 (en)1985-08-091993-04-09Noritoshi Nakabachi
WO1993018007A1 (en)1992-03-131993-09-16Tokyo Tanabe Company LimitedNovel carbostyril derivative
WO1993019750A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating allergic or inflammatory diseases
WO1993019749A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating allergic and inflammatory diseases
WO1993019751A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating inflammatory diseases and inhibiting production of tumor necrosis factor
WO1994017090A1 (en)1993-01-201994-08-04Glaxo Group Limited2,6-diaminopurine derivatives
US5451700A (en)1991-06-111995-09-19Ciba-Geigy CorporationAmidino compounds, their manufacture and methods of treatment
WO1996002543A1 (en)1994-07-141996-02-01Glaxo Group Limited2,3-dihydroxy cyclopentane derivatives of purines
WO1996002553A2 (en)1994-07-141996-02-01Glaxo Group LimitedAMINO PURINE-β-D-RIBOFURANURONAMIDE DERIVATIVES
WO1998018796A1 (en)1996-10-281998-05-07Novartis AgNaphthyridine derivatives
WO1998028319A1 (en)1996-12-241998-07-02Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999016766A1 (en)1997-10-011999-04-08Kyowa Hakko Kogyo Co., Ltd.Benzodioxole derivatives
WO1999024450A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdensine a1 receptor agonists
WO1999024451A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdenosine a1 receptor agonists
WO1999024449A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdenosine a1 receptor agonists
WO1999038877A2 (en)1998-01-311999-08-05Glaxo Group Limited2-(PURIN-9-yl)-TETRAHYDROFURAN-3,4-DIOL DERIVATIVES
WO1999041267A1 (en)1998-02-141999-08-19Glaxo Group Limited2-(purin-9-yl) -tetrahydrofuran-3, 4-diol derivatives
WO1999064035A1 (en)1998-06-081999-12-16Advanced Medicine, Inc.β2-ADRENERGIC RECEPTOR AGONISTS
WO1999067266A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067265A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067263A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067264A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO2000000531A1 (en)1998-06-302000-01-06The Dow Chemical CompanyPolymer polyols and a process for the production thereof
WO2000023457A1 (en)1998-10-162000-04-27Pfizer LimitedAdenine derivatives
EP1052264A2 (en)1999-05-112000-11-15Pfizer Products Inc.Process for the synthesis of nucleoside analogs
WO2000075114A1 (en)1999-06-042000-12-14Novartis AgBeta2-adrenoceptor agonists
WO2000077018A2 (en)1999-06-152000-12-21Pfizer LimitedPurine derivatives
WO2000078774A2 (en)1999-06-182000-12-28University Of Virginia Patent FoundationInduction of pharmacological stress with adenosine receptor agonists
WO2001004118A2 (en)1999-07-142001-01-18Almirall Prodesfarma S.A.Quinuclidine derivatives and their use as muscarinic m3 receptor ligands
WO2001013953A2 (en)1999-08-212001-03-01Byk Gulden Lomberg Chemische Fabrik GmbhSynergistic combination of pde inhibitors and beta 2 adrenoceptor agonist
WO2001023399A1 (en)1999-09-302001-04-05Pfizer Products Inc.Compounds for the treatment of ischemia
WO2001027130A1 (en)1999-10-142001-04-19Pfizer LimitedPurine derivatives
WO2001027131A1 (en)1999-10-142001-04-19Pfizer LimitedPurine derivatives
WO2001042193A1 (en)1999-12-082001-06-14Theravance, Inc.β2-ADRENERGIC RECEPTOR AGONISTS
WO2001060835A1 (en)2000-02-182001-08-23Pfizer LimitedPurine derivatives
WO2001083462A1 (en)2000-04-272001-11-08Boehringer Ingelheim Pharma KgNovel, slow-acting betamimetics, a method for their production and their use as medicaments
WO2001094368A1 (en)2000-06-062001-12-13Pfizer Limited2-aminocarbonyl-9h-purine derivatives
WO2002000679A2 (en)2000-06-282002-01-03Novartis Ag9.alpha.-chloro-6.alpha.-fluoro-17.alpha.-hydroxy-16-methyl-17-beta-methoxycarbonyl-androst-1,4-dienes esterified in position 17.alpha. by a cyclic acyl group
WO2002000652A1 (en)2000-06-272002-01-03Laboratorios S.A.L.V.A.T., S.A.Carbamates derived from arylalkylamines
WO2002000676A1 (en)2000-06-272002-01-03Pfizer LimitedPurine derivatives
WO2002010143A1 (en)2000-07-282002-02-07Schering AktiengesellschaftNon-steroidal inflammation inhibitors
WO2002012266A1 (en)2000-08-052002-02-14Glaxo Group Limited17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
WO2002022630A1 (en)2000-09-152002-03-21Pfizer LimitedPurine derivatives
US20020055651A1 (en)1999-06-022002-05-09Moran Edmund J.Beta2-adrenergic receptor agonists
WO2002042298A1 (en)2000-11-212002-05-30Novartis AgAminothiazoles and their use as adenosine receptor antagonists
WO2002051841A1 (en)2000-12-222002-07-04Almirall Prodesfarma AgQuinuclidine carbamate derivatives and their use as m3 antagonists
WO2002053564A2 (en)2000-12-282002-07-11Almirall Prodesfarma AgQuinuclidine derivatives and their use as m3 antagonists
WO2002066422A1 (en)2001-02-142002-08-29Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2002070490A1 (en)2001-03-082002-09-12Glaxo Group LimitedAgonists of beta-adrenoceptors
EP1241176A1 (en)2001-03-162002-09-18Pfizer Products Inc.Purine derivatives for the treatment of ischemia
WO2002076933A1 (en)2001-03-222002-10-03Glaxo Group LimitedFormailide derivatives as beta2-adrenoreceptor agonists
WO2002088167A1 (en)2001-04-302002-11-07Glaxo Group LimitedAnti-inflammatory 17.beta.-carbothioate ester derivatives of androstane with a cyclic ester group in position 17.alpha
WO2002096462A1 (en)2001-05-252002-12-05Pfizer Inc.An adenosine a2a receptor agonist and an anticholinergic agent in combination for treating obstructive airways diseases
WO2002100879A1 (en)2001-06-122002-12-19Glaxo Group LimitedNovel anti-inflammatory 17.alpha.-heterocyclic-esters of 17.beta.carbothioate androstane derivatives
WO2003000840A2 (en)2001-06-212003-01-03Diversa CorporationNitrilases
WO2003024439A1 (en)2001-09-142003-03-27Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2003033495A1 (en)2001-10-172003-04-24Ucb, S.A.Quinuclidine derivatives, processes for preparing them and their uses as m2 and/or m3 muscarinic receptor inhibitors
WO2003035668A2 (en)2001-10-202003-05-01Glaxo Group LimitedNovel anti-inflammatory androstane derivatives -17-carboxy-lactone substituted steroids with an aryl-carboxylic ester in position 17.alpha
WO2003039544A1 (en)2001-11-052003-05-15Novartis AgNaphtyridine derivatives, their preparation and their use as phosphodiesterase isoenzyme 4 (pde4) inhibitors
WO2003042164A1 (en)2001-11-132003-05-22Theravance, IncAryl aniline beta-2 adrenergic receptor agonists
WO2003042160A1 (en)2001-11-132003-05-22Theravance, Inc.Aryl aniline beta-2 adrenergic receptor agonists
WO2003042214A2 (en)2001-11-092003-05-22Cv Therapeutics, Inc.A2b adenosine receptor antagonists
WO2003048181A1 (en)2001-12-012003-06-12Glaxo Group Limited17.alpha. -cyclic esters of 16-methylpregnan-3,20-dione as anti-inflammatory agents
WO2003053966A2 (en)2001-12-202003-07-03Laboratorios S.A.L.V.A.T., S.A.1-alkyl-1-azoniabicyclo [2.2.2] octane carbamate derivatives and their use as muscarinic receptor antagonists
WO2003062259A2 (en)2002-01-212003-07-31Glaxo Group LimitedNon-aromatic 17.alpha.-esters of androstane-17.beta.-carboxylate esters as anti-inflammatory agents
WO2003064445A1 (en)2002-01-312003-08-07Glaxo Group Limited17-alpha-heterocyclic esters of androstane derivatives having anti-inflammatory activity
WO2003072592A1 (en)2002-01-152003-09-04Glaxo Group Limited17.alpha-cycloalkyl/cycloylkenyl esters of alkyl-or haloalkyl-androst-4-en-3-on-11.beta.,17.alpha.-diol 17.beta.-carboxylates as anti-inflammatory agents
WO2003072539A1 (en)2002-02-282003-09-04Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2003082787A1 (en)2002-03-262003-10-09Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003082280A1 (en)2002-03-262003-10-09Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003086294A2 (en)2002-04-112003-10-23Merck & Co., Inc.1h-benzo[f]indazol-5-yl derivatives as selective glucocorticoid receptor modulators
WO2003086408A1 (en)2002-04-102003-10-23University Of Virginia Patent FoundationUse of a2a adenosine receptor agonists for the treatment of inflammatory diseases
WO2003087094A2 (en)2002-04-162003-10-23Almirall Prodesfarma AgPyrrolidinium derivatives as antagonists of m3 muscarinic receptors
WO2003091204A1 (en)2002-04-252003-11-06Glaxo Group LimitedPhenethanolamine derivatives
WO2003099807A1 (en)2002-05-292003-12-04Almirall Prodesfarma S.A.New indolylpiperidine derivatives as potent antihistaminic and antiallergic agents
WO2003099764A1 (en)2002-05-282003-12-04Theravance, Inc.ALKOXY ARYL β2 ADRENERGIC RECEPTOR AGONISTS
WO2003101932A2 (en)2002-05-292003-12-11Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003104204A1 (en)2002-06-052003-12-18Merck Patent GmbhPyridazine derivatives
WO2003104195A1 (en)2002-06-062003-12-18Boehringer Ingelheim Pharmaceuticals, Inc.4-(aryl or heteroaryl) -2-butylamine derivatives and their use as glucocorticoid ligans
WO2003104205A1 (en)2002-06-102003-12-18Merck Patent GmbhAryloximes
WO2004000814A1 (en)2002-06-252003-12-31Merck Frosst Canada & Co.8-(biaryl) quinoline pde4 inhibitors
WO2004000839A1 (en)2002-06-192003-12-31Merck Patent GmbhThiazole derivatives as phosphodiesterase iv inhibitors
WO2004005285A1 (en)2002-07-022004-01-15Almirall Prodesfarma AgNew quinuclidine amide derivatives
WO2004005258A1 (en)2002-07-022004-01-15Merck Frosst Canada & Co.Di-aryl-substituted-ethane pyridone pde4 inhibitors
WO2004005229A1 (en)2002-07-082004-01-15Pfizer Products Inc.Modulators of the glucocorticoid receptor
WO2004016578A2 (en)2002-07-252004-02-26Glaxo Group LimitedArylethanolamine beta2-adrenoreceptor agonist compounds
WO2004016601A1 (en)2002-08-092004-02-26Novartis AgBenzothiazole derivatives having beta-2-adrenoreceptor agonist activity
WO2004018465A2 (en)2002-08-172004-03-04Altana Pharma AgBenzonaphthyridines with pde 3/4 inhibiting activity
WO2004018450A1 (en)2002-08-102004-03-04Altana Pharma AgPiperidine-n-oxide-derivatives
WO2004018429A2 (en)2002-08-212004-03-04Boehringer Ingelheim Pharmaceuticals, Inc.Substituted hihydroquinolines as glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2004018422A1 (en)2002-08-232004-03-04Ranbaxy Laboratories LimitedFluoro and sulphonylamino containing 3,6-disubstituted azabicyclo (3.1.0) hexane derivatives as muscarinic receptor antagonists
WO2004018457A1 (en)2002-08-102004-03-04Altana Pharma AgPyrrolidinedione substituted piperidine-phthalazones as pde4 inhibitors
WO2004018449A1 (en)2002-08-102004-03-04Altana Pharma AgPiperidine-derivatives as pde4 inhibitors
WO2004018451A1 (en)2002-08-102004-03-04Altana Pharma AgPyridazinone-derivatives as pde4 inhibitors
WO2004018431A2 (en)2002-08-172004-03-04Altana Pharma AgNovel phenanthridines
WO2004019935A1 (en)2002-08-292004-03-11Boehringer Ingelheim Pharmaceuticals, Inc.-3 (sulfonamidoethyl) -indole derivaties for use as glucocorticoid mimetics in the treatment of inflammatory, allergic and proliferative diseases
WO2004019944A1 (en)2002-08-292004-03-11Altana Pharma Ag2-hydroxy-6-phenylphenanthridines as pde-4 inhibitors
WO2004019945A1 (en)2002-08-292004-03-11Altana Pharma Ag3-hydroxy-6-phenylphenanthridines as pde-4 inhibitors
WO2004022547A1 (en)2002-09-062004-03-18Glaxo Group LimitedPhenethanolamine derivatives and their use in the treatment of respiratory diseases
WO2004026248A2 (en)2002-09-202004-04-01Merck & Co., Inc.Octahydro-2-h-naphtho[1,2-f] indole-4-carboxamide derivatives as selective glucocorticoid receptor modulators
WO2004026841A1 (en)2002-09-182004-04-01Sumitomo Pharmaceuticals Co., Ltd.Novel 6-substituted uracil derivative and therapeutic agent for allergic disease
JP2004107299A (en)2002-09-202004-04-08Japan Energy Corp Novel 1-substituted uracil derivatives and therapeutic agents for allergic diseases
WO2004033412A1 (en)2002-10-042004-04-22Boehringer Ingelheim Pharma Gmbh & Co. KgNovel beta mimetics with extended duration of action, method for production and use thereof as medicaments
WO2004032921A1 (en)2002-10-112004-04-22Pfizer LimitedIndole derivatives as beta-2 agonists
WO2004037805A1 (en)2002-10-232004-05-06Glenmark Pharmaceuticals Ltd.Novel tricyclic compounds useful for the treatment of inflammatory and allergic disorders: process for their preparation and pharmaceutical compositions containing them
WO2004037773A1 (en)2002-10-282004-05-06Glaxo Group LimitedPhenethanolamine derivative for the treatment of respiratory diseases
WO2004037768A2 (en)2002-10-282004-05-06Glaxo Group LimitedPhenethanolamine derivatives
WO2004037807A2 (en)2002-10-222004-05-06Glaxo Group LimitedMedicinal arylethanolamine compounds
WO2004039827A2 (en)2002-10-302004-05-13Glaxo Group LimitedGlucocorticosteroids having a specific 17a-sidechain useful as antiinflammatory agents
WO2004039762A1 (en)2002-11-012004-05-13Glaxo Group LimitedPhenethanolamine derivatives for the treatment of respiratory diseases
WO2004039766A1 (en)2002-11-012004-05-13Glaxo Group LimitedPhenylethanolamine derivatives for the treatment of respiratory diseases
WO2004046083A1 (en)2002-11-152004-06-03Boehringer Ingelheim Pharma Gmbh & Co. KgNovel dihydroxy-methylphenyl derivatives, method for the production and use thereof as medicaments
WO2004045607A1 (en)2002-11-152004-06-03Elbion AgNovel hydroxyindoles, use as inhibitors of phosphodiesterase 4 and method for production thereof
WO2004045618A2 (en)2002-11-152004-06-03Boehringer Ingelheim Pharma Gmbh & Co. KgNovel medicaments for the treatment of chronic obstructive pulmonary diseases
DE10261874A1 (en)2002-12-202004-07-08Schering Ag Nonsteroidal anti-inflammatories
EP1440966A1 (en)2003-01-102004-07-28Pfizer LimitedIndole derivatives useful for the treatment of diseases
WO2004063197A1 (en)2003-01-092004-07-29Fujisawa Pharmaceutical Co., Ltd.Pyrrolopyridazine derivatives
WO2004066920A2 (en)2003-01-212004-08-12Merck & Co. Inc.17-carbamoyloxy cortisol derivatives as selective glucocorticoid receptor modulators
US20040167167A1 (en)2003-02-142004-08-26Mathai MammenBiphenyl derivatives
EP1460064A1 (en)2003-03-142004-09-22Pfizer LimitedIndole-2-carboxamide derivatives useful as beta-2 agonists
WO2004087142A1 (en)2003-04-042004-10-14Novartis AgQuinoline-2-one-derivatives for the treatment of airways diseases
WO2004089892A2 (en)2003-04-012004-10-21Theravance, Inc.Diarylmethyl and related compounds having beta2 andrenergic receptor agonist and muscarinic receptor antagonist activity
WO2004096800A2 (en)2003-05-022004-11-11Novartis AgQuinuclidine derivatives binding to mucarinic m3 receptors
EP1477167A1 (en)2003-05-152004-11-17Pfizer Limited[(2-hydroxy-2-(4-hydroxy-3-hydroxymethylphenyl)-ethylamino)-propyl] phenyl derivatives as beta2 agonists
US20040229904A1 (en)2003-05-152004-11-18Pfizer IncCompounds useful for the treatment of diseases
WO2004103998A1 (en)2003-05-212004-12-02Glaxo Group LimitedQuinoline derivatives as phosphodiesterase inhibitors
US20040242622A1 (en)2003-05-282004-12-02Mathai MammenAzabicycloalkane compounds
WO2004108720A1 (en)2003-06-112004-12-16Merck Frosst Canada Ltd.7- (1, 3-thiazol-2-yl)thio!-coumarin derivatives and their use as leukotriene biosynthesis inhibitors
WO2004108675A1 (en)2003-06-042004-12-16Pfizer Limited2-amino-pyridine derivatives as beta-2 adrenoreceptor agonists
WO2004108676A1 (en)2003-06-042004-12-16Pfizer Limited2-(6-amino-pyridin-3-yl)-2-hydroxyethylamine derivatives as beta 2-adrenoceptors agonists
WO2004111044A1 (en)2003-06-172004-12-23Glenmark Pharmaceuticals Ltd.Tricyclic compounds useful for the treatment of inflammatory and allergic disorders:process for their preparation
WO2005005452A1 (en)2003-07-112005-01-20Glaxo Group LimitedSpecific glucocorticosteroid compound having anti- inflammatory activity
WO2005012252A1 (en)2003-07-312005-02-10Altana Pharma AgNovel 6-phenylphenantridines
WO2005012253A1 (en)2003-07-312005-02-10Altana Pharma AgNovel 6-phenylphenanthridines
WO2005013995A1 (en)2003-07-162005-02-17Zambon Group S.P.A.4-substituted 7-alkoxy-2-aminosulfonyl-1,2-dihydro-phtalazin derivatives as phosphodiesterase 4 inhibitors for the treatment of allergies and inflammations
WO2005030212A1 (en)2003-09-272005-04-07Glaxo Group Limited4-aminoquinoline-3-carboxamide derivatives as pde4 inhibitors
WO2005030725A1 (en)2003-09-272005-04-07Glaxo Group LimitedDerivatives of 3-aminocarbonylquinoline, pharmaceutical compositions containing them and processes and intermediates for their preparation
WO2005033121A2 (en)2003-10-032005-04-14King Pharmaceuticals Research & Development, Inc.Synthesis of 2-aralkyloxyadenosines, 2-alkoxyadenosines, and their analogs
WO2005040103A1 (en)2003-10-222005-05-06Glaxo Group LimitedMedicinal compounds
WO2005044787A1 (en)2003-10-242005-05-19Glaxo Group LimitedPhenetanolamine derivatives
US20050133417A1 (en)2003-12-192005-06-23Bhan Opinder K.Systems, methods, and catalysts for producing a crude product
WO2005058867A1 (en)2003-12-172005-06-30Glaxo Group LimitedBenzothiophene-and thiochromene containing phenethanolamine derivatives for the treatment of respiratory disorders
JP2005187357A (en)2003-12-252005-07-14Nippon Suisan Kaisha Ltd Method for producing benzothiazolone derivative having selective β2 receptor agonist activity
WO2005066140A1 (en)2004-01-092005-07-21Boehringer Ingelheim International Gmbh3-hydroxymethyl-4-hydroxy-phenyl derivatives for the treatment of respiratory illnesses
WO2005065650A2 (en)2003-10-092005-07-21Glaxo Group LimitedAerosol formulations comprising a carboxylic acid surfactant
WO2005070908A1 (en)2004-01-232005-08-04Boehringer Ingelheim International GmbhNovel long-working beta-2-agonists and use thereof as medicaments
US20050171147A1 (en)2004-01-222005-08-04Brown Alan D.Sulfonamide derivatives for the treatment of diseases
US20050182092A1 (en)2004-02-132005-08-18Theravance, Inc.Crystalline form of a biphenyl compound
WO2005074924A1 (en)2004-02-092005-08-18Novartis AgCombination of benzothiazol-2-one beta2 adrenoceptor agonists and corticosteroids for the treatment of respiratory diseases
US20050182091A1 (en)2004-01-222005-08-18Brown Alan D.Sulfonamide derivatives for the treatment of diseases
WO2005077361A1 (en)2004-02-142005-08-25Boehringer Ingelheim International GmbhNovel, sustained-action beta-2-agonists and their use as medicaments
EP1574501A1 (en)2004-03-112005-09-14Pfizer LimitedQuinolinone derivatives, pharmaceutical compositions containing them and their use
US20050209227A1 (en)2004-03-172005-09-22Boehringer Ingelheim International GmbhNovel long acting betamimetics for the treatment of respiratory diseases
WO2005087744A1 (en)2004-03-102005-09-22Altana Pharma AgNovel thio-containing hydroxy-6-phenylphenanthridines and their use as pde4 inhibitors
WO2005087749A1 (en)2004-03-152005-09-22Kyowa Hakko Kogyo Co., Ltd.2-aminoquinazoline derivative
WO2005087745A1 (en)2004-03-102005-09-22Altana Pharma AgNovel amido-substituted hydroxy-6-phenylphenanthridines and their use as pde4 inhibitors
WO2005090287A2 (en)2004-03-172005-09-29Pfizer LimitedPhenylethanolamine derivatives as beta-2 agonists
WO2005090345A1 (en)2004-03-172005-09-29Altana Pharma AgNovel n- (alkoxyalkyl) carbamoyl - substituted 6-phenyl-benzonaphthyridine derivatives and their use as pde3/4 inhibitors
US20050215590A1 (en)2004-03-232005-09-29Brown Alan DFormamide derivatives for the treatment of diseases
US20050215542A1 (en)2004-03-232005-09-29Pfizer IncCompounds for the treatment of diseases
WO2005090288A1 (en)2004-03-172005-09-29Pfizer LimitedPhenylaminoethanol derivates as beta2 receptor agonists
WO2005092860A1 (en)2004-03-232005-10-06Pfizer LimitedCompounds for the treatment of diseases
WO2005092887A1 (en)2004-03-232005-10-06Pfizer LimitedCompounds for the treatment of diseases
WO2005095328A1 (en)2004-04-022005-10-13Glaxo Group LimitedChemical process and new crystalline form
US20050239778A1 (en)2004-04-222005-10-27Boehringer Ingelheim International GmbhNovel medicament combinations for the treatment of respiratory diseases
WO2005102350A1 (en)2004-04-222005-11-03Boehringer Ingelheim International GmbhBenzoxazine for treating respiratory tract diseases
US20050256115A1 (en)2004-05-142005-11-17Boehringer Ingelheim International GmbhAerosol formulation for the inhalation of beta-agonists
US20050256114A1 (en)2004-05-142005-11-17Boehringer Ingelheim International GmbhNovel long acting bronchodilators for the treatment of respiratory diseases
US20050272769A1 (en)2004-06-032005-12-08Theravance, Inc.Diamine beta2 adrenergic receptor agonists
US20050277632A1 (en)2004-05-132005-12-15Boehringer Ingelheim International GmbhBeta agonists for the treatment of respiratory diseases
US20060019991A1 (en)2004-07-212006-01-26Theravance, Inc.Diaryl ether beta2 adrenergic receptor agonists
WO2006008173A2 (en)2004-07-222006-01-26Glaxo Group LimitedPharmaceutical formulations for inhalation
US20060035933A1 (en)2004-08-162006-02-16Mathai MammenCompounds having beta adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2006023475A1 (en)2004-08-202006-03-02Celanese International CorporationFluid loss concentrate for hydraulic cement
US20060058530A1 (en)2004-09-102006-03-16Theravance, Inc.Amidine substituted aryl aniline compounds
WO2006048225A1 (en)2004-11-022006-05-11Novartis AgQuinuclidine derivatives and their use as muscarinic m3 receptor antagonists
WO2006056471A1 (en)2004-11-292006-06-01Novartis Ag5-hydroxy-benzothiazole derivatives having beta-2-adrenorecptor agonist activity
WO2006074897A1 (en)2005-01-112006-07-20Glaxo Group LimitedCinnamate salts of a beta-2 adrenergic agonist
WO2009140128A2 (en)*2008-05-132009-11-19Irm LlcCompounds and compositions as kinase inhibitors
WO2010048314A1 (en)*2008-10-222010-04-29Array Biopharma Inc.SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS
WO2011006074A1 (en)*2009-07-092011-01-13Array Biopharma Inc.SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS

Patent Citations (190)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US3714357A (en)1968-07-151973-01-30Rech D Applic Scient Sogeras SPharmaceutical compositions comprising quinuclidinol derivatives
EP0147719A2 (en)1983-12-241985-07-10Tanabe Seiyaku Co., Ltd.Novel carbostyril derivative and process for preparing same
JPH0525045B2 (en)1985-08-091993-04-09Noritoshi Nakabachi
EP0409595A2 (en)1989-07-191991-01-23Glaxo Group LimitedProcess for the preparation of a carbocyclic nucleoside analogue
EP0424021A1 (en)1989-10-191991-04-24Pfizer LimitedAntimuscarinic bronchodilators
US5171744A (en)1989-10-191992-12-15Pfizer Inc.Antimuscarinic bronchodilators
WO1992019594A1 (en)1991-05-021992-11-12Smithkline Beecham CorporationPyrrolidinones
US5451700A (en)1991-06-111995-09-19Ciba-Geigy CorporationAmidino compounds, their manufacture and methods of treatment
WO1993018007A1 (en)1992-03-131993-09-16Tokyo Tanabe Company LimitedNovel carbostyril derivative
WO1993019750A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating allergic or inflammatory diseases
WO1993019749A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating allergic and inflammatory diseases
WO1993019751A1 (en)1992-04-021993-10-14Smithkline Beecham CorporationCompounds useful for treating inflammatory diseases and inhibiting production of tumor necrosis factor
WO1994017090A1 (en)1993-01-201994-08-04Glaxo Group Limited2,6-diaminopurine derivatives
WO1996002543A1 (en)1994-07-141996-02-01Glaxo Group Limited2,3-dihydroxy cyclopentane derivatives of purines
WO1996002553A2 (en)1994-07-141996-02-01Glaxo Group LimitedAMINO PURINE-β-D-RIBOFURANURONAMIDE DERIVATIVES
WO1998018796A1 (en)1996-10-281998-05-07Novartis AgNaphthyridine derivatives
WO1998028319A1 (en)1996-12-241998-07-02Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999016766A1 (en)1997-10-011999-04-08Kyowa Hakko Kogyo Co., Ltd.Benzodioxole derivatives
WO1999024450A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdensine a1 receptor agonists
WO1999024451A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdenosine a1 receptor agonists
WO1999024449A2 (en)1997-11-081999-05-20Glaxo Group LimitedAdenosine a1 receptor agonists
WO1999038877A2 (en)1998-01-311999-08-05Glaxo Group Limited2-(PURIN-9-yl)-TETRAHYDROFURAN-3,4-DIOL DERIVATIVES
WO1999041267A1 (en)1998-02-141999-08-19Glaxo Group Limited2-(purin-9-yl) -tetrahydrofuran-3, 4-diol derivatives
WO1999064035A1 (en)1998-06-081999-12-16Advanced Medicine, Inc.β2-ADRENERGIC RECEPTOR AGONISTS
WO1999067266A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067263A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067264A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO1999067265A1 (en)1998-06-231999-12-29Glaxo Group Limited2-(purin-9-yl)-tetrahydrofuran-3,4-diol derivatives
WO2000000531A1 (en)1998-06-302000-01-06The Dow Chemical CompanyPolymer polyols and a process for the production thereof
WO2000023457A1 (en)1998-10-162000-04-27Pfizer LimitedAdenine derivatives
EP1052264A2 (en)1999-05-112000-11-15Pfizer Products Inc.Process for the synthesis of nucleoside analogs
US20020055651A1 (en)1999-06-022002-05-09Moran Edmund J.Beta2-adrenergic receptor agonists
WO2000075114A1 (en)1999-06-042000-12-14Novartis AgBeta2-adrenoceptor agonists
WO2000077018A2 (en)1999-06-152000-12-21Pfizer LimitedPurine derivatives
WO2000078774A2 (en)1999-06-182000-12-28University Of Virginia Patent FoundationInduction of pharmacological stress with adenosine receptor agonists
WO2001004118A2 (en)1999-07-142001-01-18Almirall Prodesfarma S.A.Quinuclidine derivatives and their use as muscarinic m3 receptor ligands
WO2001013953A2 (en)1999-08-212001-03-01Byk Gulden Lomberg Chemische Fabrik GmbhSynergistic combination of pde inhibitors and beta 2 adrenoceptor agonist
WO2001023399A1 (en)1999-09-302001-04-05Pfizer Products Inc.Compounds for the treatment of ischemia
WO2001027130A1 (en)1999-10-142001-04-19Pfizer LimitedPurine derivatives
WO2001027131A1 (en)1999-10-142001-04-19Pfizer LimitedPurine derivatives
WO2001042193A1 (en)1999-12-082001-06-14Theravance, Inc.β2-ADRENERGIC RECEPTOR AGONISTS
WO2001060835A1 (en)2000-02-182001-08-23Pfizer LimitedPurine derivatives
WO2001083462A1 (en)2000-04-272001-11-08Boehringer Ingelheim Pharma KgNovel, slow-acting betamimetics, a method for their production and their use as medicaments
WO2001094368A1 (en)2000-06-062001-12-13Pfizer Limited2-aminocarbonyl-9h-purine derivatives
WO2002000652A1 (en)2000-06-272002-01-03Laboratorios S.A.L.V.A.T., S.A.Carbamates derived from arylalkylamines
WO2002000676A1 (en)2000-06-272002-01-03Pfizer LimitedPurine derivatives
WO2002000679A2 (en)2000-06-282002-01-03Novartis Ag9.alpha.-chloro-6.alpha.-fluoro-17.alpha.-hydroxy-16-methyl-17-beta-methoxycarbonyl-androst-1,4-dienes esterified in position 17.alpha. by a cyclic acyl group
WO2002010143A1 (en)2000-07-282002-02-07Schering AktiengesellschaftNon-steroidal inflammation inhibitors
WO2002012266A1 (en)2000-08-052002-02-14Glaxo Group Limited17.beta.-carbothioate 17.alpha.-arylcarbonyloxyloxy androstane derivative as anti-inflammatory agents
WO2002022630A1 (en)2000-09-152002-03-21Pfizer LimitedPurine derivatives
WO2002042298A1 (en)2000-11-212002-05-30Novartis AgAminothiazoles and their use as adenosine receptor antagonists
WO2002051841A1 (en)2000-12-222002-07-04Almirall Prodesfarma AgQuinuclidine carbamate derivatives and their use as m3 antagonists
WO2002053564A2 (en)2000-12-282002-07-11Almirall Prodesfarma AgQuinuclidine derivatives and their use as m3 antagonists
WO2002066422A1 (en)2001-02-142002-08-29Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2002070490A1 (en)2001-03-082002-09-12Glaxo Group LimitedAgonists of beta-adrenoceptors
EP1241176A1 (en)2001-03-162002-09-18Pfizer Products Inc.Purine derivatives for the treatment of ischemia
WO2002076933A1 (en)2001-03-222002-10-03Glaxo Group LimitedFormailide derivatives as beta2-adrenoreceptor agonists
WO2002088167A1 (en)2001-04-302002-11-07Glaxo Group LimitedAnti-inflammatory 17.beta.-carbothioate ester derivatives of androstane with a cyclic ester group in position 17.alpha
WO2002096462A1 (en)2001-05-252002-12-05Pfizer Inc.An adenosine a2a receptor agonist and an anticholinergic agent in combination for treating obstructive airways diseases
WO2002100879A1 (en)2001-06-122002-12-19Glaxo Group LimitedNovel anti-inflammatory 17.alpha.-heterocyclic-esters of 17.beta.carbothioate androstane derivatives
WO2003000840A2 (en)2001-06-212003-01-03Diversa CorporationNitrilases
WO2003024439A1 (en)2001-09-142003-03-27Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2003033495A1 (en)2001-10-172003-04-24Ucb, S.A.Quinuclidine derivatives, processes for preparing them and their uses as m2 and/or m3 muscarinic receptor inhibitors
WO2003035668A2 (en)2001-10-202003-05-01Glaxo Group LimitedNovel anti-inflammatory androstane derivatives -17-carboxy-lactone substituted steroids with an aryl-carboxylic ester in position 17.alpha
WO2003039544A1 (en)2001-11-052003-05-15Novartis AgNaphtyridine derivatives, their preparation and their use as phosphodiesterase isoenzyme 4 (pde4) inhibitors
WO2003042214A2 (en)2001-11-092003-05-22Cv Therapeutics, Inc.A2b adenosine receptor antagonists
WO2003042164A1 (en)2001-11-132003-05-22Theravance, IncAryl aniline beta-2 adrenergic receptor agonists
WO2003042160A1 (en)2001-11-132003-05-22Theravance, Inc.Aryl aniline beta-2 adrenergic receptor agonists
WO2003048181A1 (en)2001-12-012003-06-12Glaxo Group Limited17.alpha. -cyclic esters of 16-methylpregnan-3,20-dione as anti-inflammatory agents
WO2003053966A2 (en)2001-12-202003-07-03Laboratorios S.A.L.V.A.T., S.A.1-alkyl-1-azoniabicyclo [2.2.2] octane carbamate derivatives and their use as muscarinic receptor antagonists
WO2003072592A1 (en)2002-01-152003-09-04Glaxo Group Limited17.alpha-cycloalkyl/cycloylkenyl esters of alkyl-or haloalkyl-androst-4-en-3-on-11.beta.,17.alpha.-diol 17.beta.-carboxylates as anti-inflammatory agents
WO2003062259A2 (en)2002-01-212003-07-31Glaxo Group LimitedNon-aromatic 17.alpha.-esters of androstane-17.beta.-carboxylate esters as anti-inflammatory agents
WO2003064445A1 (en)2002-01-312003-08-07Glaxo Group Limited17-alpha-heterocyclic esters of androstane derivatives having anti-inflammatory activity
WO2003072539A1 (en)2002-02-282003-09-04Glaxo Group LimitedPhenethanolamine derivatives for treatment of respiratory diseases
WO2003082787A1 (en)2002-03-262003-10-09Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003082280A1 (en)2002-03-262003-10-09Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003086408A1 (en)2002-04-102003-10-23University Of Virginia Patent FoundationUse of a2a adenosine receptor agonists for the treatment of inflammatory diseases
WO2003086294A2 (en)2002-04-112003-10-23Merck & Co., Inc.1h-benzo[f]indazol-5-yl derivatives as selective glucocorticoid receptor modulators
WO2003087094A2 (en)2002-04-162003-10-23Almirall Prodesfarma AgPyrrolidinium derivatives as antagonists of m3 muscarinic receptors
WO2003091204A1 (en)2002-04-252003-11-06Glaxo Group LimitedPhenethanolamine derivatives
WO2003099764A1 (en)2002-05-282003-12-04Theravance, Inc.ALKOXY ARYL β2 ADRENERGIC RECEPTOR AGONISTS
WO2003099807A1 (en)2002-05-292003-12-04Almirall Prodesfarma S.A.New indolylpiperidine derivatives as potent antihistaminic and antiallergic agents
WO2003101932A2 (en)2002-05-292003-12-11Boehringer Ingelheim Pharmaceuticals, Inc.Glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2003104204A1 (en)2002-06-052003-12-18Merck Patent GmbhPyridazine derivatives
WO2003104195A1 (en)2002-06-062003-12-18Boehringer Ingelheim Pharmaceuticals, Inc.4-(aryl or heteroaryl) -2-butylamine derivatives and their use as glucocorticoid ligans
WO2003104205A1 (en)2002-06-102003-12-18Merck Patent GmbhAryloximes
WO2004000839A1 (en)2002-06-192003-12-31Merck Patent GmbhThiazole derivatives as phosphodiesterase iv inhibitors
WO2004000814A1 (en)2002-06-252003-12-31Merck Frosst Canada & Co.8-(biaryl) quinoline pde4 inhibitors
WO2004005285A1 (en)2002-07-022004-01-15Almirall Prodesfarma AgNew quinuclidine amide derivatives
WO2004005258A1 (en)2002-07-022004-01-15Merck Frosst Canada & Co.Di-aryl-substituted-ethane pyridone pde4 inhibitors
WO2004005229A1 (en)2002-07-082004-01-15Pfizer Products Inc.Modulators of the glucocorticoid receptor
WO2004016578A2 (en)2002-07-252004-02-26Glaxo Group LimitedArylethanolamine beta2-adrenoreceptor agonist compounds
WO2004016601A1 (en)2002-08-092004-02-26Novartis AgBenzothiazole derivatives having beta-2-adrenoreceptor agonist activity
WO2004018457A1 (en)2002-08-102004-03-04Altana Pharma AgPyrrolidinedione substituted piperidine-phthalazones as pde4 inhibitors
WO2004018450A1 (en)2002-08-102004-03-04Altana Pharma AgPiperidine-n-oxide-derivatives
WO2004018451A1 (en)2002-08-102004-03-04Altana Pharma AgPyridazinone-derivatives as pde4 inhibitors
WO2004018449A1 (en)2002-08-102004-03-04Altana Pharma AgPiperidine-derivatives as pde4 inhibitors
WO2004018465A2 (en)2002-08-172004-03-04Altana Pharma AgBenzonaphthyridines with pde 3/4 inhibiting activity
WO2004018431A2 (en)2002-08-172004-03-04Altana Pharma AgNovel phenanthridines
WO2004018429A2 (en)2002-08-212004-03-04Boehringer Ingelheim Pharmaceuticals, Inc.Substituted hihydroquinolines as glucocorticoid mimetics, methods of making them, pharmaceutical compositions, and uses thereof
WO2004018422A1 (en)2002-08-232004-03-04Ranbaxy Laboratories LimitedFluoro and sulphonylamino containing 3,6-disubstituted azabicyclo (3.1.0) hexane derivatives as muscarinic receptor antagonists
WO2004019935A1 (en)2002-08-292004-03-11Boehringer Ingelheim Pharmaceuticals, Inc.-3 (sulfonamidoethyl) -indole derivaties for use as glucocorticoid mimetics in the treatment of inflammatory, allergic and proliferative diseases
WO2004019944A1 (en)2002-08-292004-03-11Altana Pharma Ag2-hydroxy-6-phenylphenanthridines as pde-4 inhibitors
WO2004019945A1 (en)2002-08-292004-03-11Altana Pharma Ag3-hydroxy-6-phenylphenanthridines as pde-4 inhibitors
WO2004022547A1 (en)2002-09-062004-03-18Glaxo Group LimitedPhenethanolamine derivatives and their use in the treatment of respiratory diseases
WO2004026841A1 (en)2002-09-182004-04-01Sumitomo Pharmaceuticals Co., Ltd.Novel 6-substituted uracil derivative and therapeutic agent for allergic disease
WO2004026248A2 (en)2002-09-202004-04-01Merck & Co., Inc.Octahydro-2-h-naphtho[1,2-f] indole-4-carboxamide derivatives as selective glucocorticoid receptor modulators
JP2004107299A (en)2002-09-202004-04-08Japan Energy Corp Novel 1-substituted uracil derivatives and therapeutic agents for allergic diseases
WO2004033412A1 (en)2002-10-042004-04-22Boehringer Ingelheim Pharma Gmbh & Co. KgNovel beta mimetics with extended duration of action, method for production and use thereof as medicaments
WO2004032921A1 (en)2002-10-112004-04-22Pfizer LimitedIndole derivatives as beta-2 agonists
WO2004037807A2 (en)2002-10-222004-05-06Glaxo Group LimitedMedicinal arylethanolamine compounds
WO2004037805A1 (en)2002-10-232004-05-06Glenmark Pharmaceuticals Ltd.Novel tricyclic compounds useful for the treatment of inflammatory and allergic disorders: process for their preparation and pharmaceutical compositions containing them
WO2004037773A1 (en)2002-10-282004-05-06Glaxo Group LimitedPhenethanolamine derivative for the treatment of respiratory diseases
WO2004037768A2 (en)2002-10-282004-05-06Glaxo Group LimitedPhenethanolamine derivatives
WO2004039827A2 (en)2002-10-302004-05-13Glaxo Group LimitedGlucocorticosteroids having a specific 17a-sidechain useful as antiinflammatory agents
WO2004039762A1 (en)2002-11-012004-05-13Glaxo Group LimitedPhenethanolamine derivatives for the treatment of respiratory diseases
WO2004039766A1 (en)2002-11-012004-05-13Glaxo Group LimitedPhenylethanolamine derivatives for the treatment of respiratory diseases
WO2004046083A1 (en)2002-11-152004-06-03Boehringer Ingelheim Pharma Gmbh & Co. KgNovel dihydroxy-methylphenyl derivatives, method for the production and use thereof as medicaments
WO2004045607A1 (en)2002-11-152004-06-03Elbion AgNovel hydroxyindoles, use as inhibitors of phosphodiesterase 4 and method for production thereof
WO2004045618A2 (en)2002-11-152004-06-03Boehringer Ingelheim Pharma Gmbh & Co. KgNovel medicaments for the treatment of chronic obstructive pulmonary diseases
DE10261874A1 (en)2002-12-202004-07-08Schering Ag Nonsteroidal anti-inflammatories
WO2004063197A1 (en)2003-01-092004-07-29Fujisawa Pharmaceutical Co., Ltd.Pyrrolopyridazine derivatives
EP1440966A1 (en)2003-01-102004-07-28Pfizer LimitedIndole derivatives useful for the treatment of diseases
WO2004066920A2 (en)2003-01-212004-08-12Merck & Co. Inc.17-carbamoyloxy cortisol derivatives as selective glucocorticoid receptor modulators
US20040167167A1 (en)2003-02-142004-08-26Mathai MammenBiphenyl derivatives
WO2004074812A2 (en)2003-02-142004-09-02Theravance IncLibrary of biphenyl derivatives useful for identifying compounds having both beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2004074246A2 (en)2003-02-142004-09-02Theravance Inc.Biphenyl derivatives having beta2 adrenergic receptor agonist and muscarinic receptor antagonist activity
EP1460064A1 (en)2003-03-142004-09-22Pfizer LimitedIndole-2-carboxamide derivatives useful as beta-2 agonists
WO2004080964A1 (en)2003-03-142004-09-23Pfizer LimitedIndole derivatives useful for the treatment of diseases
WO2004089892A2 (en)2003-04-012004-10-21Theravance, Inc.Diarylmethyl and related compounds having beta2 andrenergic receptor agonist and muscarinic receptor antagonist activity
WO2004087142A1 (en)2003-04-042004-10-14Novartis AgQuinoline-2-one-derivatives for the treatment of airways diseases
WO2004096800A2 (en)2003-05-022004-11-11Novartis AgQuinuclidine derivatives binding to mucarinic m3 receptors
EP1477167A1 (en)2003-05-152004-11-17Pfizer Limited[(2-hydroxy-2-(4-hydroxy-3-hydroxymethylphenyl)-ethylamino)-propyl] phenyl derivatives as beta2 agonists
US20040229904A1 (en)2003-05-152004-11-18Pfizer IncCompounds useful for the treatment of diseases
WO2004103998A1 (en)2003-05-212004-12-02Glaxo Group LimitedQuinoline derivatives as phosphodiesterase inhibitors
US20040242622A1 (en)2003-05-282004-12-02Mathai MammenAzabicycloalkane compounds
WO2004108675A1 (en)2003-06-042004-12-16Pfizer Limited2-amino-pyridine derivatives as beta-2 adrenoreceptor agonists
WO2004108676A1 (en)2003-06-042004-12-16Pfizer Limited2-(6-amino-pyridin-3-yl)-2-hydroxyethylamine derivatives as beta 2-adrenoceptors agonists
WO2004108720A1 (en)2003-06-112004-12-16Merck Frosst Canada Ltd.7- (1, 3-thiazol-2-yl)thio!-coumarin derivatives and their use as leukotriene biosynthesis inhibitors
WO2004111044A1 (en)2003-06-172004-12-23Glenmark Pharmaceuticals Ltd.Tricyclic compounds useful for the treatment of inflammatory and allergic disorders:process for their preparation
WO2005005452A1 (en)2003-07-112005-01-20Glaxo Group LimitedSpecific glucocorticosteroid compound having anti- inflammatory activity
WO2005013995A1 (en)2003-07-162005-02-17Zambon Group S.P.A.4-substituted 7-alkoxy-2-aminosulfonyl-1,2-dihydro-phtalazin derivatives as phosphodiesterase 4 inhibitors for the treatment of allergies and inflammations
WO2005012252A1 (en)2003-07-312005-02-10Altana Pharma AgNovel 6-phenylphenantridines
WO2005012253A1 (en)2003-07-312005-02-10Altana Pharma AgNovel 6-phenylphenanthridines
WO2005030212A1 (en)2003-09-272005-04-07Glaxo Group Limited4-aminoquinoline-3-carboxamide derivatives as pde4 inhibitors
WO2005030725A1 (en)2003-09-272005-04-07Glaxo Group LimitedDerivatives of 3-aminocarbonylquinoline, pharmaceutical compositions containing them and processes and intermediates for their preparation
WO2005033121A2 (en)2003-10-032005-04-14King Pharmaceuticals Research & Development, Inc.Synthesis of 2-aralkyloxyadenosines, 2-alkoxyadenosines, and their analogs
WO2005065650A2 (en)2003-10-092005-07-21Glaxo Group LimitedAerosol formulations comprising a carboxylic acid surfactant
WO2005040103A1 (en)2003-10-222005-05-06Glaxo Group LimitedMedicinal compounds
WO2005044787A1 (en)2003-10-242005-05-19Glaxo Group LimitedPhenetanolamine derivatives
WO2005058867A1 (en)2003-12-172005-06-30Glaxo Group LimitedBenzothiophene-and thiochromene containing phenethanolamine derivatives for the treatment of respiratory disorders
US20050133417A1 (en)2003-12-192005-06-23Bhan Opinder K.Systems, methods, and catalysts for producing a crude product
JP2005187357A (en)2003-12-252005-07-14Nippon Suisan Kaisha Ltd Method for producing benzothiazolone derivative having selective β2 receptor agonist activity
WO2005066140A1 (en)2004-01-092005-07-21Boehringer Ingelheim International Gmbh3-hydroxymethyl-4-hydroxy-phenyl derivatives for the treatment of respiratory illnesses
US20050171147A1 (en)2004-01-222005-08-04Brown Alan D.Sulfonamide derivatives for the treatment of diseases
US20050182091A1 (en)2004-01-222005-08-18Brown Alan D.Sulfonamide derivatives for the treatment of diseases
WO2005070908A1 (en)2004-01-232005-08-04Boehringer Ingelheim International GmbhNovel long-working beta-2-agonists and use thereof as medicaments
WO2005074924A1 (en)2004-02-092005-08-18Novartis AgCombination of benzothiazol-2-one beta2 adrenoceptor agonists and corticosteroids for the treatment of respiratory diseases
US20050182092A1 (en)2004-02-132005-08-18Theravance, Inc.Crystalline form of a biphenyl compound
WO2005077361A1 (en)2004-02-142005-08-25Boehringer Ingelheim International GmbhNovel, sustained-action beta-2-agonists and their use as medicaments
WO2005087745A1 (en)2004-03-102005-09-22Altana Pharma AgNovel amido-substituted hydroxy-6-phenylphenanthridines and their use as pde4 inhibitors
WO2005087744A1 (en)2004-03-102005-09-22Altana Pharma AgNovel thio-containing hydroxy-6-phenylphenanthridines and their use as pde4 inhibitors
EP1574501A1 (en)2004-03-112005-09-14Pfizer LimitedQuinolinone derivatives, pharmaceutical compositions containing them and their use
WO2005087749A1 (en)2004-03-152005-09-22Kyowa Hakko Kogyo Co., Ltd.2-aminoquinazoline derivative
US20050209227A1 (en)2004-03-172005-09-22Boehringer Ingelheim International GmbhNovel long acting betamimetics for the treatment of respiratory diseases
WO2005090288A1 (en)2004-03-172005-09-29Pfizer LimitedPhenylaminoethanol derivates as beta2 receptor agonists
WO2005090287A2 (en)2004-03-172005-09-29Pfizer LimitedPhenylethanolamine derivatives as beta-2 agonists
WO2005090345A1 (en)2004-03-172005-09-29Altana Pharma AgNovel n- (alkoxyalkyl) carbamoyl - substituted 6-phenyl-benzonaphthyridine derivatives and their use as pde3/4 inhibitors
US20050215590A1 (en)2004-03-232005-09-29Brown Alan DFormamide derivatives for the treatment of diseases
US20050215542A1 (en)2004-03-232005-09-29Pfizer IncCompounds for the treatment of diseases
WO2005092860A1 (en)2004-03-232005-10-06Pfizer LimitedCompounds for the treatment of diseases
WO2005092887A1 (en)2004-03-232005-10-06Pfizer LimitedCompounds for the treatment of diseases
WO2005095328A1 (en)2004-04-022005-10-13Glaxo Group LimitedChemical process and new crystalline form
US20050239778A1 (en)2004-04-222005-10-27Boehringer Ingelheim International GmbhNovel medicament combinations for the treatment of respiratory diseases
WO2005102350A1 (en)2004-04-222005-11-03Boehringer Ingelheim International GmbhBenzoxazine for treating respiratory tract diseases
US20050277632A1 (en)2004-05-132005-12-15Boehringer Ingelheim International GmbhBeta agonists for the treatment of respiratory diseases
US20050256115A1 (en)2004-05-142005-11-17Boehringer Ingelheim International GmbhAerosol formulation for the inhalation of beta-agonists
US20050256114A1 (en)2004-05-142005-11-17Boehringer Ingelheim International GmbhNovel long acting bronchodilators for the treatment of respiratory diseases
US20050272769A1 (en)2004-06-032005-12-08Theravance, Inc.Diamine beta2 adrenergic receptor agonists
US20060019991A1 (en)2004-07-212006-01-26Theravance, Inc.Diaryl ether beta2 adrenergic receptor agonists
WO2006008173A2 (en)2004-07-222006-01-26Glaxo Group LimitedPharmaceutical formulations for inhalation
US20060035933A1 (en)2004-08-162006-02-16Mathai MammenCompounds having beta adrenergic receptor agonist and muscarinic receptor antagonist activity
WO2006023475A1 (en)2004-08-202006-03-02Celanese International CorporationFluid loss concentrate for hydraulic cement
US20060058530A1 (en)2004-09-102006-03-16Theravance, Inc.Amidine substituted aryl aniline compounds
WO2006048225A1 (en)2004-11-022006-05-11Novartis AgQuinuclidine derivatives and their use as muscarinic m3 receptor antagonists
WO2006056471A1 (en)2004-11-292006-06-01Novartis Ag5-hydroxy-benzothiazole derivatives having beta-2-adrenorecptor agonist activity
WO2006074897A1 (en)2005-01-112006-07-20Glaxo Group LimitedCinnamate salts of a beta-2 adrenergic agonist
WO2009140128A2 (en)*2008-05-132009-11-19Irm LlcCompounds and compositions as kinase inhibitors
WO2010048314A1 (en)*2008-10-222010-04-29Array Biopharma Inc.SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS
WO2011006074A1 (en)*2009-07-092011-01-13Array Biopharma Inc.SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINE COMPOUNDS AS TRK KINASE INHIBITORS

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JEAN JACQUES, ANDRE COLLET, SAMUEL H. WILEN: "Enantiomers, Racemates and Resolutions", 1981, JOHN WILEY AND SONS, INC.
SAULNIER ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 4, 1994, pages 1985
T. W. GREENE: "Protecting Groups in Organic Chemistry", 1999, JOHN WILEY AND SONS, INC.
T.W. GREENE, P. G. M. WUTS: "Protective Groups in Organic Chemistry", 1991, JOHN WILEY AND SONS

Cited By (181)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
US10011604B2 (en)2008-09-222018-07-03Array Biopharma, Inc.Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US9795611B2 (en)2008-09-222017-10-24Array Biopharma, Inc.Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US9796723B2 (en)2008-09-222017-10-24Array Biopharma, Inc.Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US10590139B2 (en)2008-09-222020-03-17Array Biopharma Inc.Method of treatment using substituted imidazo[1,2b]pyridazine compounds
US9227975B2 (en)2008-09-222016-01-05Array Biopharma, Inc.Method of treatment using substituted imidazo[1,2B]pyridazine compounds
US10774085B2 (en)2008-10-222020-09-15Array Biopharma Inc.Method of treatment using substituted pyrazolo[1,5-A] pyrimidine compounds
US9447104B2 (en)2008-10-222016-09-20Array Biopharma, Inc.Method of treatment using substituted pyrazolo[1,5-a]pyrimidine compounds
US10047097B2 (en)2008-10-222018-08-14Array Biopharma Inc.Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US10005783B2 (en)2008-10-222018-06-26Array Biopharma Inc.Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US9127013B2 (en)2008-10-222015-09-08Array Biopharma, Inc.Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US9676783B2 (en)2008-10-222017-06-13Array Biopharma, Inc.Method of treatment using substituted pyrazolo[1,5-A] pyrimidine compounds
US11267818B2 (en)2008-10-222022-03-08Array Biopharma Inc.Method of treatment using substituted pyrazolo[1,5-a] pyrimidine compounds
US9782415B2 (en)2009-07-092017-10-10Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US8791123B2 (en)2009-07-092014-07-29Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US9682979B2 (en)2009-07-092017-06-20Array Biopharma, Inc.Substituted pyrazolo [1,5-A] pyrimidine compounds as TRK kinase inhibitors
US10758542B2 (en)2009-07-092020-09-01Array Biopharma Inc.Substituted pyrazolo[l,5-a]pyrimidine compounds as Trk kinase inhibitors
US10251889B2 (en)2009-07-092019-04-09Array BioPharm Inc.Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US9796724B2 (en)2009-07-092017-10-24Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyrimidine compounds as Trk kinase inhibitors
US9902741B2 (en)2010-05-202018-02-27Array Biopharma Inc.Macrocyclic compounds as TRK kinase inhibitors
US9840519B2 (en)2010-05-202017-12-12Array Biopharma, Inc.Macrocyclic compounds as TRK kinase inhibitors
US9493476B2 (en)2010-05-202016-11-15Array Biopharma, Inc.Macrocyclic compounds as trk kinase inhibitors
US10647730B2 (en)2010-05-202020-05-12Array Biopharma Inc.Macrocyclic compounds as TRK kinase inhibitors
US8933084B2 (en)2010-05-202015-01-13Array Biopharma Inc.Macrocyclic compounds as Trk kinase inhibitors
US9750744B2 (en)2010-05-202017-09-05Array Biopharma, Inc.Macrocyclic compounds as Trk kinase inhibitors
US9718822B2 (en)2010-05-202017-08-01Array Biopharma, Inc.Macrocyclic compounds as Trk kinase inhibitors
EP2914622B1 (en)*2012-11-052023-06-07Foundation Medicine, Inc.Novel fusion molecules and uses thereof
EP3628749A1 (en)2013-07-302020-04-01Blueprint Medicines CorporationNtrk2 fusions
WO2015017533A1 (en)2013-07-302015-02-05Blueprint Medicines CorporationNtrk2 fusions
US10407509B2 (en)2013-07-302019-09-10Blueprint Medicines CorporationNTRK2 fusions
US10246466B2 (en)2014-01-242019-04-02Tp Therapeutics, Inc.Diaryl macrocycles as modulators of protein kinases
US10618912B2 (en)2014-01-242020-04-14Turning Point Therapeutics, Inc.Diaryl macrocycles as modulators of protein kinases
US12404281B2 (en)2014-01-242025-09-02Turning Point Therapeutics, Inc.Diaryl macrocycles as modulators of protein kinases
JP2017507980A (en)*2014-03-172017-03-23レクシコン ファーマシューティカルズ インコーポレイテッド Inhibitor of adapter-related kinase 1, composition containing the same, and method of use thereof
US10751341B2 (en)2014-11-062020-08-25Lysosomal Therapeutics Inc.Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US10786508B2 (en)2014-11-062020-09-29Lysosomal Therapeutics Inc.Substituted imidazo[1,5-A]-pyrimidines and their use in the treatment of medical disorders
US11091492B2 (en)2014-11-062021-08-17Bial—R&D Investments, S.A.Substituted pyrazolo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US11351173B2 (en)2014-11-062022-06-07Bial—R&D Investments, S.A.Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US11400095B2 (en)2014-11-062022-08-02Bial—R&D Investments, S.A.Substituted imidazo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US10570135B2 (en)2014-11-062020-02-25Lysosomal Therapeutics Inc.Substituted pyrazolo[1,5-A]pyrimidines and their use in the treatment of medical disorders
US11932645B2 (en)2014-11-062024-03-19Bial—R & D Investments, S.A.Substituted pyrazolo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US9782414B2 (en)2014-11-162017-10-10Array Biopharma, Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10799505B2 (en)2014-11-162020-10-13Array Biopharma, Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10813936B2 (en)2014-11-162020-10-27Array Biopharma, Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-YL)-pyrazolo[1,5-A]pyrimidin-3-YL)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10172861B2 (en)2014-11-162019-01-08Array Biopharma Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
US10285993B2 (en)2014-11-162019-05-14Array Biopharma Inc.Crystalline form of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
CN107207514B (en)*2014-12-152020-01-24康联制药有限公司 Fused ring heteroaryl compounds and their use as TRK inhibitors
EP4420732A3 (en)*2014-12-152024-11-27CMG Pharmaceutical Co., Ltd.Fused ring heteroaryl compounds and their use as trk inhibitors
CN107207514A (en)*2014-12-152017-09-26康联制药有限公司 Fused ring heteroaryl compounds and their use as TRK inhibitors
RU2708674C2 (en)*2014-12-152019-12-11СиЭмДжи ФАРМАСЬЮТИКАЛ КО., ЛТД.Condensed ring heteroaryl compounds and use thereof as trk inhibitors
WO2016097869A1 (en)2014-12-152016-06-23Cmg Pharmaceutical Co., Ltd.Fused ring heteroaryl compounds and their use as trk inhibitors
JP2017538766A (en)*2014-12-152017-12-28シーエムジー ファーマシューティカル カンパニー,リミテッド Fused ring heteroaryl compounds and uses as TRK inhibitors
US9701681B2 (en)2014-12-152017-07-11Cmg Pharmaceutical Co., Ltd.Fused ring heteroaryl compounds and their use as Trk inhibitors
USRE50634E1 (en)2015-01-232025-10-14Turning Point Therapeutics, Inc.Diaryl macrocycles as modulators of protein kinases
EP3722441A1 (en)2015-06-012020-10-14Loxo Oncology Inc.Method of diagnosing a cancer for a treatment with a trk inhibitor
US10316044B2 (en)2015-07-022019-06-11Tp Therapeutics, Inc.Chiral diaryl macrocycles as modulators of protein kinases
US12187739B2 (en)2015-07-062025-01-07Turning Point Therapeutics, Inc.Diaryl macrocycle polymorph
US10138243B2 (en)2015-07-162018-11-27Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10174028B2 (en)2015-07-162019-01-08Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10174027B2 (en)2015-07-162019-01-08Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10023570B2 (en)2015-07-162018-07-17Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
WO2017011776A1 (en)2015-07-162017-01-19Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10017512B2 (en)2015-08-262018-07-10Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to NTRK
US11046697B2 (en)2015-08-262021-06-29Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to NTRK
US10370727B2 (en)2015-10-262019-08-06Loxo Oncology, Inc.Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10655186B2 (en)2015-10-262020-05-19Loxo Oncology, Inc.Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10724102B2 (en)2015-10-262020-07-28Loxo Oncology, Inc.Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10378068B2 (en)2015-10-262019-08-13Loxo Oncology, Inc.Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
WO2017075107A1 (en)2015-10-262017-05-04Nanda NishaPoint mutations in trk inhibitor-resistant cancer and methods relating to the same
US10907215B2 (en)2015-10-262021-02-02Loxo Oncology, Inc.Point mutations in TRK inhibitor-resistant cancer and methods relating to the same
US10370379B2 (en)2015-11-192019-08-06Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to NTRK
US11059827B2 (en)2015-11-192021-07-13Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to NTRK
JP2019500328A (en)*2015-11-192019-01-10ブループリント メディシンズ コーポレイション Compounds and compositions useful for the treatment of disorders associated with NTRK
WO2017087778A1 (en)2015-11-192017-05-26Blueprint Medicines CorporationCompounds and compositions useful for treating disorders related to ntrk
US10668072B2 (en)2016-04-042020-06-02Loxo Oncology, Inc.Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US11191766B2 (en)2016-04-042021-12-07Loxo Oncology, Inc.Methods of treating pediatric cancers
US10137127B2 (en)2016-04-042018-11-27Loxo Oncology, Inc.Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10588908B2 (en)2016-04-042020-03-17Loxo Oncology, Inc.Methods of treating pediatric cancers
US11484535B2 (en)2016-04-042022-11-01Loxo Oncology, Inc.Liquid formulations of (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a] pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
US10045991B2 (en)2016-04-042018-08-14Loxo Oncology, Inc.Methods of treating pediatric cancers
WO2017176751A1 (en)2016-04-042017-10-12Loxo Oncology, Inc.Liquid formulations of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
WO2017176744A1 (en)2016-04-042017-10-12Loxo Oncology, Inc.Methods of treating pediatric cancers
US10787454B2 (en)2016-04-062020-09-29BIAL—BioTech Investments, Inc.Imidazo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US9840510B1 (en)2016-04-062017-12-12Lysosomal Therapeutics Inc.Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US9920061B2 (en)2016-04-062018-03-20Lysosomal Therapeutics Inc.Imidazo[1,5-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US12116369B2 (en)2016-04-062024-10-15Bial—R&D Investments, S.A.Substituted pyrazolo[1,5-a]pyrimidines as glucocerebrosidase activators
US11453675B2 (en)2016-04-062022-09-27Bial—R&D Investments, S.A.Imidazo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US11192892B2 (en)2016-04-062021-12-07Bial—R&D Investments, S.A.Substituted pyrazolo[1,5-a]pyrimidines for the treatment of medical disorders
US10934298B2 (en)2016-04-062021-03-02BIAL—BioTech Investments, Inc.Substituted pyrazolo[1,5-a]pyrimidines for the treatment of medical disorders
US11124516B2 (en)2016-04-062021-09-21BIAL-BioTech Investments, Inc.Pyrrolo[1,2-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US11345698B2 (en)2016-05-052022-05-31Bial—R&D Investments, S.A.Substituted imidazo[1,2-a]pyridines, substituted imidazo[1,2-a]pyrazines, related compounds, and their use in the treatment of medical disorders
US9868742B2 (en)2016-05-052018-01-16Lysosomal Therapeutics Inc.Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b] pyridazines, related compounds, and their use in the treatment of medical disorders
US11168087B2 (en)2016-05-052021-11-09Bial—R&D Investments, S.A.Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b]pyridazines, related compounds, and their use in the treatment of medical disorders
US11878979B2 (en)2016-05-052024-01-23Bial—R&D Investments, S.A.Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b]pyridazines, related compounds, and their use in the treatment of medical disorders
US11214571B2 (en)2016-05-182022-01-04Array Biopharma Inc.Process for the preparation of (S)-N-(5-((R)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide and salts thereof
US10112942B2 (en)2016-10-102018-10-30Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10172851B2 (en)2016-10-102019-01-08Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
WO2018071447A1 (en)2016-10-102018-04-19Andrews Steven WSubstituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10555944B2 (en)2016-10-102020-02-11Eli Lilly And CompanySubstituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10137124B2 (en)2016-10-102018-11-27Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
US10144734B2 (en)2016-10-102018-12-04Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11998545B2 (en)2016-10-102024-06-04Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
WO2018071454A1 (en)2016-10-102018-04-19Andrews Steven WSubstituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10172845B2 (en)2016-10-102019-01-08Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
EP4144735A1 (en)2016-10-102023-03-08Array Biopharma, Inc.Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10441581B2 (en)2016-10-102019-10-15Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11648243B2 (en)2016-10-102023-05-16Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US10953005B1 (en)2016-10-102021-03-23Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
EP3753939A1 (en)2016-10-102020-12-23Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
US10881652B2 (en)2016-10-102021-01-05Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyridine compounds as RET kinase inhibitors
US11091486B2 (en)2016-10-262021-08-17Array Biopharma, IncProcess for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof
KR20190067913A (en)*2016-10-282019-06-17치아타이 티안큉 파마수티컬 그룹 주식회사 Aminopyrazolopyrimidine compounds used as inhibitors of tyrosine kinase receptors which are neurotrophic factors
AU2017348826B2 (en)*2016-10-282021-12-02Centaurus Biopharma Co., Ltd.Amino pyrazolopyrimidine compound used as neurotrophic factor tyrosine kinase receptor inhibitor
US10829492B2 (en)2016-10-282020-11-10Chia Tai Tianqing Pharmaceutical Group Co., Ltd.Amino pyrazolopyrimidine compound used as neurotrophic factor tyrosine kinase receptor inhibitor
EP3533796A4 (en)*2016-10-282020-06-03Chia Tai Tianqing Pharmaceutical Group Co., Ltd.Amino pyrazolopyrimidine compound used as neurotrophic factor tyrosine kinase receptor inhibitor
KR102616249B1 (en)2016-10-282023-12-21치아타이 티안큉 파마수티컬 그룹 주식회사 Amino pyrazolopyrimidine compounds used as neurotrophic factor tyrosine kinase receptor inhibitors.
RU2764523C2 (en)*2016-10-282022-01-18Чиа Тай Тянцин Фармасьютикал Груп Ко., Лтд.Aminopyrazolopyrimidine compound used as inhibitor of tyrosine kinase receptor of neurotrophic factor
CN108003161B (en)*2016-10-282020-10-09正大天晴药业集团股份有限公司Neurotrophic factor tyrosine kinase receptor inhibitors
CN108003161A (en)*2016-10-282018-05-08正大天晴药业集团股份有限公司Neurotrophic factor tyrosine kinase receptor inhibitor
US11168090B2 (en)2017-01-182021-11-09Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyrazines as RET kinase inhibitors
WO2018136661A1 (en)2017-01-182018-07-26Andrews Steven WSUBSTITUTED PYRAZOLO[1,5-a]PYRAZINE COMPOUNDS AS RET KINASE INHIBITORS
US11851434B2 (en)2017-01-182023-12-26Array Biopharma Inc.Substituted pyrazolo[1,5-A]pyrazine compounds as ret kinase inhibitors
WO2018136663A1 (en)2017-01-182018-07-26Array Biopharma, Inc.Ret inhibitors
WO2018170381A1 (en)2017-03-162018-09-20Andrews Steven WMacrocyclic compounds as ros1 kinase inhibitors
US10966985B2 (en)2017-03-162021-04-06Array Biopharma Inc.Macrocyclic compounds as ROS1 kinase inhibitors
US10688100B2 (en)2017-03-162020-06-23Array Biopharma Inc.Macrocylic compounds as ROS1 kinase inhibitors
WO2019075108A1 (en)2017-10-102019-04-18Metcalf Andrew TCrystalline forms
WO2019075114A1 (en)2017-10-102019-04-18Mark ReynoldsFormulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
WO2019084285A1 (en)2017-10-262019-05-02Qian ZhaoFormulations of a macrocyclic trk kinase inhibitor
US11098046B2 (en)2017-11-102021-08-24Angex Pharmaceutical, Inc.Macrocyclic compounds as TRK kinase inhibitors and uses thereof
JP2021502413A (en)*2017-11-102021-01-28エンジェクス ファーマシューティカル インコーポレイテッド Macrocycles and their use as TRK kinase inhibitors
CN111819179A (en)*2017-12-152020-10-23金字塔生物科技公司5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3- (1H-pyrazol-1-yl) pyrazolo [1,5-a ] pyrimidine derivatives and related compounds as TRK kinase inhibitors for the treatment of cancer
CN111819179B (en)*2017-12-152023-09-01金字塔生物科技公司Related compounds as TRK kinase inhibitors for the treatment of cancer
WO2019143994A1 (en)2018-01-182019-07-25Array Biopharma Inc.Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
US11603374B2 (en)2018-01-182023-03-14Array Biopharma Inc.Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
WO2019143977A1 (en)2018-01-182019-07-25Array Biopharma Inc.Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
US11524963B2 (en)2018-01-182022-12-13Array Biopharma Inc.Substituted pyrazolo[3,4-d]pyrimidines as RET kinase inhibitors
US11472802B2 (en)2018-01-182022-10-18Array Biopharma Inc.Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
AU2019233204B2 (en)*2018-03-142022-02-03Fochon Biosciences, Ltd.Substituted (2-azabicyclo (3.1.0) hexan-2-yl) pyrazolo (1, 5-a) pyrimidine and imidazo (1, 2-b) pyridazine compounds as TRK kinases inhibitors
CN111936500A (en)*2018-03-142020-11-13重庆复创医药研究有限公司Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
JP7294677B2 (en)2018-03-142023-06-20フォチョン・ファーマシューティカルズ・リミテッド Substituted (2-azabicyclo[3.1.0]hexane-2-yl)pyrazolo[1,5-a]pyrimidine compounds and substituted (2-azabicyclo[3.1.0]hexane-2 as TRK kinase inhibitors -yl)imidazo[1,2-b]pyridazine compounds
CN111936500B (en)*2018-03-142023-09-01重庆复尚源创医药技术有限公司Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidines and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
US11459332B2 (en)2018-03-142022-10-04Fochon Biosciences, Ltd.Substituted 2-azabicyclo[3.1.0]hexanes as TRK kinases inhibitors
RU2781618C2 (en)*2018-03-142022-10-14Фочон Байосайенсис, Лтд.Substituted (2-azabicyclo[3.1.0]hexane-2-yl)pyrazolo[1.5-a]pyrimidine and imidazo[1.2-b]pyridazine compounds as trk kinase inhibitors
WO2019174598A1 (en)2018-03-142019-09-19Fochon Pharmaceuticals, Ltd.SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
JP2021517136A (en)*2018-03-142021-07-15フォチョン・ファーマシューティカルズ・リミテッドFochon Pharmaceuticals, Ltd. Substitution as TRK kinase inhibitor (2-azabicyclo [3.1.0] hexane-2-yl) pyrazolo [1,5-a] pyrimidine compound and substitution (2-azabicyclo [3.1.0] hexane-2) -Il) Imidazo [1,2-b] pyridazine compound
WO2019191659A1 (en)2018-03-292019-10-03Loxo Oncology, Inc.Treatment of trk-associated cancers
US11597729B2 (en)2018-06-082023-03-07Jiangsu Vcare Pharmatech Co., Ltd.Tropomyosin receptor kinase inhibitor, preparation method therefor and use thereof
CN110294761A (en)*2018-06-082019-10-01南京雷正医药科技有限公司Substituted pyrazolo [1,5-a] pyrimidine compound as Trk kinase inhibitor
CN110577532A (en)*2018-06-082019-12-17江苏威凯尔医药科技有限公司Tropomyosin receptor kinase inhibitor and preparation method and application thereof
WO2019233461A1 (en)*2018-06-082019-12-12江苏威凯尔医药科技有限公司Tropomyosin receptor kinase inhibitor, preparation method therefor and use thereof
CN110577532B (en)*2018-06-082022-06-03江苏威凯尔医药科技有限公司Tropomyosin receptor kinase inhibitor and preparation method and application thereof
WO2020028258A1 (en)2018-07-312020-02-06Loxo Oncology, Inc.Spray-dried dispersions and formulations of (s)-5-amino-3-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)-1-(1,1,1-trifluoro propan-2-yl)-1h-pyrazole-4-carboxamide
US11964988B2 (en)2018-09-102024-04-23Array Biopharma Inc.Fused heterocyclic compounds as RET kinase inhibitors
WO2020055672A1 (en)2018-09-102020-03-19Array Biopharma Inc.Fused heterocyclic compounds as ret kinase inhibitors
WO2020131674A1 (en)2018-12-192020-06-25Array Biopharma Inc.7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020131627A1 (en)2018-12-192020-06-25Array Biopharma Inc.Substituted pyrazolo[1,5-a]pyridine compounds as inhibitors of fgfr tyrosine kinases
CN111718351B (en)*2019-03-192021-10-12华中师范大学 Oxygen-substituted pyrazolopyrimidine compound and pharmaceutical composition and application thereof
CN111718351A (en)*2019-03-192020-09-29华中师范大学 Oxygen-substituted pyrazolopyrimidine compound and pharmaceutical composition and application thereof
WO2021047584A1 (en)*2019-09-112021-03-18Fochon Pharmaceuticals, Ltd.SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS
CN114630829A (en)*2019-09-112022-06-14重庆复创医药研究有限公司Substituted (2-azabicyclo [3.1.0] hex-2-yl) pyrazolo [1,5-a ] pyrimidine and imidazo [1,2-b ] pyridazine compounds as TRK kinase inhibitors
CN110804059B (en)*2019-09-302024-03-12郑州泰基鸿诺医药股份有限公司Carbamate compound, pharmaceutical composition and application thereof
CN110804059A (en)*2019-09-302020-02-18郑州泰基鸿诺医药股份有限公司Carbamate compound, pharmaceutical composition and application thereof
CN111138437A (en)*2019-12-042020-05-12杭州华东医药集团新药研究院有限公司Substituted pyrazolo [1,5-a ] pyrimidine amino acid derivatives and uses thereof
CN111138437B (en)*2019-12-042021-03-05杭州华东医药集团新药研究院有限公司Substituted pyrazolo [1,5-a ] pyrimidine amino acid derivatives and uses thereof
CN115551859B (en)*2020-05-152024-08-27石药集团中奇制药技术(石家庄)有限公司Aza condensed ring amide compound and application thereof
JP7495998B2 (en)2020-05-152024-06-05石薬集団中奇制薬技術(石家庄)有限公司 Aza-condensed cyclic amide compounds and their uses
RU2811975C1 (en)*2020-05-152024-01-19СиЭсПиСи ЧЖУНЦИ ФАРМАСЬЮТИКАЛ ТЕКНОЛОДЖИ (ШИЦЗЯЧЖУАН) КО., ЛТД.Condensed aza-heterocyclic amide compound and its use
WO2021228248A1 (en)2020-05-152021-11-18石药集团中奇制药技术(石家庄)有限公司Fused aza-heterocyclic amide compound and use thereof
JP2023525380A (en)*2020-05-152023-06-15石薬集団中奇制薬技術(石家庄)有限公司 Aza-fused cyclic amide compound and use thereof
CN115551859A (en)*2020-05-152022-12-30石药集团中奇制药技术(石家庄)有限公司Aza condensed ring amide compound and application thereof
RU2811975C9 (en)*2020-05-152024-05-13СиЭсПиСи ЧЖУНЦИ ФАРМАСЬЮТИКАЛ ТЕКНОЛОДЖИ (ШИЦЗЯЧЖУАН) КО., ЛТД.Condensed aza-heterocyclic amide compound and its use
CN113278022B (en)*2020-07-072023-12-12南京纳丁菲医药科技有限公司Pyrazolopyrimidine compound, pharmaceutical composition and application thereof
CN113278022A (en)*2020-07-072021-08-20南京纳丁菲医药科技有限公司Pyrazolopyrimidine compounds, pharmaceutical compositions and uses thereof
WO2022262671A1 (en)*2021-06-152022-12-22中国医药研究开发中心有限公司Macro heterocyclic compound and medical use thereof
CN116120322A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司Salt of aza-condensed ring amide compound, crystal form and use thereof
CN116115618A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司 a drug for treating tumors
CN116120323A (en)*2021-11-152023-05-16石药集团中奇制药技术(石家庄)有限公司Solid form aza-condensed ring amide compound and use thereof
WO2023083356A1 (en)*2021-11-152023-05-19石药集团中奇制药技术(石家庄)有限公司Nitrogen fused-heterocyclic amide compound in solid form and use thereof
CN116115618B (en)*2021-11-152025-07-08石药集团中奇制药技术(石家庄)有限公司Medicine for treating tumor
WO2023083362A1 (en)*2021-11-152023-05-19石药集团中奇制药技术(石家庄)有限公司Drug for treating tumors
WO2023083357A1 (en)*2021-11-152023-05-19石药集团中奇制药技术(石家庄)有限公司Salt of nitrogen-containing fused heterocyclic amide compound, crystal form thereof, and use thereof

Similar Documents

PublicationPublication DateTitle
WO2012034095A1 (en)Compounds and compositions as trk inhibitors
US8637516B2 (en)Compounds and compositions as TRK inhibitors
EP2678338B1 (en)Pyrazolo[1,5-a]pyridines as trk inhibitors
EP2300469B1 (en)Fused nitrogen containing heterocycles and compositions thereof as kinase inhibitors
US8722692B2 (en)Compounds and compositions as Syk kinase inhibitors
WO2022132200A1 (en)Azaquinazoline pan-kras inhibitors
JP2025122005A (en) Degradation of Bruton&#39;s Tyrosine Kinase (BTK) by Conjugation of BTK Inhibitors with E3 Ligase Ligands and Methods of Use
CN106459057B (en) Analogues of 4H-pyrazolo[1,5-α]benzimidazole compounds as PARP inhibitors
CN104974161B (en)The analog of 4H- pyrazolo [1,5- α] benzimidazole compound as PARP inhibitor
WO2020247298A2 (en)1-pyrazolyl, 5-, 6- disubstituted indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
CA3200524A1 (en)Ppar.gamma. modulators and methods of use
TW202334135A (en)Compounds, compositions, and methods

Legal Events

DateCodeTitleDescription
121Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number:11758347

Country of ref document:EP

Kind code of ref document:A1

NENPNon-entry into the national phase

Ref country code:DE

122Ep: pct application non-entry in european phase

Ref document number:11758347

Country of ref document:EP

Kind code of ref document:A1


[8]ページ先頭

©2009-2025 Movatter.jp