NZ718073B2 - Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease - Google Patents

Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease Download PDF

Info

Publication number
NZ718073B2
NZ718073B2 NZ718073A NZ71807314A NZ718073B2 NZ 718073 B2 NZ718073 B2 NZ 718073B2 NZ 718073 A NZ718073 A NZ 718073A NZ 71807314 A NZ71807314 A NZ 71807314A NZ 718073 B2 NZ718073 B2 NZ 718073B2
Authority
NZ
New Zealand
Prior art keywords
phenoxyphenyl
nicotinamide
mmol
esi
title compound
Prior art date
Application number
NZ718073A
Other versions
NZ718073A (en
Inventor
Xiangyang Chen
Yingxiang Gao
Chong Liu
Mark Mulvihill
Haihong Ni
Original Assignee
Guangzhou Innocare Pharma Tech Co Ltd
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 Guangzhou Innocare Pharma Tech Co Ltd filed Critical Guangzhou Innocare Pharma Tech Co Ltd
Priority claimed from PCT/US2014/058084 external-priority patent/WO2015048662A2/en
Publication of NZ718073A publication Critical patent/NZ718073A/en
Publication of NZ718073B2 publication Critical patent/NZ718073B2/en

Links

Abstract

Disclosed are substituted nicotinamide derivatives as inhibitors of Bruton's Tyrosine Kinase (BTK) and are useful in treating associated diseases including cancer, inflammation, and autoimmune diseases.

Description

TUTED NICOTINIMIDE TORS OF BTK AND THEIR PREPARATION AND USE IN THE TREATMENT OF CANCER, INFLAMMATION AND AUTOIMMUNE DISEASE CROSS-REFERENCE TO RELATED APPLICATIONS This ation claims priority under 35 U.S.C. 119(e) to US. Provisional Patent ation No. 61/884,958, filed on September 30, 2013 and entitled "Kinase ting Compounds," and under 35 U.S.C. 1 19(a) to Chinese Patent Application No. 4850481, filed on October 16, 2013 and entitled tic Amide Derivative, Method of its Preparation, and its Application on Medicine." The entire contents of each of the above-referenced applications are incorporated herein by reference.
FIELD AND BACKGROUND The present invention relates to chemical compounds, pharmaceutical compositions including these compounds, and their use in treatment of disease. In particular, the present invention relates to the use of substituted nicotinimides as irreversible inhibitors of tyrosine kinases useful in the treatment of diseases mediated by Bruton’s Tyrosine Kinase (BTK) including cancer, inflammation and autoimmune disease.
BTK is a Tec family ceptor protein kinase which plays a role in multiple signal- transduction ys regulating survival, activation, proliferation, and differentiation of B- lineage lymphoid cells. BTK is overexpressed and active in several B-lineage lymphoid malignancies. BTK is expressed in ant cells in humans with B-Cell Precursor (BCP)— Acute Lymphoblastic Leukemia (ALL), c Lymphocytic Leukemia (CLL), and Non- Hodgkin’s Lymphoma (NHL). BTK is an upstream activator of anti-apoptotic ing molecules and networks, including: Signal Transducer and Activator of Transcription 5 (STAT5) protein, Phosphatidylinositol (PI), 3-kinase/AKT/Mammalian Target of Rapamycin (mTOR) pathway, and Nuclear Factor kappa B (NF-KB). D’Cruz, Osmond J., OncoTargets and Therapy 2013:6,161-176. to De Man et al. describes that BTK is expressed in B cells and myeloid cells, and is a terminal enzyme in the B-Cell antigen Receptor (BCR) signaling pathway. ons in human BTK leads to X-Linked Agammaglobulinemia (XLA), an immunodeficiency disease d to a failure to generate mature B cells leading to reduced immunoglobulin in serum. BTK is therefore implicated in regulation of the tion of auto-antibodies in autoimmune diseases. Furthermore, BTK may play a role in treatment of autoimmune diseases terized by production of flammatory cytokines and chemokines by B cells due to BTK's position in the BCR pathway. BTK inhibitors may be used for treatment of B cell lymphomas due to BTK' s involvement in the regulation of proliferation and apoptosis of B cells. tion of BTK is relevant in ular for B cell lymphomas due to chronic active BCR ing. Davis et al., Nature, 463 (2010), 88-94.
Adaptive immune responses may involve B lymphocyte tion and absence of B lymphocyte tion is an indication of autoimmune disease. Treatment of autoimmune e, such as Rheumatoid Arthritis (RA), with Rituximab, an anti-CD20 therapy, trates that B cell therapies are effective. Additionally, treatment with Rituximab has been shown to improve disease symptoms in Relapsing Remitting Multiple Sclerosis (RRMS) and Systemic Lupus Erythematosus (SLE) patients. Accordingly, targeting B cell immunity is effective for treatment of autoimmune diseases.
SUMMARY The present invention includes certain substituted compounds described herein, their pharmaceutically acceptable salts, solvates and hydrates, preparation of the compounds, intermediates, pharmaceutical compositions and ations thereof, and methods of treating disease including cancers, inflammation, and autoimmune diseases therewith.
It is an object of the present invention to overcome or ameliorate at least one of the disadvantages of the prior art, or to provide a useful alternative.
The present invention includes compounds of Formula I and pharmaceutically acceptable salts thereof as provided below and further defined herein: In some aspects, compounds of the invention are irreversible inhibitors of kinases, including Bruton’s Tyrosine Kinase (BTK). In some aspects, compounds ofthe invention are selective tors of BTK.
In some s, the invention includes methods of treating proliferative disease, particularly cancers, conditions g inflammation, and autoimmune diseases mediated at least in part by BTK, alone or in combination regimens with other therapies.
Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful as inhibitors of protein kinases. In some embodiments, the compounds are effective as tors of Bruton’s ne Kinase (BTK). In some aspects, the invention includes pharmaceutically acceptable salts of the compounds of a I: wherein: A is ed from C3_12cycloalkyl, C3_12heterocycloalkyl, C3_12aryl, or C3_12heteroaryl, any of which is optionally substituted with G1 substituents; B1 is selected from C3_12cycloalkyl—C0_1zalky1—, C34gheterocycloalkyl—C0_12alkyl—, aryl— C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lzheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_1zheterocycloalky1—, any h is ally substituted with G2 substituents; B2 is selected from C0_12alkyl, C3-12cycloalkyl—Co_12alkyl—, C3-12heterocycloalkyl—Co_ 12alkyl—, aryl—C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_12heterocycloalkyl—, heteroaryl—C0_ 12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_lzheterocycloalkyl—, any of which is optionally substituted with G3 substituents; L1 is selected from —C0_2alkyl—, —CR5R6—, —C0_3alkyl(R5)(OH)—, —C(O)—, —CH20—, — OCH2—, —CF2—, —SCR5R6—, —CR5R6S—, —N(R5)—, —N(R5)C(O)—, —C(O)N(R5)—, — N(R5)C(0)N(R")—, —0—, —S—, —S(0)mi—, —N(R5)S(O)mi—, or —S(0)miN(R5)—; L2 is selected from —C0_4alkyl—, , —N(R7)—, —N(R7)C(O)—, or —N(R7)S(O)m2—; X is selected from C342cycloalkyl—C0_12alkyl—, C34gheterocycloalkyl—Cdlzalkyl—, aryl— C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lgheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_1gheterocycloalkyl—, any ofwhich is optionally substituted with G4 tuents; Y is selected from —C(O)—, —N(R8)—, —N(R8)C(O)—, g—, or S(O)m3—; R1 is selected from —C(O)R9,—C(O)NR9R1°,—C(O)OR9, C14alkynyl, 0R9, S(O)m4R9R10, or —CN; R2, R3, and R4 are each independently selected from C0_12alkyl, —CN, halo, C3- 12cycloalkyl—C0_1zalkyl—, C3-1gheterocycloalkyl—C0_12alkyl—, aryl—C0_12alkyl—, aryl—C3_ oalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_lzcycloalkyl—, or heteroaryl—C3_lzheterocycloalkyl—, any ofwhich is optionally substituted with G5 substituents; R5, R6, R7, R8, R9, and R10 are each independently selected from Co_12alkyl, C3- 12cycloalkyl—C0_1zalkyl—, eterocycloalkyl—C0_12alkyl—, aryl—C0_12alkyl—, 3_ 12cycloalkyl—, aryl—C3_1zheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_1chcloalkyl—, or heteroaryl—C3_lzheterocycloalkyl—, any of which is optionally substituted with G6 substituents; G1, G2, G3, G4, G5, and G6 are each independently selected from one or more of C0. lzalkyl, —C2_12alkenyl, —C2_12alkynyl, D, —CD3, —OCD3, halo, CN, OX0 , CF3, OCFg, OCHFz, —N02, —B(OH)2, —1>(0)c0_3 alkyl, —PO(OR")2, —PO(OR")R12, ycloalkyl—C0_ lgalkyl—, C34gheterocycloalkyl—C0_1zalkyl—, aryl—Co_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_ lzheterocycloalkyl—, aryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, heteroaryl—C3_ lzheterocycloalkyl—, —C0_6alkleR11,—OC(O)NR"R12, —C(O)OR",—C(O)NR"R12,—C(O)R", — NRUR", —NR"C(O)R12, —NR13C(0)NR"R", —S(O)m5R", and O)m5R12, any ofwhich is optionally substituted with Q1 substituents; Q1 is selected from one or more of C0_1zalkyl—, —C2_12alkenyl, —C2_12alkynyl, D, halo, — CN, —CD3, —OCD3, —oxo—, —CF3, —OCF3, —OCHF2, —N02, —B(OH)2, —PO(OR14)2, — PO(OR14)R15,NR14R15, —C(O)NR14OH, —C0_6alkleR14, aryl—C0_12alkyl—, heteroaryl—C0_12alkyl— , C34gcycloalkyl—C0_12alkyl—, C3-12heterocycloalkyl—Co_12alkyl—, aryl—C0_1gcycloalkyl—, heteroaryl—C3_12cycloalkyl—, terocycloalkyl—C3_12cycloalkyl—, C3_12cycloalkyl—C3_ 12cycloalkyl—, C3.12heterocycloalkyl—C3_12heterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_12heterocycloalkyl—, —C(O)-C(O)NR14R15, —C(O)-C(O)OR14, —OC(O)R14, — NR14C(O)R15, —NR14S(O)m6R15, —(CR15R16)n1C(O)R14, —(CR15R16)n1C(O)OR14, — 16)H1C(O)NR14R17, —(CR15R16)H1S(O)m6NR14R17, —(CR15R16)H1NR14R17, —(CR15R16)H10R14, —(CR15R16)H18(O)m6R14, —NR16C(O)NR14R15, and —NR16S(O)m6NR14R15, any of which is optionally substituted with independently ed E1 substituents; E1 is selected from one or more of C0_12alkyl—, —C2_12alkenyl, —C2_12alkynyl, D, halo, — CN, —oxo—, —CD3, —OCD3, —CF3, —OCF3, —OCHF2, —N02, —B(OH)2, —PO(OR18)2, — PO(OR18)R19, R180H, —C(O)NR18R19, —C0_12alkleR18, aryl—C0_12alkyl—, heteroaryl—C0_ lzalkyl—, C34gcycloalkyl—C0_12alkyl—, C3-1gheterocycloalkyl—Cmgalkyl—, aryl—C0_12cycloalkyl—, heteroaryl—C3_12cycloalkyl—, C34gheterocycloalkyl—C3_12cycloalkyl—, C3_12cycloalkyl—C3_ lzcycloalkyl—, C34zheterocycloalkyl—C3_1zheterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_12heterocycloalkyl—, —C(O)-C(O)NR18R19, —C0_12alkle(O)OR18, —C(O)- C(O)OR18, —OC(O)R18, —NR18C(O)R19, —NR18C(O)OR19, —NR188(O)m7R19, — (CR19R2°)n2C(O)R18, —(CR19R20)n2C(O)OR18,—(CR19R2°)n2C(O)NR18R21, — )HZS(O)m7NR18R21, —(CR19R20)n2NR18R21, —(CR19R20)HZOR18, —(CR19R20)HZS(O)m7R18’ _ NR2°C(O)NR18R19, and —NR2°S(O)m7NR18R19 substituents; R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, and R21 are each independently ed from H, C1_5alkyl—, C3_gcycloalkyl—C0_6alkyl—, C3_gheterocycloalkyl—C0_6alkyl—, aryl—Co_5alkyl—, aryl—C3_gcycloalkyl—, 3_gheterocycloalkyl—, heteroaryl—C1_6alkyl—, heteroaryl—C3_ 8cycloalkyl—, or heteroaryl—C3-8heterocycloalkyl—g R3 and R4 are taken together with the carbon atoms to which they are attached to form a 3-12 membered partially ted or unsaturated ring, wherein said ring optionally includes one or more onal heteroatoms selected from O, N, or S(O)m8; ml, m2, m3, m4, m5, m6, m7, m8, nl, and n2 are each independently selected from 0, l, or 2; or a pharmaceutically able salt, solvate or a prodrug thereof.
In some aspects of Formula 1, compounds of the present invention are a subgenus of Formula I, having the Formula Ia: wherein: B1 is selected from C3-12cycloalkyl—C0_1zalky1—, C3_12heterocycloalkyl—C0_12alkyl—, aryl— C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lgheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_1zheterocycloalkyl—, any ofwhich is ally substituted with G2 substituents; B2 is selected from C0_12alkyl, ycloalkyl—C0_12alkyl—, eterocycloalkyl—C0_ 12alkyl—, aryl—C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lzheterocycloalkyl—, heteroaryl—C0_ 12alkyl—, aryl—C3_12cycloalkyl—, or heteroaryl—C3.12heterocycloalkyl—, any of which is ally substituted with G3 substituents; L1 is selected from —C0_2alkyl—, —CR5R6—, —C0_3alkyl(R5)(OH)—, —C(O)—, —CH20—, — OCH2—, —CF2—, 6—, —CR5R6S—, —N(R5)—, —N(R5)C(O)—, —C(O)N(R5)—, — N(R5)C(O)N(R6)—i -O—i —S-i _S(O)m1_a _N(R5)S(O)m1_a 0r —S(O)m1N(R5)-; L2 is selected from —C0_4alkyl—, —C(O)—, —N(R7)—, —N(R7)C(O)—, or —N(R7)S(O)m2—; X is selected from C3_12cycloalkyl—C0_12alkyl—, C34gheterocycloalkyl—Cdlzalkyl—, aryl— C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lgheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroal'yl—C3_1gheterocycloalkyl—, any ofwhich is optionally substituted with G4 substituents; Y is selected from , —N(R8)—, —N(R8)C(O)—, —S(O)m3—, or —N(R8)S(O)m3—; Z1 is (CRa)o-1; Z2 is selected from CRb, NRb, O, or S; Z3 is selected from C or N; Z4 is selected from CRC, NR°, O, or S; R1 is selected from —C(O)R9,—C(O)NR9R10,—C(O)OR9, C1_4alkynyl, 0R9, S(O)m4R9R1°, or —CN; R2, R3, R4, Ra, Rb, and R0 are each independently selected from C0_12alkyl, —CN, halo, C3- 12cycloalkyl—C0_1zalkyl—, C3_1gheterocycloalkyl—C0_12alkyl—, aryl—C0_12alkyl—, aryl—C3_ 12cycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—Co_12all(0)c0_3 alkyl, —PO(OR")2, —PO(OR")R12, C3_12cycloalkyl—C0_ lzalkyl—, C3_1gheterocycloalkyl—C0_lzalky1—, aryl—C0_12alkyl—, 3_12cycloalky1—, aryl—C3_ 12heterocycloalkyl—, heteroaryl—C0_12alkyl—, aryl—C3_1zcycloalkyl—, heteroaryl—C3_ lzheterocycloalkyl—, —C0_6a1kyIOR11,—OC(O)NR"R12, —C(O)OR", —C(O)NR"R12, —C(O)R", — NRUR", —NR"C(O)R12, —NR"C(0)NR"R", —S(O)m5R", and —NR"S(O)m5R12, any ofwhich is optionally substituted with independently selected Q1 substituents; Q1 is selected from one or more of C0_12alky1—, —C2_12alkenyl, —C2_12alkyny1, D, halo, — CN, —CD3, —OCD3, —oxo—, —CF3, —OCF3, —OCHF2, —N02, —B(OH)2, 14)2, — PO(OR14)R15,NR14R15, —C(O)NR14OH, —C0_6alky10R14, aryl—Co_12alkyl—, heteroaryl—C0_12alkyl— , ycloalkyl—C0_12alkyl—, C3-12heterocycloalkyl—C0_12alkyl—, aryl—C0_1zcycloalkyl—, heteroaryl—C3_12cycloalky1—, C34zheterocycloalkyl—C3_12cycloalkyl—, C3_12cycloalkyl—C3_ 12cycloalkyl—, C34gheterocycloalkyl—C3_12heterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_lzheterocycloalkyl: —C(O)-C(O)NR14R15, —C(O)-C(O)OR14, R14, — NR14C(O)R15, —NR14S(O)m6R15, —(CR15R16)H1C(O)R14, —(CR15R16)H1C(O)OR14, — (CR15R16)H1C(O)NR14R17, —(CR15R16)H1S(O)m6NR14R17, —(CR15R16)n1NR14R17, —(CR15R16)H10R14, R16)HIS(O)m6R14, —NR16C(O)NR14R15, and —NR16S(O)m6NR14R15, any of which is optionally substituted with independently selected El substituents; E1 is ed from one or more of C0_12alky1—, —C2_12alkenyl, —C2_12alkynyl, D, halo, — CN, —oxo—, —CD3, —OCD3, —CF3, —OCF3, , —N02, —B(OH)2, —PO(OR18)2, — PO(OR18)R19, —C(O)NR180H, R18R19, —C0_12alkleR18, aryl—C0_12alkyl—, heteroaryl—C0_ lzalkyl—, C342cycloalkyl—C0_12alkyl—, C34zheterocycloalkyl—C0_1zalkyl—, aryl—C0_1zcycloalkyl—, heteroaryl—C3_ucycloalkyl—, C3.12heterocycloalkyl—C3_12cycloalkyl—, C3_12cycloalkyl—C3_ 12cycloalkyl—, C34gheterocycloalkyl—C3_12heterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_lzheterocycloalkyl—, —C(O)—C(O)NR18R19, alkle(O)OR18, —C(O)- C(O)OR18, —OC(O)R18, —NR18C(O)R19, —NR18C(O)OR19, —NRISS(O)m7R19, — (CR19R2°)n2C(O)R18, —(CR19R20)n2C(O)OR18,—(CR19R2°)n2C(O)NR18R21, — (CR19R20)H28(O)m7NR18R21, —(CR19R20)n2NR18R21, —(CR19R20)HZOR18, —(CR19R20)HZS(O)m7R18, _ O)NR18R19, and —NRZOS(O)m7NR18R19 substituents; R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, and R21 are each independently selected from H, C1_6alkyl—, C3_gcycloalkyl—C0_6alkyl—, C3_gheterocycloalkyl—C0_6alkyl—, aryl—C0_6alkyl—, aryl—C3_gcycloalkyl—, aryl—C3_gheterocycloalkyl—, heteroaryl—C1_5alkyl—, heteroaryl—C3_ gcycloalkyl—, or heteroaryl—C3_gheterocycloalkyl—; R3 and R4 are taken together with the carbon atoms to which they are attached to form a 3-12 ed partially saturated or unsaturated ring, wherein said ring ally includes one or more additional heteroatoms selected from O, N, or S(O)mg; ml, m2, m3, m4, m5, m6, m7, m8, n1, and n2 are each independently selected from 0, l, or 2; or a pharmaceutically acceptable salt, e or a prodrug thereof.
In some aspects of Formula 1, compounds of the present invention are a subgenus of a I selected from one of Formulas Ib-Ii: R4 o \ leB2 R3\H\ / L2 A\ / Y/ \X N R2 and R4 N \ L1\B2 1 / R3 L2 / Y/ \x 0 wherein: B1 is selected from C3-12cycloalkyl—C0_1zalky1—, C3_12heterocycloalkyl—C0_12alkyl—, aryl— C0_lzalkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lzheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_1gheterocycloalkyl—, any ofwhich is ally substituted with G2 substituents; B2 is selected from C0_12alkyl, ycloalkyl—C0_12alkyl—, C3-12heterocycloalkyl—C0_ 12alkyl—, aryl—C0_12alkyl—, 3_12cycloalkyl—, aryl—C3_lzheterocycloalkyl—, heteroaryl—C0_ 12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_12heterocycloalkyl—, any of which is optionally substituted with G3 substituents; L1 is selected from lkyl—, —CR5R6—, —C0_3alkyl(R5)(OH)—, —C(O)—, —CHzO—, — OCH2—, —CF2—, —SCR5R6—, —CR5R6S—, —N(R5)—, —N(R5)C(O)—, —C(O)N(R5)—, — N(R5)C(0)N(R6)-a , -S-» -S(0)m1-, -N(R5)S(O)m1-, 0r -S(0)m1N(R5)-; L2 is selected from —C0_4alkyl—, , —N(R7)—, —N(R7)C(O)—, or —N(R7)S(O)m2—; X is selected from C3_1zcycloalkyl—C0_12alkyl—, C34gheterocycloalkyl—C0_lzalkyl—, aryl— C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_lgheterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, or heteroaryl—C3_1zheterocycloalkyl—, any ofwhich is optionally tuted with G4 substituents; Y is selected from —C(O)—, —N(R8)—, —N(R8)C(O)—, —S(O)m3—, or —N(R8)S(O)m3—; R1 is selected from —C(O)R9,—C(O)NR9R10,—C(O)OR9, nyl, 0R9, S(O)m4R9R1°, or —CN; R2, R3, and R4 are each independently selected from C0_12alkyl, —CN, halo, C3- 12cycloalkyl—C0_1zalkyl—, C34gheterocycloalkyl—C0_12alkyl—, aryl—C0_12alkyl—, aryl—C3_ 12cycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C0_12alkyl—, aryl—C3_lgcycloalkyl—, or heteroaryl—C3_12heterocycloalkyl—, any of which is optionally substituted with G5 substituents; R5, R6, R7, R8, R9, and R10 are each ndently selected from C0_12alkyl, C3- 12cycloalkyl—C0_1galkyl—, C3.1gheterocycloalkyl—Co_1zalky1—, aryl—C0_12alkyl—, aryl—C3_ 12cycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C0_12alkyl—, aryl—C3_lzcycloalkyl—, or heteroaryl—C3_1zheterocycloalkyl—, any of which is optionally substituted with G6 substituents; G1, G2, G3, G4, G5, and G6 are each independently selected from one or more of C0- 12alkyl, —C2_12alkenyl, —C2_12alkynyl, D, —CD3, —OCD3, halo, CN, OX0 , CF3, OCFg, OCHFZ, —N02, —B(OH)2, —1>(0)c0_3 alkyl, —PO(OR")2, ")R12, C3_12cycloalkyl—C0_ lzalkyl—, C34gheterocycloalkyl—C0_1Zalkyl—, aryl—C0_12alkyl—, aryl—C3_12cycloalkyl—, aryl—C3_ 12heterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, heteroaryl—C3_ 12heterocycloalkyl—, —C0_6alkleR11,—OC(O)NR"R12, —C(O)OR", —C(O)NR"R12, —C(O)R", — NRllRlz, —NR"C(O)R12, 0)NR"R", —S(O)m5R", and —NR"S(O)m5R12, any h is optionally tuted with independently selected Q1 substituents; Q1 is selected from one or more of Co_1zalkyl—, —C2_12alkenyl, —C2_12alkynyl, D, halo, — CN, —CD3, —OCD3, —oxo—, —CF3, —OCF3, —OCHF2, —N02, —B(OH)2, —PO(OR14)2, — PO(OR14)R15,NR14R15, —C(O)NR14OH, —C0_6alkleR14, aryl—C0_12alkyl—, heteroaryl—C0_12alkyl— , ycloalkyl—C0_12alkyl—, C3-12heterocycloalkyl—C0_lzalkyl—, aryl—C0_12cycloalkyl—, heteroaryl—C3_12cycloalkyl—, C34gheterocycloalkyl—C3_12cycloalkyl—, C3_1zcycloalkyl—C3_ 12cycloalkyl—, C34gheterocycloalkyl—C3_12heterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_12heterocycloalkyl—, —C(O)-C(O)NR14R15, —C(O)-C(O)OR14, —OC(O)R14, — NR14C(O)R15, —NR14S(O)m6R15, —(CR15R16)H1C(O)R14, —(CR15R16)n1C(O)OR14, — (CR15R16)H1C(O)NR14R17, —(CR15R16)H1S(O)m6NR14R17, —(CR15R16)H1NR14R17, R16)n10R14, —(CR15R16)nlS(O)m6R14, —NR16C(O)NR14R15, (O)m6NR14R15, any ofwhich is optionally substituted with independently selected E1 substituents; E1 is selected from one or more of C0_12alkyl—, —C2_12alkenyl, —C2_12alkynyl, D, halo, — CN, —oxo—, —CD3, —OCD3, —CF3, —OCF3, —OCHF2, —N02, —B(OH)2, —PO(OR18)2, — PO(OR18)R19, —C(O)NR180H, —C(O)NR18R19, —C0_12alkleR18, aryl—C0_12alkyl—, heteroaryl—C0_ 12alkyl—, C3_1gcycloalkyl—C0_1zalkyl—, C34gheterocycloalkyl—Cmzalkyl—, aryl—C0_12cycloalkyl—, heteroaryl—C3_12cycloalkyl—, C34gheterocycloalkyl—C3_12cycloalkyl—, C3_1zcycloalkyl—C3_ 12cycloalkyl—, C34gheterocycloalkyl—C3_12heterocycloalkyl—, aryl—C3_1gheterocycloalkyl—, heteroaryl—C3_12heterocycloalkyl—, —C(O)-C(O)NR18R19, —C0_12alkle(O)OR18, —C(O)- C(O)OR18, —OC(O)R18, —NR18C(O)R19, —NR18C(O)OR19, —NR18S(0)m7R19, — (CR19R2°)n2C(O)R18, —(CR19R20)HZC(O)OR18, R20)n2C(O)NR18R21, — (CR19R20)HZS(O)m7NR18R21, —(CR19R20)n2NR18R21, —(CR19R20)HZOR18, —(CR19R20)HZS(O)m7R18, _ NR20C(O)NR18R19, and —NRZOS(O)m7NR18R19 substituents; R11, R12, R13, R14, R15, R16, R17, R18, R19, R20, and R21 are each independently selected from H, C1_6alkyl—, C3_gcycloalkyl—C0_6alkyl—, C3_gheterocycloalkyl—C0_6alkyl—, aryl—C0_6alkyl—, aryl—C3_gcycloalkyl—, aryl—C3_8heterocycloalkyl—, heteroaryl—C1_6alkyl—, heteroaryl—C3_ gcycloalkyl—, or heteroaryl—C3_gheterocycloalkyl—; R3 and R4 are taken together with the carbon atoms to which they are attached to form a 3-12 membered lly saturated or unsaturated ring, wherein said ring optionally es one or more additional atoms selected from O, N, or S(O)mg; ml, m2, m3, m4, m5, m6, m7, m8, n1, and n2 are each independently selected from 0, 1, or 2; or a ceutically acceptable salt, solvate or a prodrug thereof.
In some ments of Formulas I, and Ia-Ii, B1 is selected from C4_gcycloalkyl—C0_ l—, C4_gheterocycloalkyl—C0_12alkyl—, C4_garyl—C0_12alkyl—, or C4_gheteroaryl—C0_12alkyl—, any of which is optionally substituted with G2 substituents.
In some embodiments of Formulas I, and Ia-Ii, B2 is selected from C4_gcycloalkyl—Co_ 12alkyl—, C4_gheterocycloalkyl—C0_12alkyl—, C4_garyl—C0_12alkyl—, or C4_gheteroaryl—C0_12alkyl—, any of which is optionally substituted with G3 substituents.
In some embodiments of Formulas I, and Ia-Ii, L1 is selected from —C0_2alkyl—, —CR5R6—, —C0_3alkyl(R5)(OH) , C(O) , CHZO , OCH2 , CF2 , N(R5) , N(R5)C(O)—, —C(O)N(R5)—, —O—, or —S(O)m1—.
In some ments of Formulas I, and Ia-Ii, L1 is selected from —C0_2alkyl—, —CR5R6—, —C1_2alkyl(R5)(OH)—, —C(O)—, —CF2—, —N(R5)—, —N(R5)C(O)—, —C(O)N(R5)—, —o—, or 1—.
In some embodiments of Formulas I, and Ia-Ii, L2 is selected from —C0_2alkyl—, , or —N(R7)—.
In some embodiments of Formulas I, and Ia-Ii, X is selected from C4_8cycloalkyl—C0_ 12alkyl—, C4_gheterocycloalkyl—C0_12alkyl—, C4_garyl—C0_12alkyl—, or C4_gheteroaryl—C0_12alkyl—.
In some embodiments of as I, and Ia-Ii, Y is selected from —C(O)—, —N(R8)—, — N(R8)C(O)—, or —S(O)m3—.
In some embodiments of Formulas I, and Ia—Ii, R1 is selected from —C(O)R9, — C(O)NR9R10, —C(O)OR9, C1_4alkynyl, or —CN.
In some embodiments of as I, and Ia-Ii, R2, R3, and R4 are each independently selected from C0_12alkyl, —CN, halo, C3_6cycloalkyl—C0_12alkyl—, C3_6heterocycloalkyl—C0_1galkyl— of which is optionally substituted with G5 tuents. , any In some embodiments of Formulas I, and Ia-Ii, G1, G2, G3, G4, G5, and G6 are each independently selected from one to three of C0_12alkyl, —C2_12alkenyl, —C2_12alkynyl, D, —CD3, — OCD3, halo, CN, oxo , CF3, OCF3, OCHFZ, —N02, —B(OH)2, —P(O)C0_3 alkyl, — PO(OR")2, —PO(OR")R12, C34zcycloalkyl—C0_12alkyl—, C3-12heterocycloalkyl—C0_12alkyl—,aryl— C0_12alkyl—, aryl—C3_1zcycloalkyl—, aryl—C3_12heterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, heteroaryl—C3-12heterocycloalkyl—, —C0_6all In some ments of Formulas I, and Ia-Ii, G1, G2, G3, G4, G5, and G6 are each independently selected from one to two of C0_12alkyl, —C2_12alkenyl, —C2_12alkynyl, D, —CD3, — OCD3, halo, CN, oxo , CF3, OCF3, OCHFZ, —N02, —B(OH)2, —P(O)C0_3 alkyl, — PO(OR")2, —PO(OR")R12, C34zcycloalkyl—C0_12alkyl—, C3_12heterocycloalkyl—C0_12alkyl—,aryl— C0_12alkyl—, aryl—C3_1zcycloalkyl—, 3_12heterocycloalkyl—, heteroaryl—C0_12alkyl—, heteroaryl—C3_12cycloalkyl—, aryl—C3_12heterocycloalkyl—, —C0_6all In some aspects, the t invention includes a nd selected from the group consisting of: 6-(3-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; 6-(4-acryloylpiperazinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(4-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; O N N O 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; O NH2 N N crylamidopiperidinyl)(4-phenoxyphenyl)nicotinamide; O N N N O 6-(3-acrylamidopyrrolidinyl)(4-phenoxyphenyl)nicotinamide; HN N N 1-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N N O 1-(1-acryloylazetidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O 1-(4-acrylamidophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O 1-(3-acrylamidophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N N N O (S)(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N O N N (1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N N O 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)pyrimidinecarboxamide; N NH2 N N (4-(4-(dimethylamino)butenoyl)piperazinyl)(4- phenoxyphenyl)nicotinamide; N N N O 6-(4-acryloylpiperazinyl)(4-(cyclohexyloxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(3-methoxymethylphenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)-6'-phenoxy-2,3'-bipyridinecarboxamide; N N N O 6-(4-acryloylpiperazinyl)(4-(pyridinyloxy)phenyl)nicotinamide; N N O N 1-(1-(4-(dimethylamino)butenoyl)pyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazole- 4-carboxamide; O N N O 6-(4-acryloylpiperazinyl)(4-(3-fluorophenoxy)phenyl)nicotinamide; N N O F 6-(4-acryloylpiperazinyl)(3-fluorophenoxyphenyl)nicotinamide; N N cryloylpiperazinyl)(4-(4-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(4-(2-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(2-fluorophenoxyphenyl)nicotinamide; N N F O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; O S N N cryloylpyrrolidinyl)(4-phenoxyphenyl)thiazolecarboxamide; N N O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; O O N N O 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)oxazolecarboxamide; N N 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)thiazolecarboxamide; N S 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; O N N S O cryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; N O 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)oxazolecarboxamide; O O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; S O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; N O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; O N N O O -(1-acryloylpiperidinyl)-4'-phenoxybiphenylcarboxamide; -(4-acryloylpiperazinyl)-4'-phenoxybiphenylcarboxamide; 6-(1-acryloylpiperidinyl)(4-(hydroxy(phenyl)methyl)phenyl)nicotinamide; O OH (1-(2-cyanobutenoyl)azetidinyl)(4-phenoxyphenyl)-1H-pyrazole carboxamide; N O 6-(4-acryloylpiperazinyl)(4-((4,4-difluorocyclohexyl)oxy)phenyl)nicotinamide; N N F cryloylpiperidinyl)(4-(phenylcarbamoyl)phenyl)nicotinamide; N N O O 2-(4-phenoxyphenyl)(4-(vinylsulfonyl)piperazinyl)nicotinamide; N N S O 2-(4-phenoxyphenyl)(1-(vinylsulfonyl)piperidinyl)nicotinamide; S O or a pharmaceutically acceptable salt thereof.
In some aspects, the present invention includes a compound selected from the group consisting of: 2-(4-phenoxyphenyl)(piperidinyl)nicotinamide; 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide; N N tidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O 1-(3-aminophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; H2N N O 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)pyridinecarbonitrile; 6-(4-acryloylpiperazinyl)(4-phenoxyphenyl)pyridinecarboxylic acid; N N 2-(4-acryloylpiperazinyl)-N-methyl(4-phenoxyphenyl)thiazolecarboxamide; N O O .
In some aspects, the present ion includes a compound having a Formula IId, or a pharmaceutically acceptable salt or solvate thereof: R3 X Z R2 O O Formula IId N N N N wherein X is , , , or ; Z is C or N; G1, G2, and G3 are selected from the group ting of one or more H, D (deuterium), C1-12 alkyl, halogen, and CF3; R2 and R3 are each independently selected from the group consisting of H, D, C1-12 alkyl, halogen, CN, and CF3, wherein C1-12 alkyl is ally substituted with NR11R12; R4 is selected from the group consisting of H, D, C1-12 alkyl, halogen, CN, and CF3; and R11 and R12 are independently H or C1-6 alkyl.
In some aspects, the t invention includes a pharmaceutical ition for treating a disease mediated by Bruton’s tyrosine Kinase (BTK), comprising a compound of Formula I or a nd of Formula IId described herein, and a pharmaceutically acceptable salt thereof.
In some aspects, the present invention includes a use of a compound described herein in the manufacture of a medicament for treating of at least one of cancer, chronic inflammation, and autoimmune e mediated at least in part by BTK.
In some aspects, the present invention includes a ceutical composition including the compound or salt of any one of the compounds of Formula I, formulated with or without one or more pharmaceutical carriers.
In some aspects, the present invention includes a method for the treatment of at least one of cancer, chronic inflammation, and autoimmune disease mediated at least in part by BTK including administering to a subject in need thereof a therapeutically effective amount of a compound or salt of the compound of Formula I.
In some aspects, the present invention includes a method of treating cancer, chronic inflammation, or autoimmune disease in a mammal including administering a therapeutically effective amount of a compound of Formula I, or a pharmaceutically able salt thereof, to the mammal in need thereof.
In some aspects, the present invention includes a method of irreversibly ting tyrosine s, the method including administering to a patient a therapeutically effective amount of a tyrosine kinase inhibitor including a compound of Formula I.
In some aspects, the present invention includes a method of irreversibly inhibiting BTK, the method including administering to a patient a therapeutically effective amount of a BTK inhibitor ing a compound according to Formula I.
The methods described herein include administering to a subject in need a composition containing a therapeutically effective amount of one or more BTK tor compounds described herein. t being bound by theory, the diverse roles played by BTK signaling in s hematopoietic cell functions, e.g., B-cell receptor activation, suggests that small molecule BTK inhibitors are useful for ng the risk of or treating a variety of es affected by or affecting many cell types of the hematopoetic lineage including, e.g., autoimmune diseases, heteroimmune ions or diseases, inflammatory diseases, cancer ( e.g., B-cell proliferative disorders), and thromboembolic disorders. Further, the BTK inhibitor nds described herein can be used to inhibit a small subset of other tyrosine kinases that share homology with BTK by having a cysteine e (including a Cys 481 residue) that can form a [Followed by page 15]. covalent bond with the tor. Thus, a subset of tyrosine s other than BTK may be usefiil as therapeutic targets in a number of health conditions.
The methods described herein can be used to treat an autoimmune disease, which includes, but is not d to, rheumatoid arthritis, psoriatic tis, osteoarthritis, Still's disease, juvenile arthritis, lupus, diabetes (type I and type II), myasthenia gravis, oto's thyroiditis, Ord's thyroiditis, Graves' disease Sjogren's syndrome, multiple sclerosis, Guillain- Barre syndrome, acute disseminated encephalomyelitis, Addison's disease, opsoclonus- myoclonus me, ankylosing spondylitisis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, c disease, sture's syndrome, idiopathic ocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener‘s granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, dysautonomia, endometriosis, interstitial cystitis, neuromyotonia, scleroderma, and vulvodynia.
The methods described herein can be used to treat heteroimmune conditions or diseases, which include, but are not limited to graft versus host e, transplantation, usion, anaphylaxis, allergies (e. g., allergies to plant pollens, latex, drugs, foods, insect poisons, animal hair, animal dander, dust mites, or cockroach calyx), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, allergic asthma, and atopic dermatitis.
The methods described herein can be used to treat an inflammatory disease, which includes, but is not limited to asthma, inflammatory bowel disease, icitis, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, ystitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatitis, omyositis, alitis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, hepatitis, hidradenitis suppurativa, laryngitis, mastitis, meningitis, is myocarditis, myositis, tis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, rditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, pneumonia, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, itis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, and vulVitis.
The methods described herein can be used to treat a cancer, e. g., B-cell proliferative disorders, which include, but are not limited to diffuse large B cell lymphoma, ular lymphoma, chronic lymphocytic lymphoma, chronic lymphocytic leukemia, B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, cytoma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary on lymphoma, t lymphoma/leukemia, and lymphomatoid granulomatosis.
The methods described herein can be used to treat thromboembolic disorders, which include, but are not limited to myocardial infarct, angina pectoris (including unstable angina), reocclusions or restenoses after angioplasty or oronary bypass, stroke, transitory ischemia, peripheral arterial occlusive ers, pulmonary embolisms, and deep venous thromboses.
Disclosed herein is a method for ng a hematological malignancy in an dual in need thereof, comprising: administering to the individual a composition containing a eutic amount of at least one compound having the structure of as I, Ia-Ii or IIa-IId.
The hematological malignancy is a chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high risk CLL, or a non-CLL/SLL lymphoma. In some embodiments, the hematological malignancy is follicular lymphoma, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma, Waldenstrom's lobulinemia, multiple a, marginal zone lymphoma, Burkitt's lymphoma, rkitt high grade B cell ma, or extranodal marginal zone B cell lymphoma. In some embodiments, the logical malignancy is acute or chronic myelogenous (or myeloid) leukemia, myelodysplastic syndrome, or acute lymphoblastic leukemia. In some embodiments, the hematological malignancy is relapsed or refractory diffuse large B-cell lymphoma (DLBCL), relapsed or refractory mantle cell lymphoma, relapsed or refractory follicular lymphoma, relapsed or refractory CLL; relapsed or refractory SLL; relapsed or refractory multiple myeloma.
In some embodiments, the hematological malignancy is a hematological malignancy that is classified as high-risk. In some embodiments, the hematological malignancy is high risk CLL or high risk SLL.
B-cell proliferative disorders (BCLDs) are neoplasms of the blood and encompass, inter alia, non-Hodgkin lymphoma, multiple myeloma, and leukemia. BCLDs can originate either in the lymphatic tissues (as in the case of lymphoma) or in the bone marrow (as in the case of leukemia and myeloma), and they all are involved with the uncontrolled growth of lymphocytes or white blood cells. There are many subtypes of BCLD, e.g., chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). The disease course and treatment of BCLD is dependent on the BCLD e; however, even within each subtype the clinical presentation, morphologic appearance, and response to y is heterogeneous.
Malignant lymphomas are neoplastic transformations of cells that reside predominantly within lymphoid tissues. Two groups of malignant lymphomas are Hodgkin's lymphoma and non-Hodgkin's lymphoma (NHL). Both types of lymphomas infiltrate reticuloendothelial s.
However, they differ in the neoplastic cell of origin, site of disease, presence of systemic symptoms, and response to treatment (Freedman et al., "Non—Hodgkin's Lymphomas" Chapter 134, Cancer ne, (an approved publication of the American Cancer Society, B.C. Decker Inc., Hamilton, Ontario, 2003).
Disclosed herein is a method for treating a non-Hodgkin's lymphoma in an individual in need f, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the ure of Formulas I, Ia-Ii or Ila-11d.
Further disclosed herein, is a method for treating relapsed or refractory non-Hodgkin's lymphoma in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia—Ii or Ila-11d. In some ments, the non-Hodgkin's lymphoma is relapsed or refractory diffuse large B-cell lymphoma (DLBCL), relapsed or refractory mantle cell lymphoma, or relapsed or refractory follicular ma.
Non-Hodgkin lymphomas (NHL) are a diverse group of malignancies that are predominately of B-cell origin. NHL may p in any organs associated with lymphatic system such as spleen, lymph nodes or tonsils and can occur at any age. NHL is often marked by enlarged lymph nodes, fever, and weight loss. NHL is classified as either B-cell or T-cell NHL.
Lymphomas related to lymphoproliferative disorders following bone marrow or stem cell transplantation are usually B-cell NHL. In the Working Formulation classification scheme, NHL has been divided into low-, ediate-, and high-grade categories by virtue of their natural histories (see "The Non-Hodgkin's Lymphoma Pathologic Classification Project," Cancer 49 (1982):2112—2135). The low-grade lymphomas are indolent, with a median survival of 5 to 10 years (Homing and Rosenberg (1984) N. Engl. J. Med. 311:1471-1475). Although herapy can induce ions in the majority of indolent lymphomas, cures are rare and most patients eventually e, ing r y. The intermediate- and high-grade lymphomas are more sive tumors, but they have a greater chance for cure with chemotherapy. However, a significant proportion of these ts will relapse and require further treatment.
A miting list of the B-cell NHL includes Burkitt's lymphoma (e.g., Endemic Burkitt's Lymphoma and Sporadic Burkitt's Lymphoma), Cutaneous B-Cell ma, ous al Zone Lymphoma (MZL), Diffiise Large Cell Lymphoma (DLBCL), Diffuse Mixed Small and Large Cell Lympoma, Diffuse Small Cleaved Cell, e Small Lymphocytic Lymphoma, odal Marginal Zone B-cell lymphoma, follicular lymphoma, Follicular Small Cleaved Cell (Grade 1), Follicular Mixed Small Cleaved and Large Cell (Grade 2), Follicular Large Cell (Grade 3), Intravascular Large B—Cell Lymphoma, Intravascular Lymphomatosis, Large Cell Immunoblastic Lymphoma, Large Cell Lymphoma (LCL), Lymphoblastic Lymphoma, MALT Lymphoma, Mantle Cell Lymphoma (MCL), immunoblastic large cell lymphoma, precursor B—lymphoblastic lymphoma, mantle cell lymphoma, chronic cytic leukemia (CLL)/small lymphocytic lymphoma (SLL), extranodal marginal zone B-cell lymphoma-mucosa-associated lymphoid tissue (MALT) lymphoma, Mediastinal Large B-Cell Lymphoma, nodal marginal zone B-cell lymphoma, splenic marginal zone B-cell lymphoma, primary mediastinal B-cell ma, lymphoplasmocytic lymphoma, hairy cell leukemia, Waldenstrom's Macroglobulinemia, and primary l nervous system (CNS) lymphoma.
Additional non-Hodgkin's lymphomas are contemplated within the scope of the present invention and apparent to those of ordinary skill in the art.
Disclosed herein is a method for treating a DLCBL in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia-Ii or Ila-11d.
As used herein, the term "Diffuse large B-cell lymphoma (DLBCL)" refers to a neoplasm of the germinal center B lymphocytes with a diffuse growth n and a high-intermediate proliferation index. DLBCLs represent approximately 30% of all lymphomas and may present with several morphological variants including the blastic, immunoblastic, T-cell/histiocyte rich, stic and plasmoblastic subtypes. Genetic tests have shown that there are different subtypes of DLBCL. These subtypes seem to have ent outlooks (prognoses) and responses to treatment. DLBCL can affect any age group but occurs mostly in older people (the average age is mid-60$).
Disclosed herein is a method for treating diffuse large B-cell lymphoma, activated B cell- like subtype (ABC-DLBCL), in an individual in need thereof, comprising: administering to the individual an irreversible BTK inhibitor in an amount from 300 mg/day up to, and including, 1000 . The ABC subtype of diffuse large B-cell lymphoma (ABC-DLBCL) is t to arise from post germinal center B cells that are ed during plasmatic differentiation. The ABC subtype of DLBCL (ABC-DLBCL) accounts for approximately 30% total DLBCL diagnoses. It is considered the least curable of the DLBCL molecular subtypes and, as such, patients sed with the ABC-DLBCL typically display significantly d survival rates compared with individuals with other types of DLCBL. ABC-DLBCL is most ly associated with chromosomal translocations deregulating the germinal center master regulator BCL6 and with mutations inactivating the PRDMl gene, which encodes a transcriptional repressor required for plasma cell differentiation.
A particularly relevant signaling pathway in the pathogenesis ofABC-DLBCL is the one mediated by the nuclear factor (NF)-1 The dependence ofABC DLBCLs on NF-KB s on a signaling pathway upstream of IkB kinase comprised of CARDl l, BCLlO and MALTl (the CBM complex). Interference with the CBM pathway extinguishes NF-KB ing in ABC DLBCL cells and induces apoptosis. The molecular basis for constitutive ty of the NF-KB y is a subject of current investigation but some somatic alterations to the genome ofABC DLBCLs clearly invoke this pathway. For example, somatic ons of the coiled—coil domain of CARDll in DLBCL render this signaling scaffold protein able to spontaneously nucleate protein-protein interaction with MALTl and BCL10, causing IKK activity and NF-KB activation. Constitutive activity of the B cell receptor signaling y has been implicated in the activation ofNF-KB in ABC DLBCLs with wild type CARDl l, and this is associated with mutations within the cytoplasmic tails of the B cell receptor subunits CD79A and CD79B. Oncogenic activating mutations in the ing adapter MYD88 activate NF-KB and synergize with B cell receptor signaling in ning the survival ofABC DLBCL cells. In addition, inactivating mutations in a negative regulator of the NF-KB pathway, A20, occur almost ively in ABC DLBCL.
Indeed, genetic alterations ing multiple components of the NF-KB signaling pathway have been recently identified in more than 50% DLBCL patients, where these lesions promote constitutive NF-1 An even larger fraction of cases (30%) carry biallelic genetic lesions inactivating the negative NF-KB regulator A20. Further, high levels of expression ofNF-KB target genes have been observed in ABC-DLBCL tumor samples. See, e.g., U. Klein et al., (2008), Nature Reviews Immunology 8:22-23; R. E. Davis et al., (2001), Journal of Experimental Medicine 61- 1874; G. Lentz et al., (2008), Science 319:1676-1679; M. Compagno et al., (2009), Nature 459:712-721; and L. Srinivasan et al., (2009), Cell 139:573—586).
Disclosed herein is a method for treating a follicular lymphoma in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia-Ii or IIa-IId.
As used herein, the term "follicular ma" refers to any of several types of non- Hodgkin's lymphoma in which the lymphomatous cells are clustered into nodules or follicles.
The term follicular is used because the cells tend to grow in a circular, or nodular, pattern in lymph nodes. The average age for people with this lymphoma is about 60.
Disclosed herein is a method for treating a CLL or SLL in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia-Ii or d. c lymphocytic leukemia and small cytic lymphoma (CLL/SLL) are commonly thought as the same disease with slightly ent manifestations. Where the cancerous cells gather determines whether it is called CLL or SLL. When the cancer cells are primarily found in the lymph nodes, lima bean shaped structures of the lymphatic system (a system primarily of tiny vessels found in the body), it is called SLL. SLL accounts for about 5% to 10% of all lymphomas. When most of the cancer cells are in the tream and the bone marrow, it is called CLL.
Both CLL and SLL are slow-growing diseases, although CLL, which is much more common, tends to grow slower. CLL and SLL are treated the same way. They are usually not considered curable with standard treatments, but depending on the stage and growth rate of the disease, most patients live longer than 10 years. Occasionally over time, these slow—growing mas may transform into a more aggressive type of lymphoma.
Chronic lymphoid leukemia (CLL) is the most common type of leukemia. It is ted that 100,760 people in the United States are living with or are in remission from CLL. Most (>75%) people newly diagnosed with CLL are over the age of 50. Currently CLL treatment focuses on lling the disease and its symptoms rather than on an outright cure. CLL is treated by chemotherapy, radiation therapy, biological therapy, or bone marrow transplantation.
Symptoms are sometimes treated surgically (splenectomy removal of enlarged spleen) or by radiation therapy ("de-bulking" n lymph nodes). Though CLL progresses slowly in most cases, it is considered generally incurable. Certain CLLs are classified as high-risk. As used herein, "high risk CLL" means CLL characterized by at least one of the following 1) ; 2) 11q22—; 3) unmutated IgVH together with ZAP-70+ and/or CD3 8+; or 4) trisomy 12.
CLL treatment is typically stered when the t's clinical symptoms or blood counts indicate that the disease has progressed to a point where it may affect the patient's y of life.
Small lymphocytic leukemia (SLL) is very similar to CLL described supra, and is also a cancer of B-cells. In SLL the abnormal lymphocytes mainly affect the lymph nodes. However, in CLL the abnormal cells mainly affect the blood and the bone marrow. The spleen may be affected in both conditions. SLL accounts for about 1 in 25 of all cases of non-Hodgkin lymphoma. It can occur at any time from young adulthood to old age, but is rare under the age of 50. SLL is considered an indolent lymphoma. This means that the disease sses very , and patients tend to live many years after diagnosis. However, most patients are diagnosed with advanced disease, and although SLL responds well to a variety of chemotherapy drugs, it is generally considered to be incurable. Although some cancers tend to occur more often in one gender or the other, cases and deaths due to SLL are evenly split n men and women. The average age at the time of diagnosis is 60 years.
Although SLL is indolent, it is persistently progressive. The usual pattern of this disease is one of high response rates to radiation y and/or chemotherapy, with a period of disease remission. This is followed months or years later by an inevitable relapse. Re-treatment leads to a response again, but again the e will relapse. This means that although the short-term prognosis of SLL is quite good, over time, many patients develop fatal complications of recurrent disease. Considering the age of the individuals typically diagnosed with CLL and SLL, there is a need in the art for a simple and effective treatment of the disease with minimum side- effects that do not impede on the patient's quality of life. The t invention fulfills this long standing need in the art.
Disclosed herein is a method for treating a Mantle cell lymphoma in an individual in need thereof, comprising: administering to the dual a composition containing a therapeutic amount of at least one compound having the structure of as I, Ia-Ii or IIa-IId.
As used herein, the term, "Mantle cell lymphoma" refers to a subtype of B-cell lymphoma, due to CD5 positive antigen-naive minal center B-cell within the mantle zone that surrounds normal germinal center follicles. MCL cells generally over-express cyclin D1 due to a t(l l;l4) chromosomal translocation in the DNA. More specifically, the translocation is at t(l l;l4)(ql3;q32). Only about 5% of lymphomas are of this type. The cells are small to medium in size. Men are affected most often. The average age of patients is in the early 605. The lymphoma is usually read when it is diagnosed, involving lymph nodes, bone marrow, and, very often, the spleen. Mantle cell ma is not a very fast growing lymphoma, but is difficult to treat.
Disclosed herein, in n embodiments, is a method for treating a al zone B-cell lymphoma in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia—Ii or Ila-11d.
As used herein, the term "marginal zone B-cell ma" refers to a group of related B- cell neoplasms that involve the lymphoid tissues in the marginal zone, the patchy area outside the follicular mantle zone. Marginal zone lymphomas account for about 5% to 10% of lymphomas.
The cells in these lymphomas look small under the microscope. There are 3 main types of marginal zone lymphomas including extranodal marginal zone B-cell lymphomas, nodal marginal zone B-cell lymphoma, and c marginal zone lymphoma.
Disclosed herein, in n embodiments, is a method for ng a MALT in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia—Ii or IIa—IId.
The term "mucosa-associated lymphoid tissue (MALT) lymphoma", as used , refers to extranodal manifestations of marginal-zone lymphomas. Most MALT lymphoma are a low grade, although a ty either manifest initially as ediate-grade non-Hodgkin lymphoma (NHL) or evolve from the low-grade form. Most of the MALT lymphoma occur in the h, and roughly 70% of gastric MALT lymphoma are associated with Helicobacter pylori infection. Several cytogenetic abnormalities have been identified, the most common being trisomy 3 or t(l l;18). Many of these other MALT lymphoma have also been linked to infections with bacteria or viruses. The average age of patients with MALT ma is about 60.
Disclosed herein, in certain embodiments, is a method for ng a nodal marginal zone B-cell lymphoma in an individual in need thereof, comprising: administering to the dual a ition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia—Ii or Ila-11d.
The term "nodal marginal zone B-cell lymphoma" refers to an indolent B-cell lymphoma that is found mostly in the lymph nodes. The disease is rare and only accounts for 1% of all Non- Hodgkin's mas (NHL). It is most commonly diagnosed in older patients, with women more susceptible than men. The disease is classified as a marginal zone lymphoma because the mutation occurs in the marginal zone of the B-cells. Due to its confinement in the lymph nodes, this disease is also classified as nodal.
Disclosed herein, in certain embodiments, is a method for treating a splenic marginal zone B-cell lymphoma in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia-Ii or Ila-11d.
The term "splenic al zone B-cell lymphoma" refers to specific low-grade small B- cell lymphoma that is incorporated in the World Health Organization classification.
Characteristic features are splenomegaly, moderate lymphocytosis with villous morphology, intrasinusoidal pattern of involvement of various organs, especially bone marrow, and ve indolent course. Tumor progression with increase of blastic forms and aggressive behavior are observed in a minority of patients. Molecular and netic s have shown heterogeneous s probably because of the lack of standardized diagnostic ia.
Disclosed herein, in certain embodiments, is a method for ng a Burkitt lymphoma in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia- Ii or IIa-Hd.
The term "Burkitt lymphoma" refers to a type ofNon-Hodgkin Lymphoma (NHL) that commonly affects children. It is a highly aggressive type of B-cell lymphoma that often starts and involves body parts other than lymph nodes. In spite of its fast—growing , Burkitt's lymphoma is often curable with modern intensive therapies. There are two broad types of Burkitt's lymphoma—the sporadic and the endemic varieties: Endemic Burkitt's lymphoma and Sporadic Burkitt's lymphoma.
Endemic Burkitt's lymphoma involves children much more than adults, and is related to Epstein Barr Virus (EBV) infection in 95% cases. It occurs primarily in equatorial Africa, where about half of all childhood cancers are Burkitt's lymphoma. It characteristically has a high chance of involving the jawbone, a rather distinctive feature that is rare in ic Burkitt's. It also commonly involves the abdomen.
Sporadic Burkitt's lymphoma is a type of Burkitt's lymphoma that affects the rest of the world, including Europe and the as. Here too, it's mainly a disease in children. The link between Epstein Barr Virus (EBV) is not as strong as with the endemic variety, though direct evidence of EBV infection is present in one out of five patients. More than the involvement of lymph nodes, it is the abdomen that is notably affected in more than 90% of the children. Bone marrow involvement is more common than in the sporadic variety.
Disclosed herein, in n embodiments, is a method for treating a Waldenstrom macroglobulinemia in an dual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the ure of Formulas I, Ia—Ii or Ila-11d.
The term "Waldenstrom macroglobulinemia", also known as lymphoplasmacytic ma, is cancer involving a subtype of white blood cells called cytes. It is characterized by an uncontrolled clonal proliferation of terminally entiated B lymphocytes.
It is also characterized by the lymphoma cells making an antibody called immunoglobulin M (IgM). The IgM antibodies circulate in the blood in large amounts, and cause the liquid part of the blood to thicken, like syrup. This can lead to decreased blood flow to many organs, which can cause problems with Vision (because of poor circulation in blood vessels in the back of the eyes) and neurological problems (such as headache, dizziness, and confiJsion) caused by poor blood flow within the brain. Other symptoms can include feeling tired and weak, and a tendency to bleed easily. The underlying etiology is not fully understood but a number of risk factors have been identified, including the locus 6p21.3 on some 6. There is a 2- to 3-fold risk increase of ping WM in people with a personal history of autoimmune diseases with autoantibodies and particularly elevated risks associated with hepatitis, human immunodeficiency Virus, and rickettsiosis.
Disclosed herein, in n embodiments, is a method for treating a myeloma in an dual in need f, comprising: stering to the individual a composition containing a therapeutic amount of at least one nd having the ure of as I, Ia-Ii or IIa-IId.
Multiple a, also known as MM, a, plasma cell myeloma, or as Kahler's disease (after Otto Kahler) is a cancer of the white blood cells known as plasma cells. A type of B cell, plasma cells are a crucial part of the immune system responsible for the production of antibodies in humans and other vertebrates. They are produced in the bone marrow and are transported through the lymphatic system.
Disclosed herein, in certain embodiments, is a method for treating a leukemia in an individual in need thereof, comprising: administering to the individual a composition containing a therapeutic amount of at least one compound having the structure of Formulas I, Ia—Ii or d.
Leukemia is a cancer of the blood or bone marrow characterized by an abnormal increase of blood cells, usually leukocytes (white blood cells). Leukemia is a broad term covering a spectrum of diseases. The first division is n its acute and chronic forms: (i) acute leukemia is characterized by the rapid increase of immature blood cells. This crowding makes the bone marrow unable to produce healthy blood cells. Immediate treatment is required in acute leukemia due to the rapid progression and accumulation of the malignant cells, which then spill over into the bloodstream and spread to other organs of the body. Acute forms of leukemia are the most common forms of leukemia in children; (ii) chronic leukemia is distinguished by the excessive build up of relatively mature, but still abnormal, white blood cells. Typically taking months or years to progress, the cells are produced at a much higher rate than normal cells, resulting in many abnormal white blood cells in the blood. Chronic leukemia mostly occurs in older people, but can theoretically occur in any age group. Additionally, the diseases are ided according to which kind of blood cell is ed. This split divides leukemias into lymphoblastic or lymphocytic leukemias and myeloid or myelogenous leukemias: (i) in lymphoblastic or lymphocytic leukemias, the cancerous change takes place in a type of marrow cell that normally goes on to form lymphocytes, which are infection-fighting immune system cells; (ii) in myeloid or myelogenous leukemias, the cancerous change takes place in a type of marrow cell that normally goes on to form red blood cells, some other types of white cells, and platelets.
Within these main categories, there are several subcategories including, but not limited to, Acute blastic leukemia (ALL), Acute myelogenous leukemia (AML), Chronic myelogenous leukemia (CML), and Hairy cell leukemia (HCL).
Symptoms, diagnostic tests, and prognostic tests for each of the above—mentioned conditions are known in the art. See, e. g., Harrison's Principles of al ne®," 16th ed., 2004, The McGraw-Hill Companies, Inc. Dey et a1. (2006), Cytojournal 3(24), and the "Revised European American Lymphoma" (REAL) classification system (see, e.g., the website maintained by the al Cancer Institute).
A number of animal models are useful for establishing a range of therapeutically effective doses of BTK inhibitor compounds for treating any of the ing diseases.
For example, dosing of BTK tor compounds for treating an autoimmune disease can be assessed in a mouse model ofrheumatoid arthitis. In this model, arthritis is induced in Balb/c mice by administering anti-collagen antibodies and lipopolysaccharide. See Nandakumar et al. (2003), Am. J. Pathol 163:1827-1837.
In another example, dosing of BTK inhibitors for the treatment of B-cell proliferative disorders can be examined in, e.g., a to-mouse xenograft model in which human B-cell lymphoma cells (e. g. Ramos cells) are implanted into defficient mice (e.g., "nude" mice) as described in, e.g., Pagel et al. (2005), Clin Cancer Res ll(l3):4857-4866.
Animal models for treatment of thromboembolic disorders are also known.
The eutic efficacy of the compound for one of the foregoing diseases can be zed during a course of treatment. For example, a subject being treated can undergo a diagnostic evaluation to correlate the relief of disease ms or pathologies to inhibition of in vivo BTK activity achieved by administering a given dose of a BTK inhibitor. Cellular assays known in the art can be used to determine in vivo activity ofBTK in the ce or absence of an BTK inhibitor. For example, since activated BTK is orylated at tyrosine 223 (Y223) and tyrosine 551 (Y551), phospho-specific immunocytochemical staining of P—Y223 or P-Y551- positive cells can be used to detect or quantify activation of BTK in a population of cells (e.g., by FACS analysis of stained vs unstained cells). See, e.g., Nisitani et al. (1999), Proc. Natl. Acad.
Sci, USA 1-2226. Thus, the amount of the BTK inhibitor compound that is administered to a subject can be increased or decreased as needed so as to maintain a level of BTK inhibition optimal for treating the subject's disease state.
Compounds disclosed herein irreversibly inhibit BTK and may be used to treat mammals suffering from Bruton's tyrosine kinase-dependent or ‘s tyrosine kinase ed conditions or diseases, including, but not limited to, cancer, autoimmune and other inflammatory diseases. Compounds sed herein have shown efficacy in a wide variety of diseases and conditions that are described herein.
A further aspect resides in the use of compounds of Formulas I, Ia-Ii or IIa-IId or a ceutically acceptable salt thereof for the manufacture of a medicament to be used for the treatment of chronic B cell disorders in which T cells play a prominent role.
In yet r aspect, the compounds of Formulas I, Ia-Ii or IIa-IId are used for the manufacture of a medicament to be used for the treatment of BTK-mediated diseases or conditions. These include, but are not limited to, the treatment of B cell mas resulting from chronic active B cell receptor signaling.
BTK mediated disorders or BTK mediated conditions as used herein, mean any disease state or other deleterious condition in which B cells, mast cells, myeloid cells or lasts play a central role. These diseases include but are not limited to, , autoimmune and inflammatory diseases, allergies, infectious diseases, bone resorption disorders and proliferative diseases.
Immune, autoimmune and inflammatory diseases that can be treated or prevented with the compounds of Formulas I, Ia-Ii or d further include rheumatic diseases (e.g. infectious arthritis, progressive chronic arthritis, ing arthritis, traumatic arthritis, gouty tis, osteoporosis, Reiter's syndrome, polychondritis, acute synovitis and spondylitis), glomerulonephritis (with or without nephrotic syndrome), autoimmune hematologic disorders (e.g. hemolytic , aplasic anemia, idiopathic ocytopenia, and neutropenia), and autoimmune inflammatory bowel diseases (e.g. ulcerative colitis and Crohn's disease), host versus graft disease, allograft ion, chronic thyroiditis, schleroderma, primary billiary cirrhosis, systemic lupus erythematosis, contact dermatitis, eczema, skin sunbums, chronic renal insufficiency, Stevens-Johnson syndrome, inflammatory pain, idiopathic sprue, cachexia, sarcoidosis, kerato conjunctivitis, otitis media, periodontal disease, pulmonary interstitial fibrosis, pneumoconiosis, pulmonary ciency me, pulmonary emphysema, pulmonary fibrosis, silicosis, chronic inflammatory pulmonary disease (e.g. chronic obstructive pulmonary disease) and other inflammatory or obstructive disease on airways.
Allergies that can be treated or prevented include, among others, ies to foods, food additives, insect s, dust mites, pollen, animal materials and contact allergans, type I hypersensitivity allergic asthma, allergic conjunctivitis.
Infectious diseases that can be treated or prevented e, among others, , septic shock, endotoxic shock, sepsis by Gram-negative bacteria, shigellosis, meningitis, al malaria, pneumonia, tuberculosis, viral myocarditis, viral hepatitis (hepatitis A, hepatitis B and hepatitis C), HIV infection, retinitis caused by cytomegalovirus, influenza, herpes, treatment of infections associated with severe burns, myalgias caused by infections, cachexia secondary to infections, and nary viral infections such as lentivirus, caprine arthritic virus, visna-maedi virus, feline immunodeficiency virus, bovine immunodeficiency virus or canine immunodeficiency virus.
In some embodiments of Formula I, compounds are present as a material in substantially pure form.
In some embodiments of a 1, compounds are selected from any one of the Examples herein or a pharmaceutically acceptable salt f.
Each variable definition above includes any subset thereof and the compounds of Formula I include any combination of such les or variable s.
The present ion es the compounds and salts thereof, their physical forms, preparation of the compounds, useful intermediates, and pharmaceutical compositions and ations thereof.
The compounds of the present invention and the term und" in the claims include any pharmaceutically acceptable salts or solvates, and any amorphous or crystal forms, or tautomers, Whether or not specifically recited in context.
The present invention includes all isomers of the compounds. Compounds may have one or more asymmetric carbon atoms and can exist as two or more isomers. Where a compound of the invention contains an alkenyl or alkenylene group, geometric cis/trans (or Z/E) isomers are possible. Where the compound contains, for example, a keto or oxime group or an ic moiety, tautomeric isomerism ('tautomerism') can occur. A single compound may t more than one type of isomerism.
The present invention includes any isomers, even if not specifically shown, individually as well as mixtures, geometric isomers, and pharmaceutically acceptable salts thereof, including compounds exhibiting more than one type of isomerism,. Where a compound or stereocenter is described or shown without definitive stereochemistry, it is to be taken to embrace all possible individual isomers, configurations, and mixtures thereof. Thus, a material sample containing a mixture of stereoisomers would be embraced by a recitation of either of the stereoisomers or a recitation without definitive stereochemistry. Also contemplated are any cis/trans isomers or ers of the compounds described. When a tautomer of the compound of a I exists, the compound of Formula I of the present ion includes any possible tautomers and pharmaceutically acceptable salts thereof, and mixtures thereof, except where specifically stated otherwise.
DETAILED DESCRIPTION OF THE INVENTION Described herein are compounds of Formula I, which include compounds of Formulas Ia- Ii and IIa-IId, and compositions and formulations containing such compounds, and methods of using and making such compounds. These compounds are useful in treating es or ions modulated at least in part by BTK.
In an embodiment, a compound according to Formula I and above embodiments is provided, wherein the nd of Formula I is ented by the nd of Formula 11a: /21 R1 | 62 x/ \24 R3 L2 wherein, Rl-R4, G2, G3, L1, L2, X, Y, and 21-24, are as previously described for a compound of a I and B1 and B2 are independently selected from oalkyl, C6heterocycloalkyl, C6aryl, or C6heteroaryl.
In an embodiment, a compound according to Formula I and above embodiments is provided, wherein the nd of Formula I is represented by the compound of Formula 11b: 2/21 R1 23 L1 R2 n 26/ \24 31/ \82 R3 Y\L2/Z wherein, Rl-R4, B1, B2, L1, L2, Y, and 21-24, are as previously described for a compound of Formula I, Z5 and Z6 are each independently selected from C(Ra) or N, where Ra is alkyl or H, and n and m are each independently selected from 0, l, or 2.
In an embodiment, a compound according to Formula I and above embodiments is provided, wherein the compound of Formula I is represented by the compound of Formula IIc: /z1 R1 22 I (P9326 z3 L1 / \Z4 / Bl \BZ n, Rl-R4, B1, B2, L1, L2, and 21-24, are as previously bed for a compound of Formula I, Z5 and Z6 are each independently selected from C or N, and n and m are each independently selected from 0, l, or 2.
In an embodiment, a compound according to Formula I and above embodiments is provided, wherein the compound of Formula I is represented by the compound of Formula 11d: | 62 G3 R2 0 (W326 \Z4 \ Z5 R3 L2 L1 wherein, Rl-R4, L1, L2, G2, G3, and Zl—Z4, are as previously described for a compound of Formula I, Z5 and Z6 are each independently selected from C or N, B1 and B2 are independently selected from C6cycloalkyl, C6heterocycloalkyl, C6aryl, or C6heteroaryl, and n and m are each independently selected from 0, l, or 2.
The t invention includes the compounds, intermediates, es and synthetic methods described herein. Compounds of Formula I are prepared according to reaction schemes described herein. Unless otherwise indicated, the substituents in the schemes are defined as above.
Synthetic Methods: Compounds of the present invention include the intermediates, examples, and synthetic methods described herein. Synthetic methods provided herein are generally preceded by their respective tic schemes. Where a procedure for an intermediate or example refers to an analogous procedure for an analogous intermediate or example, such reference includes the procedure for that analogous ediate or example, the associated synthetic scheme as well as the procedures and s utilized for the synthesis of the analogous intermediate or example.
The compounds of Formulas I, Ia-Ii and IIa-d may be prepared by the methods described below, together with synthetic methods known in the art of organic chemistry, or modifications and derivatizations that are ar to those of ordinary skill in the art. The starting materials used herein are commercially ble or may be prepared by e methods known in the art [such as those methods sed in standard reference books such as the Compendium of Organic tic Methods, Vol. I-VI (Wiley-Interscience); or the hensive Organic Transformations, by RC. Larock (Wiley-Interscience)]. Preferred methods include, but are not limited to, those described below.
During any of the following synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This can be achieved by means of conventional protecting groups, such as those described in T. W. , Protective Groups in Organic Chemistry, John Wiley & Sons, 1981; T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons, 1991, and T. W. Greene and P. G.
M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons, 1999, which are hereby orated by reference.
Compounds of Formulas I, Ia—Ii and Ila-d, or their pharmaceutically acceptable salts, can be prepared according to the on schemes discussed hereinbelow and utilizing ordinary skill in the art. Unless otherwise indicated, the substituents in the schemes are defined as above.
Isolation and ation of the products is accomplished by standard procedures, which are known to a chemist of ordinary skill.
When a general or exemplary synthetic procedure is referred to, one skilled in the art can readily determine the appropriate ts, if not indicated, extrapolating from the general or exemplary procedures. Some of the general procedures are given as examples for general preparation of compounds. One skilled in the art can readily adapt such procedures to the synthesis of other specific compounds. Representation of an unsubstituted on in ures shown or referred to in the general procedures is for convenience and does not preclude substitution as bed elsewhere herein. For specific groups that can be present, either as groups in the general procedures or as optional tuents not shown, refer to the descriptions in the remainder of this nt, including the claims and detailed description.
A general procedure for the synthesis of compounds of Formulas I, Ia-Ii and Ila-d is shown in the General Scheme, below. l Scheme: CN 02NXB/ —B\L’I_B2 NH2 o N /L CouplingSuzuki OZNX/ 2 \X/ SuzukiCoupling B1 \Bz 2)acetaldoxime CuCI2 MKKUN'G (c) R2 O o o )LNHZ R3 \ 2LNH2 Pd/C, H2 R4 R4 —> 1 —>RWMXH 1 B1’L\B2 3 /L H2N\X/ Base '31 \32 LG = Leaving Group R2 0 (d) (e) Where A, B1, B2, L1, X, R2, R3, and R4 are as defined previously for a compound of Formulas I, Ia-Ii and IIa-d and LG is equal to a suitable leaving group such as triflate, mesylate, tosylate, HATU, Cl, Br or I.
In a typical preparation of a compound of Formulas I, Ia-Ii and Ila-d, a compound of Formula (a) was reacted under Suzuki coupling conditions with a suitable boronic acid. Suitable conditions include, but are not limited to, treating nds of Formula (a) with a suitable base, such as CszC03 or K2C03, and a suitable palladium catalyst, such as Pd(dppf)C12'DCM. le solvents for use in the above sis include 1,4-dioxane, water, DME and mixtures thereof. The mixture was ed with nitrogen six times and refluxed for about 16 h under nitrogen here to afford the compound of Formula (b). The above process was carried out at temperatures between about 50° C and about 150° C. ably, the reaction was carried out at about 100° C. The above process was ably carried out at or about atmospheric pressure although higher or lower pressures may be used if desired. Substantially equimolar amounts of reactants were preferably used although higher or lower amounts may be used if desired.
The compound of Formula (b) was d with a suitable boronic acid under Suzuki coupling conditions. Suitable conditions include, but are not limited to, treating compounds of Formula (b) with a suitable base, such as Cs2C03 or K2C03, and a le palladium catalyst, such as Pd(dppt)C12‘DCM or Pd(PPh3)4. Suitable solvents for use in the above synthesis include 1,4-dioxane, water, DME and mixtures thereof. The mixture was degassed with nitrogen six times and refluxed for about 5 h under nitrogen atmosphere to afford the compound of a (c). The above process was carried out at temperatures between about 40° C and about 120° C. ably, the reaction was carried out at about 900 C. The above process was preferably carried out at or about atmospheric pressure although higher or lower pressures may be used if d.
Substantially equimolar amounts of reactants were preferably used although higher or lower amounts may be used if d.
The compound of a (c) was d under hydrogen atmosphere with palladium on carbon, preferably ing the mixture with hydrogen about 6 times. Suitable solvents for use in the above synthesis include ethyl e and methanol. The above process was carried out at temperatures between about 10° C and about 60° C, or ably at ambient temperature, to afford the compound of Formula (d).The above process was preferably carried out at or about atmospheric pressure although higher or lower pressures may be used if desired. Substantially equimolar amounts ofreactants were preferably used although higher or lower amounts may be used if desired.
The compound of Formula (d) was reacted with a suitable acryloyl chloride with a suitable base. Suitable bases include organic bases such as TEA or DIPEA. The above s was carried out at temperatures between about -10° C and about ambient temperature, or preferably at 0° C, to afford the compound of Formula (d). The above process was preferably carried out at or about atmospheric pressure although higher or lower pressures may be used if desired. ntially equimolar amounts of reactants were preferably used although higher or lower amounts may be used if desired.
Examples: Preparations and Intermediates Scheme 1 08,0H 0 (m \ \ 2 l l/ NH2 CNPdd / \ (PP)fCl2 C$2CO i 3 N Cl acetaldoxime, CuCl2 N CI —> —'> CI N CI 14-gioxane/H20 THF/H20,11O°C 110 0.161121% No2 16h, 84% N02 1 3 4 Pd2(dba)3 5C32003 £20—>©Pd/C H2 | : NH2 14-dioxane/HZO EA, r..t, 16h O N 0 110°C 16h 60% TEA, DCM 61):) 0°C 10min Example1 2-chlor0(3-nitr0phenyl)nicotinonitrile (3). To a solution of ophenylboronic acid 2 (5.19 g, 30 mmol), CSQCOg (19.56 g, 60 mmol) and 2,6-dichloronicotinonitrile 1 (5.51 g, 33 mmol) in dry 1,4-dioxane (100 mL) was added Pd(dppf)Clg’DCM (2.4 g, 3.0 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then refluxed for 16 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the e was purified by flash chromatography, eluting with 20:1 to 3:1 PE/EA to afford the title compound as a yellow solid (1.6 g, 21%). 2-chlor0(3-nitr0phenyl)nic0tinamide (4). To a solution of 2-chloro(3- nitrophenyl)nicotinonitrile 3 (259 mg, 1.0 mmol) and doxirne (88 mg, 1.5 mmol) in ydrofuran (5 mL) and water (5 mL) was added CuClz (15 mg, 0.1 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and stirred for 16 h under en atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography, eluting with 20:1 DCM/MeOH to afford the title compound as a yellow solid (240 mg, 84%). MS (ESI): m/Z =277.9 [M+H]+. 6-(3-nitrophenyl)(4-phenoxyphenyl)nic0tinamide (6). To a solution of 2-chloro—6-(3- nitrophenyl)nicotinamide 4 (240 mg, 0.87 mmol), Cs2C03 (567 mg, 1.74 mmol) and biphenyl ylboronic acid 5 (203 mg, 0.95 mmol) in 1,4—dioxane (10 mL) and water (2.5 mL) was added Pd2(dba)3 (80 mg, 0.09 mmol) under nitrogen here, and the mixture was degassed with nitrogen 6 times, then refluxed for 16 h under nitrogen atmosphere. After cooling to room temperature, the solvent was ated and the crude product was purified by flash chromatography eluting with 150:1 DCM/MeOH to afford the title compound as a yellow solid (110 mg, crude). 6-(3-aminophenyl)—2—(4-phen0xyphenyl)nicotinamide (7). To a solution of 6-(3-nitrophenyl)- 2-(4-phenoxyphenyl)nicotinamide 6 (110 mg, 0.27 mmol) in ethyl acetate (5 mL) was added Pd/C (10 mg) under hydrogen here, and the e was degassed with hydrogen 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude product as red oil (61 mg, 60%). MS (ESI): m/z = 382.1 [M+H]+.
Example 1 6-(3-acrylamidophenyl)(4-phen0xyphenyl)nicotinamide (0&0NH2| : HN\n/\ To a solution of 6-(3—aminophenyl)—2-(4-phenoxyphenyl)nicotinamide 7 (38 mg, 0.1 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and acryloyl chloride 8 (9 mg, 0.1 mmol) at 0 oC. The mixture was stirred at 0 0C for 10 s. The solvent was removed and the residue was purified by Prep-TLC g with 25 :1 DCM/MeOH to afford the title compound (30 mg, 71%) as white solid. 1H NMR (400 MHz, DMSO) 8 10.36 (s, 1H), 8.43 (s, 1H), 7.72—7.97 (m, 7H), 7.49 (s, 1H), 7.33—7.52(m, 3H), 7.24(t, J: 6.9 Hz, 1H), 13(m, 4H), 6.45 (dd, J: 16.9, 10.0 Hz, 1H), 6.30 (d, J: 16.9 Hz, 1H), 5.79 (d, J: 10.5 Hz, 1H), MS (ESI, method A): m/Z = 436.0 [M + H]+, tR=1.553 min., HPLC: 97.5% (214nm), 98.0% (254nm).
Scheme 2 (1 or"O O N(H2304concszo \ \ N...2 Pd(dppf)Clz 052003 I ""2 —> | —> / 90°C 1h 73% N/ 1,4-dioxane/HZO Cl N CI Cl 110°C, 16h, 53% o0 1 2 4 Dc ,0 I PM: H2 Pd(PPh3)4, ch03 N/ DME/HZO EA rt 16h BOC/ 90°C, 5h 84% O O VLCI \ \ NH2 I TFA, DCM, | 9 N r1, 1h, 71% TEA DCM HN NHZOQ O 0°C,10min 44% Example3 2,6-dichloronicotinamide (2). To 2,6-dichloronicotinonitrile 1 (l .73 g, 10 mmol) was added conc H2804 (10 mL) and water (2 mL). The e was heated to 90°C and stirred for l h.
After cooling to room temperature, the solution was poured into ice—cold water, then adjusted to PH = 8 with ammonia water. The precipitate was filtered, washed with water (20 mL) and dried under vacuum to afford the title compound as an brown solid (1.4 g, 73%). MS (ESI): m/z = 191.1 [M+H]+. 6-chlor0(4-phenoxyphenyl)nicotinamide (4). To a solution of 2,6-dichloronicotinamide 2 (668 mg, 3.5 mmol), C82C03 (1.14 g, 7 mmol) and 4—phenoxyphenylboronic acid 3 (749 mg, 3.5 mmol) in 1,4-dioxane (30 mL) was added Pd(dppf)C12‘DCM (285 mg, 0.35 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then refluxed for 16 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the e was purified by flash chromatography, eluting with 20:] to 3:1 petroleum ethyl acetate (PE/EA) to afford the title nd as a yellow solid (611 mg, 53%). MS (ESI): m/Z = 325.0 [M+H]*. utyl(5-carbamoyl(4-phenoxyphenyl)pyridinyl)—5,6—dihydropyridine—1(2H)— carboxylate (6). To a solution of 4-(4,4,5,5—tetramethyl-1,3,2-dioxaborolanyl)-1 ,2,3,6- tetrahydropyridine 5 (232 mg, 0.75 mmol), K2C03 (207 mg, 1.50 mmol) and 6-chloro—2-(4— phenoxyphenyl)nicotinamide 4 (162 mg, 0.50 mmol) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was added 3)4 (115 mg, 0.10 mmol) under en atmosphere, and the mixture was ed with nitrogen 6 times, then heated to 90°C and stirred for 5 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography, eluting with 70:1 DCM/MeOH to afford the title compound as a white solid (232 mg, crude). tert-butyl 4-(5-carbamoyl—6—(4-phenoxyphenyl)pyridinyl)piperidine—l-carboxylate (7).
To a solution of 4-(5 -carbamoyl(4-phenoxyphenyl)pyridinyl)—5,6-dihydropyridine-1(2H)- carboxylate 6 (232 mg, 0.75 mmol) in ethyl acetate (5 mL) was added Pd/C (10 mg) under hydrogen atmosphere, and the mixture was degassed with hydrogen 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude t as brown solid (215 mg, 60% for two steps). MS (ESI): m/z = 474.1 [M+H]+. 2-(4-phenoxyphenyl)(piperidinyl)nicotinamide le 2) (8). To a solution of tert- butyl 4-(5 -carbamoyl(4-phenoxyphenyl)pyridinyl)piperidine— 1 -carboxylate 7 (215 mg, 0.45 mmol) in dry dichloromethane (6 mL) was added TFA (2 mL), and the resulting mixture was stirred for 1 h at ambient temperature. The solvent was removed and the residue was partitioned between saturated aqueous sodium onate (30 mL) and ethyl acetate (20 mL).
The organic phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated.
The crude residue was purified by flash chromatography, eluting with 5 :1 DCM/MeOH to afford the title compound as a white solid (120 mg, 71%). MS (ESI): m/z = 374.2 [M+H]+.
Example 3 6-(1-acryloylpiperidinyl)—2-(4-phenoxyphenyl)nic0tinamide To a on of 2-(4-phenoxyphenyl)—6-(piperidin-4—yl)nicotinamide 8 (26 mg, 0.07 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and acryloyl chloride 9 (7 mg, 0.07 mmol) at 0 °C. The mixture was stirred at 0 °C for 10 minutes. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 :1 OH to afford the title compound (13 mg, 44%) as white solid. 1H NMR (400 MHz, CDC13) 8 7.90 (d, J= 8.0 Hz, 1H), 7.71—7.62 (m, 2H), 7.42—7.31 (m, 2H), 7.21—7.11 (m, 2H), 7.10— 7.01 (m, 4H), 6.60 (dd, J: 16.8, 10.5 Hz, 1H), 6.26 (dd, J: 16.8, 2.0 Hz, 1H), 5.84 (s, 1H), 5.68 (dd, J: 10.5, 2.0 Hz, 1H), 5.54 (s, 1H), 4.79 (d, J: 13.3 Hz, 1H), 4.13 (d,J= 12.8 Hz, 1H), 3.17 (t, J: 12.3 Hz, 1H), 3.39 (t, J: 11.7 Hz, 1H), 2.76 (t, J: 11.7 Hz, 1H), 2.03 (br, 2H), 1.80 (ddd, J: 25.5, 12.5, 4.2 Hz, 2H). MS (ESI, method A): m/z = 428.0 [M + H]+, 80 min. HPLC: 96.7% (214nm), 99.3% (254nm).
Scheme 3 [f] 0 \ \ I NQI H2804((conc)/H20 | NHz Cl N/ CI K2CO3, ethanol 9,o°c 1h, 94% (\N N Cl reflux 4h, 62% BOG/N HNd 1 3 4 (1 03""O O \ NH2 Pd(dppf)C| 2! Cs co2 3 IN/ | . HQN N/ TEA DCM, (\N 1,4-dloxane/H20 110°C, 16h, 58% N\2 6 0°C,10min N O Example 4 tert-butyl 4-(6-ch10rocyan0pyridiny])piperazine-l-carboxylate (3). To a solution of 2,6- dichloronicotinonitrile 1 (519 mg, 3.0 mmol) and tert—butyl zine-l-carboxylate 2 (558 mg, 3.0 mmol) in ethanol (15 mL) was added K2C03 (636 mg, 6.0 mmol), and the resulting solution was refluxed for 4 h. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography, eluting with 5:1 to 2:1 PE/EA to afford the title compound as a white solid (600 mg, 62%). MS (ESI): m/z = 345.1 [M + Na]+. 6-chlor0(4—phen0xyphenyl)nicotinamide (4). To tert-butyl-(6-chloro-5—cyanopyridin y1)piper azine—l-carboxylate 3(600 mg, 10 mmol) was added H2S04 (60716., 5 mL) and water (1 mL). The mixture was heated to 90°C and d for 1 h. After cooling to room temperature, the solution was poured into ice—cold water, then adjusted to PH = 8 with ammonia water. The precipitate was filtered, washed with water (20 mL) and dried under vacuum to afford the title compound as an off-white solid , 94%). MS (ESI): m/z = 241.0 [M + H]+. 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide (Example 4) (6). To a solution of 6— chloro(4-phenoxyphenyl)nicotinamide 4 (420 mg, 1.75 mmol), K2C03 (483 mg, 3.5 mmol) and 4-phenoxyphenylboronic acid 5 (374 mg, 1.75 mmol) in 1,4-dioxane (15 mL) and water (3 mL) was added Pd(PPh3)4 (231 mg, 0.20 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and stirred for 5 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting with 70:1 DCM/MeOH to afford the title compound as a white solid (260 mg, 58%). MS (ESI): m/z = 375.0 [M + H]+.
Example 5 6-(4-acryloylpiperazin-l-y])—2-(4-phenoxyphenyl)nicotinamide Nominee|\/ NH2 To a on of 2-(4-phenoxyphenyl)—6-(piperazin-1—yl)nicotinamide 6 (59 mg, 0.16 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and yl chloride 7 (14 mg, 0.16 mmol) at 0°C. The e was stirred at 0°C for 10 minutes. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 :1 DCM/MeOH to afford the title compound (27 mg, 40%) as light yellow solid. 1H NMR (400 MHz, CDClg) 8 8.04 (d, J: 8.7 Hz, 1H), 7.64 (d, J= 8.2 Hz, 2H), 7.40 (t, J: 7.6 Hz, 2H), 7.18 (t, J: 7.3 Hz, 1H), 7.08 (dd, J: 12.0, 8.4 Hz, 4H), 6.66 (d, J= 8.5 Hz, 1H), .56 (m, 1H), 6.37 (d, J= 16.7 Hz, 1H), 5.77 (d, J= 10.6 Hz, 1H), .57 (s, 1H), 5.33 (s, 1H), 3.81 (s, 4H), 3.73 (s, 4H).MS (ESI, method A): m/z = 429.2 [M + H]+, tR=1.454 min., HPLC: 97.5% (214nm), 98.0% (254nm).
Scheme 4 Z\/ NH2 HCIIEtOH CIJK/ Example 6 6-chlor0(4-phenoxyphenyl)nic0tinamide (1). 6-chloro(4-phenoxyphenyl)nicotinamide 1 was synthesized by using the same procedure above providing a yellow solid (611 mg, 53%).
MS (ESI): m/z = 325.0 [M+H]+. tert-butyl(5-carbamoyl(4-phenoxyphenyl)pyridinyl)—2,S-dihydro-lH-pyrrole-l- carboxylate (3). The title compound was synthesized using a procedure analogous to the procedure described in tert-butyl 4-(5-carbamoyl-6—(4-phenoxypheny1)pyridin-2—y1)-5,6— dihydropyridine-1(2H)- carboxylate ed as white solid (300 mg, crude). MS (ESI): m/z = 458.2 [M+H]+. tert-butyl(5-carbam0yl—6-(4-phenoxyphenyl)pyridin-2—yl)pyrrolidine-l-carboxylate (4).
The title compound was synthesized using a procedure analogous to the procedure described in tert-butyl 4-(5—carbamoyl(4—phenoxyphenyl)pyridinyl)piperidinecarboxylate provided as white solid (240 mg, 45% for two steps). MS (ESI): m/z = 460.1 [M+H]+. 2-(4-phenoxyphenyl)(pyrrolidinyl)nic0tinamide(5). The title compound was synthesized using a procedure analogous to the procedure described in 2-(4-phenoxyphenyl)—6-(piperidin otinamide as white solid (190 mg, . MS (ESI): m/z = 360.1 .
Example 6 6-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)nic0tinamide N K) The title compound was synthesized using a procedure analogous to the procedure described in Example 3 as a white solid (18 mg, 29%). 1H NMR (300 MHZ, CD30D) 8 7.85 (d, J: 7.9 Hz, 1H), 7.72 (d, J: 8.8 Hz, 2H), 7.37 (m, 3H), 7.15 (t, J: 7.4 Hz, 1H), 7.02 (m, 4H), 6.65 (dd, J: 16.7, 10.6 Hz, 1H), 6.28 (m, 1H), 5.74 (dd, J: 9.5, 7.4 Hz, 1H), 3.98 (m, 2H), 3.69 (m, 3H), 2.34 (dtd, J: 30.9, 12.6, 8.0 Hz, 2H). MS (ESI, method A): m/z = 414.2 [M + H]+, tR=1.421 min.
HPLC: 100% (214nm), 100% (254nm).
Scheme 5 03‘OH |\ NHZ / / N WOO—*0 O N O O Pd(dppf)C]2, 032003 OZN o 1, 4--dioxane/HZO 110°C, 16h 58% O O VLCI Pd/C H2 5 EA, r..,t 90%H16h,ZINON\4020 TEA, DCM, 02 0):) °OC, 10min Example? tert-butyl 4-(6-chlorocyanopyridin-Z-priperazine-l-carboxylate (3). To a solution of 6- chloro(4-phenoxyphenyl)nicotinamide 1 (130 mg, 0.4 mmol) and 4-nitrophenylboronic acid 2 (67 mg, 0.4 mmol) in DME (10 mL) /water (3 mL), was added K2C03 (110 mg, 0.8 mmol) and Pd(dppf)Clz (33 mg, 0.04 mmol) under en atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and d for 5 h under nitrogen atmosphere. After cooling to room temperature, the t was evaporated and the crude product was purified by flash chromatography eluting with 70:1 DCM/MeOH to afford the title compound as a white solid (60 mg, crude). MS (ESI): m/z = 412.1 [M+H]+. 6-(4-aminophenyl)(4-phenoxyphenyl)nic0tinamide (4). To a solution of tert-butyl 4-(6— cyanopyridinyl)piperazinecarboxylate 3 (60 mg, 0.15 mmol) in ethyl acetate (5 mL) was added Pd/C (6 mg) under en atmosphere, and the mixture was degassed with hydrogen 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude product as brown solid (42 mg, 90%). MS (ESI): m/Z = 382.0 [M+H]+.
Example 7 6-(4-acrylamidophenyl)(4-phen0xyphenyl)nicotinamide | \/ NH2 0 O N O Q leu 0 To a solution of 6-(4-aminophenyl)—2-(4-phenoxyphenyl)nicotinamide 4 (42 mg, 0.11 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and acryloyl chloride 5 (10 mg, 0.11 mmol) at 0 oC. The mixture was stirred at 0 0C for 10 minutes. The t was removed and the residue was purified by Prep-TLC eluting with 25 :1 DCM/MeOH to afford the title compound (2.5 mg, %) as white solid. 1H NMR (300 MHz, CD3OD) 8 8.17—8.11 (m, 1H), 7.98—7.92 (m, 1H), 7.90—7.84 (m, 1H), 7.81 (s, 3H), 7.69— 7.60 (m, 2H), 7.58—7.52 (m, 1H), 7.42—7.34 (m, 2H), 7.18—7.12 (m, 1H), .00 (m, 3H), 6.47— 6.38 (m, 2H), 5.82—5.76 (m, 1H). MS (ESI, method A): m/z = 435.9 [M + H]+, tR=1.540 min., HPLC: 97.5% (214nm), 98.7% (254nm).
Scheme 6 fo ,Tf <3 i?"3:3? ‘9? (To LDA B\o THF -78°C-rt (JCT Pd(dppf)C[2 KOAC1 4-dioxane Boc 6h, 85% B06 85°C 16h, 54% Bee 1 5 \ \ | NHZ Boc I I CI N/ N Nit—>00Pd/C H2 N Pd(dPPf)012 052003 O EA r.t. 16h 14--dioxaneIH20 Bee Bee 7 6 90°C, 16h, 71% 8 \ NH2 I TFA | / 1—> N TEA DCM DCM 0°C 1h , , N 0 0 0 10mino 41% H 9 34% utyl 5-(trifluoromethylsulfonyloxy)—3,4-dihydr0pyridine-1(2H)—carb0xylate (3). To a solution of tert-butyl 3-oxopiperidine-l-carboxylate l (495 mg, 5.0 mmol) in tetrahydrofuran (10 mL) in a three-necked flask was added m diisopropylamide (2.0M, 2.5 mL, 5.0 mmol) at - 78 °C. After stirring for 2 h at -78 °C, a solution of trifluoro—N-phenyl-N— (trifluoromethylsulfonyl)methanesulfonamide 2 (1.8 g, 5.0 mmol) in tetrahydrofuran (5 mL) was added, and the solution was stirred for another 30 min at this temperature, then warmed to room temperature and stirred for 3 h. Water (30 mL) was added to quench the reaction, and the on was extracted with ethyl acetate (3 X 30 mL). The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with eum ether to afford the title compound as brown oil (1.4 g, 85%). tert-butyl-S-(4,4,5,5-tetramethyl—1,3,2-dioxab0rolanyl)-3,4-dihydr0pyridine-1(2H)- carboxylate (5). To a solution of 4,4,4',4',5,5,5',5'—octamethyl-2,2'—bi(1,3,2—dioxaborolane) 4 (559 mg, 2.2 mmol), KOAc (588 mg, 6.0 mmol) and tert-butyl 5-(trifluoromethylsulfonyloxy)— 3,4-dihydropyridine-1(2H)-carboxylate 3 (662 mg, 2.0 mmol) in dry l,4-dioxane (10 mL) was added Pd(dppf)Clz'DCM (326 mg, 0.4 mmol) under en atmosphere, and the mixture was ed with nitrogen 6 times, then heated to 85°C and stirred overnight under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by silica gel column chromatography, g with 100:1 to 20:1 PE/EA to afford the title compound as brown oil (336 mg, 54%). tert-butyl(5-carbamoyl-6—(4-phenoxyphenyl)pyridin-2—yl)—3,4—dihydr0pyridine-1(2H)— ylate (7). To a solution of tert-butyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl) -3,4- dihydropyridine-1(2H)- carboxylate 5 (260 mg, 0.84 mmol), C82CO3 (456 mg, 1.4 mmol) and 6- chloro(4-phenoxyphenyl)nicotinamide 6 (227 mg, 0.7 mmol) in 1,4-dioxane (10 mL) was added Pd(dppf)Clz'DCM (57 mg, 0.07 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and stirred overnight under nitrogen atmosphere. After cooling to room ature, the solvent was evaporated and the crude product was d by silica gel column chromatography eluting with 100:1 to 20:1 DCM/MeOH to afford the title compound as white solid (240 mg, 71%). MS (ESI): m/Z = 472.2 [M + Hr. tert-butyl 3-(5-carbam0yl—6—(4-phenoxyphenyl)pyridinyl)piperidine-l-carboxylate (8).
To a solution of tert-butyl 5-(5 -carbamoyl(4-phenoxyphenyl)pyridinyl)-3,4- dihydropyridine-1(2H)- carboxylate 7 (240 mg, 0.5 mmol) in ethyl acetate (5 mL) was added Pd/C (24 mg) under hydrogen atmosphere, and the mixture was degassed with hydrogen 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude product as brown solid (230 mg, crude). MS (ESI): m/Z = 474.2 [M + Hr. 2-(4-phenoxyphenyl)(piperidinyl)nicotinamide (9). To a solution of tert—butyl 3-(5- carbamoyl(4-phenoxyphenyl)pyridinyl)piperidinecarboxylate 8 (150 mg, 0.32 mmol) in dry dichloromethane (6 mL) was added TFA (2 mL), and the resulting mixture was stirred for 1 h at ambient ature. The solvent was removed and the residue was partitioned between ted aqueous sodium bicarbonate (30 mL) and ethyl acetate (20 mL). The organic phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated. The crude residue was purified by silica gel column tography eluting with 5:1 DCM/MeOH to afford the title compound as a white solid (77 mg, 71%). MS (ESI): m/z =374.2 [M + H]+.
Example 8 6-(1-acryloylpiperidinyl)—2-(4-phenoxyphenyl)nicotinamide To a solution of 2-(4-phenoxyphenyl)—6-(piperidin-3—yl)nicotinamide 9 (19 mg, 0.05 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and acryloyl chloride 10 (5 mg, 0.05 mmol) at 0°C. The mixture was stirred at 0°C for 10 minutes. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 :1 DCM/MeOH to afford the title nd (7 mg, 34%) as white solid. 1H NMR (300 MHZ, CDClg) 8 7.97 (d, J: 8.9 Hz, 1H), 7.69 (d, J: 8.1 Hz, 2H), 7.38 (t, J: 7.8 Hz, 2H), 7.24—7.15 (m, 2H), 7.12—7.01 (m, 4H), 6.62 (dd, J: 16.6, 10.6 Hz, 1H), 6.28 (d, J= 16.4 Hz, 1H), 5.75—5.60 (m, 2H), 5.46 (s, 1H), 4.89—4.55 (m, 1H), 4.30—3.90 (m, 1H), 3.50 (t, J: 12.2 Hz, 0.5H), 3.24—3.06 (m, 1H), 2.99 (d, J: 10.7 Hz, 1H), 2.80 (t, J: 12.2 Hz, 0.5H), 2.13 (s, 1H), 1.87 (d, J: 13.4 Hz, 2H), 1.64 (s, 1H). MS (ESI, method A): m/z = 428.0 [M + H]+, tR=1.526 min., HPLC: 93.3% (214nm), 99.8% (254nm).
Scheme 7 HN§>—NH3°C 0 elm?" K200: ethanol \ NH2 N N/ H2804(conc)/H20 CI l —’ / 90°C, 1h. 96% DN N Cl reflux 6h 70% HN Boc HZN 3 4 E103:" O f)C|2, 052003 1 ,4—dioxane/HZO GIN: Who—,0 \jLNO": H2N TEA, DCM, NHZOQ 110°C, 1611, 31% 0°C,10min Example9 utyl 1-(6-chlorocyanopyridiny])piperidin-4—ylcarbamate (3). To a on of 2,6- dichloronicotinonitrile 1 (346 mg, 2.0 mmol) in ethanol (10 mL) was added tert-butyl piperidin- 4-ylcarbamate 2 (400 mg, 2.0 mmol) and K2C03 (552 mg, 4.0 mmol). The mixture was heated to reflux stirred for 4 h. After g to room temperature, the solvent was evaporated and the crude product was purified by silica gel column chromatography eluting with 20:1 to 5 :1 PE/EA to afford the title compound as a white solid (470 mg, 70%). MS (ESI, method A): m/Z = 337.2 [M + H].+ 6-(4-aminopiperidinyl)chloronicotinamide (4). To tert—butyl 1-(6-chlorocyanopyridin- 2-yl) piperidinylcarbamate 3 (470 mg, 1.4 mmol) was added H2804 (conc., 5 mL) and water (1 mL). The mixture was heated to 90°C and stirred for 1 h. After cooling to room temperature, the solution was poured into ice-cold water, then adjusted to PH = 8 with ammonia water. The precipitate was filtered, washed with water (10 mL) and dried under vacuum to afford the title compound as a white solid (340 mg, 96%). MS (ESI): m/z = 255.0 [M + H]+. 6-(4-amin0piperidinyl)—2—(4-phenoxyphenyl)nicotinamide (6). To a solution of 6-(4- aminopiperidinyl)—2-chloronicotinamide 4 (51 mg, 0.2 mmol), K2C03 (55 mg, 0.4 mmol) and 4-phenoxyphenylboronic acid 5 (43 mg, 0.2 mmol) in 1,4-dioxane (15 mL) and water (3 mL) was added Pd(dppf)Clz (18 mg, 0.02 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and stirred for 5 h under nitrogen atmosphere. After cooling to room ature, the solvent was evaporated and the crude t was purified by silica gel column chromatography eluting with 70:1 DCM/MeOH to afford the title compound as a white solid (24 mg, 31%). MS (ESI): m/Z = 389.2 [M + H]+.
Example 9 crylamid0piperidinyl)(4-phenoxyphenyl)nic0tinamide VLN ONO/ D To a solution of 6-(4—aminopiperidin—1-yl)(4-phenoxyphenyl)nicotinamide 6 (62 mg, 0.16 mmol) in DCM (5 mL) was added TEA (0.05 mL, 0.4 mmol) and acryloyl chloride 7 (15 mg, 0.16 mmol) at 0°C. The mixture was d at 0°C for 10 minutes. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 :1 OH to afford the title compound (14 mg, 20%) as white solid. 1H NMR (300 MHz, CDC13) 8 7.98 (d, J = 8.8 Hz, 1H), 7.62 (d, J: 8.5 Hz, 2H), 7.38 (t, J: 7.8 Hz, 2H), 7.16 (t, J: 7.4 Hz, 1H), 7.12—7.00 (m, 4H), 6.65 (d, J: 8.9 Hz, 1H), 6.31 (d, J: 16.9 Hz, 1H), 6.07 (dd, J=16.9, 10.2 Hz, 1H), 5.66 (d, J: .2 Hz, 1H), 5.52—5.48 (m, 1H), 5.28 (s, 1H), 4.44 (d, J= 13.4 Hz, 2H), 4.24—4.08 (m, 1H), 3.08 (t, J: 11.9 Hz, 2H), 2.08 (d, J: 10.7 Hz, 2H), 1.62 (s, 1H), 1.47 (dt, .1: 11.3, 6.1 Hz, 2H).
MS (ESI, method A): m/z = 443.2 [M + H]+, tR=1.434 min., HPLC: 99.1% (214nm), 99.3% (254nm).
Scheme8 HNQ/ ‘Boc 0 2 \ \ NH2 \ | 1 K2003, ethanol / H2804(C0nC)/H20 / | N N Clo—o> N N CI CI N CI 9091111964 reflux, 6h, 70% Boc—‘NH H2N 1 4 CL 0°" 0 |\ NH2 Pd(dPPf)C|2.Cszcos N N —> NHo/Q 1,4—dioxane/H20 H N H%:lN\TEA, DCM 110°C, 16h, 31% 5 0°C 10min % \0 Example 10 tert-butyl 1-(6-chlor0cyanopyridin-Z-yl)pyrrolidinylcarbamate (3). To a solution of 2, 6-dichloronicotinonitrile 1 (346 mg, 2.0 mmol) in ethanol (10 mL) was added tert-butyl piperidinylcarbamate 2 (372 mg, 2.0 mmol) and K2C03 (552 mg, 4.0 mmol). The mixture was heated to reflux and stirred for 4 h. After cooling to room temperature, the solvent was evaporated and the crude product was purified by silica gel column chromatography eluting with :1 to 5:1 PE/EA to afford the title compound as a white solid (365 mg, 57%). MS (ESI, method A): m/Z = 345.1 [M + Nar. min0piperidinyl)—2—chloronicotinamide (4). To tert-butyl l-(6-chlorocyanopyridin- 2-yl) pyrrolidiny1carbamate 3 (357 mg, 1.1 mmol) was added H2804 (00:10., 5 mL) and water (1 mL). The mixture was heated to 90°C and stirred for 1 h. After cooling to room temperature, the solution was poured into ld water, then adjusted to PH = 8 with ammonia water. The precipitate was filtered, washed with water (10 mL) and dried under vacuum to afford the title compound as brown solid (192 mg, 72%). MS (ESI, method A): m/z = 241.0 [M + H]+. minopyrrolidinyl)(4-phenoxyphenyl)nic0tinamide (6). To a solution of 6-(4- iperidin-l-yl)—2-chloronicotinamide 4 (192 mg, 0.79 mmol), K2C03 (220 mg, 1.6 mmol) and 4-phenoxyphenylboronic acid 5 (171 mg, 0.8 mmol) in 1,4-dioxane (15 mL) and water (3 mL) was added Pd(dppf)Clz (69 mg, 0.08 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90°C and stirred for 5 h under en atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting with 70:1 DCM/MeOH to afford the title compound as a white solid (92 mg, 31%). MS (ESI): m/z = 375.2 [M + H]+.
Example 10 6-(3-acrylamid0pyrrolidinyl)(4-phen0xyphenyl)nicotinamide l NH2 Q N Oo To a on of 6-(3—aminopyrrolidin-l-yl)(4-phenoxyphenyl)nicotinamide 6 (92 mg, 0.25 mmol) in DCM (5 mL) was added TEA (0.14 mL, 1.0 mmol) and acryloyl chloride 7 (23 mg, 0.25 mmol) at 0 oC. The mixture was stirred at 0 0C for 10 s. The solvent was removed and the residue was purified by Prep—TLC g with 25 :1 DCM/MeOH to afford the title compound (58 mg, 54%) as white solid. 1H NMR (300 MHz, CDCl3) 8 7.95 (d, J = 8.7 Hz, 1H), 7.62 (d, J= 8.5 Hz, 2H), 7.38 (t, J= 7.8 Hz, 2H), 7.16 (t, J= 7.4 Hz, 1H), 7.05 (dd, J= 12.6, 8.3 Hz, 4H), 6.40—625 (m, 1H), 6.10 (dd, J: 16.8, 10.2 Hz, 2H), 5.67 (d, J: 10.2 Hz, 1H), 5.48 (s, 1H), 5.31 (s, 1H), 4.71 (d, J: 4.4 Hz, 1H), 3.85 (dd, J: 11.2, 6.0 Hz, 1H), 3.75—340 (m, 4H), 2.40—2.25 (m, 1H), 2.10—1.90 (m, 1H). MS (ESI, method A): m/z = 429.0 [M + H]+, tR=l.443 min., HPLC: 100% (214nm), 100% (254nm).
Example 1 1 1-(1-acryloylpiperidinyl)(4-phenoxyphenyl)—1H-pyrazole-4—carb0xamide PhO0"):o ""2 Refer to Example 34 schemes and associated experimental procedures and data bed below.
Example 12 1-(1-acryloylpiperidinyl)—3-(4-phen0xyphenyl)-1H-pyrazole-4—carboxamide PhDM0 "HZ Refer to Example 34 schemes and associated experimental procedures and data described below.
Example 13 cryloylpyrrolidinyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarb0xamide PhD 0 ""2 Refer to Example 34 schemes and associated experimental ures and data described below.
Example 14 1-(azetidinyl)—3-(4—phenoxyphenyl)—1H-pyrazolecarb0xamide PhOMo NH2 Refer to Example 34 scheme and associated experimental procedures and data described below.
Scheme 9 N'NH HO TsC|,Pyridine Tso d C 3"0 b— 3 OE. G LiOH/EtOH , ‘NQN’BOC N D —.N . overni9 ht .
BOG BOG 052003 DMF o 100°C; 3h NH4CLHATU O HC|,DCM,rt ‘N’CN’BOC —— DIEA,DMF 7 Example 15 tert-butyl 3-(tosyloxy)azetidine—l-carboxylate (2). To a solution of tert-butyl 3- hydroxyazetidinecarboxylate 1 (2.0 g, 11.5 mmol) in pyridine (25 mL) was added TsCl (2.64 g, 13.8 mmol), and the resulting solution was stirred for 16 h at rt. The solvent was evaporated and the crude residue was purified by silica gel column, eluting with 5 :1 petroleum ether/ethyl acetate to afford the title compound (3.2 g, 85%) as colorless oil. MS (ESI): m/z = 350.0 [M+Na]+.
Ethyl(1-(tert-butoxycarbonyl)azetidin-3—yl)—3-(4-phenoxyphenyl)—1H—pyrazole carboxylate (4). To a solution of utyl yloxy)azetidine-l-carboxylate 2 (1.3 g, 3.88 mmol) and CsC03 (2.12 g, 6.48 mmol) in DMF (25 mL) was added ethyl 3-(4-phenoxyphenyl)- 1H-pyrazolecarboxylate 3 (1.0 g, 3.24 mmol), and the resulting solution was d for 16 h at 100°C. The solvent was evaporated and the crude residue was purified by silica gel column g with 3:1 petroleum ether/ethyl acetate to afford the title compound (1.39 g, 93%) as white solid. MS (ESI): m/Z = 464.0 . 1-(1-(tert—butoxycarbonyl)azetidinyl)—3-(4-phenoxyphenyl)—1H-pyrazole—4-carboxylic acid (5). To a solution of ethyl 1-(1-(tert-butoxycarbonyl)azetidinyl)—3-(4-phenoxyphenyl)- lH—pyrazole-4—carboxylate 4 (1.39 g, 3.0 mmol) in EtOH (20 mL) and water (2 mL) was added LiOH (630 mg, 15.0 mmol), and the resulting solution was stirred for 16 h at rt. The solvent was evaporated, and the e was dissolved in water (5 mL) and the resulting solution was acidified with 2 N hydrochloric acid to pH = 6. The precipitate was filtered, washed with water (15 mL) and dried under vacuum to afford the title compound (1.2 g, 92%) as a white solid. MS (ESI): m/Z = 436.0 [M+H]+. tert-butyl(4-carbamoyl(4-phenoxyphenyl)—1H-pyrazol—1-yl)azetidine—l-carboxylate (6). To a solution of 1-(1-(tert-butoxycarbonyl)azetidinyl)(4-phenoxyphenyl)-1H-pyrazole- 4-carboxylic acid 5 (800 mg, 1.84 mmol), NH4C1 (120 mg, 2.21 mmol) and HATU (1.04 mg, 2.76 mmol) in dry DMF (10 mL) was added DIPEA (957 mg, 7.36 mmol), and the resulting solution was stirred overnight at rt. After the reaction was completed, the solution was concentrated, d with ethyl acetate (30 mL) and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium e, filtered and trated.
The residue was purified by silica gel column eluting with 40:1 OH to afford the title compound (700 mg, 82%) as white solid. MS (ESI): m/z = 435.0 [M+H]+. 1-(azetidinyl)(4-phen0xyphenyl)-1H-pyrazolecarb0xamide (7). To a solution of 3-(4- carbamoyl(4-phenoxyphenyl)-1H-pyrazol—1-yl)azetidinecarboxylate 6 (700 mg, 1.61 mmol) in DCM (20 mL) was added conc. HCl (5 mL) and the reaction mixture was stirred at rt for 1 h. After the reaction was completed, the solution was concentrated to afford the title compound (600 mg, 100%) as white solid. MS (ESI): m/z = 335.0 [M + H]+.
Example 15 1-(1-acryloylazetidinyl)—3-(4-phen0xyphenyl)—1H-pyrazole—4-carb0xamide /N\N’CN/<; To a solution of tidinyl)—3-(4-phenoxyphenyl)—1H—pyrazolecarboxamide 7 (300 mg, 0.81 mmol) in dry DCM (15 mL) were added DIEA (316 mg, 2.43 mmol) and acryloyl chloride (88 mg, 0.97 mmol) at 0°C, and the resulting solution was stirred at 0°C for 10 min. Water (10 mL) was added to quench the reaction. The mixture was diluted with DCM (20 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by Prep—HPLC zO = -70, 0.1%FA) to afford the title compound (160 mg, 51%) as a white solid. 1H NMR (400 MHz, DMSO) 5 8.38 (s, 1H), 7.81 (d, J: 8.7 Hz, 2H), .38 (m, 3H), 7.20—7.14 (m, 1H), 7.11—6.99 (m, 5H), 6.43—6.34 (m, 1H), 6.20—6.12(m, 1H), 5.76—5.70 (m, 1H), 5.38—5.29 (m, 1H), 4.78—4.68 (m, 1H), 4.59—4.49 (m, 1H), 4.48—4.40 (m, 1H), 4.32—4.22 (m, 1H). MS (ESI, Method A): m/Z = 389.1 [M + H]+, tR = 1.374 min. HPLC: 99.7% ), 99.7% (254nm).
Example 16 1-(4-acrylamid0phenyl)—3-(4-phenoxyphenyl)—1H—pyrazolecarb0xamide HN’<§ PhOMo NH2 Refer to Example 34 schemes and associated experimental procedures and data bed below.
Example 17 1-(3-aminophenyl)—3-(4-phen0xyphenyl)—1H-pyrazolecarboxamide PhDMo ""2 Refer to Example 34 schemes and associated experimental procedures and data described below.
Example 18 1-(3-acrylamid0phenyl)(4-phen0xyphenyl)-1H-pyrazolecarb0xamide Refer to Example 34 schemes and associated experimental procedures and data described below.
Example 19 (S)(1-acryloylpyrrolidinyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarboxamide Refer to Example 34 schemes and associated experimental procedures and data described below.
Example 20 (R)(1-acryloylpyrrolidinyl)(4-phen0xyphenyl)—1H-pyrazole—4-carboxamide N’ [\I PhDMo ""2 Refer to Example 34 schemes and associated experimental ures and data described below.
Scheme 10 0 OH O C] gOH "Hz 3H PCI5, POC13 g NH3 in dioxane gm CL 0‘OH 0 N?" N/N —’ 115 °C, overnight Y 0 °c to ,1 NYN Pd(dppf)C[2, K2003, OH Cl C] dioxane—HZO (8/1 V) 60 O 1 2 c, 3 h it Pd(PPh3)4,K2003 N \ 0 c1 N —» I DME-HZO (8/1 V) N I 85 °C, ght Boc Boc’N OO NI \ 0 Pd/C, H2 CF3COOH —> 000 EtOAc, rt, overnight N Boc’ O)\N/ DCM, It 2 h \JLCI DCM 0 °C 1 h q©fi$oQ e 21 2,4-dichloropyrimidine—S-carbonyl chloride (2). To a on of 2,4-dihydroxypyrimidine carboxylic acid (5.0 g, 32.0 mmol) in POC13 (50 mL) was added PC15 (23.9 g, 115.2 mmol), and the resulting solution was heated to 115 °C and stirred for 12 h. The solvent was evaporated and the crude residue was d with ethyl acetate (100 mL). The solid was filtered and the filtrate was concentrated to give the title compound as brown oil (6.2 g, 92%). 2,4-dichloropyrimidine-S-carboxamide (3). To a solution of 2,4-dichloropyrimidine carbonyl de (6.2 g, 29.3 mmol) in 1,4-dioxane (50 mL) was added ammonia (1.0 g, 58.3 mmol) in 1,4-dioxane (50 mL) dropwise at 0 0C, and the resulting solution was stirred for 12 h at ambient temperature. The mixture was diluted with ethyl acetate (50 mL), and washed with water (2 X 80 mL) and brine (2 X 80 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound as a white solid (4.5 g, 80%). MS (ESI): m/Z = 191.8 [M + Hr. 2-chloro(4—phenoxyphenyl)pyrimidine—S-carboxamide (4). To a solution of 4- phenoxyphenylboronic acid (1.0 g, 4.7 mmol), K2C03 (1.4 g, 10.4 mmol) and 2,4- dichloropyrimidine-5—carboxamide (1.0 g, 5.2 mmol) in 1,4—dioxane (24 mL) and water (3 mL) was added Pd(dppf)C12 (380 mg, 0.52 mmol) under en atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 60 °C and stirred for 3 h under nitrogen atmosphere. After cooling to room temperature, the solution was poured into water (50 mL), and then extracted with ethyl acetate (3 X 40 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column tography, eluting with 100 : l dichloromethane/methanol to afford the title compound as a brown solid (410 mg, 24%). MS (ESI): m/Z = 325.9 [M + H]+. tert-butyl 4-(5-carbamoyl-4—(4-phenoxyphenyl)pyrimidinyl)—5,6-dihydr0pyridine—1(2H)- carboxylate (6). To a solution of 2-chloro-4—(4-phenoxyphenyl)pyrimidine—5-carboxamide 4 (410 mg, 1.26 mmol), K2C03 (521.6 mg, 3.78 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl —1,3,2- dioxaborolan—2—yl)-5,6—dihydropyridine-1(2H)—carboxylate 5 (583 mg, 1.89 mmol) in DME (30 mL) and water (5 mL) was added Pd(PPh3)4 (146 mg, 0.126 mmol) under nitrogen atmosphere, and the mixture was degassed with en 6 times, and then heated to 85 °C and stirred for 12 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography g with 80 : 1 DCM/MeOH to afford the title compound as a brown solid (420 mg, 71%). MS (ESI): m/Z = 472.8 [M + H]+. tert-butyl 4-(5-carbam0yl—4—(4-phenoxyphenyl)pyrimidinyl)piperidine—l-carboxylate (7).
To a solution of tert—butyl 4-(5-carbamoyl-4—(4-phenoxyphenyl)pyrimidin-2—yl)—5,6— dihydropyridine-1(2H)- carboxylate 6 (100 mg, 0.212 mmol) in ethyl acetate (5 mL) was added Pd/C (20 mg) under hydrogen atmosphere, and the mixture was ed with hydrogen 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The on was filtered and the filtrate was evaporated to the crude product as brown solid (96 mg). MS (ESI): m/Z = 474.8 [M+H]+. 4-(4-phen0xyphenyl)(piperidinyl)pyrimidinecarb0xamide (8). To a solution of tert- butyl 4-(5-carbamoyl—4-(4-phenoxyphenyl)pyrimidinyl) piperidine-l-carboxylate 7 (96 mg, crude) in dry dichloromethane (2 mL) was added TFA (2 mL), and the resulting mixture was stirred for 1 h at ambient temperature. The solvent was removed and the residue was partitioned between saturated aqueous sodium bicarbonate (30 mL) and ethyl e (20 mL). The c phase was separated, dried over anhydrous sodium sulfate, d and concentrated. The crude residue was purified by flash chromatography, eluting with 5 : l DCM/MeOH to afford the title compound as a white solid (70 mg, 88% for two . MS (ESI): m/z = 374.9 [M+H]+.
Example 21 2-(1-acryloylpiperidinyl)—4-(4-phenoxyphenyl)pyrimidinecarb0xamide N \ O O"I / W" O To a solution of 4-(4-phenoxyphenyl)(piperidin-4—yl)pyrimidinecarboxamide 8 (70 mg, 0.187 mmol) in DCM (3 mL) was added TEA (56.7 mg, 0.56 mmol) and yl chloride (25.3 mg, 0.28 mmol) at 0°C. The mixture was stirred at 0°C for 10 min. The t was removed and the residue was purified by Prep-TLC eluting with 25 : 1 DCM/MeOH to afford the title compound (50 mg, 63%) as light yellow solid. 1H NMR (400 MHz, CDCl3) 5 8.95 (s, 1H), 7.82 (d, J= 8.8 Hz, 2H), 7.49—7.36 (m, 2H), 7.27—7.17 (m, 1H), 7.16—7.04 (m, 4H), 6.64 (dd, J: 16.8, 10.6 Hz, 1H), 6.29 (dd,.]= 16.8, 1.9 Hz, 1H), 5.94 (s, 1H), 5.77 (s, 1H), 5.71 (dd,.]= 10.6, 1.9 Hz, 1H), 4.76 (d, J: 12.5 Hz, 1H), 4.14 (d, J: 14.1 Hz, 1H), 3.37—3.19 (m, 2H), 2.89 (t, J: 11.5 Hz, 1H), 2.16 (d, J= 11.0 Hz, 2H), 1.97 (d, J= 9.9 Hz, 2H). MS (ESI, method F): m/z = 428.8 [M + H]+, tR=1.407 min., HPLC: 99.8% (214nm), 99.6% (254nm).
Scheme 11 OTf 0335:? 3% NH2 fig PhN(CF3802)2 / >§(N:I)NI\N,O N| \ NH2 CF3COOH CI OXN/ N 0 DCMrch ODCMO°C1h\O [’3/ o 7 Example 22 1-(tert-but0xycarbonyl)—2,5-dihydro-1H-pyrrol—3-yl trifluoromethanesulfonate (2). To a solution of tert—butyl 3-oxopyrrolidinecarboxylate 1 (5.0 g, 27.0 mmol) in tetrahydrofuran (130 mL) in a three-necked flask was added lithium diisopropylamide (2.0M, 16.2 mL, 32.4 mmol) at -78 °C. After stirring for 2 h at -78 °C, a solution of trifluoro-N-phenyl-N— (trifluoromethylsulfonyl) methanesulfonamide (10.1 g, 28.4 mmol) in tetrahydrofiiran (20 mL) was added, and the solution was stirred for r 30 min at this temperature, then warmed to room temperature and stirred for 3 h. Water (30 mL) was added to quench the reaction, and the solution was extracted with ethyl acetate (3 X 200 mL). The c layers were combined, dried over anhydrous sodium e, filtered and trated. The residue was purified by silica gel column chromatography, eluting with petroleum ether to afford the title compound as a brown oil (4.2 g, 49%).MS (ESI): m/z = 318.1 [M + H]+. tert-butyl-(4,4,5,5-tetramethyl—1,3,2-dioxabor01anyl)-2H-pyrrole-1(5H)-carb0xylate (3).
To a solution of bis(pinacolato)diborone (4.0 g, 15.9 mmol), KOAc (2.59 g, 26.4 mmol) and 1- (tert—butoxycarbonyl)—2,5-dihydro-1H—pyrrolyl trifluoromethanesulfonate 2 (4.2 g, 13.2 mmol) in dry 1,4-dioxane (10 mL) was added Pd(dppf)C12 (965 mg, 1.32 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 85 OC and stirred overnight under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude t was ed by silica gel column chromatography, eluting from 100 : 1 to 20 : 1 eum ether/ethyl e to afford the title compound as brown oil (1.2 g, 31%). MS (ESI): m/Z = 296.1 [M + H. tert-butyl(5-carbamoyl-4—(4-phenoxyphenyl)pyrimidinyl)-2H-pyrrole—1(5H)— carboxylate (5). To a solution of tert—butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl)—2H— pyrrole-1(5H)- carboxylate 3 (136 mg, 0.46 mmol), K2C03 (127 mg, 0.92 mmol) and 2-chloro (4-phenoxyphenyl)pyrimidinecarboxamide 4 (100 mg, 0.31 mmol) in 1,4-dioxane (15 mL) and water (3 mL) was added Pd(PPh3)4 (35.5 mg, 0.031 mmol) under nitrogen here. The mixture was degassed with nitrogen 6 times, then heated to 90 CC and stirred for 12 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting with 70 : 1 DCM/MeOH to afford the title compound as a white solid (90 mg, 64%). MS (ESI): m/Z = 458.8 [M + H]+. tert-butyl(5-carbamoyl-4—(4-phenoxyphenyl)pyrimidinyl)pyrrolidine—l-carboxylate (6). To a solution of tert-butyl 3-(5-carbamoyl(4-phenoxyphenyl)pyrimidinyl)-2H-pyrrole- 1(5H) -carboxy1ate 5 (90 mg, 0.20 mmol) in ethyl acetate (3 mL) and MeOH (3 mL) was added Pd/C (20 mg) under hydrogen atmosphere, and the mixture was degassed with en 6 times, then stirred for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude product as brown solid (80 mg, 88%). MS (ESI): m/Z = 461.1 [M+H]+. 4-(4-phenoxyphenyl)—2-(pyrrolidinyl)pyrimidine—S-carboxamide (7). To a solution of tert- butyl 3-(5 -carbamoyl(4-phenoxyphenyl)pyrimidinyl)pyrrolidine-l- carboxylate 6 (80 mg, 0.174 mmol) in dry dichloromethane (2 mL) was added TFA (2 mL), and the resulting mixture was stirred for 1 h at ambient temperature. The solvent was removed and the residue was partitioned n saturated aqueous sodium onate (30 mL) and ethyl acetate (20 mL).
The c phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound as a white solid (70 mg, . MS (ESI): m/z = 360.8 [M+H]+.
Example 22 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)pyrimidine—5-carb0xamide To a solution of 4-(4-phenoxyphenyl)(pyrrolidinyl)pyrimidinecarboxamide 7 (70 mg, crude) in DCM (5 mL) was added TBA (59 mg, 0.58 mmol) and acryloyl chloride (26 mg, 0.29 mmol) at 0°C. The mixture was stirred at 0°C for 10 min. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 : 1 DCM/MeOH to afford the title compound (20 mg, 25%) as a white solid. 1H NMR (400 MHZ, CDClg) 5 8.95 (d, J: 6.2 Hz, 1H), 7.82 (d, J = 6.6 Hz, 2H), 7.42 (t, J: 7.8 Hz, 2H), 7.22 (t, J: 7.4 Hz, 1H), 7.10 (t, J: 7.9 Hz, 4H), 6.55— 6.47 (m, 1H), 6.39 (d,J= 16.8, Hz, 1H), 5.84 (s, 2H), 5.71 (d, J: 10.1 Hz, 1H), 4.14—3.96 (m, 2H), .78 (m, 2H), 3.76—3.62 (m, 1H), .36 (m, 2H). MS (ESI, method F): m/z = 414.8 [M + H]+, tR=1.382 min, HPLC: 99.0% (214nm), 98.4% ).
Scheme 12 3’0 3)4. \ 0 | K2003 / —> + / CI N N Q DME-HZO (8/1 V) \ 85 Oc, 16 h Boo o 1 2 \ NH \ ""2 2 Pd/C, H2 | CFscOOH I —> N —> N MeOH/EtOAc. rt, overnight DCM, rt, 2 h HN 0 N O , 0 4 5 d \ NH2 CI l —’ N DCM,O°C, 10 min N O / 0 Example 23 tert-butyl—3-(5-carbamoyl(4-phen0xyphenyl)pyridinyl)—2H—pyrrole-1(5H)-carb0xylate (3). To a solution of tert—butyl 3—(4,4,5,5-tetramethyl—1,3,2-dioxaborolanyl)—2H—pyrrole- 1(5H)—carboxylate 1 (136 mg, 0.0.46 mmol), K2C03 (127.5 mg, 0.92 mmol) and 6-chloro(4- phenoxyphenyl) nicotinamide 2 (100 mg, 0.31 mmol) in 1,2-dimethoxyethane (5 mL) and water (1 mL) was added Pd(PPh3)4 (35.6 mg, 0.031 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 85 °C and stirred for 12 h under nitrogen atmosphere. After cooling to room temperature, the t was evaporated and the crude product was purified by flash chromatography, eluting with 50 : 1 DCM/MeOH to afford the title nd as a white solid (84 mg, 60%). MS (ESI): m/z = 458.1 [M+H]+. tert-butyl 3-(5-carbamoyl(4-phenoxyphenyl)pyridin-Z-yl)pyrrolidine-l-carboxylate (4).
To a solution of tert—butyl 3-(5-carbamoyl(4-phenoxyphenyl)pyridinyl)—2H—pyrrole-1(5H)- ylate 3 (84 mg, 0.18 mmol) in ethyl acetate (5 mL) and MeOH (5 mL) was added Pd/C (40 mg) under hydrogen atmosphere, and the mixture was degassed with hydrogen 6 times, then stirred for 12 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to the crude t as brown solid (80 mg, 95%). MS (ESI): m/z = 459.8 [M+H]+. 2-(4-phenoxyphenyl)—6-(pyrrolidinyl)pyridinecarb0xamide (5). To a on of tert- butyl 3-(5 moyl(4-phenoxyphenyl)pyridinyl)pyrrolidinecarboxylate 4 (80 mg, 0.17 mmol) in dry dichloromethane (2 mL) was added TFA (2 mL), and the resulting mixture was stirred for 1 h at ambient temperature. The solvent was removed and the residue was partitioned between saturated aqueous sodium bicarbonate (30 mL) and ethyl acetate (20 mL). The organic phase was separated, dried over anhydrous sodium sulfate, d and concentrated to afford the title compound as a white solid (70 mg, crude). MS (ESI): m/z = 359.9 [M+H]+.
Example 23 6-(1-acryloylpyrrolidinyl)—2-(4-phenoxyphenyl)pyridine—3-carboxamide To a solution of 2-(4-phenoxyphenyl)(pyrrolidinyl)pyridine-3—carboxamide 5 (70 mg, crude) in DCM (5 mL) was added TBA (59 mg, 0.58 mmol) and acryloyl chloride (26.4 mg, 0.29 mmol) at 0 OC. The e was stirred at 0 °C for 10 min. The solvent was removed and the residue was purified by Prep-TLC eluting with 25 : l DCM/MeOH to afford the title compound (18 mg, 25% for two steps) as white solid. 1H NMR (400 MHz, CDC13) 8 8.06 (s, 1H), 7.72 (s, 2H), 7.41 (t, J: 7.8 Hz, 2H), 7.28—7.24 (m, 1H), 7.19 (t, J: 7.4 Hz, 1H), 7.11—7.09 (m, 4H), 6.55—6.47 (m, 1H), 6.41 (d,J= 16.8, Hz, 1H), 5.71 (d,J= 10.1 Hz, 1H), 5.65 (br, 1H), 5.45 (br, 1H), 4.14—3.96 (m, 1H), .78 (m, 2H), 3.76—3.62 (m, 2H), 2.55—2.36 (m, 2H). MS (ESI, method F): m/z = 413.8 [M + H]+, tR=1.393 min, HPLC: 97.3% ), 99.5% (254nm).
Scheme 13 0% O \ o CN ' Va" Boc’N 100 0C’ 3 h o HN o/O DCM, 0 DC, 10 min 1 2 Ar"%l/" 0Q Example 24 2-(4-phen0xyphenyl)—6-(piperidinyl)pyridinecarbonitrile (2). A solution of tert—butyl 4- (5 -carbamoyl(4-phenoxyphenyl)pyrimidinyl)piperidine- 1 xylate (100 mg, 0.21 mmol) in POCl3 (3 mL) was heated to 100 °C and stirred for 3 h. After cooling to room temperature, the reaction mixture was poured into water (3 mL), and basified by saturate aqueous NaHC03 to PH=10. Then the solution was extracted with ethyl acetate (3 X 60 mL). The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated. The crude residue was d by preparative TLC eluting with 8 : 1 DCM/MeOH to afford the title compound as brown oil (10 mg, 13%). MS (ESI): m/z = 356.1 [M + H]+.
Example 24 6-(1-acryloylpiperidinyl)—2-(4-phen0xyphenyl)pyridinecarb0nitrile Ar"0%l/N 0 To a solution of 2-(4-phenoxyphenyl)(piperidinyl)pyridinecarbonitrile 2 (10 mg, 0.028 mmol) in DCM (2 mL) was added TEA (8.5 mg, 0.084 mmol) and acryloyl chloride (3.8 mg, 0.042 mmol) at 0 oC. The mixture was stirred at 0 0C for 10 min. The t was removed and the residue was purified by Prep-TLC eluting with 25 : 1 DCM/MeOH to afford the title compound (13 mg, 44%) as white solid. 1H NMR (400 MHz, CDCl3) 5 8.01 (d, J = 8.1 Hz, 1H), 7.97 (dd, J: 92,23 Hz, 2H), 7.41 (t, J: 7.9 Hz, 2H), 7.24—7.17 (m, 2H), 7.16—7.09 (m, 4H), 6.64 (dd, .1: 16.8, 10.6 Hz, 1H), 6.32 (dd, .1: 16.8, 1.8 Hz, 1H), 5.73 (dd, J: 10.6, 1.9 Hz, 1H), 4.85—4.79 (m, lH),4.21—4.16(m, 25—4.18(m, 1H), 3.12 (tt,J= 12.0, 3.8 Hz, 1H), 2.84— 2.78 (m, 1H), 2.09—2.06 (m, 2H), 1.93—1.82 (m, 2H). MS (ESI, method F): m/Z = 409.8 [M + Hr, tR=1.669 min, HPLC: 98.7% (214nm), 98.8% (254nm).
Scheme 14 WE] \ H? K2003 | "Hz / sto4/H20 |\ —> N N CI / N N CI CI , p EtOH reflux 3h Boo/Nd 90°C,40mm HNQ 2 3 4 ' F’d2(dba)3, 05003 I +11 <1B\0H 1 OH HATU DIPEA 1,4-dioxaneIH20 "NJ o0+w —. o Hg 0 DMF,rt.12h 90°C, 16h | : NH2 " O Example 25 tert-butyl 4-(6-chloro—S-cyanopyridinyl)piperazine-l-carboxylate (3). To a solution of 2,6- dichloronicotinonitrile 1 (1.0 g, 5.78 mmol) and tert—butyl piperazine-l-carboxylate 2 (1.08 g, .78 mmol) in ethanol (20 mL) was added K2C03 (636 mg, 6.0 mmol). The ing solution was refluxed for 3 h. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting from 5:1 to 2:1 PE/EA to afford the title compound as a white solid (800 mg, 43%). MS (ESI): m/z = 345.1 [M + Na]+. 6-chlor0(4—phenoxyphenyl)nicotinamide (4). To tert—butyl-(6-chlorocyanopyridin yl)piper azine-l-carboxylate 3(800 mg, 2.5 mmol) was added conc. H2S04 (5 mL) and water (1 mL). The mixture was heated to 90°C and stirred for 40 min. After cooling to room temperature, the solution was poured into ice-cold water, then adjusted to PH = 8 by ammonia water. The precipitate was d, washed with water (20 mL) and dried under vacuum to afford the title compound as an off-white solid (580 mg, 96%). MS (ESI): m/z = 241.1 [M + H]+. 2-(4-phen0xyphenyl)—6-(piperazinyl)nic0tinamide (6). To a on of 6-chloro(4- phenoxyphenyl)nicotinamide 4 (580 mg, 2.41 mmol), Cs2C03 (2.35 g, 7.23 mmol) and 4— phenoxyphenylboronic acid 5 (619 mg, 2.89 mmol) in l,4-dioxane (15 mL) and water (3 mL) was added Pd2(dba)3 (220 mg, 0.24 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90 °C and stirred for 12 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting with 10:1 DCM/MeOH to afford the title compound as a white solid (480 mg, 52%). MS (ESI): m/z = 375.1 [M + H]+.
Example 25 (E)—6-(4-(4-(dimethylamino)but—2-enoyl)piperazinyl)(4-phen0xyphenyl)nic0tinamide ©| : NH2 A mixture of 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide 6 (100 mg, 0.27 mmol), (E)- 4-(dimethylamino)but—2-enoic acid hydrochloride 7 (53 mg, 0.32 mmol), HATU (152 mg, 0.4 mmol), and DIPEA (172.5 mg, 1.34 mmol) in DMF (10 mL) was stirred at rt for 12 h. The solution was poured into water (50 mL), and then extracted with ethyl acetate (3 X 50 mL). The ed organic layers were dried over anhydrous sodium sulfate, filtered and concentrated.
The residue was d by Prep-TLC eluting with 10:1 DCM/MeOH to afford the title compound (30 mg, 23%) as a white solid. 1H NMR (400 MHz, MeOD) 8 7.78 (d, J = 8.7 Hz, 1H), 7.69 (d, J: 8.6 Hz, 2H), 7.40 (t, J: 7.9 Hz, 2H), 7.17 (d, J: 7.4 Hz, 1H), 7.04 (dd, J: .3, 8.3 Hz, 4H), 6.93 (d, J: 15.3 Hz, 1H), 6.82 (d, J: 8.7 Hz, 1H), 6.77 (m, 1H), 3.89—3.68 (m, 10H), 2.76 (s, 6H). MS (ESI, method F): m/z = 485.9 [M + H]+, tR=1.249 min, HPLC: 93.3% (214nm), 93.0% (254nm).
Scheme 15 \ EHH Pd(dppf)C|2 K2003 (\N N Cl 1 4--dioxane/HZO Boc’N Boc’N 110°C 12h con.HC|, sealed tube IN/ EtN, CH Cl 115°c,5h HNQ OHOQ 3 2 2’ min HCI 4 e 26 2-(4-phen0xyphenyl)—6-(piperazinyl)nic0tinamide (3). To a solution of tert—butyl 4-(6— chlorocyanopyridinyl)piperazinecarboxylate 1 (300 mg, 0.93 mmol), K2C03 (385 mg, 2.79 mmol) and 4-phenoxyphenylboronic acid 2 (297 mg, 1.39 mmol) in 1,4-dioxane (12 mL) and water (2 mL) was added Pd(dppf)Clz (68 mg, 0.093 mmol) under en atmosphere. The mixture was degassed with nitrogen 6 times, then heated to 90 CC and d for 12 h under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by flash chromatography eluting with 20:1 DCM/MeOH to afford the title compound as a white solid (260 mg, 58%). MS (ESI): m/z = 457.1 [M + H]+. 2-(4-phenoxyphenyl)—6-(piperazin-l-yl)nicotinic acid hydrochloride (4). In a 20 mL sealed tube, was placed a solution of 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide 3 (140 mg, 0.31 mmol) in come. HCl (5 mL). The mixture was heated to 115 °C and stirred for 4 h. After cooling to room temperature, the solution was concentrated under vacuum to afford the title compound as an off—white solid (120 mg, 95%). MS (ESI): m/z = 376.1 [M + H]+.
Example 26 cryloylpiperazinyl)(4-phenoxyphenyl)pyridine—3-carb0xylic acid To a solution of 2-(4—phenoxyphenyl)(piperazinyl)nicotinic acid hydrochloride 4 (120 mg, 0.32 mmol) in DCM (5 mL) was added TEA (162 mg, 1.6 mmol) and acryloyl de (35 mg, 0.38 mmol) at 0°C. The mixture was stirred at 0 0C for 10 minutes. The solvent was removed and the residue was d by Prep-TLC eluting with 20:1 DCM/MeOH to afford the title compound (15 mg, 11%) as a white solid. 1H NMR (400 MHz, CDClg) 8 8.17 (d, J= 8.9 Hz, 1H), 7.55 (d, J: 8.3 Hz, 2H), 7.38 (t, J: 7.7 Hz, 2H), 7.16 (t, J: 7.4 Hz, 1H), 7.11 (d, J: 8.0 Hz, 2H), 7.03 (d, J: 8.3 Hz, 2H), 6.65—6.55(m, 2H), 6.38 (d, J: 16.7 Hz, 1H), 5.78 (d, J: 10.6 Hz, 1H), 3.86—375 (m, 8H). MS (ESI, method F): m/z = 429.8 [M + H]+, tR=1.477 min., HPLC: 93.5% (214nm), 93.0% (254nm).
Scheme 16 OH H o K2003 DMF Pd(dppf)C|2 KOAc + 1150c:N/O:OBDMFreflux ,o 1 2 4 O O \ NH2 \ | | NH2 Pd2(dba)3, CSZCO3 N N/ pN N/ dioxane/HZO, 120°C p Et3N CHZCIQO0°C HN O N 06% /\n,01 /j (\N N’ 01 HNd 5 Example 27 4-(4-bromophenoxy)benzonitrile (3). The e of 4-bromophenol 1 (0.50 g, 2.9 mmol), 4- fluorobenzonitrile 2 (0.28 g, 2.31 mmol), K2C03 (0.80 g, 5.8 mmol) and DMF (4 mL) was stirred at 115 °C for 16 h. After cooled to It, the mixture was concentrated in vacuo. The residue was purified by silica gel chromatography eluting with 100:] petroleum ether/EtOAc to afford the title compound (0.59 g, 74%) as white solid. 1H NMR (400 MHZ, DMSO-d6) 8 7.86 (d, J = 8.7 Hz, 2H), 7.64 (d, J: 6.0 Hz, 2H), 7.13 (dd, J: 5.1, 4.7 Hz, 4H). 4-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl)phenoxy)benzonitrile (4). The mixture of 4-(4-bromophenoxy)benzonitrile 3 (0.59 g, 2.15 mmol), acolato diboron (1.09 g, 4.3 mmol), KOAc (0.63 g, 6.45 mmol), f)Clz (0.157 g, 0.215 mmol) and DMF (3.5 mL) was degassed with N2 6 times and then stirred under reflux for 16 h. After cooled to rt, the mixture was concentrated in vacuo. The residue was purified by silica gel chromatography eluting with 50:1 petroleum ether/EtOAc to afford the title compound (0.55 g, 79%) as white solid. 4-cyan0phenoxy)phenyl)—6-(piperazinyl)nic0tinamide (6). The title compound was obtained using a procedure analogous to the ure described in 2-(4- xy(phenyl)methyl)phenyl)(piperidin—4-yl)nicotinamide (see Scheme 48) as yellow solid (0.15 g, 37%). MS (ESI): m/z = 400.1 [M + H.
Example 27 6-(4-acryloylpiperazinyl)—2-(4-(4-cyanophenoxy)phenyl)nicotinamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (40 mg, 23%). 1H NMR (400 MHz, DMSO—d6) 8 7.88 (d, J = 8.7 Hz, 2H), 7.73 (d, J: 8.6 Hz, 2H), 7.68 (d, J: 8.7 Hz, 1H), 7.59 (s, 1H), 7.26 (s, 1H), 7.21—7.14 (m, 4H), 6.86 (dd, J= 17.8, 9.3 Hz, 2H), 6.16 (dd, J= 16.7, 2.2 Hz, 1H), 5.73 (dd, J= 10.4, 2.2 Hz, 1H), 3.66—3.67 (m, 8H). MS (ESI, method A): m/z = 454.1 [M + H]+, tR=1.685 (min). HPLC: 97.1% (214nm), 99.7% (254nm).
Scheme 17 I O OH Cul,1,10-phenanthroline Pd(dppf)C[2 KOAc ngco3 toluene 1200c 0/0of»DMF reflux + 8’0 1 2 4 O O \ \ Ed2(dba)3,ngcog I NH2 I NH2 E N CH Cl 000 / dloxane/H20,120 0C (\N N t3, 2 2. (\N N 0 HNd O O Nd rr" Arc} 0 0 / (‘N N’c 6 Example 28 1-br0mo(cyclohexyloxy)benzene (3). The mixture of 1-bromo-4—iodobenzene l (2.83 g, 10 mmol), cyclohexanol 2 (5.0 g, 50 mmol), CuI (0.381 g, 2.0 mmol), 1,10-phenanthroline (0.793 g, 4.0 mmol), CS2C03 (8.15 g, 25 mmol) and toluene (5 mL) was stirred at 120 °C in a sealed tube under N2 for 16 h. After cooled to rt, the mixture was filtered over celite. The filtrate was dried over NazSO4, concentrated in vacuo and purified by silica gel chromatography eluting with 20:1 eum ether/EtOAc to afford the title compound (1.17 g, 46%) as colorless oil. 1H NMR (400 MHz, CDC13) 5 7.40—7.33 (m, 2H), 6.83—6.77 (m, 2H), 4.26—4.16 (m, 1H), 2.04—1.92 (m, 2H), 1.87—1.75 (m, 2H), 1.65—1.47 (m, 3H), 1.45—1.25 (m, 3H). 2-(4-(cyclohexyloxy)phenyl)—4,4,5,5—tetramethyl-1,3,2-di0xab0rolane (4). The title nd was obtained using a procedure analogous to the procedure described in 4-(4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolanyl)phenoxy)benzonitrile (see Scheme 16) as brown oil (1.04 g, 75%). 1H NMR (400 MHz, CDClg) 6 7.75 (d, J: 8.5 Hz, 2H), 6.91 (d, J: 8.5 Hz, 2H), 4.40— 4.25 (m, 1H), 2.09—1.94 (m, 2H), 1.89—1.76 (m, 2H), 1.66—1.49 (m, 4H), 1.48—1.23 (m, 14H). 2-(4-(cyclohexyloxy)phenyl)(piperazinyl)nicotinamide (6). The title compound was obtained using a procedure analogous to the ure described in 6-(3 -nitrophenyl)(4- yphenyl)nicotinamide (see Scheme 1) as brown gum (0.157 g, 41%). MS (ESI): m/z = 381.1 [M + H.
Example 28 6-(4-acryloylpiperazinyl)(4-(cyclohexyloxy)phenyl)nicotinamide The title compound was obtained using a procedure analogous to the procedure described in e 1 as white solid (61 mg, 34%). 1H NMR(400 MHZ, CD30D) 5 7.76 (d, J = 8.7 HZ, 1H), 7.63 (d, J= 8.7 Hz, 2H), 6.96 (d, J: 8.7 Hz, 2H), 6.87—6.76 (m, 2H), 6.27 (dd, J: 16.8, 1.8 Hz, 1H), 5.80 (dd, J: 10.6, 1.8 Hz, 1H), 4.43—4.35 (m, 1H), .75 (m, 8H), 2.07—1.96 (m, 2H), 1.89—1.78 (m, 2H), 1.63—1.34 (m, 6H). MS (ESI, method A): m/z = 435.2 [M + H]+, tR=1.800 (min). HPLC: 98.2% (214nm), 98.5% (254nm).
Scheme 18 NNHZ Pd2(()3dba,C52003 ()ZHO28\©/\O\ dioxane/HZO 120°C \ NH2 + I HO / p / O N N CI \ Et3N, , o 00 | / O\ MCI O O 0 Z Example 29 2-(3-methoxy—4-methylphenyl)—6-(piperazinyl)nicotinamide (3). The title nd was obtained using a procedure analogous to the procedure described in 2-(4- (hydroxy(phenyl)methyl)phenyl)(piperidinyl)nicotinamide (see Scheme 48) as brown solid (0.238 g, 58%). MS (ESI): m/z = 327.1 [M + H.
Example 29 6-(4-acryloylpiperazinyl)—2-(3-meth0xy—4-methylphenyl)nicotinamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (115 mg, 43%). 1H NMR (400 MHZ, CDC13) 5 7.77 (d, J: 8.7 HZ, 1H), 7.23 (s, 1H), 7.21—7.15 (m, 2H), 6.88—6.78 (m, 2H), 6.26 (dd, J: 16.8, 1.9 Hz, 1H), 5.80 (dd, J: 10.6, 1.9 Hz, 1H), 3.88 (s, 3H), 3.75—3.78 (m, 8H), 2.24 (s, 3H).MS (ESI, method A): m/z = 381.1 [M + H]+, tR=l.513 (min). HPLC: 95.9% (214nm), 98.7% (254nm).
Scheme 19 Pd2(()3dba,CSZC03 | NH2 ()2H023\©[:K dioxane/HZO 120°C \ | NH2 (\N / / O N Cl (\N N \ Example 30 2-(4-flu0r0methoxyphenyl)—6-(piperazinyl)nic0tinamide (3). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 -nitrophenyl)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as brown gum (0.264 g, 72%). MS (ESI): m/z = 331.1 [M + H. e 30 6-(4-acryloylpiperazinyl)—2-(4-flu0rometh0xyphenyl)nicotinamide The title compound was obtained using a procedure analogous to the procedure described in e 1 as White solid (45 mg, 15%). 1H NMR (400 MHz, CDgOD) 5 7.78 (d, J: 8.7 Hz, 1H), 7.41 (dd, J: 8.3, 2.0 Hz, 1H), 7.28—7.22 (m, 1H), 7.16—7.12(m,= 1H), 6.88—6.77 (m, 2H), 6.27 (dd, J: 16.8, 1.9 Hz, 1H), 5.80 (dd, J: 10.6, 1.9 Hz, 1H), 3.93 (s, 3H), 3.87—3.71 (m, 8H).
MS (ESI, method A): m/z = 385.1 [M + H]+, tR=1.527 (min). HPLC: 99.6% (214nm), 99.7% (254nm).
Scheme 20 0H ch03 DMF d(,dppf)C|2 KOAc \ DMF reflux + | 115°C B’0 1 2 3 4 o o \ NH \ NH Pd2(dba)3,C32003 | 2 | 2 / / dioxane/H20,120°C (\N N |\ EtsN,CHzC|2,0°C (\N N l\ o HNJ N/ 0 0 We] Nd N/ O (\N N/ 01 6 Example31 -br0m0phen0xypyridine (3). The title compound was ed using a procedure analogous to the procedure described in romophenoxy)benzonitrile (see Scheme 16) as yellow solid (3.85 g, 81%). 2-phenoxy—5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-Z-yl)pyridine (4). The title nd was obtained using a procedure ous to the procedure described in 4-(4-(4,4,5,5- tetramethyl-l,3,2-dioxaboroIanyl)phenoxy)benzonitrile (see Scheme 16) as yellow solid (0.536 g, 90%). 6'-phen0xy-6—(piperazinyl)-2,3'-bipyridine—3-carb0xamide (6). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 -nitrophenyl)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as brown gum (0.14 g, 55%). MS (ESI): m/z = 376.1 [M + H.
Example 3 1 6-(4-acryloylpiperazin-l-yl)-6'-phen0xy—2,3 '-bipyridine—3-carb0xamide (\N \ 0}" | N’ 0 / G The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (50 mg, 31%). 1H NMR (400 MHz, CDgOD) 5 8.43 (d, J: 2.4 Hz, 1H), 8.10 (dd, J: 8.6, 2.4 Hz, 1H), 7.80 (d, J: 8.7 Hz, 1H), 7.49—7.41 (m, 2H), 7.29—7.22 (m, 1H), 7.16 (d, J: 7.9 Hz, 2H), 6.97 (d, J: 8.6 Hz, 1H), 6.88—6.77 (m, 2H), 6.27 (dd, J: 16.8, 1.8 Hz, 1H), 5.80 (dd, J: 10.6, 1.8 Hz, 1H), 3.87—3.69 (m, 8H). MS (ESI, method A): m/Z = 430.1 [M + H]+, tR=1.620 (min). HPLC: 95.1% (214nm), 96.2% (254nm).
Scheme 21 F K2003 DMF f)C|2 KOAc Pd2(dba)s.052003 115 DMF reflux dioxane/HZO, 120 °C + \N DC Br {NJ/03m11'1 0% 11 4 HO N CI | NH2 (\N N/ Et3N CHZCIZ 0°C N/ 0 To \ \ 6 Example 32 2-(4-br0mophen0xy)pyridine (3). The title compound was obtained using a procedure analogous to the procedure described in 4-(4-bromophenoxy)benzonitrile (see Scheme 16) as yellow solid (2.83 g, 57%). MS (ESI): m/z = 250.1 [M + H]+. 2-(4-(4,4,5,5-tetramethyl-1,3,2-dioxab0r0]anyl)phenoxy)pyridine (4). The title compound was obtained using a procedure analogous to the procedure described in 4,4,5,5- tetramethyl-1,3,2-dioxaborolanyl)phenoxy)benzonitrile (see Scheme 16) as brown solid (0.882 g, 74%). MS (ESI): m/z = 298.1 [M + Hr. 6-(piperazinyl)(4-(pyridin-Z-yloxy)phenyl)nic0tinamide (6). The title compound was obtained using a procedure analogous to the ure described in 2-(4- (hydroxy(phenyl)methyl)phenyl)(piperidin—4-yl)nicotinamide (see Scheme 48) as brown gum (0.198 g, 53%). MS (ESI): m/z = 376.0 [M + Hr.
Example 32 6-(4-acryloylpiperazinyl)—2-(4-(pyridinyloxy)phenyl)nic0tinamide |\ NH2 N N The title compound was obtained using a procedure analogous to the procedure bed in Example 1 as white solid (41 mg, 18%). 1H NMR (400 MHz, CDgOD) 5 8.18 (dd, J: 5.0, 1.4 Hz, 1H), 7.86 (ddd, J: 8.4, 7.3, 2.0 Hz, 1H), 7.82—7.74 (m, 3H), 7.20—7.14(m, 3H), 7.01 (d, J: 8.3 Hz, 1H), 6.88—6.79 (m, 2H), 6.27 (dd, J: 16.8, 1.9 Hz, 1H), 5.80 (dd, J: 10.6, 1.9 Hz, 1H), 3.86—3.71 (m, 8H). MS (ESI, method A): m/z = 430.1 [M + H]+, tR=1.560 (min). HPLC: 100% (214nm), 100% (254nm).
Scheme 22 O O Pd2(dba)3, C52C03 \ NH, (HO)2B dioxaneiH20,120°C \ | NH2 ET3NVCH2C'2yOOC | + —> —> (\N / / N CI CI (\N N m" HNd HNd 0 1 2 3 |\ NH2 (\N N/ Oj/N\2 CI Example33 2-(4-chlor0phenyl)(piperazinyl)nic0tinamide (3). The title compound was obtained using a procedure analogous to the procedure described in 2—(4-(hydroxy(phenyl)methyl)phenyl)—6- (piperidinyl)nicotinamide (see Scheme 48) as brown gum (0.23 g, 72%).
Example 33 6-(4-acryloylpiperazinyl)(4-chlorophenyl)nic0tinamide Oj/Q / N N The title compound was obtained using a ure ous to the procedure described in e 1 as white solid (50 mg, 18%). 1H NMR (400 MHZ, CDgOD) 5 7.78 (d, J: 8.7 Hz, 1H), 7.72—7.64 (m, 2H), 7.45—7.39 (m, 2H), 6.88—6.76 (m, 2H), 6.27 (dd, J: 16.8, 1.9 Hz, 1H), .80 (dd, J: 10.6, 1.9 Hz, 1H), 3.87—3.69 (m, 8H). MS (ESI, method A): m/z = 371.0 [M + H]+, tR=1.645 (min). HPLC: 95.1% (214nm), 95.3% (254nm).
Scheme 23 o o o o o 00<0E02 DMF-DMA O/\ N2H4.Et0H —> —> | —> PhO NaH, e PhOWOA PhO N/ 1 2 3 ,R‘ ,R‘ N’N N’N TsO-RBoc orF-RNO2 ' / ‘ / LiOH, EtOH HATU, NHs. DMF a)052003 DMF 0 OH Pho O Pho or b) 052003 NMP \\ O 4 s 7 ,R ,R ,R N/N a)HC|/EtOH DOM N—N . N—N or b) Zn NH4C| EtOH I / acwlovlchlorlde I / TEA, DCM PhD 0 "Hz PhO 0 "Hz C: 9 1o . N'w w'w (7 Q r: O O Boc 30¢ Boc l|30c (1 0:1 b c e @ 3:9 M 1'4 E: (5W NH (1,, 0 (SR) HNO f0 JO f (£0 a b c d e\ f 9 h Ethyl 3-0x0(4-phen0xyphenyl)propanoate (2). To a on of diethyl carbonate (14 g, 120 mmol) in toluene (100 mL) was added NaH (60%, 4.7 g, 12 mmol) at 0 °C, and the resulting solution was heated to 90 CC, then a solution of l-(4—phenoxyphenyl)ethanone 1 (10 g, 47 mmol) in toluene (50 mL) was added drop-wise over 30 min, and the solution was refluxed for 20 min.
After cooling to room temperature, AcOH/HZO (55 mL/275 mL) was added. Toluene was removed under vacuum, and the crude e was diluted with water (500 mL). Then the mixture was extracted with dichloromethane (4 X 800 mL). The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was d by silica gel column chromatography eluting with 9:1 PE/EA to get the title compound as brown oil (11.5 g, 79%). MS (ESI): m/Z = 285.0 [M + H] i Ethyl 3-(dimethylamino)—2-(4-phenoxybenzoyl)acrylate (3). A solution of ethyl 3—oxo-3—(4- phenoxyphenyl)propanoate 2 (9.6 g, 34 mmol) in DMA-DMF (100 mL) was stirred at 100 0C for 1 h. The resulting solution was concentrated in high vacuum to get the title compound (12 g, 100%) as brown thick oil. MS (ESI): m/Z = 340.1 [M + Hr.
Ethyl 3-(4-phenoxyphenyl)—1H-pyrazole—4—carboxylate (4). To a solution of ethyl 3- (dimethylamino)(4-phenoxybenzoyl)acrylate 3 (6 g, 17 mmol) in ethanol (30 mL) was added hydrazine hydrate (875 mg, 17 mmol), and the resulting on was heated to 85 °C and stirred for 3 h. The solvent was evaporated and the crude residue was purified by silica gel column chromatography eluting with 4:1 PE/EA to get the title compound as brown oil (4.3 g, 78%). MS (ESI): m/Z = 309.0 [M + H.
Ethyl(1-(tert-but0xycarb0nyl)pyrrolidinyl)(4-phen0xyphenyl)—lH-pyrazole—4- carboxylate (6c). C52C03 (1.08 g, 3.3 mmol) was added to a solution of ethyl 3-(4- phenoxyphenyl)-1H-pyrazolecarboxylate 4 (500 mg, 1.6 mmol) and tert-butyl 3— (tosyloxy)pyrrolidinecarboxylate 5c (1.1 g, 3.2 mmol) in DMF (20 mL) and the resulting mixture was stirred at 100 CC for 16 h. The reaction mixture was concentrated in high vacuum and the crude e was purified by silica gel column chromatography eluting with 20:1 DCM/EA to get the title compound (780 mg, 100%) as colorless thick oil. MS (ESI): m/z = 478.1 [M + Hr.
Similar procedures were used to prepare the following compounds: 6a, 6b, 6d, 6g, 6h. tert-butyl(4-(ethoxycarbonyl)(4-phenoxyphenyl)—1H-pyrazolyl)piperidine—1- carboxylate (6a) = 492.1 [M + H]+. , thick ess oil, 340 mg, 36%. MS (ESI): m/z tert-butyl(4-(ethoxycarbonyl)—3-(4-phenoxyphenyl)—1H-pyrazolyl)piperidine—1- carboxylate (6b), thick oil 557 mg, 71%. MS (ESI): m/z = 492.1 [M + H]+.
Ethyl(1-(tert-butoxycarbonyl)azetidinyl)—3-(4-phen0xyphenyl)—1H—pyrazole—4- ylate (6d), white solid, 1.7 g, 61%. MS (ESI): m/z = 464.1 [M + H]+. hyl(1-(tert-but0xycarb0nyl)pyrrolidinyl)—3-(4-phenoxyphenyl)—1H-pyrazole—4- carboxylate (6g), colorless oil, 1.5 g, 99%. MS (ESI): m/z = 478.1 [M + H]+.
(R)—ethyl(1-(tert-but0xycarb0nyl)pyrrolidinyl)(4—phenoxyphenyl)—1H-pyrazole—4- ylate (6h), yellow oil, 2 g, 100%. MS (ESI): m/z = 478.1 [M + H]+.
Ethyl 1-(4-nitrophenyl)(4-phenoxyphenyl)—1H-pyrazole—4-carboxylate (6e). To a solution of ethyl 3-(4-phenoxyphenyl)—1H—pyrazolecarboxylate 4 (300 mg, 1.0 mmol) and 1-fluoro nitrobenzene 5e (140 mg, 1.0 mmol) in 1-methylpyrrolidone (8 mL) was added cesium carbonate (950 mg, 3.0 mmol), and the resulting solution was heated to 130 oC and stirred for 3 h. After cooling to ambient temperature, the solvent was evaporated and the crude residue was purified by silica gel column chromatography eluting with 4:1 PE/EA to get the title compound as a yellow solid (290 mg, 68%). MS (ESI): m/Z = 430.1 [M + H]+.
Similar procedures were used to prepare compound of 6f: ethyl 1-(3-nitr0phenyl)—3-(4-phenoxyphenyl)—1H-pyrazole—4-carboxylate (61), yellow solid , 290 mg, 40%. MS (ESI): m/Z = 430.1 [M + H]+. 1-(1-(tert-but0xycarb0nyl)pyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazole—4—carb0xylic acid (7c). To a solution of ethyl 1-(1—(tert—butoxycarbonyl)pyrrolidin—3-yl)—3—(4- phenoxyphenyl)—1H- lecarboxylate 6c (390 mg, 0.8 mmol) in EtOH (8 mL) was added 1N LiOH/HZO (4 mL). the reaction mixture was stirred at 75 °C for 1 h. Then to the solution was added 1N HCl/HgO to pH = 4—5, trated to give title compound (690 mg, crude) as white solid contented LiCl salt. MS (ESI): m/z = 450.1 [M + H]+.
Similar procedures were used to prepare the following compounds: 7a, 7b, 7d, 7e, 7f, 7g, 7h. tert—butoxycarb0nyl)piperidinyl)—3-(4-phen0xyphenyl)—lH—pyrazolecarb0xylic acid (7a), white solid, 310 mg, 100%. MS (ESI): m/z = 464.1 [M + H]+. 1-(1-(tert—butoxycarb0nyl)piperidinyl)—3-(4-phen0xyphenyl)—1H-pyrazolecarb0xylic acid (7b), yellow solid, 525 mg, 99%. MS (ESI): m/z = 464.1 [M + H]+. 1-(1-(tert—butoxycarbonyl)azetidinyl)—3-(4-phenoxyphenyl)—1H-pyrazole—4-carb0xylic acid (7d), yellow solid, 300 mg, 80%. MS (ESI): m/Z = 436.1 [M + Hr. 1-(4-nitr0phenyl)—3-(4-phen0xyphenyl)—1H-pyrazole—4-carb0xylic acid (7e), white solid, 280 mg, 99%. MS (ESI): m/z = 402.1 [M + Hr. itr0phenyl)—3-(4-phe0nxyphenyl)—1H-pyrazole—4-carb0xylic acid (71), yellow oil, 260 mg, 100%. MS (ESI): m/Z = 402.1 [M + H]+.
(S)(1-(tert—butoxycarbonyl)pyrrolidinyl)(4-phen0xyphenyl)—lH-pyrazole carboxylic acid (7g) = 450.1 [M + H]+. , yellow foam, 1.2 g, 99%. MS (ESI): m/z (1-(tert—butoxycarbonyl)pyrrolidinyl)—3-(4-phen0xyphenyl)—1H-pyrazole carboxylic acid (7h), yellow oil, 1.2 g, 99%. MS (ESI): m/z = 450.1 [M + H]+.
Tert-butyl(4-carbamoyl(4-phenoxyphenyl)—lH-pyrazolyl)pyrrolidine-l-carboxylate (80). To a solution of 1-(1-(tert-butoxycarbonyl)pyrrolidinyl)—3-(4-phenoxyphenyl)-1H- pyrazole carboxylic acid 7c (630 mg, crude, 0.67 mmol) in DMF (20 mL) was added HATU (510 mg, 1.33 mmol) and stirred at rt for 30 minutes, the resulting on was bubbled with NH3 gas for 10 minutes. After ng at rt for 1 h, the on was concentrated in high vacuum and the crude residue was diluted with DCM (50 mL), washed with water (3 X 20 mL), dried over , concentrated to give a residue which was purified by silica gel column chromatography eluting with 100:1 DCM/MeOH to get the title compound (120 mg, 33%) as colorless foam. MS (ESI): m/z = 471.2 [M + Na]+.
Similar procedures were used to prepare the following compounds: 8a, 8b, 8d, 8e, 8f, 8g, 8h tert-butyl(4-carbamoyl(4-phen0xyphenyl)-1H-pyrazol-l-yl)piperidine-l-carboxylate (83), colorless oil, 310 mg, 100%. MS (ESI): m/z = 463.1 [M + H]+. tert-butyl(4-carbamoyl(4-phen0xyphenyl)-1H-pyrazol-l-yl)piperidine-l-carboxylate (8b), yellow solid, 320 mg, 63%. MS (ESI): m/Z = 463.1 [M + Hr. tert-butyl(4-carbam0yl(4-phenoxyphenyl)—1H-pyrazolyl)azetidine-l-carboxylate (8d), yellow solid, 300 mg, 99%. MS (ESI): m/Z = 435.1 [M + H]+. 1-(4-nitrophenyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarb0xamide (8e), yellow solid, 230 mg, 99%. MS (ESI): m/Z = 401.1 [M + Hr. 1-(3-nitrophenyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarb0xamide (81), yellow solid, 260 mg, 100%. MS (ESI): m/Z = 401.0 [M + H]+. rt—butyl(4-carbam0yl(4-phenoxyphenyl)—1H-pyrazolyl)pyrrolidine carboxylate (8g), white solid, 1.2 g, 100%. MS (ESI): m/z = 449.0 [M + H]+.
(R)-tert-butyl(4-carbamoyl(4-phen0xyphenyl)—lH-pyrazolyl)pyrrolidine-l- carboxylate (8h), white solid, 960 mg, 80%. MS (ESI): m/z = 449.0 [M + H]+. 3-(4-phenoxyphenyl)(pyrrolidinyl)-1H-pyrazolecarboxamide. To a solution of tert- butyl 3 -(4-carbamoyl(4-phenoxyphenyl)-1H-pyrazolyl)pyrrolidinecarboxylate 80 (120 mg, 0.27 mmol) in DCM (10 mL) was added 6 N HCl/EtOH (5 mL, 30 mmol), stirred at rt for 1h, the resulting solution was concentrated in high vacuum and the crude residue was purified by Prep-HPLC to give title compound (86 mg, 91%) as white solid. 1H NMR (400 MHZ, CDgOD): 5 8.20 (s, 1H), 7.75 (d, J: 8.6 Hz, 2H), 7.43—7.36 (m, 2H), 7.20—7.13 (m, 1H), .00 (m, 4H), .32—5.27 (m, 1H), 3.87—3.72 (m, 3H), 3.59—3.50 (m, 1H), 2.67—2.56 (m, 1H), 2.52—2.43 (m, 1H). MS (ESI, Method A): m/z = 349.1 [M + H]+, tR= 1.242 min. HPLC: 99.3% (214nm), 99.5% (254nm).
Similar procedures were used to prepare the following compounds: 9a, 9b, 9d, 9g, 9e, and 9f. 3-(4-phen0xyphenyl)—1-(piperidinyl)—1H—pyrazolecarboxamide (921), white solid, 240 mg, 99%. 1H NMR (400 MHz, CD30D): 8 8.23 (s, 1H), 7.73 (d, J: 8.6 Hz, 2H), 7.48—7.31 (m, 2H), 7.21—7.12 (m, 1H), .97 (m, 4H), 4.76—4.65 (m, 1H), 3.74—3.57 (m, 2H), 3.43—3.36 (m, 1H), 3.28—3.15 (m, 1H), 2.39—2.02 (m, 3H), 2.01—1.84 (m, 1H). MS (ESI, Method A): m/Z = 363.2 [M + H]+, tR= 1.255 min. HPLC: 100% (214nm), 100% (254nm). 3-(4-phen0xyphenyl)—1-(piperidin-4—yl)—1H—pyrazolecarboxamide (9b), light yellow solid, 3.1 mg, 40%. 1H NMR (300 MHz, CD30D)I 5 8.18 (s, 1H), 7.67 (d, J: 8.7 Hz, 2H), 7.43—7.31 (m, 2H), 7.19—7.09 (m, 1H), 7.07—6.95 (m, 4H), .55 (m, 1H), 3.64—3.54 (m, 2H), 3.29— 3.17(m, 2H), 2.42—2.23 (m, 4H). MS (ESI, Method A): m/z = 363.0 [M + H]+, tR= 1.288 min.
HPLC: 100% (214nm), 100% (254nm). 1-(azetidinyl)—3-(4—phenoxyphenyl)—1H-pyrazolecarboxamide (Example 14) (9d), yellow solid, 300 mg, 100%. 1H NMR (300 MHz, CD30D)I 8 8.20—8.14 (m, 1H), 7.82—7.66 (m, 2H), 7.44—7.30 (m, 2H), 7.18—7.11(m, 1H), 7.07—6.96 (m, 4H), 5.52—5.40 (m, 1H), 4.64—4.55 (m, 4H). MS (ESI, Method A): m/Z = 335.1 [M + H]+, tR= l.235-l.255 min. HPLC: 100% ), 100% (254nm).
(S)—3-(4-phenoxyphenyl)—1-(pyrrolidinyl)—1H-pyrazole—4-carboxamide (9g), white solid, 90 mg, 83%. 1H NMR (400 MHz, CD3OD)I 6 8.25 (s, 1H), .68 (m, 2H), 7.45—7.33 (m, 2H), 7.20—7.11 (m, 1H), 7.11—6.98 (m, 4H), 5.38—5.25 (m, 1H), 3.88—3.69 (m, 3H), 3.62—3.49 (m, 1H), 2.68—2.54 (m, 1H), 2.54—2.40 (m, 1H). MS (ESI, Method A): m/z = 349.1 [M + H]+, tR= 1.219 min. HPLC: 100% (214nm), 100% ). (4-phen0xyphenyl)—1-(pyrrolidinyl)-1H-pyrazole—4-carboxamide (9h), yellow oil, 100mg, 100%. MS (ESI, Method A): m/z = 349.1 [M + H]+. 1-(4-amin0phenyl)—3-(4-phen0xyphenyl)—1H-pyrazolecarboxamide (9e). To a solution of 1-(4—nitrophenyl)-3—(4-phenoxyphenyl)-1H-pyrazolecarboxamide 8e (200 mg, 0.50 mmol) in ethanol (8 mL) was added saturated aqueous NH4Cl (4 mL), followed by the addition of zinc powder (260 mg, 4.0 mmol) in n over 5 min, then the resulting solution was stirred for 10 h at ambient temperature. The mixture was filtered and the filtrate was diluted with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound as a yellow solid (150 mg, 81%). 1H NMR (400 MHz, cogoD): 8 8.47 (s, 1H), 7.78 (d, J: 8.7 Hz, 2H), 7.51 (d, J: 8.7 Hz, 2H), 7.44—7.36 (m, 2H), 7.16 (s, 1H), 7.19—7.13 (m, 4H), 6.83 (d, J: 8.7 Hz, 2H). MS (ESI, Method A): m/z = 371.1 [M + H]+, tR= 1.468 min. HPLC: 97.8% (214nm), 98.0% (254nm).
Similar ures were used to prepare compound 8f. minophenyl)—3-(4-phenoxyphenyl)—1H-pyrazole—4-carb0xamide (81), white solid, 105 mg, 47%. 1H NMR (400 MHz, CD30D)C 8 8.60 (s, 1H), 7.81 (d, J: 8.6 Hz, 2H), 7.44—7.36 (m, 2H), 7.28—7.03 (m, 8H), 6.75 (d, J: 7.9 Hz, 1H). MS (ESI, Method A): m/z = 371.1 [M + H]+, tR= 1.507 min. HPLC: 96.9% (214nm), 98.2% ). 1-(1-acryloylpyrrolidinyl)—3-(4-phen0xyphenyl)-1H-pyrazolecarboxamide (Example 13) (10c). To a solution of 3-(4-phenoxyphenyl)—1-(pyrrolidinyl)—1H-pyrazole-4—carboxamide 9c (43 mg, 0.12 mmol) and TEA (50 mg, 0.5 mmol) in DCM (10 mL) was added acryloyl chloride (22 mg, 0.24 mmol) and stirred at 75 °C for 1 h, the resulting was concentrated and purified by Prep-TLC eluting with 10:1 DCM/MeOH to get the title compound (17 mg, 35%) as white solid. 1H NMR (400 MHz, CD3OD)I 8 8.18 (d, J= 8.7 Hz, 1H), 7.70 (d, J= 8.6 Hz, 2H), 7.43—7.34 (m, 2H), 7.18—7.08 (m, 1H), 7.03 (dd, J: 15.5, 8.3 Hz, 4H), .58 (m, 1H), 6.32 (dd, J: 15.9, 4.0 Hz, 1H), 5.83—5.73 (m, 1H), 5.18—5.04 (m, 1H), 4.21—3.65 (m, 4H), 2.63—2.45 (m, 2H). MS (ESI, Method A): m/Z = 403.1 [M + H]+, tR= 1.414 min. HPLC: 95.3% (214nm), 95.3% (254nm).
Similar procedures were used to e the following compounds: 10a, 10b, 10d, 10e, 10f, 10g and 10h. 1-(1-acryloylpiperidiny])(4-phenoxyphenyl)—1H-pyrazole-4—carb0xamide (Example 11) (1021). White solid, 39 mg, 23%. 1H NMR (400 MHz, CDgOD) 8 8.20 (d, J: 5.4 Hz, 1H), 7.71 (d, J: 8.3 Hz, 2H), 7.44—7.35 (m,2H),7.19—7.12(m, 1H), 7.04 (dd, J: 12.3, 8.4 Hz, 4H), 6.89—6.73 (m, 1H), 63—612 (m, 1H), 5.76 (dd, J: 25.5, 10.7 Hz, 1H), 4.71 (d, J: 11.5 Hz, 0.5 H), 4.45—4.17 (m, 2H), 4.08 (d, J: 14.3 Hz, 0.5 H), .73 (m, 0.5 H), 3.45—3.34 (m, 1H), 3.27—3.14 (m, 0.5 H), 2.39—2.18 (m, 2H), 2.06—1.92 (m, 1H), 1.71—1.61 (m, 1H). MS (ESI, Method A): m/z = 417.2 [M + H]+, tR= 1.468 min. HPLC: 97.7% (214nm), 98.7% (254nm). 1-(1-acryloylpiperidinyl)—3-(4-phen0xyphenyl)—1H-pyrazole-4—carb0xamide (Example 12) (10b). White solid, 4.7 mg, 20.8%. 1H NMR (400 MHz, CD30D) 8 8.19 (s, 1H), 7.69 (d, J: 8.5 Hz, 2H), 7.45—7.33 (m, 2H), 7.25—7.12 (m, 1H), 7.04 (dd, J: 12.2, 8.5 Hz, 4H), 6.84 (dd, J: 16.8, 10.7 Hz, 1H), 6.24 (dd,J= 16.8, 1.5 Hz, 1H), 5.79 (dd, J: 10.7, 1.5 Hz, 1H), 4.77—4.64 (m, 1H), 4.61—4.46 (m, 1H), 4.35—4.23 (m, 1H), 3.38 (d, J: 13.0 Hz, 1H), 3.03—2.92 (m, 1H), 2.34—2.17(m, 2H), 2.12—1.96 (m, 2H). MS (ESI, Method A): m/z = 417.1 [M + H]+, tR=1.421 min. HPLC: 97.5% (214nm), 97.8% ). 1-(1-acryloylazetidinyl)—3-(4-phen0xyphenyl)—1H-pyrazole—4-carb0xamide (10d). White solid, 19 mg, 18%. 1H NMR (300 MHz, CD30D) 5 8.21 (s, 1H), 7.71 (d, J= 9.1 Hz, 2H), 7.42— 7.31 (m, 2H), 7.20—7.09 (m, 1H), 7.06—6.94 (m, 4H), 6.46—6.22 (m, 2H), 5.78 (dd, J: 9.9, 2.3 Hz, 1H), 5.33 (td, J: 8.0, 4.0 Hz, 1H), 4.83—4.65 (m, 2H), 4.62—4.41 (m, 2H). MS (ESI, Method A): m/Z = 389.1 [M + H]+, tR=1.388-1.401 min. HPLC: 95.1% ), 96.1% (254nm). 1-(4-acrylamid0phenyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarboxamide (Example 16) (10e). White solid, 52 mg, 46%. 1H NMR (400 MHz, CD3OD) 8 8.67 (s, 1H), 7.89—7.78 (m, 6H), 7.44—7.36 (m, 2H), 7.20—7.12 (m, 1H), .02 (m, 4H), .37 (m, 2H), 5.82 (dd, J: 9.4, 2.4 Hz, 1H). MS (ESI, Method A): m/z = 425.2 [M + H]+, tR=1.521 min. HPLC: 97.3% (214nm), 97.0% (254nm). 1-(3-acrylamid0phenyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarb0xamide (Example 18) (101). White solid, 24 mg, 24%. 1H NMR (400 MHz, CD30D) 8 8.70 (s, 1H), 8.35 (s, 1H), 7.84 (d, J: 8.7 Hz, 2H), 7.60 (d, J: 8.1 Hz, 2H), .48 (m, 1H), 7.44—7.36 (m, 2H), 7.11—7.03 (m, 1H), 7.20—7.13(m, 4H), 6.53—6.38(m, 2H), 5.83 (dd, J: 9.2, 2.6 Hz, 1H). MS (ESI, Method A): m/z = 425.1 [M + H]+, tR=1.547 (min). HPLC: 97.6% (214nm), 98.6% (254nm).
(S)(1-acryloylpyrrolidinyl)—3-(4-phenoxyphenyl)—1H-pyrazolecarboxamide (Example 20) (10g). Gray solid, 120 mg, 27%. 1H NMR (400 MHz, CD30D) : 8 8.21 (d, J = 6.6 Hz, 1H), 7.73—7.64 (m, 2H), 7.43—7.34 (m, 2H), 7.20—7.10 (m, 1H), 7.10—6.96 (m, 4H), 6.75— 6.55 (m, 1H), 6.32 (ddd, J: 16.8, 4.8, 1.9 Hz, 1H), 5.86—5.72 (m, 1H), 5.21—5.03 (m, 1H), 4.22— 4.07 (m, 1H), 4.05—3.67 (m, 3H), 2.64—2.43 (m, 2H). MS (ESI, Method A): m/z = 403.2 [M + H]+, tR= 1.421 min. HPLC: 99.0% (214nm), 99.0% (254nm).
(R)(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)—1H-pyrazole—4-carboxamide (Example 19) (10h). White solid, 20 mg, 6%. 1H NMR (300 MHz, CDgOD) : 8 8.16 (d, J: 5.9 Hz, 1H), 7.67 (d, J: 8.7 Hz, 2H), 7.42—7.30 (m, 2H), 7.17—6.92 (m, 5H), 6.71—6.56 (m, 1H), 6.34—6.25 (m, 1H), .72 (m, 1H), 5.18—5.02 (m, 1H), 4.20—4.05 (m, 1H), 4.02—3.65 (m, 3H), 2.62—2.45 (m, 2H). MS (ESI, Method A): m/z = 403.1 [M + H]+, tR= 1409 min. HPLC: 95.0% (214nm), 97.0% (254nm).
Scheme 24 0 I HomN\ 1 1 N‘N N—N M HATU, TEA, DMF M PhD 0 ""2 PhD o ""2 Example 34 Example 34 1-(1-(4-(dimethylamin0)but—2-enoyl)pyrrolidinyl)(4—phenoxyphenyl)—1H-pyrazole—4- carboxamide 0 | PhDMo ""2 To a solution of 4-(dimethylamino)butenoic acid 11 (22 mg, 0.17 mmol) and HATU (98 mg, 0.26 mmol) in DMF (10 mL) was added 3-(4-phenoxyphenyl)—1-(pyrrolidinyl)-lH-pyrazole- 4-carboxamide 90 (Scheme 23) (60 mg, 0.17 mmol) and DIPEA (22 mg, 0.5 mmol). The reaction mixture was stirred at rt for 1 h. The solution was concentrated in high vacuum and the residue was diluted in DCM (10 mL), washed with water (3 X 10 mL), dried over NazSO4, concentrated to give a residue which was purified by LC eluting with 10:1 OH to get the title compound (26 mg, 37%) as brown solid. 1H NMR (300 MHz, CD3OD)I 8 8.20 (d, J: 9.0 Hz, 1H), 7.73—7.56 (m, 2H), 7.41—7.27 (m, 2H), 7.11 (t, J: 7.4 Hz, 1H), 7.05—6.91 (m, 4H), .66 (m, 2H), 5.22—5.02 (m, 1H), 4.24—4.06 (m, 1H), 4.05—3.76 (m, 4.5H), 3.76—3.61 (m, 0.5H), 2.86 (d, J: 6.1 Hz, 6H), 2.66—2.39 (m, 2H). MS (ESI, Method A): m/z = 459.8 [M + H]+, tR= 1.244 min. HPLC: 97.5% (214nm), 98.2% (254nm).
Scheme 25 OH )2, Et3N Pd(dppf)Cl2, KOAc 4A MS 0 DMF, reflux cH Cl rt —> —B(L:)12_> go i‘B-B\0% O 0 Br O )‘—/\ 1 2 4 \ \ Pd2(dba)3 052003 | NH2 / / dioxane/HZO 120°C IN HNQ Et3N CHZCIZ 0°C (\N N /\n/C' Example 35 4-(4,4,5,5-tetramethyl-1,3,2-di0xab0rolany1)phenol (2). The title compound was obtained using a procedure analogous to the procedure described in 4—(4-(4,4,5,5-tetramethyl-1,3,2- dioxaboro1any1)phenoxy)benzonitri1e (see Scheme 16) as white solid (10.2 g, 80%). 2-(4-(3-flu0r0phenoxy)phenyl)-4,4,5,5-tetramethyl—1,3,2-di0xab0rolane (4). The mixture of 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl)phenol 2 (1.0g, 4.5mmol), 3-fluorophenylboronic acid 3 (0.70 g, 5.0 mmol), Cu(OAc)2 (1.0 g, 5.0 mmol), Eth (2.75 g, 27.3 mmol), 4A lar sieves (1 g) and CHzClz (15 mL) was d at rt for 16 h. Then the mixture was concentrated in vacuo and the e was purified by silica gel chromatography eluting with 20:1 petroleum ether/EtOAc to afford the title compound (0.45 g, 32%) as white solid. 2-(4-(3-fluor0phen0xy)phenyl)—6-(piperazinyl)nicotinamide (6). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 -nitropheny1)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as white solid (0.15 g, 41%). MS (ESI): m/z = 393.1 [M + H.
Example 35 6-(4-acryloylpiperaziny])—2-(4-(3-flu0rophenoxy)phenyl)nicotinamide (\N N/ o Nd j 0 The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (50 mg, 27%). 1H NMR (400 MHz, CDgOD) 5 7.79 (d, J = 8.7 Hz, 1H), 7.77—7.70 (m, 2H), 7.42—7.33 (m, 1H), 7.13—7.06 (m, 2H), 6.93—6.75 (m, 5H), 6.27 (dd, J: 16.8, 1.9 Hz, 1H), 5.80 (dd, J: 10.6, 1.9 Hz, 1H), 3.84 —3.72 (m, 8H). MS (ESI, method A): m/z = 447.1 [M + H]+, tR=1.746 (min). HPLC: 95.9% (214nm), 96.4% (254nm).
Scheme 26 OH )2, Et3N B(OH)2 4A Ms Pd(dppf)C[2 KOAc Pd2(dba)3 052003 e/HZO 120°C + 1 o Br \ClIN 1 2 CH2C12,rt©/E|3r—>]©\DMF reflux 5 HNJ0N O O \ \ NH2 (\N N/ Et3N,CH2CI2,o°C (\N N/ HN\/‘ ON F6 2,Cl j FE) Example 36 4-brom0flu0ro-l-phenoxybenzene (3). The title compound was obtained using a procedure analogous to the procedure described in 2-(4-(3-fluorophenoxy)phenyl)—4,4,5,5-tetramethyl— 1,3,2-dioxaborolane (see Scheme 25) as yellow oil (1.5 g, 21%). MS (ESI): m/z = 267.1 [M + 2-(3-flu0r0phenoxyphenyl)—4,4,5,5-tetramethyl—1,3,2-di0xaborolane (5). The title compound was obtained using a procedure analogous to the procedure described in 4-(4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolanyl)phenoxy)benzonitrile (see Scheme 16) as yellow oil (1.04 g, 73%). 2-(3-fluor0phen0xyphenyl)—6-(piperazinyl)nic0tinamide (7). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 -nitrophenyl)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as brown gum (0.194 g, 49%). MS (ESI): m/z = 393.1 [M + H.
Example 36 6-(4-acryloylpiperaziny])—2-(3-flu0r0phenoxyphenyl)nic0tinamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (38 mg, 17%). 1H NMR (400 MHZ, 6) 8 (m, 2H), 7.59 (d, J = 11.4 Hz, 1H), 7.48 (d, J: 8.2 Hz, 1H), 7.42 (t, J: 7.7 Hz, 2H), 7.35—7.28 (m, 1H), 7.24—7.15 (m, 2H), 7.04 (d, J: 7.9 Hz, 2H), 6.95—6.81 (m, 2H), 6.16 (d, J: 16.6 Hz, 1H), 5.73 (d, J: 10.2 Hz, 1H), 3.79—3.50 (m, 8H). MS (ESI, method A): m/z = 447.1 [M + H]+, tR=1.753 (min).
HPLC: 98.6% (214nm), 98.4% (254nm).
Scheme 27 F Cuu(OAc)2, £th 4A MS Pd2(dba)s 082C03 N/ dioxane/HZO 120°C + CH2C|2,rt 474 B(0H)2 <10; Ad\IN 1 2 3 5 Eth, CHZCIZ, 0 00 e 37 2-(4-(4-flu0r0phen0xy)phenyl)-4,4,5,5-tetramethyl—1,3,2-di0xab0rolane (3). The title compound was obtained using a ure analogous to the procedure described in 2—(4-(3— fluorophenoxy)phenyl)-4,4,5,5-tetramethy1—1,3,2-dioxaborolane (see Scheme 25) as White solid (0.45 g, 32%). 2-(4-(4-fluorophenoxy)phenyl)—6-(piperazinyl)nic0tinamide (5). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 phenyl)(4- phenoxypheny1)nicotinamide (see Scheme 1) as brown gum (0.18 g, 41%).
Example 37 6-(4-acryloylpiperazinyl)(4-(4-flu0rophenoxy)phenyl)nic0tinamide \ NH2 ‘/\N N/ The title compound was obtained using a procedure analogous to the procedure described in Example 1 as white solid (80 mg, 44%). 1H NMR (400 MHZ, CD30D) 5 7.77 (d, J = 8.7 Hz, 1H), 7.69 (d, J: 8.7 Hz, 2H), .05 (m, 4H), 7.00 (d, J: 8.7 Hz, 2H), 6.87—6.76 (m, 2H), 6.26 (dd, J: 16.8, 1.8 Hz, 1H), 5.80 (dd, J: 10.6, 1.8 Hz, 1H), 3.87—3.71 (m, 8H). MS (ESI, method A): m/z = 447.0 [M + H]+, 39 (min). HPLC: 99.8% (214nm), 100% (254nm).
Scheme 28 052003, DMF Fe NH4C| CHZIZ, amyl nitrite OM32 + CE: —’E:E: 120 °c ONOZ EtOH 70°C a:ONHZ CHgCN 75°C F OH 1 2 Pd2(dban 052co Pd(dppf)C|2 KOAc | 0 o / . 03 DMF reflux dioxane/HZO, 120 C ‘/\N N Wit-1m".OB‘o O HNd F I o HN\) Pg Et3N, CHZCIZ, 0 °C KN Example 38 l-flu0r0(4-nitr0phen0xy)benzene (3). The mixture of 1-fluoronitrobenzene 1 (10.0 g, 70.8 mmol), 2-fluorophenol 2 (9.54 g, 85.0 mmol), CS2C03 (34.6 g, 106 mrnol) and DMF (80 mL) was heated to 120 °C and stirred for 2 h. After being cooled to rt, the mixture was diluted with water (150 mL) and extracted with EtOAc (200 mL X 2). The ed organic phase was washed with brine (100 mL X 3), dried over Na2S04 and filtered. The filtrate was concentrated in vacuo. The residue was recrystallized from MeOH (20 mL X 2) to afford the title compound as light yellow solid (14.6 g, 88%). uorophenoxy)benzenamine (4). To the mixture of 1-fluoro-2—(4-nitrophenoxy)benzene 3 (16.2 g, 69.3 mmol), saturated aqueous NH4Cl solution (30 mL) and EtOH (150 mL) was added iron powder (19.4 g, 347 mmol) slowly and then the resulting mixture was heated to 70 CC for 3.5 h. After being cooled to rt, the mixture was d. The filtrate was concentrated in vacuo.
The residue was dissolved in EtOAc (160 mL) and washed successively with water (100 mL X 2) and brine (100 mL X 2). The organic phase was dried over Na2S04, d and concentrated in vacuo to afford the title compound (13.6 g, 96%) as yellow solid. 1H NMR (400 MHz, DMSO- d6) 8 7.35—7.28 (m, 1H), 7.14—7.00 (m, 2H), 6.93—6.85 (m, 1H), 6.80—6.73 (m, 2H), .56 (m, 2H), 4.99 (brs, 2H). 1-flu0r0(4-iodophenoxy)benzene (5). The mixture of 4-(2—fluorophenoxy)benzenamine 4 (7.94 g, 39.1 mmol), CHZIZ (29.0 g, 136.8 mmol) and CH3CN (120 mL) was heated to 55 °C and then amyl nitrite (11.6 g, 97.7 mmol) was added. The resulting mixture was heated to 75 °C for 3.5 h. After being cooled to rt, the mixture was diluted with EtOAc and then washed successively with 10% NazSgOg and brine. The organic phase was dried over Na2S04, filtered, concentrated in vacuo and purified by silica gel chromatography eluting with petroleum ether to afford the title compound (4.12 g, 51%) as light yellow oil. 1H NMR (400 MHZ, DMSO) 8 7.78— 7.63 (m, 2H), 7.44—7.37 (m, 1H), 7.34—7.14 (m, 3H), 6.87—6.74 (m, 2H). 2-(4-(2-flu0r0phenoxy)phenyl)-4,4,5,5-tetramethyl—1,3,2-di0xab0rolane (7). The title compound was obtained using a procedure analogous to the procedure described in 4-(4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolanyl)phenoxy)benzonitrile (see Scheme 16) as yellow oil (0.40 g, 13%). 2-(4-(2-fluorophenoxy)phenyl)—6-(piperazinyl)nic0tinamide (9). The title compound was ed using a ure analogous to the procedure described in 6-(3 -nitrophenyl)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as white solid (0.16 g, 31%). MS (ESI): m/z = 393.1 [M + Hr.
Example 38 6-(4-acryloylpiperaziny])(4-(2-flu0rophenoxy)phenyl)nicotinamide O / N The title compound was ed using a procedure analogous to the procedure described in Example 1 as white solid (28 mg, 24%). 1H NMR (400 MHZ, CDgOD) 5 7.77 (d, J = 8.7 Hz, 1H), 7.71—7.68 (m, 2H), 7.34—7.13 (m, 4H), 6.99 (d, J: 8.7 Hz, 2H), 6.88—6.79 (m, 2H), 6.26 (dd, J: 16.8, 1.9 Hz, 1H), 5.80 (dd, J: 10.6, 1.9 Hz, 1H), 3.89—3.71 (m, 8H). MS (ESI, method A): m/z = 447.1 [M + H]+, tR=1.723 (min). HPLC: 98.6% (214nm), 98.8% (254nm).
Scheme 29 Cu(OAc)2, Et3N f)C|2 KOAc 4A MS Pd2(dba)3, 052003 DMF reflux ,1 dioxane/HZO, 120°C —>F CHZCIZ , , é <1 115 5 55 M 1:5 o . 511* |/\N N/ CI 1 2 4 I Et N CH3 2CI 1 21 0°C 6N N/ HN¢F 0qu O/NdFj o 11 <5 Example 39 3-fluor0(4,4,5,5-tetramethyl-1,3,2-di0xab0r01anyl)phenol (2). The title compound was obtained using a procedure analogous to the procedure described in 4-(4,4,5,5-tetramethy1—1,3,2- dioxaborolan-2—y1)phenol (see Scheme 25) as white solid (4.1 g, 66%). MS (ESI): m/z = 239.2 [M + Hr. 2-(2-fluoro-4—phenoxyphenyl)-4,4,5,5-tetramethyl-1,3,2-dioxab0rolane (4). The title nd was obtained using a procedure analogous to the procedure described in 2-(4-(3- fluorophenoxy)phenyl)-4,4,5,5-tetrarnethyl-1,3,2-dioxaborolane (see Scheme 25) as white solid ( g9 ) 2-(2-fluoro-4—phenoxyphenyl)—6-(piperazinyl)nicotinamide (6). The title compound was obtained using a procedure analogous to the procedure described in 6-(3 -nitropheny1)(4- phenoxyphenyl)nicotinamide (see Scheme 1) as brown solid (0.105 g, 21%). MS (ESI): m/z = 393.1 [M + Hr.
Example 39 6-(4-acryloylpiperazinyl)—2-(2-flu0r0phenoxyphenyl)nic0tinamide / ZIN Oj/O N F o / i) The title nd was obtained using a procedure analogous to the procedure bed in Example 1 as white solid (78 mg, 65%). 1H NMR(400 MHz, MeOD) 5 7.78 (d, J = 8.7 Hz, 1H), 7.59 (dd, J: 11.8, 2.0 Hz, 1H), 7.48 (d, J: 9.2 Hz, 1H), 7.44—7.33 (m, 2H), 7.17—7.06 (m, 2H), 7.05—6.99 (m, 2H), 6.90—6.79 (m, 2H), 6.27 (dd, J= 16.8, 1.8 Hz, 1H), 5.80 (dd, J: 10.6, 1.8 Hz, 1H), 3.86—3.70 (m, 8H). MS (ESI, method A): m/z = 447.0 [M + H]+, tR=1.770 (min).
HPLC: 96.5% (214nm), 96.9% (254nm).
Scheme 30 o O o s o CAOH )L CI 0«I/ O O NH2 Lawesson reagent 0%.
NH; "4 1NB°° —> NB°° THF/TEA/NH3‘HZO e "3°C EtOHl60°C COOH COOEt BocN BocN«3,8,3 LiOH 00—»THFH20 DMF/DIPEA CONH CONHZ 2 BOON0*? ofl’g W (DD—*09 \CONHz Example 40 tert-butyl 2-carbamoylpiperidine—l-carboxylate (2). To a solution of 1-(tert— butoxycarbonyl)piperidinecarboxylic acid 1 (5.0 g, 21.8 mmol) in dry THF (100 mL) was added TEA (3.30 g, 32.7 mmol), isobutyl carbonochloridate (3.27 g, 23.99 mmol) at 0°C and the resulting solution was d for 20 min. NH3-H20 was added and stirred at rt for 2 h. The mixture was diluted with ethyl acetate (30 mL), and washed with sat. aq. N32C03 (2 X 20 mL) and sat. aq. citric acid (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to give crude product (3.6 g, 72%) as white solid. MS (ESI): m/z = 173.0 [M — 551+. tert-butyl 2-carbamothioylpiperidinecarb0xylate (3). To a solution of tert—butyl 2- oylpiperidine-l-carboxylate 2 (2.0 g, 8.77 mmol) in dry toluene (20 mL) was added Lawesson’s reagent (2.13 g, 5.26 mmol) at N2 atmosphere. The resulting solution was stirred at 80°C for 16 h. The mixture was diluted with ethyl e (30 mL), and washed with saturated aqueous (sat. aq.) N32C03 (2 X 20 mL) and brine (2 X 20 mL). The c layer was dried over anhydrous sodium sulfate, filtered and concentrated to give crude product which was purified by silica gel column chromatography eluting with 4 : 1 PE/EA to get the title compound (330 mg, %) as white solid. MS (ESI): m/Z = 245.2 [M + H. ethyl(1-(tert-butoxycarbonyl)piperidinyl)(4-phen0xyphenyl)thiazole-5—carboxylate (5). A solution of tert-butyl 2-carbamothioylpiperidinecarboxylate 3 (60 mg, 0.239 mmol) and ethyl 2-bromooxo(4-phenoxyphenyl)propanoate 4 (250 mg, 0.7 mmol) in EtOH (10 mL) was stirred for 2 h at 60°C. The mixture was d with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium e, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with 40 : 1 PE/EA to get the title compound (120 mg, 34%) as colorless oil. MS (ESI): m/Z = 509.3[M + HT. 2-(1-(tert-butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)thiazolecarboxylic acid (6). A solution of ethyl 2-(1-(tert-butoxycarbonyl)piperidin-3 -yl)(4-phenoxyphenyl)thiazole- oxylate 5 (120 mg, 0.263 mmol ) in THF-HZO (1/1, 20 mL) and added LiOH (30 mg, 0.71 mmol) was stirred at rt for 13 h. The mixture was evaporated and diluted with water (5 mL), and the solution was acidified with 2 N hydrochloric acid to pH = 4. The residue was extracted with Ethyl Acetate (EA) (3 X 30 mL). Dried and concentrated to get crude compound (110 mg, 100%) as light red liquid. MS (ESI): m/Z = 481.0 [M + H]+. tert-butyl arbamoyl(4-phenoxyphenyl)thiazolyl)piperidine—l-carboxylate (7).
The mixture of 2-(1-(tert-butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)thiazole-S- ylic acid 6 (110 mg, 0.229 mmol), HATU (113 mg, 0.297 mmol), DIPEA (88 mg, 0.687 mmol) and dry DMF (10 mL) was bubbled with NH3 for 20 min, and the resulting solution was stirred for 3 h at ambient temperature. The mixture was diluted with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was d by Prep-TLC with 3 : 2 PE/EA to get the title compound (100 mg, 75%) as colorless oil. MS (ESI): m/z = 480.1 [M + H]+. 4-(4-phenoxyphenyl)(piperidinyl)thiazole—5-carb0xamide (8). To a solution of tert—butyl arbamoyl(4—phenoxyphenyl)thiazolyl)piperidine- l -carboxylate 7 (100 mg, 0.21 mmol) in DCM (5 mL) was added TFA (2.5 mL) at ambient temperature. The mixture was d for 3 h. The mixture was diluted with water (2 X 150 mL) and extracted with ethyl acetate (2 X 20 mL) and washed with brine (2 X 150 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to give the title compound (60 mg, 76%) as brown oil.
MS (ESI): m/Z = 380.0 [M + H.
Example 40 2-(1-acryloylpiperidinyl)(4-phen0xyphenyl)thiazolecarb0xamide .1] CONH2 M1606.
To a solution of 4-(4-phenoxyphenyl)(piperidinyl)thiazole-5—carboxamide 8 (60 mg, 0.155 mmol) in dry dichloromethane (10 mL) were added TEA (19 mg, 0.21 mmol) and acryloyl chloride 9 (32 mg, 0.316 mmol), and the resulting solution was d at rt for l h. Water (10 mL) was added to quench the reaction. The mixture was diluted with ethyl acetate (30 mL), washed with brine (2 X 20 mL). The organic layer was dried over NazSO4, filtered and concentrated to get the residue which was purified by Prep-TLC with 17:1 DCM/MeOH to get the title compound (33 mg, 50%) as a white solid. 1H NMR (400 MHz, DMSO) 8 7.76 (d, J = 8.5 Hz, 2H), 7.76 (brs, 1H),7.70 (brs, 1H), 7.44 (t, J= 8.0 Hz, 2H), 7.19 (t, J: 7.4 Hz, 1H), 7.08 (t, J = 8.6 Hz, 4H), 6.92—6.82 (m, 1H), 6.14—6.04 (m, 1H), 5.73—5.64 (m, 1H), 4.65—4.62 (m, 0.5H), .04 (m, 0.5H), 4.02—4.00 (m, 1H), 3.59—3.56 (m, 0.5H), .14 (m, 15H), 3.09—2.98 (m, 1H), 2.22—2.16 (m, 1H), .74 (m, 2H), 1.58—1.48 (m, 1H). MS (ESI, method A): m/z = 433.8 [M + H]+, tR=l.488 min. HPLC: 99.1% (214 nm), 99.2% (254 nm).
Scheme 31 Omoo 0 S \/ 0A CIOO/Y Lawessons reagent 0 0 NHZ NH2 BOON BOCN THF/TEAINH3-H20 toluene BocN 1 EtOH/60°C COO" COOEt (>4: —> BocN(>4: BocN 00 THEH20 00—,DMFIDIPEA CONH2 CONH2 CONH2 O’Qil —>BOCNO’<:\THF (Vim? BocN OHCQI tert-butlyl 3-carbam0ylpyrrolidine—l-carboxylate (2). To a solution of 1-(tert-Example 41 butoxycarbonyl)pyrrolidinecarboxylic acid 1 (5 g, 23.23 mmol) in dry THF (100 mL) was added TEA (4.69 6 mmol), isobutyl carbonochloridate (3.8 g, 27.87 mmol) at 0°C and the resulting solution was stirred for 20 min. NH3-H20 added and stirred at rt for 2 h. The mixture was diluted with ethyl acetate (30 mL), and washed with saturated (sat) Na2C03 (2 X 20 mL) and sat. citric acid (2 X 20 mL). The organic layer was dried over anhydrous sodium e, d and concentrated to give crude product (2.39 g, 49%) as yellow solid. MS (ESI): m/z = 159.0 [M + H. tert-butyl 3-carbamothioylpyrrolidine—l-carboxylate (3). To a solution of tert-butyl 3- carbamoylpyrrolidine—1-carboxylate 2 (0.76 g, 3.55 mmol) in dry toluene (20 mL) was added Lawesson’s regent (0.71 g, 1.77 mmol) at N; atmosphere . The resulting solution was stirred at 80°C for 16 h.The mixture was diluted with ethyl acetate (30 mL), and washed with sat. Na2C03 (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated to give crude product which was purified by silica gel column chromatography g with 6 : 1 PE/EA to get the title compound (230 mg, 28%) as brown oil.
MS (ESI): m/Z = 175.2 [M + Hr. ethyl 2-(1-(tert-butoxycarbonyl)pyrrolidinyl)-4—(4-phenoxyphenyl)thiazole—S-carboxylate (5). A solution of tert—butyl 3-carbamothioylpyrrolidinecarboxylate 3 (360 mg, 0.1 mmol) and ethyl 2—bromo—3-oxo—3-(4-phenoxyphenyl)propanoate 4 (230 mg, 0.1 mmol) in EtOH (10 mL) was d for 2 h at 60°C. The mixture was diluted with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, d and concentrated. The e was purified by silica gel column chromatography eluting with 30 : 1 PE/EA to get the title compound (100 mg, 20%) as colorless oil. MS (ESI): m/Z = 495.3[M + H. 2-(1-(tert-butoxycarbonyl)pyrrolidiny])(4-phenoxyphenyl)thiazole—S-carboxylic acid (6). A solution of ethyl 2-(1-(tert-butoxycarbonyl)pyrrolidin-3 -yl)(4-phenoxyphenyl)thiazole- - carboxylate 5 (100 mg, 0.2 mmol ) in THF-HZO (1/ 1, 20 mL) and added LiOH (84 mg, 2 mmol) was stirred at rt for 13 h. The e was evaporated and diluted with water (5 mL), and the solution was acidified with 2 N hydrochloric acid to pH = 4. The residue was extracted with EA (3 X 30 mL). Dried and evaporated to get crude compound (95 mg, 100%) as red oil. MS (ESI): m/Z = 411.0 [M + Hr. tert-butyl 3-(5-carbamoyl-4—(4-phenoxyphenyl)thiazol-Z-yl)pyrrolidine—l-carboxylate (7).
The mixture of 2-(1-(tert-butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)thiazole carboxylic acid 6 (950 mg, 0.2 mmol), HATU (116 mg, 0.3 mmol), DIPEA (103 mg, 0.8 mmol) and dry DMF (10 mL) was bubbled with NH3 for 20 min, and the ing solution was stirred for 3 h at ambient ature. The mixture was diluted with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, d and concentrated. The residue was purified by Prep-TLC with 2 : l PE/EA to get the title compound (70 mg, 75%) as colorless oil. MS (ESI): m/z = 466.1 [M + H]+. tert-butyl 3-(5-carbamoyl-4—(4-phenoxyphenyl)thiazol-Z-yl)pyrrolidine-l-carboxylate (8).
To a solution of tert-butyl 3-(5-carbamoyl(4-phenoxyphenyl)thiazolyl)pyrrolidine carboxylate 7 (70 mg, 0.15 mmol) in DCM (5 mL) was added TFA (2.5 mL) at ambient temperature. The mixture was stirred for 3 h. The mixture was diluted with water (2 X 150 mL) and extracted with ethyl acetate (2 X 20 mL) and washed with brine (2 X 150 mL). The organic layer was dried over ous sodium sulfate, filtered and concentrated to give the title nd (65 mg, 100%) as brown oil. MS (ESI): m/z = 366.1 [M + H]+.
Example 41 2-(1-acryloylpyrrolidiny])(4-phenoxyphenyl)thiazolecarboxamide f CONH2 O "in To a tert-butyl 3-(5-carbamoyl(4-phenoxyphenyl)thiazol—2-y1)pyrrolidine—1-carboxylate 8 (65 mg, 0.15 mmol) in dry dichloromethane (10 mL) were added TBA (50 mg, 0.45 mmol) and acryloyl chloride 9 (20.4 mg, 0.225 mmol), and the resulting solution was stirred at It for 1 h.
Water (10 mL) was added to quench the reaction. The e was diluted with ethyl acetate (30 mL), washed with brine (2 X 20 mL). The organic layer was dried over Na2S04, filtered and concentrated to get the residue which was d by Prep-TLC with 1 : 1 PE/EA to get the title compound (22 mg, 35%) as a white solid. 1H NMR (400 MHz, DMSO) 8 7.76 (d, J = 8.0 Hz, 2H), 7.76 (brs, 1H),7.70 (brs, 1H), 7.44 (t, J: 8.0 Hz, 2H), 7.19 (t, J: 7.4 Hz, 1H), 7.07 (t, J: 8.6 Hz, 4H), 6.65—6.59 (m, 1H), 6.19—6.15 (m, 1H), 5.72—5.68 (m, 1H), 4.11—4.07 (m, 0.5H), 3.99—3.95 (m, 0.5H), 3.91—3.78 (m, 2H), 3.70—3.62 (m, 1.5H), .47 (m, 0.5H), 2.44—2.36 (m, 1H), 2.28—2.25 (m, 0.5H), 2.18—2.13 (m, 0.5H). MS (ESI, method A): m/z = 419.8 [M + H]+, tR=1.429 min. HPLC: 97.2% (214 nm), 97.5% (254 nm).
Scheme 32 AJ°L J 0 0 0EtBr2, dioxane o°c~n 2h—>©\o NaH, toluene. reflux, 40 min 0 DIPEA, CH3CN rt o Q EtO AcONH4, AcOH, reflulx ° NaOH. MeOH, H20 Boc20, Nazco3 0 —> —> / —> Eto / Eto O N ° ° 0 /\g’/ 0 HATU, NH3, DMF HCI/EtOH HO / —>H2N —> / HZN / H2N / o / o /N DIPEA,DCM o /N o ’N "‘30:: N~Boc NH N 8 9 Example 42 ethyl 3-0x0(4-phenoxyphenyl)propanoate (2). To a mixture of diethyl carbonate (14 g, 120 mmol) and NaH (4.8 g, 120 mmol) in toluene (100 mL) was added 1-(4- phenoxyphenyl)ethanone 1 (10 g, 47 mmol) dropwise for 20 min. The resulting mixture was refluxed for 40 min. When it cooled to rt, which was quenched with AcOH/HZO (6 mL/30 mL), then diluted with EA (100 mL). The organic layer was separated, washed with brine, dried over sodium sulfate, filtered and trated. The residue was applied onto silica gel column g with 20:1 PE/EA to get the title compound (7.5 g, 56%) as yellow oil. MS (ESI): m/z = 285.1 [M + H]+. ethyl 2-br0m00x0(4-phen0xyphenyl)pr0panoate (3). To a mixture of ethyl 3-oxo(4- phenoxyphenyl)propanoate 2 (6.55 g, 23.1 mmol) in e (100 mL) was added Brz (3.69 g, 23.1 mmol) dropwise at 0 °C under N2. The resulting mixture was stirred at rt for 3 h. The volatile phase was removed under reduced re. This ed in crude title compound (9.5 g, overweight) as yellow oil. MS (ESI): m/Z = 3630/3650 [M + Hr. 1-tert—buty] 3-(l-ethoxy-1,3-dioxo(4-phenoxyphenyl)propanyl) piperidine-1,3- dicarboxylate (4). To a mixture of ethyl 2-bromo-3—oxo(4-phenoxyphenyl)propanoate 3 (1.0 g, 2.75 mmol) in CH3CN (20 mL) was added DIEA (0.39 g, 3.0 mmol) se. The resulting mixture was stirred at rt overnight. The volatile phase was removed under reduced pressure. The residue was applied onto silica gel column eluting with 3 :1 PE/EA to get the title nd (1.15 g, 82%) as yellow oil. MS (ESI): m/Z = 412.1 [M + H — Boer. ethyl 4-(4-phen0xyphenyl)(piperidinyl)oxazole-S-carboxylate (5). A mixture of 1-tert- butyl 3—(1-ethoxy-1,3—dioxo-3—(4-phenoxyphenyl)propanyl) piperidine-l ,3-dicarboxylate 4 (1.15 g, 2.25 mmol) and AcONH4 (0.86 g, 11.23 mmol) in AcOH (20 mL) was stirred at 120 CC for 2 h. The volatile phase was removed under reduced pressure. The residue was dissolved in EA (50 mL), which was washed with sat. NaHCOg, brine, dried over sodium sulfate, filtered and concentrated. This resulted in crude title compound (0.85 g, crude) as yellow oil. MS (ESI): m/z = 393.1 [M + H.
Ethyl(1-(tert-butoxycarbonyl)piperidinyl)—4—(4-phenoxyphenyl)oxazole-S-carboxylate (6). The title compound was obtained using a procedure analogous to the ure described in ethyl 2-(4-(tert-butoxycarbonyl)piperazin(4-phenoxyphenyl)thiazole-5 -carboxylate (see Scheme 37) as pale yellow oil (0.35 g, 33%, two steps). MS (ESI): m/z = 437.0 [M + H - 56]+. 2-(1-(tert—butoxycarb0nyl)piperidinyl)(4-phen0xyphenyl)0xazole-5—carboxylic acid (7). The title compound was obtained using a procedure analogous to the procedure described in -(1-(tert-butoxycarbony1)piperidin-4—y1)-4'-phenoxybiphenylcarboxylic acid (see Scheme 45) as yellow oil (350 mg, overweight). MS (ESI): m/z = 465.1 [M + H]+. tert-butyl 3-(5-carbamoyl(4-phenoxyphenyl)0xazolyl)piperidine-l-carboxylate (8).
The title compound was obtained using a procedure analogous to the procedure described in tert- butyl-3 -(4-carbamoyl—3-(4-phenoxyphenyl)—lH-pyrazolyl)pyrrolidine—1—carboxylate (see Scheme 23) as pale yellow solid (250 mg, overweight). MS (ESI): m/z = 408.0 [M + H - 56]+. 4-(4-phenoxyphenyl)(piperidinyl)0xazole-S-carboxamide hydrochloride (9). The title compound was obtained using a procedure analogous to the procedure bed in 1-(4- phenoxyphenyl)(piperidinyl)-1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as white solid (150 mg, overweight). MS (ESI): m/z = 364.1 [M + H]+.
Example 42 2-(1-acryloylpiperidinyl)(4-phenoxyphenyDoxazole—S-carboxamide The title compound was obtained using a ure analogous to the procedure described below in Example 43 as white solid (60 mg, 45%, four steps). 1H NMR (300 MHz, DMSO) 8 8.26 (d, J: 9.0 Hz, 2H), 7.92 (s, 1H), 7.67 (s, 1H), 7.53—7.29 (m, 2H), 7.25—7.13 (m, 1H), .99 (m, 4H), 6.85 (dd, J: 16.7, 10.5 Hz, 1H), 6.11 (dd, J: 16.7, 2.4 Hz, 1H), 5.68 (dd, J: 10.4, 2.4 Hz, 1H), 4.39—4.31 (m, 1H), 4.12—4.04 (m, 1H),3.33—3.14(m,2H),3.02—2.89(m, 1H), .06 (m, 2H), 1.90—1.65 (m, 2H). MS (ESI, method A): m/z = 418.1 [M + H]+, tR=1.488 min. HPLC: 97% (214nm), 97% (254nm).
Scheme 33 OQ o@ o@ O/Q HO N.Bon: 0 O 0 02—0 510 AcONH4,AcOH,reflulx Etc, / 3092010132903 5,0 / e0H,Hzo —> 0 —> ' ’ O N N 0 ° / THFIH o / 0 2 0 DIPEA, CH3CN, rt NH N\ N Boc 1 3 4 0 0 0 Cl / / HO / HATU. NHa. DMF HCIIEtOH HzN / q: H2" N HZN / N N —> —> —> ’ O / o I N ° o / DIPEA,DCM z) N N E NH HCI ‘Boc N‘ A Bee 0 6 7 Example 43 1-tert—butyl 3-(1-eth0xy-1,3-di0X0(4-phenoxyphenyl)propanyl) pyrrolidine-1,3- dicarboxylate (2). The title compound was obtained using a procedure analogous to the procedure described in 1-tert—buty1 3—( 1 -ethoxy- 1 ,3 (4-phenoxypheny1)propanyl) piperidine-l,3-dicarboxy1ate (see Scheme 32) as pale yellow oil (1.1 g, 80%). MS (ESI): m/z = 441.8 [M + H - 561+. ethyl 4-(4-phenoxyphenyl)(pyrrolidinyl)oxazole-S-carboxylate (3). The title compound was obtained using a procedure analogous to the procedure described in ethyl 4-(4- yphenyl)(piperidin—3-y1)oxazole-5—carboxy1ate (see Scheme 32) as yellow oil (0.80 g, crude). MS (ESI): m/Z = 420.1 [M + H + 411*. ethyl 2-(1-(tert-butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)oxazole—S-carboxylate (4). The title compound was obtained using a procedure analogous to the procedure described in ethyl 2—(4-(tert—butoxycarbonyl)piperazinyl)(4—phenoxyphenyl)thiazole—5-carboxy1ate (see Scheme 37) as pale yellow oil (0.35 g, 33%, two steps). MS (ESI): m/z = 423.1 [M + H - 56]+. 2-(1-(tert-butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)oxazole-S-carboxylic acid (5). The title compound was obtained using a procedure analogous to the procedure described in -(1-(tert-butoxycarbony1)piperidinyl)-4'-phenoxybiphenylcarboxylic acid (see Scheme 45) as yellow oil (350 mg, ight). MS (ESI): m/z = 395.0 [M + H — 56]+. tert-butyl 3-(5-carbam0yl—4—(4-phenoxyphenyl)oxazol-Z-yl)pyrrolidine-l-carboxylate (6).
The title nd was obtained using a procedure analogous to the procedure described in tert- butyl-3 —(4-carbamoyl—3-(4-phenoxyphenyl)-lH-pyrazolyl)pyrrolidine-1 -carboxylate (see Scheme 23) as pale yellow solid (250 mg, overweight). MS (ESI): m/z = 394.1 [M + H - 56]+. 4-(4-phenoxyphenyl)—2-(pyrrolidinyl)oxazole-S-carboxamide hydrochloride (7). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- phenoxyphenyl)(piperidinyl)—lH-pyrazole-3—carboxamide hydrochloride (see Scheme 46) as white solid (150 mg, overweight). MS (ESI): m/z = 350.1 [M + H]+.
Example 43 2-(1-acryloylpyrrolidinyl)—4-(4—phenoxyphenyDoxazole-S-carboxamide The title compound was ed using a procedure ous to the procedure described in e 15 as white solid (45 mg, 34%, four steps). 1H NMR (400 MHZ, DMSO) 5 8.27 (d, J = 8.8 Hz, 2H), 7.94 (d, J: 8.0 Hz, 1H), 7.70 (d, J: 8.0 Hz, 1H), 7.43 (t, J: 7.9 Hz, 2H), 7.19 (t, J = 7.9 Hz, 1H), 7.11—7.02 (m, 4H), 6.69—6.55 (m, 1H), 6.19 (d, J: 18.0 Hz, 1H), 5.70 (d, J: .4 Hz, 1H), 4.08—3.96 (m, 1H), 3.90—3.65 (m, 3H), 3.64—3.55 (m, 0.5H), 3.53—3.45 (m, 0.5H), 2.45—2.37 (m, 1H), 2.36—2.27 (m, 1H). MS (ESI, method A): m/z = 404.1 [M + H]+, tR=1.542 min. HPLC: 98% (214nm), 98% (254nm).
Scheme 34 O O NBoc O 0 . OEt 1, oxalyl chloride Iawesson s reagent, OEt—>pho HN o S —> \ 2, TEA, DCM \ NH THF 2 HC' COOEt NBoc PhD 1 2 3 NBoc 8PNBOC LiOH H20 NH3, HATU \\N —> 3.9 THF/MeOH/HZO DIPEA! DMF COOH PhO o NH2 \ TEA,DCM HCI ’ HCI/EtOH N \ s PhD 0 NHz PhD 0 NH2 Example44 tert-butyl 3-(1-eth0xy—1,3-di0x0(4-phenoxyphenyl)propan-Z-ylcarbamoyl)pyrrolidine carboxylate (2). The title compound was obtained using a procedure analogous to the ure described in tert-butyl 3-(1-ethoxy-l,3-dioxo(4-phenoxyphenyl)propan ylcarbamoyl)piperidine-l-carboxylate (see Scheme 35) as yellow oil (0.35 g, 30%). MS (ESI): m/Z = 440.7 [M — 551+. ethyl tert-butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)thiazole—4—carb0xylate (3). The title compound was obtained using a procedure analogous to the ure described in ethyl 2-(1 -(tert-butoxycarbonyl)piperidin-3 -yl)—5 -(4-phenoxyphenyl)thiazolecarboxylate (see Scheme 35) as yellow oil (0.12 g, 61%). MS (ESI): m/z = 494.7 [M + H]+. 2-(1-(tert—butoxycarb0nyl)pyrrolidinyl)(4-phenoxyphenyl)thiazolecarboxylic acid (4). The title compound was obtained using a procedure analogous to the procedure bed in -(l -(tert-butoxycarbonyl)piperidinyl)- l -(4-phenoxyphenyl)- l H-pyrazole-3 -carboxylic acid (see Scheme 46) as colorless oil (110 mg, 97%). MS (ESI): m/z = 488.7 [M + Na]+. tert-butyl 3-(4-carbamoyl-S—(4-phenoxyphenyl)thiazol-Z-yl)pyrrolidine-l-carb0xylate (5).
The title compound was obtained using a procedure analogous to the ure described in tert- butyl4-carbamoylpiperidinecarboxylate (see Scheme 42) as colorless oil (95 mg, 87%). MS (ESI): m/Z = 487.7 [M + Na]+. -(4-phenoxyphenyl)(pyrrolidinyl)thiazole-4—carboxamide hydrochloride (6). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- phenoxyphenyl)—5-(piperidinyl)-1H—pyrazolecarboxamide hydrochloride (see Scheme 46) as colorless oil (90 mg, 100%). MS (ESI): m/z = 365.8 [M + H]+.
Example 44 2-(1-acryloylpyrrolidinyl)—5-(4-phenoxyphenyl)thiazole—4-carboxamide N .0 The title compound was obtained using a procedure ous to the procedure described in Example 3 as a White solid (30 mg, 30%). 1H NMR (300 MHZ, CD30D) 5 7.52 (d, J = 9.0 Hz, 2H), 7.37 (d, J = 9.0 Hz, 2H), 7.15 (t, J = 7.4 Hz, 1H), .02 (m, 2H), 6.99—6.94 (m, 2H), 6.64 (ddd, J = 16.9, 10.4, 7.2 Hz, 1H), 6.29 (dt, J =16.8, 1.9 Hz, 1H), 5.76 (dt, J = 10.4, 1.7 Hz, 1H), 4.19—4.05 (m, 0.5H), 4.03—3.82 (m, 3H), 3.83—3.71 (m, 1H), 3.65—3.55 (m, 0.5H), 2.61— 2.23 (m, 2H). MS (ESI, method F): m/z = 419.7 [M + H]+, tR=1.487 min. HPLC: 99.3% (214nm), 98.7% (254nm).
Scheme 35 NaNOQ, ACOH Pd/C HCl/EtOH OEt H—O> 0 O O 0 1, oxalyl chloride GE 00 OEt —> Iawesson's reagent H" O 2, TEA, DCM —> S NH2 HCl Pho \ PhO THF mN COOEt NBoc PhO 4 5 NBoc NBoc HCl/EtOH LiOH H20 NH3, HATU S 8 , —> \N —> \ N THF THF/MeOH/HZO m DIPEA, DMF COOH PhO O NH2 6 7 NH N‘<— S TEA, DCM \ HCl S PhD 0 NH2 PhD 0 NH2 3 Example 45 hyl 2-(hydr0xyimin0)—3-0x0(4-phen0xyphenyl)pr0panoate (2). To a solution of ethyl 3-oxo(4-phenoxyphenyl)propanoate 1 (1 g, 3.52 mmol) in AcOH (15 mL) was added a solution ofNaNOz (0.364 g, 5.28 mmol) in water (10 mL) at 0 oC. The mixture was d at ambient temperature for 15 h. After finished, the mixture was extracted with ethyl acetate (2 X 20 mL). The organic layers were washed with water (2 X 10 mL) and brine (2 X 10 mL). The c layer was dried over NaZSO4, d and concentrated to give the title compound (1.1 g, 100%) as a yellow solid. MS (ESI): m/z = 313.8 [M + H]+. ethyl 2-amin00x0(4-phen0xyphenyl)pr0pan0ate hydrochloride (3). To the solution of (E)-ethyl 2-(hydroxyimino)—3—oxo(4—phenoxyphenyl)propanoate 2 (6 g, 19.15 mmol) in MeOH (50 mL) was added Pd/C (10%, 500 mg) and HCl/EtOH (33%, 20 mL) under hydrogen atmosphere, and the resulting solution was stirred for 24 h at ambient temperature, then filtered and the filtrate was trated. The crude product was purified by Prep-HPLC eluting with CH3CN/H20 (containing 0.5% TFA) from 10:90 to 60:40 to get the title compound (3.5 g, 61%) as yellow oil. MS (ESI): m/Z = 299.9 [M + Hr. tert-butyl 3-(1-eth0xy-1,3-di0xo(4-phenoxyphenyl)pr0panylcarbamoyl)piperidine—1- carboxylate (4). To the solution of l-(tert-butoxycarbonyl)piperidinecarboxylic acid (0.5 g, 2.18 mmol) in DCM (10 mL) was added oxalyl chloride (0.415 g, 3.27 mmol) and 5 drops of DMF. The mixture was stirred at rt for 2 h, concentrated to give the crude e. To the solution of ethyl ooxo-3—(4-phenoxyphenyl)propanoate hydrochloride 3 (0.73 g, 2.18 mmol) in DCM (20 mL) was added TEA (0.66 g, 6.54 mmol) and a solution of above mixture in DCM (10 mL). The resulting solution was d at rt for 2 h. The reaction mixture was washed with brine (2 X 10 mL), dried over NaZSO4, filtered and concentrated to get a e which was purified by silica gel column chromatography eluting with 1:1 PE/EA to get the title compound (0.16 g, 14%) as a yellow oil. MS (ESI): m/z = 454.7 [M — 55]+. ethyl 2-(1-(tert—butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)thiazole—4-carboxylate (5). To a solution of tert-butyl 3-(1-ethoxy-1,3-dioxo(4-phenoxyphenyl)propan ylcarbamoyl)piperidine-l-carboxylate 4 (0.16 g, 0.31 mmol) in THF (20 mL) was added lawesson’s reagent (0.127 g, 0.31 mmol). The mixture was degassed with N; for 3 times, then heated to reflux and stirred for 2 h. When finished, the mixture was extracted with ethyl e (2 X 20 mL). The organic layers were washed with sat. NaHC03 (2 X 20 mL) and brine (2 X 20 mL). The c layer was dried over Na2S04, filtered and concentrated to get a residue which was purified by silica gel column chromatography eluting with 2:1 PE/EA to get the title compound (90 mg, 57%) as a yellow oil. MS (ESI): m/z = 508.8 [M + H]+. 2-(1-(tert-but0xycarb0nyl)piperidinyl)(4-phenoxyphenyl)thiazolecarboxylic acid (6). The title compound was obtained using a procedure analogous to the procedure described in -(l -(tert-butoxycarbonyl)piperidinyl)- l enoxyphenyl)— l H-pyrazole-3 -carboxylic acid (see Scheme 46) as a white solid (80 mg, 94%). MS (ESI): m/z = 480.8 [M + H]+. tert-butyl 3-(4-carbamoyl(4-phenoxyphenyl)thiazolyl)piperidine—l-carboxylate (7).
The title compound was obtained using a procedure analogous to the procedure described in tert- butyl 4-carbamoylpiperidine-l-carboxylate (see Scheme 42) as colorless oil as colorless oil (40 mg, 50%). MS (ESI): m/Z = 479.7[M + Hr. henoxyphenyl)—2-(piperidinyl)thiazolecarboxamide hydrochloride (8). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- phenoxyphenyl)—5-(piperidinyl)-1H—pyrazolecarboxamide hydrochloride (see Scheme 46) as yellow oil (35 mg, 100%). MS (ESI): m/z = 379.8[M + H]+.
Example 45 2-(1-acryloylpiperidinyl)—5-(4-phenoxyphenyl)thiazolecarboxamide (fifioQ The title compound was obtained using a procedure ous to the procedure described in Example 3 as a white solid (10 mg, 27%). 1H NMR (300 MHz, CD30D) 5 7.56—7.48 (m, 2H), 7.44—7.32 (m, 2H), 7.15 (t, J: 7.4 Hz, 1H), 7.03 (d, J: 0.9 Hz, 2H), 6.99—6.94 (m, 2H), 6.84 (ddd,J= 23.2, 16.7, 10.6 Hz, 1H), 6.21 (dd,J= 16.8, 1.9 Hz, 1H), 5.75 (d,J= 10.7 Hz, 1H), 4.62 (d, J: 11.6 Hz, 0.5H), 4.27 (d, J: 13.2 Hz, 0.5H), 4.15 (d, J: 12.8 Hz, 0.5H), 4.00 (d, J: 13.6 Hz, 0.5H), 3.72 (dd, J= 13.5, 8.9 Hz, 0.5H), .33 (m, 1H), 3.29—3.15 (m, 15H), 2.35— 2.23 (m, 1H), 2.08—1.85 (m, 2H), 1.75—1.55 (m, 1H). MS (ESI, method F): m/z = 433.8 [M + H]: tR=1.557 min. HPLC: 96.7% (214nm), 98.0% (254nm).
Scheme 36 QC? 0@ 0 g o AcONH4,AcOH,reflulx EtO / Boc2O,Na2603 OC 0 —> —> N ° ) Etc O / THFIHZO DIPEA, CchN, rt 0 N N Boc H 1 2 3 o@ o@ °Q 0 0 EtO / NaOH, MEOH, H20 HATU, NHg, DMF H2N / / HCl/EtOH IN —> HO —> 03> /N —> O IN 0 Bee N Eoc o Wm / o o H2N —> o / "'2N / DIPEA,DCM Bee \ Example 46 l-tert-butyl 4-(l-ethoxy-1,3-di0X0(4-phenoxyphenyl)pr0panyl) piperidine—1,4- dicarboxylate (2). The title compound was ed using a procedure analogous to the procedure bed in l -tert—butyl 3-( l -ethoxy- l ,3 -dioxo(4-phenoxyphenyl)propanyl) piperidine-l,3-dicarboxylate (see Scheme 32) as pale yellow oil (0.85 g, 60%). MS (ESI): m/z = 456.1 [M + H — 561+. ethyl 4-(4-phenoxyphenyl)(piperidinyl)oxazole—S-carboxylate (3). The title compound was obtained using a procedure analogous to the procedure described in ethyl 4-(4- phenoxyphenyl)(piperidinyl)oxazolecarboxylate (see Scheme 32) as yellow oil (0.80 g, crude). MS (ESI): m/Z = 393.2 [M + H.
Ethyl(1-(tert-butoxycarbonyl)piperidinyl)—4—(4-phenoxyphenyl)oxazole-S-carboxylate (4). The title nd was obtained using a procedure analogous to the procedure described in ethyl 2—(4-(tert—butoxycarbonyl)piperazinyl)(4—phenoxyphenyl)thiazole—5 -carboxylate (see Scheme 37) as pale yellow oil (0.85 g, 68%, two steps). MS (ESI): m/z = 437.2 [M + H - 56]+. 2-(1-(tert—butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)0xazole-5—carboxylic acid (5). The title compound was ed using a procedure analogous to the procedure described in -(1—(tert-butoxycarbonyl)piperidin—4-yl)-4'-phenoxybipheny1—2-carboxylic acid (see Scheme 45) as yellow oil (0.8 g, 99%). MS (ESI): m/Z = 409.1 [M + H — 561+. tert-butyl 4-(5-carbamoyl-4—(4-phenoxyphenyl)0xazolyl)piperidine-l-carboxylate (6).
The title compound was obtained using a procedure analogous to the procedure bed in tert- butyl-3 -(4-carbamoyl(4-phenoxyphenyl)—1H-pyrazolyl)pyrrolidine-1—carboxylate (see Scheme 23) as pale yellow solid (390 mg, overweight). MS (ESI): m/z = 408.0 [M + H - 56]+. henoxyphenyl)(piperidinyl)oxazole-S-carboxamide hydrochloride (7). The title compound was obtained using a procedure analogous to the ure described in 1—(4- phenoxyphenyl)(piperidinyl)-1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as white solid (400 mg, overweight). MS (ESI): m/z = 364.1 [M + H]+. e 46 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)0xazolecarboxamide The title compound was obtained using a procedure analogous to the procedure described in Example 15 as white solid (45 mg, 34%, three steps). 1H NMR (300 MHz, DMSO) 8 8.26 (d, J = 9.0 Hz, 2H), 7.92 (s, 1H), 7.674 (s, 1H), 7.53—7.29 (m, 2H), 7.25—7.13 (m, 1H), 7.11—6.99 (m, 4H), 6.85 (dd, J: 16.7, 10.5 Hz, 1H), 6.11 (dd, J: 16.7, 2.4 Hz, 1H), 5.68 (dd, J: 10.4, 2.4 Hz, 1H), 4.39—4.31 (m, 1H), 4.12—4.04 (m, 1H), 3.33—3.14(m, 2H), 3.02—2.89 (m, 1H), 2.14—2.06 (m, 2H), 1.90—1.65 (m, 2H). MS (ESI, method A): m/z = 418.1 [M + H]+, tR=1.489 min. HPLC: 97% (214nm), 97% (254nm).
Scheme 37 GNi"D J 0"" (N’ Y""2 ODMO 0 J(Boc)20 STNJ [N)1. 2, THF 55°C, 1h o N 2N;3H20 ET] 0 N r..,t 3d /0 Egg/H reflux 2h TEA, THF, Boc o r.t., 2h,73% 1 3 00 00 6 OH \NH o S /—\ L'OH. |/>_N N_BOC HATU,CH3NH2 o s /—‘\ —’ N—Boc N \__/ —> | />—N THF/HZO/MeoH Q TEA, DCM, r.t., 3h,90% N 60°C, 4h, 97% 0 7 ©\0 8 1;:thr..t 1h ©\OO film—N Iii/39w TEA THF 0°C 10min Example 47 tert-butyl amothioylpiperazine—1-carb0xylate (3). To a solution of di(lH-imidazol-l- yl)methanethione 2 (2.14 g, 12 mmol) in anhydrous THF (30 mL) was added tert-butyl piperazine-l-carboxylate 1 (1.86 g, 10 mmol) at ambient temperature. The mixture was allowed to stir at ambient temperature for 2 h, then the mixture was heated at 55 °C for l h. The mixture was concentrated under vacuum to about half the volume. To the ing reaction mixture was added 2 M solution of ammonia in methanol (20 mL) and allowed to stir at ambient temperature for 3 days. The solvent was removed and the residue was purified by chromatography eluting with 50:1 DCM/MeOH to afford the title compound (1.7 g, 69%) as white solid. MS (ESI): m/z = 246.1 [M + H]+. ethyl 4-(4-phen0xyphenyl)—2-(piperazinyl)thiazolecarboxylate (5). To a solution of ethyl ooxo(4-phenoxyphenyl)propanoate 4 (362 mg, 1 mmol) in ethanol (10 mL)was added tert-butyl amothioylpiperazine—l-carboxylate 3 (245 mg, 1 mmol) at ambient ature. The e was then heated to reflux and stirred for 2 h. After cooling to ambient temperature, the solvent was removed and the residue was purified by silica gel column chromatography eluting with 40:1 to 20:1 DCM/MeOH to afford the title compound (214 mg, 52%) as brown solid. MS (ESI): m/Z = 410.1 [M + H. ethyl 2-(4-(tert-but0xycarb0nyl)piperazinyl)—4—(4-phenoxyphenyl)thiazole—5-carboxylate (6). To the compound of ethyl 4-(4-phenoxyphenyl)—2-(piperazin-1—yl)thiazolecarboxylate 5 (214 mg, 0.51 mmol) were added di-tert-butyl dicarbonate (134 mg, 0.6 mmol) and TEA (0.2 mL, 1.5 mmol) at t temperature. The mixture was then stirred at ambient temperature for 2 h. The solvent was removed and the e was purified by silica gel column chromatography eluting with 50:1 DCM/MeOH to afford the title nd (190 mg, 73%) as brown solid. MS (ESI): m/Z = 510.1 [M + Hr. 2-(4-(tert-but0xycarbonyl)piperazinyl)(4-phenoxyphenyl)thiazole—5-carboxylic acid (7). To a solution of ethyl 2-(4-(tert—butoxycarbonyl)piperazinyl)(4- yphenyl)thiazolecarboxy1ate 6 (190 mg, 0.37 mmol) in THF (50 mL)/H20 (1 mL)/MeOH (3 mL) was added lithium hydroxide (30 mg, 0.74 mmol) at ambient temperature.
The mixture was then heated to 60 °C and stirred for 2 h. After cooling to ambient temperature, the solvent was d and the residue was purified by chromatography eluting with 10:1 DCM/MeOH to afford the title compound (173 mg, 97%) as white solid. MS (ESI): m/z = 482.0 [M + Hr. tert-butyl 4-(5-(methylcarbamoyl)(4—phenoxyphenyl)thiazolyl)piperazine—1- carboxylate (8). To a solution of 2-(4-(tert-butoxycarbonyl)piperaziny1)—4-(4- phenoxyphenyl)thiazole carboxylic acid 7 (124 mg, 0.26 mmol) in DCM (10 mL) was added HATU (117 mg, 0.3 mmol) and methanamine (0.26 ml, 0.26 mmol) at ambient temperature. The mixture was stirred at ambient temperature for 3 h. The solvent was removed and the residue was d by silica gel column chromatography eluting with 1:5 EA/PE to afford the title compound (115 mg, 90%) as colorless oi1. MS (ESI): m/z = 495.2 [M + H]+.
N-methyl(4-phenoxyphenyl)(piperazinyl)thiazolecarb0xamide (9). To a solution of tert—butyl 4-(5 -(methylcarbamoy1)—4-(4-phenoxypheny1)thiazo1-2—y1)piperazineCarboxy1ate 8 (115 mg, 0.23 mmol) in DCM (5 mL) was added TFA (1 mL) at t temperature. The mixture was stirred for 1 h at ambient temperature. The solvent was removed and the residue was purified by silica gel column chromatography eluting with 10:1 OH to afford the title compound (117 mg, 100%) as colorless oil.
Example 47 2-(4-acryloylpiperazin-l-y])—N-methyl—4-(4—phenoxyphenyl)thiazole-S-carboxamide AWOSEQD../ To a solution of N—methyl(4-phenoxyphenyl)(piperazinyl)thiazole—5-carboxamide 9 (117 mg, 0.23 mmol) in DCM (5 mL) were added TEA (0.1 mL, 0.7 mmol) and acryloyl chloride 10 (0.21 mg, 0.23 mol) at 0 CC. The mixture was stirred at 0 CC for 10 minutes. The solvent was removed and the e was purified by Prep-TLC eluting with 50:1 to 20:1 DCM/MeOH to afford the title nd (50 mg, 49%) as white solid. 1H NMR (400 MHz, CDCl3) 5 7.57 (d, J: 8.6 Hz, 2H), 7.40 (t, J: 7.9 Hz, 2H), 7.19 (t, J: 7.4 Hz, 1H), 7.09 (d, J: 8.4 Hz, 4H), 6.60 (dd, J: 16.8, 10.5 Hz, 1H), 6.37 (d, J: 16.7 Hz, 1H), 5.79 (d, J: 11.8 Hz, 1H), 5.59 (d, J: 4.1 Hz, 1H), 3.78 (d, J: 48.5 Hz, 4H), 3.62 (s, 4H), 2.80 (d, J: 4.8 Hz, 3H), MS (ESI, method A): m/z = 449.1 [M + H]+, 28 min. HPLC: 96.9% (214nm), 96.9% (254nm).
Scheme 38 O O NBoc NBoc O O 1, oxalyl chloride. 12, Pth LIOH H20 OEt—>Ph0 "N o —> O O \ —> \ \ N \ N NH 2, TEA, DCM TEA, DCM THF/MeOH/HZO 2 HCI coca COOH NBOC PhO PhD 1 2 3 4 NBoc NH NH3: HATU HCI/E OHt O O \N TEA,DCM \N HCI O DIPEA‘ DMF m THF m \ N PhD 0 NH2 PhD 0 NH2 PhD 0 6 Example 48 tert-butyl(1-eth0xy—1,3-di0x0(4-phenoxyphenyl)pr0panylcarbamoyl)pyrrolidine carboxylate (2). The title compound was obtained using a procedure analogous to the procedures described in the General Scheme and e 15 as yellow oil (0.35 g, 30%). MS (ESI): m/Z = 440.7 [M — 551+.
Ethyl-2—(1-(tert-butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)0xazole carboxylate (3). To a solution of Pth (159 mg, 0.604 mmol) in DCM (20 mL) was added 12 (153 mg, 0.604 mmol). The resulting mixture was degassed with N2 3 times and stirred at ambient temperature for 2 h. TEA (122 mg, 1.208 mmol) was added and stirred for 10 min, then a solution of tert-butyl 3-(1-ethoxy-1,3-dioxo—3-(4-phenoxyphenyl)propan-2— ylcarbamoy1)pyrrolidinecarboxy1ate 2 (150 mg, 0.302 mmol) was added and d for 15 h.
The mixture was diluted with ethyl acetate (50 mL), and washed with sat. Na2S203 (20 mL), brine (3 X 20 mL). The organic layer was dried over NazSO4, filtered and concentrated to get a residue which was purified by silica gel column chromatography eluting with 1:1 PE/EA to get the title compound (0.11 g, 76%) as a ess oil. MS (ESI): m/z = 422.8 [M — 55]+. tert—butoxycarbonyl)pyrrolidinyl)(4-phenoxyphenyl)oxazole—4-carboxylic acid (4). The title compound was obtained using a procedure analogous to the ure described in -(1 -(tert-butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)— 1 zole-3 -carboxylic acid (see Scheme 46) as yellow oil (110 mg, 100%). MS (ESI): m/z = 394.8 [M — 55]+ tert-butyl arbam0yl-5—(4-phenoxyphenyl)oxazol-Z-yl)pyrrolidine-l-carboxylate (5).
The title compound was obtained using a procedure ous to the procedure described in tert- butyl 4-carbamoylpiperidinecarboxylate (see Scheme 42) as colorless oil (95 mg, 92%). MS (ESI): m/z = 393.8 [M — 551+. -(4-phenoxyphenyl)(pyrrolidinyl)0xazole—4—carb0xamide hydrochloride (6). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- phenoxyphenyl)—5-(piperidinyl)-1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as a white solid (75 mg, 92%). MS (ESI): m/z = 349.9 [M + H]+.
Example 48 2-(1-acryloylpyrrolidinyl)(4—phenoxyphenyl)0xazolecarboxamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as an off white solid (50 mg, 64%). 1H NMR (400 MHz, DMSO) 5 8.27—8.22 (m, 2H), 7.61 (s, 1H), 7.58 (s, 1H), 7.45 (t, J: 7.9 Hz, 2H), 7.21 (t, J: 7.3 Hz, 1H), 7.14—7.06 (m, 4H), 6.67—6.58 (m, 1H), 6.16 (d, J: 16.7 Hz, 1H), 5.70 (d, J: 10.4 Hz, 1H), 4.05—4.01 (m, 0.5H), 3.97—3.92 (m, 0.5H), 3.90—3.59 (m, 3.5H), 3.53—3.45 (m, 0.5H), 2.45—2.21 (m, 2H). MS (ESI, method A): m/z = 404.1 [M + H]+, tR=1.484 min. HPLC: 99.5% (214nm), 99.3% (254nm) Scheme 39 O O NBoc NBoc 1 oxalyl chloride HN O on's rea ent LiOH H 0 08—»Pho —g> —2> S \ NHz HCI 2, TEA DCM PhO Et OHiHZOmNCOOH N PhO 1 2 NBoc NH3. HATU HCI/EtOH TEA DCM S —> \ —> sp\ HCI '—>Pho DIPEA, DMF m" THF MHZ PhD 0 NH2 NH PhD 0 6 Example 49 tert-butyl—4-(1-ethoxy—1,3-di0X0(4-phenoxyphenyl)propanylcarbamoyl)piperidine carboxylate (2). The title compound was obtained using a procedure analogous to the procedures described in the General Scheme and Example 15 as colorless oil (0.18 g, 16%). MS (ESI): m/Z = 454.8[M — 55]+ ethyl 2-(1-(tert—butoxycarb0nyl)piperidinyl)—5-(4-phenoxyphenyl)thiazolecarboxylate (3). The title compound was obtained using a ure analogous to the procedure described in tert-butyl 2-carbamothioylpiperidinecarboxylate (see Scheme 30) as colorless oil (0.12 g, 67%). MS (EST): m/Z = 508.8 [M + H]+. 2-(1-(tert—butoxycarb0nyl)piperidinyl)(4-phenoxyphenyl)thiazolecarboxylic acid (4). The title compound was obtained using a procedure analogous to the procedure described in -(1 -(tert-butoxycarbonyl)piperidin-4—yl)(4-phenoxyphenyl)— 1 H-pyrazole-3 -carboxylic acid (see Scheme 46) as yellow oil (120 mg, 100%). MS (ESI): m/z = 480.8 [M + H]+. tert-butyl 4-(4-carbam0yl-5—(4-phenoxyphenyl)thiazolyl)piperidine—l-carboxylate (5).
The title compound was obtained using a procedure analogous to the procedure described in tert- butyl arbamoyl—4—(4-phenoxyphenyl)thiazol-2—yl)piperidine-1—carboxylate (see Scheme ) as colorless oil (100 mg, 88%). MS (ESI): m/z = 479.8[M + H]+. -(4-phenoxyphenyl)(piperidinyl)thiazole—4-carboxamide hydrochloride (6). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- yphenyl)—5-(piperidinyl)—1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as a white solid (80 mg, 92%). MS (ESI): m/z = 379.8[M + H]+.
Example 49 2-(1-acryloylpiperidinyl)—5-(4-phenoxyphenyl)thiazole—4-carboxamide PhD SQ O \ / The title compound was obtained using a procedure ous to the procedure described in Example 51 as an off white solid (20 mg, 24%). 1H NMR (400 MHz, DMSO) 8 7.68 (s, 1H), 7.56 (d, J: 8.7 Hz, 2H), 7.50 (s, 1H), 7.47—7.42 (m, 2H), 7.21 (t, J: 7.4 Hz, 1H), 7.10 (d, J: 7.7 Hz, 2H), 7.00 (d, J= 8.7 Hz, 2H), 6.85 (dd, J= 16.7, 10.5 Hz, 1H), 6.12 (dd, J= 16.7, 2.4 Hz, 1H), .69 (dd, J: 10.5, 2.4 Hz, 1H), 4.49 (d, J: 12.8 Hz, 1H), 4.15 (d, J: 12.8 Hz, 1H), 3.33—3.19 (m, 2H), 2.88 (d, J: 11.5 Hz, 1H), 2.12 (d, J: 12.8 Hz, 2H), 1.73—1.55 (m, 2H). MS (ESI, method A): m/z = 434.0 [M + H]+, tR=1.606 min. HPLC: 95.0% (214nm), 96.6% (254nm) Scheme 40 OO o O "Gk/IN O O ‘N,.CNH N / ~N,CN/ Scheme 41 C D 0 0 J NH ~ ~ . SW (1 .r 0/‘0 m mB..
O SYN N \ [ 1 ' 2 THF 55°C 1h N IN (300120 ’ ‘ ’ E 4 0 o \SYN{I N 2. NH H O, r.t., 3d EtOH, reflux, 2h, face 69%3 2 '1 TEAvTHE 3°C at, 2h, 73% 1 3 00 5 QOG LiOH />_N/—\—NBoc HATU NH3 THFiHZO/MeOH TEA DCM, rt 3h 100% |:\/:/\,>—NN-Boc 60°C 4h 97% l:/—\/>—N >~N Ego/M rt 1h goo TEA THF 0°C 10mim Example 51 tert-butyl 4-carbamothioylpiperazine-l-carb0xylate (3). To a solution of di(1H—irnidazol yl)methanethione 2 (2.14 g, 12 mmol) in anhydrous THF (30 mL) was added tert-butyl zine-l—carboxylate 1 (1.86 g, 10 mmol) at t temperature. The e was allowed to stir at ambient temperature for 2 h, and then the mixture was heated to 55 °C for 1 h. The mixture was concentrated in vacuum to about half the volume. To the remaining reaction mixture was added a 2 M ammonia in methanol (20 mL) and was d to stir at ambient temperature for 3 days. The t was removed and the residue was purified by silica gel column chromatography eluting with 50:1 DCM/MeOH to afford the title compound (1.7 g, 69%) as white solid. MS (ESI): m/Z = 246.1 [M + H]+. ethyl 4-(4-phen0xyphenyl)(piperazinyl)thiazolecarboxylate (5). To a solution of ethyl 2-bromooxo(4-phenoxyphenyl)propanoate 4 (362 mg, 1 mmol) in ethanol (10 l 1 1 mL)was added tert-butyl 4-carbamothioylpiperazine—1-carboxy1ate 3 (245 mg, 1 mmol) at ambient ature. The mixture was then heated to reflux and stirred for 2 h. After cooling to ambient temperature, the solvent was removed and the residue was d by silica gel column chromatography eluting with 40:1 to 20:1 DCM/MeOH to afford the title nd (214 mg, 52%) as brown solid. MS (ESI): m/z = 410.1 [M + H]+.
Ethyl(4-(tert-butoxycarbonyl)piperazinyl)-4—(4-phenoxyphenyl)thiazole-S-carboxylate (6). To the compound of ethyl 4-(4-phenoxyphenyl)(piperazin-1—yl)thiazolecarboxylate 5 (214 mg, 0.51 mmol) was added di-tert-butyl dicarbonate (134 mg, 0.6 mmol) and TEA (0.2 mL, 1.5 mmol) at ambient temperature. The mixture was then stirred at ambient temperature for 2 h.
The solvent was removed and the residue was purified by silica gel column chromatography eluting with 50:1 DCM/MeOH to afford the title compound (190 mg, 73%) as brown solid. MS (ESI): m/Z = 510.1 [M + Hr. 2-(4-(tert-but0xycarb0nyl)piperazinyl)(4-phenoxyphenyl)thiazole—5-carb0xylic acid (7). To a solution of ethyl 2-(4-(tert-butoxycarbonyl)piperazinyl)(4- phenoxyphenyl)thiazolecarboxylate 6 (190 mg, 0.37 mmol) in THF (50 mL)/H20(1mL)/MeOH(3 mL) was added lithium hydroxide (30 mg, 0.74 mmol) at ambient temperature. The mixture was then heated to 60 CC and stirred for 2 h. After cooling to ambient temperature, the solvent was removed and the residue was purified by silica gel column chromatography eluting with 10:1 DCM/MeOH to afford the title compound (173 mg, 97%) as white solid. MS (ESI): m/Z = 482.0 [M + H]+. tert-butyl 4-(5-carbamoyl-4—(4-phenoxyphenyl)thiazol-Z-yl)piperazine—l-carboxylate (8).
To a solution of 2-(4-(tert-butoxycarbonyl)piperazinyl)(4-phenoxyphenyl)thiazole carboxylic acid 7 (173 mg, 4.6 mmol) in DCM (10 mL) was added HATU (164 mg, 0.43 mmol) at ambient temperature. The mixture was degassed three times with ammonia gas and stirred at ambient temperature under a here for 6 h. The solvent was d and the residue was purified by silica gel column chromatography eluting with 30:1 DCM/MeOH to afford the title compound (170 mg, 100%) as white solid. MS (ESI): m/z = 481.1 [M + H]+. 4-(4-phen0xypheny])—2-(piperazinyl)thiazole-S-carboxamide (9). A solution of tert-butyl arbamoyl(4-phenoxyphenyl)thiazolyl)piperazinecarboxylate 8 (170 mg, 0.35 mmol) in DCM (5 mL) and TFA (1 mL) at ambient temperature was d for 1 h. The solvent was removed and the residue was purified by silica gel column chromatography g with :1 to 10:1 DCM/MeOH to afford the title compound (110 mg, 58%) as white solid.
Example 51 cryloylpiperazinyl)(4-phenoxyphenyl)thiazole—5-carboxamide 0 S /—\ 0 lN/>_N\__/NJ<= To a solution of 4-(4-phenoxyphenyl)—2-(piperazin-l—yl)thiazolecarboxamide 9 (110 mg, 0.22 mmol) in DCM (5 mL) was added TEA (0.1 mL, 0.66 mmol) and acryloyl chloride 10 (0.20 mg, 0.22 mmol) at 0 OC. The mixture was stirred at 0 °C for 10 minutes. The solvent was removed and the residue was purified by Prep-TLC g with 25 :1 DCM/MeOH to afford the title compound (70 mg, 73%) as white solid. 1H NMR (400 MHz, MeOD) 8 7.66—7.61 (m, 2H), 7.37— 7.43 (m, 2H), 7.18 (t, J: 7.4 Hz, 1H), 7.10—7.04 (m, 4H), 6.83 (dd, J: 16.8, 10.6 Hz, 1H), 6.27 (dd,J=16.8, 1.8 Hz, 1H), 5.81 (dd,J= 10.6, 1.9 Hz, 1H), 3.87—3.78 (m, 4H), 3.69—3.61 (m, 4H). MS (ESI, method A): m/z = 435.0 [M + H]+, tR=1.539 min., HPLC: 95.5% (214nm), 96.4% (254nm).
Scheme 42 HO s 60 Q mBr 4 HATU NH3 DMF 150 Lawessons reagent 0 O OEt toluene, reflux, 2h EtOH, reflux 2 3 o@ o@ o@ o 0 "0 / NHS’HATU'DMF EtO / EC / Boc20,NaZCO3 NaOH,MeOH,H20 N —’ —> 3% N —> s THF/H20 3% E/ N N Boc H Boc e 7 o o H2N / / N HCI/EtOH HZN "I H2N S / 3 / i S /N E DIPEA,DCM N N Example 52 tert-butyl 4-carbamoylpiperidine-l-carboxylate (2). To a mixture of l-(tert- butoxycarbonyl)piperidinecarboxylic acid 1 (2.29 g, 10 mmol), DIEA (3.87 g, 30 mmol) and HATU (4.18 g, 11 mmol) in DMF (50 mL) was added NH3 by bubbling for 20 min. The resulting mixture was d at rt overnight. The e was diluted with water (200 mL) and ted with EA (2 X 50 mL). The organic layers were combined, washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was applied onto silica gel column g with EA to get the title compound (1.3 g, 56%) as white solid. MS (ESI): m/Z = 173.2 [M + H — 561+. tert-butyl 4-carbamothioylpiperidinecarb0xylate (3). The title compound was obtained using a procedure analogous to the procedure described in cyclopentanecarbothioamide (see Scheme 30) as pale yellow solid (0.35 g, 29%). MS (ESI): m/z = 245.2 [M + H]+. ethyl 4-(4-phenoxyphenyl)—2-(piperidinyl)thiazolecarb0xylate (5). The crude of the title compound was obtained using a procedure analogous to the procedure described in ethyl 2- cyclopentyl-4—(4-phenoxyphenyl)thiazolecarboxylate (see Scheme A-2) as yellow oil (0.90 g, overweight). MS (ESI): m/Z = 408.8 [M + H]+. ethyl 4-(4-phenoxyphenyl)—2-(piperidinyl)thiazolecarboxylate (6). The title compound was obtained using a procedure analogous to the procedure described in ethyl 2-(4-(tert- butoxycarbony1)piperazinyl)-4—(4-phenoxyphenyl)thiazolecarboxylate (see Scheme 37) as yellow solid (0.30 g, 30%, two . MS (ESI): m/Z = 509.1 [M + H]+. 2-(1-(tert-but0xycarb0nyl)piperidinyl)(4-phenoxyphenyl)thiazolecarboxylic acid (7). The title compound was obtained using a procedure analogous to the procedure bed in -(1-(tert-butoxycarbonyl)piperidiny1)-4'-phenoxybiphenylcarboxylic acid (see Scheme 45) as yellow oil (250 mg, overweight). MS (ESI): m/z = 481.0 [M + H]+. tert-butyl 4-(5-carbamoyl(4-phenoxyphenyl)thiazolyl)piperidine—l-carboxylate (8).
The title nd was obtained using a procedure analogous to the procedure described in tert- butyl-3 -(4-carbamoyl(4-phenoxyphenyl)-1H-pyrazoly1)pyrrolidinecarboxylate (see Scheme 23) as pale yellow solid (250 mg, overweight). MS (ESI): m/Z = 480.1 [M + H]+. 4-(4-phenoxyphenyl)(piperidinyl)thiazole—5-carb0xamide (9). The title compound was obtained using a procedure ous to the procedure described in 1-(4-phenoxyphenyl) (piperidinyl)—1H-pyrazolecarboxamide hloride (see Scheme 46) as white solid (300 mg, overweight). MS (ESI): m/z = 380.1 [M + H]+.
Example 52 2-(1-acryloylpiperidinyl)(4-phen0xyphenyl)thiazolecarb0xamide WNO"i \ o O o The title compound was obtained using a procedure analogous to the procedure described in Example 15 as white solid (50 mg, 23%, four steps). 1H NMR (300 MHz, DMSO) 8 .69 (m, 4H), 7.42 (t, J= 7.4 Hz, 2H), 7.19 (t, J= 7.4 Hz, 1H), 7.09—7.00 (m, 4H), 6.84 (dd, J= 16.7, .4 Hz, 1H), 6.10 (dd, J: 16.7, 2.4 Hz, 1H), 5.68 (dd, J: 10.4, 2.4 Hz, 1H), 4.52—4.41 (m, 1H), 4.19—4.08 (m, 1H), 3.32—3.15 (m, 2H), 2.90—2.78 (m, 1H), 2.14—2.06 (m, 1.73—1.46 , 2H), (m, 2H). MS (ESI, method A): m/Z = 434.0 [M + H]+, tR=1.540 min. HPLC: 96% (214nm), 96% Scheme 43 w (9 o .
EtOH,70°C / Example 50 Example 53 e 53 (E)(1-(2-cyan0cyclopropylacryloyl)azetidinyl)—3-(4-phenoxyphenyl)—1H-pyrazole carboxamide OO O / ‘N/CN o / To a solution of 1-(1—(2-cyanoacetyl)azetidinyl)—3-(4-phenoxyphenyl) -1H-pyrazole carboxamide (100 mg, 0.25 mmol) and piperidine (2 drops) in EtOH (10 mL) was added cyclopropanecarbaldehyde (70 mg, 1.0 mmol). Then the solution was stirred at 70°C for 1 h.
After the reaction was completed, the solution was concentrated and purified by Prep—TLC with :1 DCM/MeOH to give the crude product (70 mg) and then purified by PLC (ACN/H20 = 42%, ) to afford the title compound (15 mg, 13%) as a white solid. 1H NMR (400 MHZ, CDClg) 8 8.16 (s, 1H), 7.64—7.57 (m, 2H), 7.45—7.37 (m, 2H), 7.22—7.16 (m, 1H), 7.15—7.03 (m, 5H), 5.60 (s, 2H), 5.23—5.13 (m, 1H), 5.11—4.95 (m, 2H), 4.68—4.54 (m, 2H), 2.16—2.05 (m, 1H), 1.33—1.29 (m, 2H), 1.02—0.95 (m, 2H). MS (ESI, Method A): m/z = 454.1 [M + H]+, tR = 1.485 min. HPLC: 100% (214nm), 100% (254nm).
Scheme 44 O O NBOC NBOC O O 1 ,oxayc one[ | hl 'd M03 HN o A|,PhP LiOHHO (mt—who o\ —2, o\ NHz HC] 2, TEA, DCM TEA, DCM mN THF/MeOH/HZO @N NBoc COOEt COOH PhO PhO 1 2 3 4 N_<— 0QNBOC NH3, HATU HCl/EIOH 0 TEA, DCM —, \ —> QNH\ HQ] —> \ DIPEA, DMF m" THF O/jl" PhD ml 0 NHz PhO o NHz PhO o NH2 6 Example 54 tert-butyl—3-(1-eth0xy—1,3-di0x0(4-phenoxyphenyl)propanylcarbamoyl)piperidine carboxylate (2). The title compound was obtained using a procedure analogous to the procedures described in the General Scheme and Example 15 as yellow oil (0.4 g, 36%). MS (ESI): m/z = 454.8 [M —55]+.
Ethyl(1-(tert-butoxycarbonyl)piperidinyl)—5—(4-phenoxyphenyl)0xazole-4—carboxylate (3). The title compound was obtained using a ure ous to the ure described in ethyl 2-(1-(tert-butoxycarbonyl)pyrrolidinyl)(4—phenoxyphenyl)oxazolecarboxylate (see Scheme 44) as yellow oil (0.25 g, 65%). MS (ESI): m/z = 492.8 [M + H]+. 2-(1-(tert—butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)0xazolecarboxylic acid (4). The title compound was ed using a procedure analogous to the procedure described in -(1 -(tert-butoxycarbonyl)piperidin-4—yl)(4—phenoxyphenyl)— 1 H-pyrazole—3 -carboxylic acid (see Scheme 46) as yellow oil (0.25 g, 100%). MS (ESI): m/z = 464.8 [M + H]+. tert-butyl 3-(4-carbamoyl-5—(4-phenoxyphenyl)0xazolyl)piperidine-l-carboxylate (5).
The title compound was obtained using a procedure analogous to the procedure described in tert- butyl 3-(5-carbamoyl—4-(4—phenoxyphenyl)thiazol—2—yl)piperidinecarboxylate (see Scheme ) as yellow oil (0.2 g, 84%). MS (ESI): m/z = 407.8 [M — 55]+. -(4-phenoxyphenyl)(piperidinyl)oxazolecarboxamide hydrochloride (6). The title compound was obtained using a ure analogous to the procedure described in 1-(4- phenoxyphenyl)(piperidinyl)—1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as brown oil (180 mg, 100%). MS (ESI): m/Z = 363.8[M + H]+.
Example 54 2-(1-acryloylpiperidinyl)—5-(4—phenoxyphenyl)0xazolecarboxamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as a yellow solid (70 mg, 37%). 1H NMR (400 MHz, DMSO) 8 8.24 (d, J= 7.2 Hz, 2H), 7.57 (s, 1.5H), 7.50 (s, 0.5H), 7.45 (t, J: 7.9 Hz, 2H), 7.21 (t, J: 7.4 Hz, 1H), 7.11 (d, J: 3.1 Hz, 2H), 7.08 (d,J= 4.2 Hz, 2H), 6.96—6.78 (m, 1H), 6.13 (t, J= 18 Hz, 1H), 5.75—5.57 (m, 1H), 4.51 (d, J: 11.6 Hz, 0.5H), 4.08 (d, J: 12 Hz, 0.5H), 3.93 (d, J: 13.2 Hz, 0.5H), 3.85— 3.73 (m, 0.8H), 3.34—3.20 (m, 2H), .97 (m, 0.7H), 2.27—2.09 (m, 1H), 2.07—1.64 (m, 2H), 1.60—1.43 (m, 1H). MS (ESI, method F): m/z = 418.1 [M + H]+, tR=1.679 (min). HPLC: 100% (214nm), 100% (254nm).
Scheme 45 C')H COOH COOEt Boc-N::>—Blo Cl HO‘B Cl / EtOH st04 Cl ‘o / 000 EtOOC N-Boc reflux gh' Pd(dppf)ClZ, chog, Pd2(dba)3, K3PO4, dioxane H20, 80°C dioxane H20,110°C COOEt COOEt COOH O/RPd/CH2 MeOHmO/Oi‘:NaOH, MeOH THF H20 CONHz CONH2 NH3 HATU DMF HCI/THF DIEA DCM Example 55 ethyl 4-br0mochlor0benzoate (2). To a mixture of 4-bromochlorobenzoic acid 1 (1.9 g, 8.1 mmol) in EtOH (50 mL) was added H2S04 (5 mL) dropwise carefully at 0 CC. The resulting mixture was refluxed overnight. The le phase d under reduced pressure. The residue was diluted with EA (100 mL), which was washed with water (2 X 50 mL) and brine, dried over sodium sulfate, filtered and concentrated. This resulted in the title compound (2.0 g, 95%) as brown oil. MS (ESI): m/Z = 263.0/2650 [M + H]+. tert-butyl 4-(3-chlor0(ethoxycarb0nyl)phenyl)-5,6-dihydr0pyridine-l(2H)—carb0xylate (3). A mixture of ethyl ochlorobenzoate 2 (0.53 g, 2.0 mmol), tert-butyl 4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolanyl)—5,6-dihydropyridine-1(2H)-carboxylate (0.74 g, 2.4 mmol), potassium carbonate (0.83 g, 6.0 mmol) and f)Cl2 (0.15 g, 0.2 mmol) in dioxane/H2O (20 mL/5 mL) was stirred at 80 °C for 4h under N2 atmosphere. The volatile phase was removed under reduced pressure. The residue was d onto silica gel column eluting with 6:1 PE/EA to get the title compound (0.7 g, 96%) as yellow oil. MS (ESI): m/z = 310.1 [M + H — 56]+. tert-butyl 4-(6-(eth0xycarb0nyl)—4'-phenoxybiphenylyl)-5,6-dihydr0pyridine—1(2H)- carboxylate (4). A e of tert-butyl 4-(3—chloro—4—(ethoxycarbonyl)phenyl)-5,6— dihydropyridine- 1(2H)-carboxylate 3 (200 mg, 0.66 mmol), 4-phenoxyphenylboronic acid (172 mg, 0.80 mmol), K3PO4 (414 mg, 2.0 mmol), tricyclohexylphosphine (40 mg, 0.14 mmol) and Pd2(dba)3 (64 mg, 0.07 mmol) in dioxane/H2O (15 mL/3 mL) was stirred at 110 CC overnight under N2 here. The volatile phase was removed under reduced pressure. The residue was applied onto silica gel column eluting with 6:1 PE/EA to get the title compound (200 mg, 61%) as yellow oil. MS (ESI): m/z = 500.0 [M + H]+. tert-butyl 4-(6-(eth0xycarb0nyl)—4'-phen0xybiphenylyl)piperidine—l-carboxylate (5). A mixture of 4-(6-(ethoxycarbonyl)-4'-phenoxybiphenylyl)—5,6-dihydropyridine-1(2H)- carboxylate 4 (180 mg, 0.36 mmol) and Pd/C (wet 10%, 18 mg) in MeOH (10 mL) was stirred at rt under H2 atmosphere for 5h. The solid was filtered off and the filtrate was concentrated under reduced pressure. This resulted in the title compound (180 mg, 99%) as colorless oil, which was used directly for the next step. MS (ESI): m/z = 524.0 [M + Na]+. -(1-(tert—butoxycarbonyl)piperidinyl)—4'-phenoxybiphenyl—Z-carboxylic acid (6). A mixture of tert-butyl 4-(6-(ethoxycarbonyl)-4'-phenoxybiphenylyl)piperidinecarboxylate 5 (180 mg, 0.36 mmol) and NaOH (72 mg, 1.8 mmol) in THF/MeOH/H2O (5 mL/5 mL/5 mL) was stirred at rt overnight. The le phase was removed under d pressure. The PH of which was adjusted to 3 by HCl (1 N), which was extracted with EA (2 X 30 mL). The organic layers were combined, washed with brine, dried over sodium e, filtered and concentrated. This resulted in the title compound (180 mg, overweight) in colorless oil, which was used directly for the next step. MS (ESI): m/Z = 374.1 [M + H - Boc]+. tert-butyl 4-(6-carbamoyl-4'-phenoxybiphenylyl)piperidine—l-carboxylate (7). The title compound was ed using a procedure analogous to the procedure bed in tert-butyl (4-carbamoyl(4-phenoxyphenyl)—1H-pyrazol-l-yl)pyrrolidine—1-carboxylate (see Scheme 23) as pale yellow solid (200 mg, overweight). MS (ESI): m/z = 416.8 [M + H - 56]+. tert-butyl 4-(6-carbam0yl-4'-phenoxybiphenylyl)piperidine—l-carboxylate (8). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- phenoxypheny1)(piperidiny1)—1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as white solid (200 mg, ight). MS (ESI): m/z = 373.1 [M + H]+.
Example 55 -(1-acryloylpiperidiny])—4'-phenoxybiphenyl-Z-carboxamide 4009 The title compound was obtained using a procedure analogous to the procedure described in Example 61 as white solid (70 mg, 20%). 1H NMR (300 MHZ, CDClg) 5 7.75 (d, J: 7.9 Hz, 1H), 7.44—7.32 (m, 4H), 7.23—7.28 (m, 1H), 7.20—7.12 (m, 2H), 7.11—6.99 (m, 4H), 6.62 (dd, J: 16.5 Hz, 10.5 Hz, 1H), 6.31 (dd, J: 16.5 Hz, 1.8 Hz, 1H), 5.73 (brs, 1H), 5.71 (dd,J= 10.5 Hz, 1.8 Hz, 1H), 5.35 (brs, 1H), 4.99—4.74 (m, 1H), 4.24—4.03 (m, 1H), 3.30—3.05 (m, 1H), 2.97— 2.58 (m, 2H), 2.04—1.88 (m, 2H), 1.83—1.58 (m, 2H). MS (ESI, method A): m/z = 427.1 [M + H]+, 63 min. HPLC: 99% (214nm), 99% (254nm).
Scheme 46 H N o 0 N _, N\ / \. 0%N w N 0 0v \ I H \ o 0 0H 8 O 9 0 o G o NH4C| HCI-EtOH —> —> Q AvofN _> :1 ‘IN m 37" HCI HN O NH2 0 ""2 MWN‘N I 2 0 1 1 12 Example 56 (4-phenoxyphenyl)hydrazine (2). To a solution of 4—phenoxybenzenamine 1 (1.0 g, 5.4 mmol) in HCl (100 mL) was added NaNOz (700 mg, 10.1 mmol) at 0°C. Then the mixture was stirred at 0°C for 1 h. A solution of SnClz (5.0 g, 22.1 mmol) in HCl (100 mL) was added to the mixture, and the resulting mixture was stirred at rt for 3 h. Aqueous NaOH (3N) was added to adjust pH to 10, and the mixture was extracted with EA (3 X 50 mL), the combined organic phase was washed with brine (20 mL), dried over NaZSO4. The solvent was d under reduced pressure to give a crude title compound (567 mg, 56%) as a yellow solid, which was used to next step without further purification. MS (ESI): m/z = 201.0 [M+H]+. ethyl 5-hydr0xy—1-(4-phen0xyphenyl)—1H-pyrazolecarb0xylate (4). To the solution of diethyl fumarate 3 (260mg, 1.5 mmol) in EtOH (25 mL) was added (4-phenoxyphenyl)hydrazine 2 (254 mg, 1.3 mmol), and the resulting solution was stirred overnight at 80°C. The solvent was evaporated under vacuum and the crude residue was diluted with water (30 mL). The solution was extracted with ethyl acetate (3 X 30 mL). The organic layers were ed, dried over ous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with 20:1 DCM/MeOH to afford the title nd (174 mg, 42%) as a yellow solid. MS (ESI): m/Z = 325.1 [M+H]+. ethyl 1-(4-phenoxyphenyl)—5—(trifluoromethylsulfonyloxy)—1H-pyrazole—3-carb0xylate (6).
To a solution of ethyl 5-hydroxy-l-(4-phenoxyphenyl)-1H-pyrazole—3-carboxylate 4 (170 mg, 0.5 mmol) in DCM (20 mL) was added trifluoromethanesulfonic anhydride 5 (295 mg, 1.0 mmol) and stirred for 1 h at -30°C under N2 atmosphere. The mixture was poured into water (60 mL), and the solution was ted with ethyl acetate (3 X 30 mL). The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with 5:1 PE/EA to afford the title compound (210 mg, 92%) as a yellow solid. MS (ESI): m/z = 457.0 [M+H]+. tert-butyl(3-(eth0xycarb0nyl)—1-(4-phen0xyphenyl)—1H—pyrazol-S-yl)—5,6- dihydropyridine— 1(2H)—carb0xylate (8). To a solution of ethyl 1-(4-phenoxyphenyl) (trifluoromethylsulfonyloxy)-1H-pyrazolecarboxylate 6 (210 mg, 0.5 mmol), K2C03 (552 mg, 4.0 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl— l ,3,2—dioxaborolan-2—yl)-5,6—dihydropyridine— 1(2H)—carboxylate 7 (201 mg, 0.6 mmol) in dioxane/H2O (10:1, 20 mL) was added Pd(dppf)Cl2 (327 mg, 0.2 mmol) under en atmosphere, and the e was degassed with nitrogen 6 times, then heated to 90°C and stirred overnight under nitrogen atmosphere. After cooling to room temperature, the solvent was evaporated and the crude product was purified by silica gel column chromatography eluting with 20:1 DCM/MeOH to afford the title compound (95 mg, 42%) as a yellow solid. MS (ESI): m/Z = 490.2[M+H]+. tert-butyl(3-(eth0xycarb0nyl)—1-(4-phen0xyphenyl)—1H—pyrazolyl)piperidine carboxylate (9). The mixture of tert—butyl 4—(3-(ethoxycarbonyl)—l—(4-phenoxyphenyl)-1H- pyrazol—5-yl)—5,6- dihydropyridine-l(2H)-carboxylate 8 (95 mg, 0.2 mmol), Pd/C (10% palladium on carbon, 56.5% water, 48 mg) and MeOH (20 mL) was degassed with H2 6 times and then d under H2 at rt for l h. Then the mixture was filtered and the filtrate was trated in vacuo to give the title compound (56 mg, 58%) as colorless oil. MS (ESI): m/z = 492.2 [M + Hr. -(1-(tert-but0xycarbonyl)piperidinyl)—1-(4-phen0xyphenyl)—lH-pyrazolecarb0xylic acid (10). To the solution of tert-butyl 4-(3-(ethoxycarbonyl)(4-phenoxyphenyl)—lH-pyrazol- -yl)piperidine-l-carboxylate 9 (166mg, 0.3 mmol) in THF/MeOH/H2O (20 : 20 : 10 mL) was added LiOH (60mg, 1.4 mmol), and the resulting solution was stirred for 15 h at ambient temperature. The e was concentrated, the residue was d with water (10 mL), and the solution was acidified with citric acid to pH = 4, ted with ethyl acetate (3 X 20 mL), washed with brine (20 mL), dried over , filtered and concentrated to get the title compound (120 mg, 76%) as a brown solid. MS (ESI): m/z = 464.2 [M + H]+ utyl(3-carbam0y](4-phenoxyphenyl)—1H—pyrazol-S-yl)piperidine—l-carboxylate (11). To a solution of 5-(1-(tert-butoxycarbonyl)piperidinyl)-l-(4-phenoxyphenyl)-1H- pyrazole ylic acid 10 (250 mg, 0.59 mmol) and HATU (147 mg, 0.38 mmol) in dry N,N—dimethyl formamide (25 mL) was added DIPEA (67 mg, 0.51 mmol), and the resulting solution was stirred for 20 min at ambient temperature, then NH4C1 (28 mg, 0.51 mmol) was added and stirred overnight. The mixture was diluted with ethyl acetate (50 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over Na2SO4, filtered and concentrated to get a residue which was purified by silica gel column chromatography eluting with 20:1 DCM/MeOH to afford the title compound (100 mg, 84%) as a yellow solid.
MS (ESI): m/Z = 463.2 [M+H]+. 1-(4-phenoxyphenyl)—5-(piperidinyl)—1H-pyrazole—3-carb0xamide hydrochloride (12). To a solution of tert-butyl 4-(3 -carbamoyl(4-phenoxyphenyl)-1H-pyrazol-5 -yl)piperidine carboxylate 11 (100mg, 0.21 mmol) in EtOH (20 mL) was added HCl/EtOH (33%, 2 mL) at ambient temperature. The mixture was stirred for 12 h at room ature. The mixture concentrated to give the title compound (70 mg, 89%) as yellow oil. MS (ESI): m/z =363.1 [M + Example 56 -(1-acryloylpiperidinyl)—1-(4-phen0xyphenyl)-lH-pyrazolecarboxamide _ <3; 7w \.~ To the solution of l-(4—phenoxyphenyl)—5-(piperidin—4-yl)-lH-pyrazolecarboxamide hloride 12 (110 mg, 0.3 mmol), DIPEA (117 mg, 0.9 mmol) and CH2C12 (10 mL) was added acryloyl chloride (28 mg, 0.3 mmol) dropwise and then the reaction mixture was stirred at 0°C for 1 h. The mixture was diluted with CHzClz (20 mL) and washed with saturated aqueous NaHC03 on (20 mL). The aqueous phase was reextracted with EtOAc (20 mL). The ed organic phase was dried over Na2S04, trated in vacuo and purified by Prep- TLC developing with 20:1 CH2C12/1V1eOH to give the title compound (66 mg, 52%) as yellow oil. 1H NMR (300 MHz, CD30D) 5 7.52—7.37 (m, 4H), 7.24—7.06 (m, 5H), 6.84—6.68 (m, 2H), 6.21 (d, J= 2.0 Hz, 0.5H), 6.15 (d, J= 2.0 Hz, 0.5H), 5.73 (dd, J= 10.6, 2.0 Hz, 1H), 4.61 (d, J = 12.8 Hz, 1H), 4.15 (d, J: 14.0 Hz, 1H), 3.04 (tt, .1: 8.0, 7.5 Hz, 2H), 2.77—2.59 (m, 1H), 2.03—1.81 (m, 2H), 1.72—1.46 (m, 2H). MS (ESI, method A): m/z = 417.1 [M + H]+, tR=1.547 min. HPLC: 98.2% (214nm), 98.8% (254nm).
Scheme 47 0 Br 0 Br 0 Br 0 Br r"\ 3°C N\_,NH3_ SOCI2 HO NH3, HATU H2N HQ —>o —> —> MeOH K2003 DMF N/fi DIPEA,DMF N F F K/N. Boo 5km.Bee 1 2 4 ch03 Pd(dppf)C|2 HCUEtOH dioxane/HZO 6DK/NBoc OK/NH TEA/DOM 7 ONO? Example 57 methyl 2-brom0flu0r0benz0ate (2). To a mixture of ofluorobenzoic acid 1 (2.19 g, mmol) in MeOH (20 mL) was added SOC12 (0.5 mL) dropwise at rt. The resulting mixture was stirred at 60°C for 10 h. The mixture was concentrated to give the title compound (2 g, 86%) as brown oil. MS (ESI): m/Z = 232.8 [M + H]+. 2-brom0(4—(tert—butoxycarbonyl)piperazinyl)benzoic acid (4). To a solution of methyl 2-bromofluorobenzoate 2 (1.8 g, 7.72 mmol) in DMSO (30 mL) was added tert-butyl piperazine-l-carboxylate 3 (1.726 g, 9.27 mmol) and K2C03 (4.26 g, 30.9 mmol). The e was stirred at 120°C for 5 h. When finished, the mixture was extracted with ethyl acetate (2 X 50 mL), and washed with water (50 mL) and brine (2 X 50 mL). The organic layer was dried over Na2S04, filtered and concentrated to get a residue which was purified by silica gel column chromatography eluting with 10:1 PE/EA to get the title compound (1.1 g, 36%) as a yellow oil.
MS (ESI): m/Z = 328.7 [M — 551+. tert-butyl 4-(3-br0m0carbamoylphenyl)piperazine-l-carboxylate (5). The title compound was obtained using a procedure ous to the procedure described in tert—butyl 3-(5- carbamoyl(4-phenoxyphenyl)thiazoly1)pyrrolidinecarboxylate (see Scheme 31) as a yellow solid (0.8 g, 73%). MS (ESI): m/Z = 327.7 [M — 551+. tert-butyl 4-(6-carbamoyl-4'-phenoxybiphenylyl)piperazine—l-carboxylate (6). To a mixture of tert-butyl 4-(3-bromocarbamoylphenyl)piperazinecarboxylate 5 (192 mg, 0.5 mmol), 4-phenoxyphenylboronic acid (102 mg, 0.5 mmol) and K2C03 (207 mg, 1.5 mmol) in dioxane/water (20/4 mL) were added Pd(dppf)2C12 (36.55 mg, 0.05 mmol). The e was degassed with N2 3 times, then heated to 100 CC and stirred for 16 h. After g to room ature, the mixture was added to ethyl acetate (30 mL), washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over NazSO4, filtered and concentrated to get a residue which was purified by silica gel column chromatography eluting with 30:1 DCM/MeOH to get the title compound (50 mg, 21%) as a yellow solid. MS (ESI): m/z = 473.8 [M + H]+ 4'-phenoxy-5—(piperazinyl)biphenylcarboxamide hydrochloride (7). The title compound was obtained using a procedure analogous to the procedure described in 1-(4- yphenyl)(piperidinyl)-1H-pyrazolecarboxamide hydrochloride (see Scheme 46) as a yellow solid (40 mg, 92%). MS (ESI): m/Z = 374.1[M + H.
Example 57 -(4-acryloylpiperazinyl)—4'-phenoxybiphenyl-Z-carboxamide The title compound was obtained using a procedure analogous to the procedure described in Example 1 as an off white solid (15 mg, 36%). 1H NMR (400 MHZ, DMSO) 5 7.52—7.30 (m, 6H), 7.25—6.93 (m, 7H), .82 (m, 2H), 6.15 (d,J= 16.7 Hz, 1H), 5.73 (d, J: 9.9 Hz, 1H), 3.70 (d, J: 12.0 Hz, 4H), 3.27 (s, 4H). MS (ESI, method A): m/z =428.1 [M + H]+, tR=1.525 min. HPLC: 96.4% (214nm), 99.3% (254nm) Scheme 48 NH2 \ Pd(dppf)C12,KZCO3 0 | \ Pd(dppf)Clz,K2CO3 \ O + I —>CI N/ —> CI N/ CI dioxane-Hzo (6/1 V) O O '3 dioxane—HZO (6/1 V) 90°C, 12 h 300 90 00, 12h 2 o 3 [foo O Pd/C H2 MeOH n Boo/N {5 040—,DCMCF3COOHrt \ O ' | / N/ N DCM 0°C 1h HN O O —>/\|OrN Example 58 2-(4-benz0ylphenyl)—6-chloronicotinamide (2). To a solution of 4-phenoxyphenylboronic acid (650 mg, 2.88 mmol), K2C03 (1.08 g, 7.86 mmol) and 2,6—dichloronicotinamide (500 mg, 2.62 mmol) in 1,4-dioxane (25 mL) and water (4 mL) was added Pd(dppf)Clg (192 mg, 0.262 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 60 CC and stirred for 12 h under nitrogen atmosphere. After cooling to room ature, the solution was poured into water (50 mL), and then extracted with ethyl acetate (2 X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated.
The crude t was purified by column tography, g with 80 : 1 dichloromethane/methanol to afford the title compound as a brown oil (868 mg, 98%). MS (ESI): m/Z = 336.9 [M + H]+. tert-butyl 4-(6-(4-benzoylphenyl)carbamoylpyridin-Z-yl)—5,6-dihydropyridine—1(2H)— carboxylate (4). To a solution of tert—butyl 4—(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl)— 5,6- dihydropyridine-1(2H)—carboxylate (1.2 g, 3.87 mmol), K2C03 (1.07 g, 7.74 mmol) and 2-(4- benzoylphenyl)—6-chloronicotinamide 2 (868 mg, 2.58 mmol) in 1,4—dioxane (24 mL) and water (3 mL) was added f)C12 (188 mg, 0.258 mmol) under nitrogen atmosphere, and the mixture was degassed with nitrogen 6 times, then heated to 90 CC and stirred for 12 h under nitrogen atmosphere. After cooling to room temperature, the solution was poured into water (50 mL), and then extracted with ethyl acetate (3 X 40 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and trated. The crude product was purified by column chromatography, eluting with 80 : l dichloromethane/methanol to afford the title compound as a brown oil (800 mg, 64%). MS (ESI): m/z = 484.0 [M + H]+. tert-butyl(5-carbamoyl—6—(4-(hydr0xy(phenyl)methyl)phenyl)pyridinyl)piperidine—1- ylate (5). To a solution of tert—butyl 4-(6-(4-benzoylphenyl)—5-carbamoylpyridinyl) - hydropyridine-1(2H)- carboxylate 4 (800 mg, 1.65 mmol) in MeOH (15 mL) was added Pd/C (150 mg) under en atmosphere, and the mixture was ed with hydrogen 6 times, then d for 16 h at ambient temperature under hydrogen atmosphere. The solution was filtered and the filtrate was evaporated to provide crude t as a white solid (500 mg, 62%).
MS (ESI): m/Z = 488.1 [M+H]+. 2-(4-(hydroxy(phenyl)methyl)phenyl)(piperidinyl)nicotinamide (6). To a solution of tert—butyl 4-(5-carbamoyl(4-(hydroxy(phenyl)methyl)phenyl)pyridinyl)piperidine- l - carboxylate 5 (500 mg, 1.03 mmol) in dry dichloromethane (5 mL) was added TFA (2 mL), and the resulting mixture was stirred for 3 h at ambient temperature. The solvent was removed and the residue was ioned between saturated aqueous sodium bicarbonate (30 mL) and ethyl acetate (20 mL). The organic phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated to afford the title compound as a white solid (350 mg, 88%). MS (ESI): m/z = 388.1 [M+H]+.
Example 58 6-(1-acryloylpiperidinyl)—2-(4-(hydroxy(phenyl)methyl)phenyl)nicotinamide \ O N O 0 0 OH To a solution of 2—(4-(hydroxy(phenyl)methyl)phenyl)-6—(piperidinyl) nicotinamide 6 (350 mg, 0.90 mmol) in DCM (10 mL) was added TEA (182 mg, 1.8 mmol) and acryloyl chloride (90 mg, 0.99 mmol) at 0 OC. The mixture was stirred at 0 °C for 10 min. The solvent was removed and the residue was purified by Prep-HPLC to afford the title compound (150 mg, 38%) as white solid. 1H NMR (300 MHz, CDClg) 8 8.03 (d, J: 7.9 Hz, 1H), 7.65 (d, J = 7.1 Hz, 2H), 7.56— 7.15 (m, 9H), 6.67—6.49 (m, 1H), 6.27 (d, J = 16.6 Hz, 1H), 5.88 (s, 1H), 5.68 (d, J: 10.1 Hz, 1H), 5.52 (d, .1: 27.1 Hz, 2H), 4.79—4.72 (m, 1H), 4.11—4.06 (m, 1H), 3.18—3.10 (m, 2H), 2.80— 2.74(m, 1H), 2.08—1.98(m, 2H), 1.78 (d, J: 10.5 Hz, 2H). MS (ESI, method F): m/Z = 441.8 [M + Hr, tR=1.268 min., HPLC: 100.0% (214nm), 100.0% (254nm).
Scheme 49 O0 O/\ O 0 O N N / ‘N'CN’KZN— / ‘N’CN :N \ \ o AcOH,45°C (>00 / NH2 NH2 Example 50 Example 59 Example 59 (1-(2-cyanobutenoyl)azetidinyl)—3-(4-phen0xyphenyl)—lH-pyrazole carboxamide : : N / \N’CN To a solution of 1-(1-(2-cyanoacetyl)azetidinyl)—3-(4-phenoxyphenyl)-1H-pyrazole amide (150 mg, 0.37 mmol) and dine (3 drops) in AcOH (10 mL) was added acetaldehyde (99 mg, 0.74 mmol), then stirred at 45°C overnight. After the reaction was completed, sat. NaOH was added to the solution to pH = 6—7. Then the solution was extracted with DCM (2 X 20 mL). The organic layer was combined and concentrated under vacuum to dryness. The residue was purified by Prep-HPLC (MeOH-HzO = 60-70, 0.1%FA) to afford the title compound (40 mg, 26%) as a white solid. 1H NMR (300 MHz, CDC13) 5 8.15 (S, 1H), 7.79— 7.68 (m, 1H), 7.62—7.54 (m, 2H), 7.43—7.34 (m, 2H), 7.21—7.13 (m, 1H), 7.13—7.04 (m, 4H), .62 (s, 2H), 5.26—5.10(m, 1H), 5.08—4.79 (m, 2H), 4.71—451 (m,2H),3.51—3.36(m,1H), 2.26—2.16 (m, 2H). MS (ESI, Method A): m/z= 428 1 [M + H], tR= 1 4.38 min HPLC: 98.4% (214nm), 98.4% (254nm).
Scheme 50 OH Cul 1 10--phenanthroline HCI (1 N) Br\©\ Q CSzCOg toluene 120°C 000OBTHF OOQ/OB + 0 o £0 1 2 3 4 DAST Pd(dppf)C|2 KOAc _>F?©/O Pd2(dba)3, 052003 DCM, rt 5Br—>\©\1,-4dioxane 100°C F70 08/0 dioxane/H20,1ZO°C 33F?#3" | 0 e ‘55 ’11* (‘N N C] O O HNQ \ NH2 \ I | NH2 FN , / N H2C|2,O°C FN N HNd O —C]> / Nd O /\n/ O (I? o E j F F F F ExampleGO 8-(4-br0m0phen0xy)—1,4-di0xaspir0[4.5]decane (3). The title compound was obtained using a ure ous to the procedure described in l-bromo(cyclohexyloxy)benzene (see Scheme 17) as colorless oil (0.272 g, 87%). 4-(4-bromophenoxy)cyclohexanone (4). A solution of 8-(4-bromophenoxy)— 1,4- dioxaspiro[4.5]decane 3 (0.272 g, 0.87 mmol) in THF (5 mL) was d with HCl (1 N, 5 mL) at 50 0C for l h. Then the mixture was neutralized with NaHCOg and extracted with EtOAc (5 mL X 3). The combined organic phase was dried over Na2S04, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with 5:1 petroleum EtOAc to give the title compound (147 mg, 63%) as white solid. 1H NMR (400 MHz, CDC13) 8 7.46—7.37 (m, 2H), 6.91—6.83 (m, 2H), 4.68 (s, 1H), 2.75—2.64 (m, 2H), 2.43—2.25 (m, 4H), 2.15—2.08 (s, 2H). 1-brom0((4,4-diflu0r0cyclohexyl)0xy)benzene (5). To the solution of 4-(4- bromophenoxy)cyclohexanone 4 (0.538 g, 2.0 mmol) in dry DCM (5 mL) was added a solution ofDAST (0.654 g, 4.0 mmol) in dry DCM (5 mL) at 0 CC and the resulting mixture was stirred at rt for 4 h. The reaction was quenched by ice water (5 mL), basified with saturated NaHC03 to pH = 8 and extracted with EtOAc (10 mL X 3). The combined organic phase was dried over Na2S04, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with 10:1 petroleum ether/EtOAc to afford the title compound (0.547 g, 94%) as yellow oil. 2-(4-((4,4-difluorocyclohexyl)oxy)phenyl)-4,4,5,5-tetramethyl—1,3,2-di0xabor0]ane (6). The mixture of 1-bromo((4,4-difluorocyclohexyl)oxy)benzene 5 (0.328 g, 1.13 mmol), bis(pinacolato)diboron (0.429 g, 1.69 mmol), Pd(dppf)C12 (0.168 g, 0.23 mmol), KOAc (0.323 g, 3.29 mmol) and dry dioxane (3 mL) was stirred at 100 CC for 3 h. After cooled to rt, the mixture was poured into water (10 mL) and extracted with EtOAc (15 mL X 3). The combined c phase was dried over Na2S04, filtered and concentrated. The residue was purified by silica gel column chromatography eluting with PE to afford the title compound (0.258 g, 68%) as a white solid. (4,4-diflu0r0cyclohexyl)0xy)phenyl)(piperazinyl)nic0tinamide (8). The title compound was obtained using a procedure analogous to the ure described in 6—(3- nitrophenyl)—2-(4-phenoxyphenyl)nicotinamide (see Scheme 1) as white solid (0.10 g, 31%). MS (ESI): m/Z = 417.1 [M + Hr.
Example 60 6-(4-acryloylpiperazinyl)(4-((4,4-difluorocyclohexyl)0xy)phenyl)nic0tinamide @115\ NH2 l F The title compound was obtained using a procedure analogous to the procedure described in e 1 as white solid (6 mg, 11%). 1H NMR (400 MHz, DMSO—d6) 5 7.88 (d, J: 8.7 Hz, 2H), 7.73 (d, J: 8.6 Hz, 2H), 7.68 (d, J: 8.7 Hz, 1H), 7.59 (s, 1H), 7.26 (s, 1H), 7.21—7.14 (m, 4H), 6.86 (dd, J: 17.8, 9.3 Hz, 2H), 6.16 (dd, J: 16.7, 2.2 Hz, 1H), 5.73 (dd, J: 10.4, 2.2 Hz, 1H), 3.66—3.67 (m, 8H). MS (ESI, method A): m/z = 471.2 [M + H]+, tR=1.753 (min). HPLC: 99.2% (214nm), 99.2% (254nm).
Scheme 51 cN NH2 \ conc)/H20 NNWZZES | —> —3> Cl N Cl 9000 m 73/ C, N/ [iiB-CN-Boo Cl Pd(dppf)Cl2 0520030 Pd(dppf)C|2 eszco3 DME/H20 90°C overnight O,DMEIH2090°C overnightB O—NHZ Pd/C/ H2/MeOH OLiOHfr—>HF/H2O ONHZQ rt. 4 days OH—>HATUDIEADMF BocN 5000 4 h BocN | NH2 TFA!DCM N/ HN NH M o HfN Example 61 2,6-dichloronicotinamide (2). The title compound was synthesized using a procedure analogous to the procedure described in 2,6-dichloronicotinamide (see Scheme 51) (9.5 g, 81%) as a brown solid. MS (ESI): m/z = 191.0 [M + Hr. methyl arbam0ylchloropyridin-Z-yl)benz0ate (4). To a solution of 2,6- dichloronicotinamide 2 (1.91 g, 10.0 mmol), 4-(methoxycarbonyl)phenylboronic acid 3 (1.8 g, .0 mmol) and Pd(dppf)Cl2 (816 mg, 1.0 mmol) in DME/H2O (20 mL/2mL) was added CS2C03 (6.5 g, 20.0 mmol). The resulting solution was degassed with N2 6 times and stirred ght at 90 0C under N2 tion. After the reaction was completed, the solution was concentrated, diluted with ethyl acetate (30 mL) and washed with water (2 X 20 mL) and brine (2 X 20 mL).
The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel column diluted with 100:1 to 60:1 DCM/MeOH to afford the title compound (1 .95g, 67%) as a white solid. MS (ESI): m/z = 291.1 [M + H]+. tert-butyl(5-carbam0y]-6—(4-(methoxycarbonyl)phenyl)pyridinyl)-5,6- dihydropyridine— 1(2H)-carboxylate (6). To a solution of methyl 4-(3-carbamoyl chloropyridin—2-yl)benzoate 4 (170 mg, 0.59 mmol), tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2— dioxaborolan—2-y1)-5,6-dihydropyridine-1(2H)- carboxylate 5 (200 mg, 0.65 mmol) and f)Cl2 (50 mg, 0.06 mmol) in DME/H2O (20 mL/2mL) was added Cs2C03 (390 mg, 1.2 mmol), the resulting solution was degassed with N2 6 times and stirred overnight at 90 0C under N2. After the reaction was completed, the on was concentrated, diluted with ethyl acetate (30 mL) and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and trated. The residue was purified by silica gel column diluted with 50:1 to 10:1 DCM/MeOH to afford the title compound (145 mg, 56%) as brown oil. MS (ESI): m/Z = 438.2 [M + Hr. utyl(5-carbam0yl—6—(4-(methoxycarbonyl)phenyl)pyridinyl)piperidine carboxylate (7). To a solution of tert-butyl 4-(5-carbamoy1(4- (methoxycarbonyl)phenyl)pyridinyl)-5,6- dihydropyridine-1(2H)—carboxylate 6 (145 mg, 0.33 mmol) in MeOH (10 mL) was added Pd/C (15 mg) and degassed with H2 6 times and stirred overnight at rt under H2. After the on was completed, the solution was filtered and the filtrate was concentrated to afford the title compound (100 mg, 69%) as brown oil. MS (ESI): m/Z = 440.2 [M + H]+. 4-(6-(1-(tert-but0xycarb0nyl)piperidinyl)—3-carbamoylpyridin-Z-yl)benzoic acid (8) To a solution of tert—butyl 4—(5-carbamoyl—6—(4-(methoxycarbonyl)phenyl)pyridin—2-yl)piperidine— 1- ylate 7 (460 mg, 1.05 mmol) in THF/HZO (20 mL/2 mL) was added LiOH (84 mg, 2.1 mmol), the resulting solution was stirred overnight at rt. After the reaction was complete, the solution was trated, added water (10 mL) and acidified with HCl (00110., 1 mL) to pH = 5- 6, diluted with ethyl acetate (30 mL) and washed with water (2 X 20 mL) and brine (2 X 20 mL).
The c layer was dried over anhydrous sodium sulfate, d and concentrated to afford the title nd (390 mg, 87%) as a yellow solid. MS (ESI): m/z = 426.2 [M + H]+. tert-butyl(5-carbamoyl(4-(phenylcarbamoyl)phenyl)pyridinyl)piperidine—1- carboxylate (9). To a solution of 4-(6-(1-(tert-butoxycarbonyl)piperidinyl)—3- carbamoylpyridin-Z-yl)benzoic acid 8 (390 mg, 0.917 mmol), aniline (102 mg, 1.101 mmol) and HATU (418 mg, 1.101 mmol) in dry DMF (10 mL) was added DIEA (355 mg, 2.751 mmol), the resulting solution was stirred overnight at rt. The mixture was diluted with ethyl acetate (30 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by Prep-TLC with :1 DCM/MeOH to get the title compound (120 mg, 27%) as a yellow solid. MS (ESI): m/z = 501.2 [M + Hr. 2-(4-(phenylcarbamoyl)phenyl)—6-(piperidinyl)nic0tinamide (10) To a solution of tert— butyl 4-(5 -carbamoyl(4-(phenylcarbamoyl)phenyl)pyridinyl)piperidinecarboxylate 9 (120 mg, 0.24 mmol) in dry DCM (10 mL) was added TFA (3 mL), the resulting solution was stirred overnight at rt. The mixture was washed with NaHC03/H20 (3 X 20 mL) and brine (3 X mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated.
The residue was concentrated to get the title compound (100 mg, 99%) as a yellow solid. MS (ESI): m/Z = 401.2 [M + Hr.
Example 61 6-(1-acryloylpiperidinyl)—2-(4-(phenylcarbamoyl)phenyl)nicotinamide 0 HNO To a solution of 2-(4-(phenylcarbamoyl)phenyl)(piperidinyl)nicotinamide 10 (100 mg, 0.24 mmol) in dry DCM (15 mL) was added DIEA (95 mg, 0.72 mmol) and acryloyl de (35 mg, 0.36 mmol) at 0 CC, and the resulting solution was stirred at 0 CC for 10 min. Water (10 mL) was added to quench the reaction. The e was diluted with DCM (20 mL), and washed with water (2 X 20 mL) and brine (2 X 20 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by Prep-HPLC (ACN—HzO = 30-90, 0.1%FA) to afford the title compound (31 mg, 29%) as a white solid. 1H NMR (300 MHz, CDC13) 8 8.59 (s, 1H), 8.04—7.58 (m, 3H), 7.75—7.54 (m, 4H), 7.37—7.27 (m, 2H), .08 (m, 2H), 6.65—6.49 (m, 1H), .10 (m, 3H), .63 (m, 1H), 4.79—4.63 (m, 1H), 4.17—3.99 (m, 1H), 3.27—2.96 (m, 2H), 2.83—2.66 (m, 1H), 2.07—1.88 (m, 2H), 1.83—1.60 (m, 2H). MS (ESI): m/z = 455.2 [M + H]+, tR= 1.316 min. HPLC: 95.3% (214nm), 93.0% (254nm).
Scheme 52 o o | NH2 0 Et3N \ NH2 + \\ /CI | ‘/\N N/ C|/\/3\\ / HN\/l 0 CHZCIZJt, 1 h 0" NJ 0 VS\\ 0 1 2 0 Example 62 Example 62 2-(4-phenoxypheny])—6-(4-(vinylsu]f0nyl)piperazinyl)nic0tinamide ' NH2 a O 0 \A’ o To a solution of 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide 1 (200 mg, 0.53 mmol) in DCM (8 mL) was added TEA (161 mg, 1.59 mmol) and 2-chloroethanesulfonyl chloride (131 mg, 0.80 mmol) at rt. The mixture was stirred at rt for 1 h. The solution was poured into water (50 mL), and then extracted with CHzClz (2 X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate, d and concentrated, and the residue was purified by Prep- TLC eluting with 100 : 1 OH to afford the title compound (17 mg, 6.9%) as a white solid. 1H NMR (300 MHz, CDC13)8 8.01 (d, J: 8.8 Hz, 1H), 7.59 (d, J: 8.6 Hz, 2H), 7.43— 7.30 (m, 2H), 7.15 (t, J: 7.4 Hz, 1H), 7.09—7.01(m, 4H), 6.64 (d, J: 8.8 Hz, 1H), 6.42 (dd, J: 16.6, 9.7 Hz, 1H), 6.26 (d, J= 16.5 Hz, 1H), 6.06 (d, J: 9.7 Hz, 1H), 5.46 (br, 1H), 5.28 (br, 1H), 3.87—3.75 (m, 4H), 3.32—3.18 (m, 4H). MS (ESI, method F): m/z = 465.1.0 [M + H]+, tR=1.546 min., HPLC: 99.5% (214nm), 98.3% (254nm).
Scheme 53 I NH2 \ W200 CIA/SO / | Q, Cl Et3N N / o \NCHZCIZ rt1 h O 1 Example 63 Example 63 2-(4-phen0xyphenyl)(1-(vinylsu]f0nyl)piperidinyl)nic0tinamide | NH2 V8" 0 To a solution of 2-(4-phenoxyphenyl)—6-(piperidin-4—yl)nicotinamide 1 (200 mg, 0.54 mmol) in DCM (8 mL) was added TEA (164 mg, 1.62 mmol) and 2-chloroethanesulfonyl chloride (131 mg, 0.80 mmol) at It. The mixture was stirred at rt for 1 h. The solution was poured into water (50 mL), and then extracted with ethyl acetate (2 X 50 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was purified by Prep-HPLC to afford the title nd (40 mg, 16%) as a white solid. 1H NMR (300 MHz, CDC13) 5 8.03 (d, J= 8.0 Hz, 1H), 7.71—7.61 (m, 2H), 7.43—7.31 (m, 2H), 7.17 (dd, J= 15.6, 7.8 Hz, 2H), 7.10—7.01(m, 4H), 6.46 (dd, J: 16.6, 9.8 Hz, 1H), 6.26 (d, J: 16.6 Hz, 1H), 6.04 (d, J = 9.9 Hz, 1H), 5.60 (b, 1H), 5.41 (b, 1H), 3.94—3.82(m, 2H), 2.97—2.86(m, 1H), 2.80—2.71(m, 2H), 2.11—1.92(m, 4H). MS (ESI, method F): m/Z = 464.0 [M + H]+, tR=1.566 min, HPLC: 99.3% (214nm), 100.0% (254nm).
The following examples can be synthesized ing to methods nced below and utilizing ordinary skill in the art. These examples are believed to be useful as inhibitors of BTK based on the biological activities of the nds described above.
Example 64 1-(1-acryloylpiperidinyl)—3-(4-(phenylcarbamoyl)phenyl)—1H-pyrazolecarb0xamide The title compound may be obtained using a procedure ous to the procedures described in the General Scheme and Example 15.
Example 65 1-(1-acryloylpiperidinyl)—3-(4-((4-(triflu0r0methyl)pyridinyl)carbamoyl)phenyl)—1H- pyrazolecarb0xamide The title compound may be obtained using a procedure analogous to the procedures described in the General Scheme and Example 15.
Example 66 6-(4-acrylamidophenyl)—2-(4-(phenylcarbamoyl)phenyl) nicotinamide The title compound may be ed using a procedure analogous to the procedures described in the General Scheme and Example 16. e 67 6-(4-acrylamid0phenyl)—2-(4-((4-(triflu0r0methyl)pyridinyl)carbamoyl)phenyl) nicotinamide The title compound may be obtained using a procedure analogous to the procedures described in the l Scheme and Example 16.
Example 68 1-(1-acryloylpiperidinyl)(4-(hydroxy(phenyl)methyl)phenyl)—1H-pyrazole—4- amide The title compound may be obtained using a procedure analogous to the procedures described in the General Scheme and Examples 15 and 58.
Example 69 1-(1-acryloylpiperidinyl)(4-(methyl(phenyl)amino)phenyl)—1H—pyrazole-4— carboxamide The title compound may be obtained using a procedure analogous to the procedures bed in the General Scheme and Example 15.
Example 70 6-(4-acrylamidophenyl)—2-(4-(1-hydr0xyphenylethyl)phenyl)nic0tinamide \. .' The title compound may be obtained using a procedure analogous to the procedures described in the General Scheme and Examples 16 and 58. e 71 6-(4-acrylamidophenyl)—2-(4-(difluor0(phenyl)methyl)phenyl)nicotinamide . 1‘ 1’? l The title compound may be obtained using a procedure analogous to the procedures described in the General Scheme and e 16.
Example 72 1-(1-acryloylpiperidinyl)(4-(phenylsulf0nyl)phenyl)—1H-pyrazolecarboxamide \\ // G o I The title compound may be ed using a procedure analogous to the procedures described in the General Scheme and Example 15.
Example 73 6-(4-acrylamidophenyl)—2-(4-(phenylsulfonyl)phenyl) nicotinamide The title compound may be obtained using a procedure analogous to the procedures described in the General Scheme and Example 16.
Scheme A-l o o HN’N PhO N/ EtOH COOEt C52C03, DMF ' COOEt 1 2 '-'OH H20 N»N NH3, HATU H2N ' / —> THF/MeOH/HZO / DIPEA DMF EN ’ N COOH (5 Analog 1 ethyl 5-(4-phen0xyphenyl)-1H-pyrazolecarb0xylate (2). The title compound was ed using a ure analogous to the procedure described in ethyl 3-(4-phenoxyphenyl)-lH- lecarboxylate (see Scheme 23) as yellow oil (0.3 g, 97%). MS (ESI): m/z = 308.8 [M + ethyl l-cyclopentyl—3-(4-phenoxyphenyl)-lH—pyrazolecarb0xylate (4). To a solution of ethyl 5—(4-phenoxyphenyl)-lH-pyrazolecarboxylate 2 (150 mg, 0.486 mmol) in DMF (10 mL) was added bromocyclopentane 3 (87 mg, 0.584 mmol) and C82C03 (475.5 mg, 1.46 mmol). The mixture was stirred at 100 °C for 3 h. After finished, the mixture was extracted with ethyl acetate (2 X 30 mL), and washed with water (2 X 10 mL) and brine (3 X 10 mL). The organic layer was dried over NaZSO4, d and concentrated to give the title compound (0.13 g, 71%) as yellow oil. MS (ESI): m/Z = 376.9 [M + H]+ 1-cyclopentyl(4-phenoxyphenyl)—1H-pyrazole-4—carboxylic acid (5). The title compound was obtained using a procedure analogous to the procedure described in 5-(1-(tert- butoxycarbonyl)piperidinyl)(4-phenoxyphenyl)—lH-pyrazole—3-carboxylic acid (see Scheme 46) as yellow oil (0.12 g, 100%). MS (ESI): m/z = 348.8 [M + H]+.
Analog 1 1-cyclopentyl(4-phen0xyphenyl)—1H-pyrazole—4—carboxamide The title nd was ed using a procedure analogous to the procedure described in tert- butyl 3-(5 -carbamoyl—4-(4-phenoxyphenyl)thiazo1-2—yl)pyrrolidine— l -carboxylate (see Scheme 31) as an offwhite solid (70 mg, 59%). 1H NMR (300 MHz, DMSO) 5 8.23 (s, 1H), 7.79 (d, J: 8.5 Hz, 2H), 7.41 (t, J: 7.0 Hz, 3H), 7.15 (t, J: 7.1 Hz, 1H), 7.05 (d, J: 7.6 Hz, 2H), 7.00 (s, 2H), 6.98 (d, J: 1.4 Hz, 1H), 4.78—4.62 (m, 1H), 2.19—1.55 (m, 8H). MS (ESI, method F): m/z = 347.9 [M + H]+, tR=1.589 (min). HPLC: 97.6% (214nm), 98.0% (254nm).
Scheme A—Z HO H N 0 5O 0 HZN S JL 1) (00002 Lawesson’s reagent QC : :Br 0 0E! —> —> 2) NH4OH toluene, reflux, 2h EtOH, reflux 1 2 3 CG OQ oJO o O NaOH, MeOH, H20 / NH3, HATU, DMF EtO —’ / N H0 / ’ HZN N 4 5 Analog 2 cyclopentanecarboxamide (2). To a e of cyclopentanecarboxylic acid 1 (2.85 g, 25 mmol) and DMF (3 drops) in DCM (60 mL) was added (COCl)2 (3.2 g, 25 mmol) dropwise carefully at 0 °C under N2. The resulting mixture was stirred at rt for 2h, then NH4OH (5 mL) was added se to this mixture at 0 0C. After the addition, the mixture was stirred at rt for another 1h, which was diluted with DCM (50 mL). It was washed with water (50 mL), brine, dried over sodium sulfate, ed and concentrated. This resulted in the title compound (1.8 g, 64%) as white solid. MS (ESI): m/Z = 114.8 [M + H]+. cyclopentanecarbothioamide (3). A mixture of cyclopentanecarboxamide 2 (1.8 g, 15.9 mmol) and lawesson’s reagent (3.2 g, 8.0 mmol) in toluene (40 mL) was stirred at 80 0C for 2h under N2 atmosphere. It was quenched with sat. NaHC03 (50 mL). which was extracted with EA (2 X 40 mL). The c layers were combined, washed with brine, dried over sodium sulfate, filtered and trated. The residue was applied onto silica gel column eluting with 3:1 PE/EA to get the title compound (0.25 g, 12%) as yellow solid. MS (ESI): m/z = 130.1 [M + H - 56]+. ethyl 2-cyclopentyl(4-phen0xyphenyl)thiazole—5-carboxylate (4). A mixture of cyclopentanecarbothioamide 3 (150 mg, 1.16 mmol) and ethyl 2-bromooxo(4— phenoxyphenyl)propanoate (422 mg, 1.16 mmol) in EtOH (15 mL) was refluxed for 2 h. The volatile phase was removed under reduced pressure. The residue was ved in EA (50 mL), which was washed with sat. NaHC03 (30 mL), brine, dried over sodium sulfate, filtered and concentrated. The residue was applied onto silica gel column eluting with 4:1 PE/EA to get the title compound (200 mg, 43%) as pale yellow oil. MS (ESI): m/z = 394.1 [M + H]+. 2-cyclopentyl(4-phenoxyphenyl)thiazole-S-carboxylic acid (5). The title compound was obtained using a procedure analogous to the procedure described in 5-(1-(tert- butoxycarbonyl)piperidinyl)-4'-phenoxybipheny1carboxy1ic acid (see Scheme 45) as pale yellow solid (190 mg, 99%). MS (ESI): m/Z = 366.1 [M + H.
Analog 2 2-cyclopentyl—4-(4-phenoxyphenyl)thiazole—5-carboxamide CW 0&3 The title compound was obtained using a procedure analogous to the procedure described in tert- butyl 4-(6-carbamoyl-4'-phenoxybiphenylyl)piperidinecarboxylate (see Scheme 45) as white solid (70 mg, 38%). 1H NMR (300 MHz, CDC13) 8 7.66—7.56 (m, 2H), 7.43—7.33 (m, 2H), 7.17 (t, J: 7.4 Hz, 1H), .00 (m, 4H), 5.73 (brs, 2H), 3.52—3.35 (m, 1H), 2.32—2.16 (m, 2H), 1.90—1.77 (m, 4H), 1.76—1.66 (m, 2H). MS (ESI, method A): m/z = 365.1 [M + H]+, tR=1.780 min. HPLC: 97% (214nm), 96% ).
Analflical conditions Unless otherwise noted, all ts, chemicals, and reagents were obtained commercially and used without purification. The lH-NMR spectra were obtained in CDClg, DMSO-d6, CD301), or acetone-d6 at 25°C at 300 MHz or 400 MHz on an OXFORD (Varian) with chemical shift (5, ppm) reported relative to TMS as an internal standard. HPLC-MS chromatograms and spectra were obtained with an Agilent 1200-6110 . Prep-HPLC instruments were Gilson GX—281 (Gilson) and P230 Preparative Gradient System ent: 95% water, 5% acetonitrile, 30-50 min gradient to 25% water, 75% acetonitrile). The microwave instrument was a CEM Discover Biological ties: IC50 ination OfBTK Inhibitors In ADP-Glo Kinase Biochemical Assay The activity of the Examples and Analogs described herein, as inhibitors of BTK are demonstrated and confirmed by pharmacological in vitro assays. Activity possessed by the compounds may be demonstrated in vivo. Those skilled in the art will appreciate that a variety of assay formats may be used to ine the activity of the compounds described herein.
Materials: ADP-GloTM Kinase Assay (cat. V9102, 10000 tests), components: 0 1><50ml oTM Reagent, o 1><100ml Kinase Detection Buffer, o 1><5ml Ultra Pure ATP, 10mM o 1><5ml ADP, 10mM Reagents and Plate: Tris.Hcl (Sigma cat. 154563), MgClz (Sigma cat.M1028), MnClz (Sigma, M3634), BSA (Sigma cat.05470), BTK Substrate (Signalchem, P61-58), DTT (Sigma, D0632), DMSO (Sigma, S5879), BTK . (1 .5mg/ml, purity 75%, 90ng/ul, made in house). 384 well assay plate (cat. 3674).
Assay conditions: 0 Enzyme concentration: l. 0 ATP concentration: 50uM o Substrate (peptide) concentration: 0.2mg/ml. 0 on buffer composition: 40mM Tris-HCl pH7.5, lOmM MgClg, 2mM MnClz, 0.1mg/mL BSA, 0.05mM DTT. 0 Test compound concentration: DMSOSO.5%.
Methods: Compound Dosage Gradient Solution Preparation: A 3-fold serial dilution of test compound was made for 10 gradient points (100, 33.33, 11.11, 3.70, 1.23, 0.41, 0.14, 0.046, 0.015, 0.005uM) in 100% DMSO. The intermediate dilution was done by adding 2ul of diluted compound into 78ul of assay buffer (containing 40mM Tris.Hcl, pH 7.5 10mM MgClz, 2mM MnCl 0.05mM DTT), making the final , 0.1mg/mL BSA, compound concentration (1000, 333.33, 111.11, 37.04, 12.35, 4.12, 1.37, 0.46, 0.15, 0.05nM) and DMSO concentration 0.5% percentage.
ADP-Glo Kinase Assay Protocol For BTK Inhibitors Testing: 1x and 2x assay buffer were made at first. BTK kinase was diluted with 1x assay buffer but substrate was diluted with 2x assay buffer. 1ul of diluted compound was transferred into 384- well assay plate, and then 2.0ul of enzyme solution was added, and spun at 2000rpm for 1min.
This mixture was incubated at 24°C for . 2ul of peptide ate/ATP mixture was added into the assay plate to start the on. The mixture was mixed thoroughly and then the 384- well plate was spun and ted at 24°C for 60mins. 5.0ul of ADP—Glo Reagent was added to stop the kinase activity and deplete the ATP unconsumed, and the plate was mixed thoroughly and incubated at 24°C for 40min. Then, 10.0ul of Kinase Detection reagent was added, and the plate was centrifuged and then kept at 24°C for 30min. The luminescence signal was read on Envision.
Data Analysis: % tion of compounds at each different concentration is calculated from Equation (1): % inhibition = 100-100*(Signal-low control)/(High control—low control). . .. Equation(1) IC50 values of compounds were calculated from a 4-parameter fit using Equation (2): Y=Bottom + (TOP-Bottom)/(1+((C/X)Ahillslope)) .............. Equation (2) In Equation 2, Y represents % inhibition, X is the log value of the test compound concentration. IC50 was the concentration of compound where half of maximal tion was achieved.
All data was analyzed with IDBS XLfit5 re (ID Business Solutions Ltd., UK). The IC50 data is ized in Table 1.
Table 1: IC50 BTK Activity Values For Examples 1-63 2, 4, 14, 27, 30, 33, 50, 53, 56 9, 24, 31 1, 3, 5, 6, 7, 8,10,11,12,13,15,16,17,18,19, 20, 21, 22, 23, 25, 26, 28, 29, 32, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 51, 52, 54, 55, 57, 58, 59, 60, 61, 62, 63 Table 2 provides a comparison of Examples 3 and 5 to Analogs 1 and 2 in terms of in Vitro BTK potency. Examples 3 and 5 demonstrate about a 100x improvement in potency over Analogs 1 and 2 when compared in head-to-head assays.
Table 2: Comparison of Examples 3 and 5 to Analogs 1 and 2 Example 3 Example 5 Analog 1 Analog 2 347.4 364.5 Examples 3 and 5 also demonstrate from 10x — 100x differences in potency between BTK and Src and are thus ive for BTK over Src.
Furthermore, selectivity of Examples 3 and 5 was determined. Table 3 provides in Vitro mical data demonstrating that Examples 3 and 5 are selective for BTK over Src.
Table 3: Selectivity Data for Examples 3 and 5 Target Example 3 Example 5 % Inhibition @ % Inhibition @ 1000 nM 1000 nM BT 99.35 99.7 SRC 23 33 Fold >100x >100x Difference To obtain the data provided in Table 3, -tagged T7 phage strains were grown in parallel in 24—well blocks in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage from a frozen stock (multiplicity of infection = 0.4) and incubated with shaking at 32°C until lysis (90-150 minutes). The lysates were fuged (6,000 x g) and filtered (0.2um) to remove cell debris. The remaining kinases were produced in HEK- 293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase . The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1 % BSA, 0.05 % Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific phage binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1x g buffer (20 % SeaBlock, 0.17x PBS, 0.05 % Tween 20, 6 mM DTT).
Example compounds were prepared as 40x stocks in 100% DMSO and directly d into the assay. All ons were performed in polypropylene 384-well plates in a final volume of 0.04 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05 % Tween 20). The beads were then re-suspended in n buffer (lx PBS, 0.05 % Tween 20, 0.5 uM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
The nd(s) were screened at 1000 nM, and results for y screen binding interactions are provided in Table 3 as "% Ctrl", where lower numbers indicate stronger hits. The % Ctrl is calculated according to Equation (1), below: Where: test compound = Example negative control = DMSO (100 % Ctrl) positive control = control compound ( 0% Ctrl) Additional ical selectivity data for Examples 3 and 5 against each member of the SRC family of protein kinases as well as EGFR are provided in Table 4, below. The data in Table 4 was obtained using the same proecedure as described above with respect to the data in Table 3.
Table 4: ivity Data for Examples 3 and 5 Example 3 Example 5 - %Inhibition @ 1000 nM % Inhibition @ 1000 nM --——_ -__— -__— Compositions: The present invention includes pharmaceutical compositions comprising a compound or pharmaceutically acceptable salt thereof of the invention, which is formulated for a desired mode of administration with or t one or more pharmaceutically acceptable and useful carriers.
The compounds can also be included in ceutical compositions in combination with one or more other therapeutically active compounds.
The pharmaceutical compositions of the invention comprise a compound of the invention (or a pharmaceutically acceptable salt thereof) as an active ingredient, optional pharmaceutically acceptable carrier(s) and optionally other therapeutic ingredients or nts. The compositions include itions suitable for oral, rectal, topical, and parenteral ding subcutaneous, intramuscular, and enous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being stered. The pharmaceutical itions can be conveniently ted in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
Compounds of the invention can be combined as the active ingredient in te admixture with a pharmaceutical carrier ing to conventional pharmaceutical compounding techniques. The carrier can take a wide y of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). Thus, the pharmaceutical compositions of the invention can be presented as discrete units suitable for oral administration such as capsules, s or tablets each containing a predetermined amount of the active ingredient. r, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non-aqueous , as an oil-in-water emulsion, or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compound represented by Formula I, or a ceutically acceptable salt thereof, can also be administered by controlled release means and/or delivery s. The compositions can be prepared by any of the methods of pharmacy. In general, such methods include a step of ng into association the active ingredient with the carrier that constitutes one or more ary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.
The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas.
Examples of solid rs include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. es of gaseous carriers include carbon dioxide and nitrogen.
A tablet containing the composition of this invention can be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. ssed tablets can be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably ns from about 0.05 mg to about 5 g of the active ingredient and each cachet or capsule preferably containing from about 0.05 mg to about 5 g of the active ingredient.
A formulation intended for the oral administration to humans may contain from about 0.5 mg to about 5 g of active agent, compounded with an riate and convenient amount of carrier material which may vary from about 5 to about 95 t of the total composition. Unit dosage forms will generally contain between from about 1 mg to about 2 g of the active ingredient, typically 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 1000 mg.
Compounds of the invention can be provided for formulation at high purity, for example at least about 90%, 95%, or 98% pure by weight.
Pharmaceutical compositions of the invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid hylene glycols, and mixtures thereof in oils. r, a preservative can be included to prevent the detrimental growth of microorganisms.
Pharmaceutical itions of the invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous ation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of cture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), ble oils, and suitable mixtures thereof. ceutical compositions of the invention can be in a form suitable for topical use such as, for example, an l, cream, ointment, lotion, dusting powder, or the like. Further, the itions can be in a form suitable for use in transdermal s. These formulations may be prepared, utilizing a compound represented by Formula I of this invention, or a pharmaceutically acceptable salt thereof, via conventional processing methods. As an example, a cream or ointment is prepared by admixing hydrophilic material and water, together with about 5 wt % to about 10 wt % of the compound, to produce a cream or ointment having a desired consistency. ceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier(s) followed by chilling and shaping in molds.
In addition to the aforementioned carrier ients, the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as ts, s, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended ent.
Compositions containing a compound described by Formula I, or pharmaceutically acceptable salts f, may also be ed in powder or liquid concentrate form.
Compounds of the present ion inhibit the activity of BTK in animals, including humans, and are useful in the ent and/or prevention ous es and conditions such as cancer, inflammation, fibrotic diseases, and autoimmune disease which are caused, mediated and/or propagated by BTK. In particular, nds of the invention, and compositions thereof, are inhibitors of BTK, and are useful in treating conditions ted, at least in part, by BTK.
In some aspects, the invention includes a method of treating cancer comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating a cancer mediated at least in part by BTK comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention includes a method of treating or a method of manufacturing a medicament for treating a cancer, such as those described herein, which is mediated at least in part by BTK, comprising administering to a mammal in need thereof a therapeutically effective amount of a nd or salt of the invention.
In some aspects, the invention includes a method of treating lymphocyte homing and inflammation mediated at least in part by BTK comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention includes a method of treating or a method of manufacturing a ment for treating lymphocyte homing and inflammation, such as those described herein, which is ed at least in part by BTK, comprising administering to a mammal in need thereof a therapeutically ive amount of a nd or salt of the invention.
In some aspects, the invention includes a method of treating neuropathic pain comprising administering to a mammal in need f a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating neuropathic pain mediated at least in part by BTK comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention includes a method of treating or a method of manufacturing a medicament for ng athic pain, such as those described herein, which is ed at least in part by BTK, comprising administering to a mammal in need thereof a therapeutically effective amount of a nd or salt of the invention.
In some aspects, the invention includes a method of treating f1brotic diseases comprising administering to a mammal in need thereof a eutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating fibrotic es mediated at least in part by BTK comprising stering to a mammal in need thereof a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention includes a method of treating or a method of manufacturing a medicament for treating a fibrotic disease, such as those described herein, which is mediated at least in part by BTK, comprising stering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating thrombosis comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating thrombosis mediated at least in part by BTK comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention includes a method of treating or a method of manufacturing a ment for treating thrombosis, such as those described herein, which is ed at least in part by BTK, comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the ion includes a method of treating cholestatic pruritus comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
In some aspects, the invention includes a method of treating cholestatic pruritus mediated at least in part by BTK comprising administering to a mammal in need f a therapeutically effective amount of a compound or salt of Formula I.
In some aspects, the invention es a method of treating or a method of manufacturing a medicament for cholestatic pruritus, such as those described herein, which is mediated at least in part by BTK, comprising stering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention.
The compounds of Formula I of the invention are usefial in the treatment of a variety of cancers, including, but not limited to, solid tumors, sarcoma, fibrosarcoma, osteoma, melanoma, blastoma, myosarcoma, glioblastoma, neuroblastoma, teratocarcinoma, hematopoietic malignancy, and ant s. More specifically, the cancers include, but not limited to, lung cancer, bladder cancer, pancreatic cancer, kidney cancer, gastric cancer, breast cancer, colon cancer, prostate cancer (including bone metastases), hepatocellular carcinoma, ovarian cancer, esophageal squamous cell carcinoma, melanoma, an anaplastic large cell ma, an inflammatory myofibroblastic tumor, and a glioblastoma.
In some aspects, the above methods are used to treat one or more of bladder, colorectal, non-small cell lung, breast, or pancreatic cancer. In some aspects, the above methods are used to treat one or more of ovarian, gastric, head and neck, prostate, hepatocellular, renal, glioma, or a cancer.
In some aspects, the invention includes a , including the above methods, wherein the nd is used to inhibit cellular epithelial to mesenchymal transition (EMT).
In some aspects, the method further comprises administering at least on additional active agent. In some aspects, the invention includes a method of treating cancer comprising administering to a mammal in need thereof a therapeutically effective amount of a compound or salt of the invention, wherein at least one additional active anti-cancer agent is used as part of the method.
In some aspects, the invention includes a method of ng the disease described herein ed at least in part by BTK comprising administering to a mammal in need thereof a therapeutically effective regimen comprising a compound or salt of Formula I and at least one additional active agent. lly, dosage levels on the order of from about 0.01 mg/kg to about 150 mg/kg of body weight per day are useful in the treatment of the above-indicated conditions, or alternatively about 0.5 mg to about 7 g per patient per day. For example, inflammation, , psoriasis, allergy/asthma, e and conditions of the immune system, disease and conditions of the l Nervous System (CNS), may be effectively treated by the administration of from about 0.01 to 50 mg of the compound per kilogram of body weight per day, or alternatively about 0.5 mg to about 3.5 g per patient per day.
It is understood, however, that the specific dose level for any particular patient will depend upon a y of factors including the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
General Definitions and Abbreviations: Except where otherwise indicated, the following general conventions and definitions apply. Unless ise indicated herein, ge and terms are to be given their broadest reasonable interpretation as understood by the skilled artisan. Any examples given are nonlimiting.
Any section headings or subheadings herein are for the reader's convenience and/or formal compliance and are miting.
A recitation of a compound herein is open to and embraces any material or composition containing the recited compound (e.g., a composition containing a racemic mixture, tautomers, epimers, isomers, impure mixtures, etc.). In that a salt, solvate, or hydrate, polymorph, or other complex of a compound es the compound itself, a recitation of a compound embraces materials containing such forms. Isotopically labeled compounds are also encompassed except where specifically excluded. For example, hydrogen is not limited to hydrogen containing zero neutrons. For example, deuterium is ed to herein as "D" and means a hydrogen atom having one n.
The term "active agent" means a compound of the invention in any salt, polymorph, crystal, solvate, or hydrated form.
The term "substituted" and substitutions contained in formulas herein refer to the replacement of one or more hydrogen radicals in a given ure with a specified radical, or, if not specified, to the replacement with any chemically feasible radical. When more than one position in a given structure can be substituted with more than one substituent selected from specified groups, the tuents can be either the same or different at every position (independently selected) unless otherwise indicated. In some cases, two positions in a given structure can be substituted with one shared substituent. It is tood that chemically impossible or highly unstable configurations are not desired or intended, as the skilled artisan would appreciate.
In descriptions and claims where subject matter (e.g., substitution at a given molecular position) is recited as being selected from a group of possibilities, the recitation is specifically intended to include any subset of the recited group. In the case of multiple variable positions or substituents, any combination of group or variable subsets is also contemplated.
Unless ted ise, a substituent, diradical or other group ed to herein can be bonded through any suitable position to a referenced subject molecule. For e, the term "indolyl" includes l-indolyl, 2-indolyl, 3-indolyl, etc.
The convention for describing the carbon content of certain moieties is "(Ca-b)" or "Ca- Cb" meaning that the moiety can contain any number of from "a" to "b" carbon atoms. Coalkyl means a single covalent chemical bond when it is a connecting moiety, and a hydrogen when it is a al moiety. Similarly, "x-y" can indicate a moiety containing from x to y atoms, e.g.,5_ 6heterocycloalkyl means a heterocycloalkyl having either five or six ring members. "CH" may be used to define number of carbons in a group. For example, "C0_1galkyl" means alkyl having 0- 12 carbons, wherein Coalkyl means a single nt chemical bond when a linking group and means hydrogen when a terminal group. C0_12alkyl includes s alternative embodiments, ing, but not 111111th to, C1_12alkyl, C2_12alkyl, C3_12alkyl, C4_12alkyl, C5_12alkyl, C6_12alkyl, C7_12alkyl, Cg_12alkyl, C9_12alkyl, C10_12alkyl, C11_12alkyl, C1_11alkyl, C1_10alkyl, C1_9alkyl, C1_ , C1_7alkyl, C1_6alkyl, kyl, C1_4alkyl, C1_3alkyl, C1_2alkyl, Clalkyl, and Coalkyl. C0_ lzalkyl further includes any combination of "a" and "b" and/or "x" and "y" number of carbon atoms including, but not d to, C2_12alkyl, C3_11alkyl, C4_10alkyl, C5_9alkyl, C6_galkyl and C7alky1.
The term "absent," as used herein to be a structural variable (e.g., "—R— is absent") means that diradical R has no atoms, and merely represents a bond between other adjoining atoms, unless ise indicated.
Unless ise indicated (such as by a connecting "-"), the connections of compound name moieties are at the rightmost recited . That is, the substituent name starts with a terminal moiety, continues with any bridging moieties, and ends with the ting moiety. For example, "heteroarylthioC1-4alkyl" is a heteroaryl group connected through a thio sulfur to a C1- 4alkyl, which alkyl connects to the chemical species bearing the substituent.
The term "aliphatic" means any hydrocarbon moiety, and can n linear, branched, and cyclic parts, and can be saturated or unsaturated.
The term " means any saturated hydrocarbon group that is straight-chain or branched. es of alkyl groups include methyl, ethyl, propyl, 2—propyl, n-butyl, iso-butyl, tert-butyl, pentyl, and the like.
The term "alkenyl" means any ethylenically unsaturated straight-chain or branched hydrocarbon group. Representative examples include, but are not limited to, ethenyl, l-propenyl, 2-propenyl, l—, 2-, or 3—butenyl, and the like.
The term "alkynyl" means any acetylenically unsaturated straight-chain or branched hydrocarbon group. Representative examples include, but are not limited to, ethynyl, l-propynyl, 2-propynyl, l—, 2-, or 3-butynyl, and the like.
The term "alkoxy" means —O-alkyl, —O-alkenyl, or ynyl. "Haloalkoxy" means an —O-(haloalkyl) group. Representative examples include, but are not limited to, trifluoromethoxy, momethoxy, and the like.
"Haloalkyl" means an alkyl, preferably lower alkyl, that is substituted with one or more same or different halo atoms.
"Hydroxyalkyl" means an alkyl, preferably lower alkyl, that is substituted with one, two, or three hydroxy groups; e.g., hydroxymethyl, l or 2-hydroxyethyl, 1,2-, 1,3-, or 2,3- dihydroxypropyl, and the like.
The term "alkanoyl" means alkyl, —C(O)-alkenyl, or —C(O)—alkynyl.
"Alkylthio" means an —S-(alkyl) or an —S-(unsubstituted cycloalkyl) group.
Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, cyclopropylthio, cyclobutylthio, cyclopentylthio, cyclohexylthio, and the like.
The term "cyclic" means any ring system with or t heteroatoms (N, O, or S(O)0_2), and which can be saturated, partially saturated, or unsaturated. Ring systems can be bridged and can include fused rings. The size of ring s may be described using terminology such as "X- ycyclic," which means a cyclic ring system that can have from x to y ring atoms. For example, the term "9.1ocarbocyclic" means a 5,6 or 6,6 fused bicyclic carbocyclic ring system which can be saturated, unsaturated or aromatic. It also means a phenyl fused to one 5 or 6 membered saturated or unsaturated carbocyclic group. Nonlimiting examples of such groups include yl, l,2,3,4 tetrahydronaphthyl, indenyl, indanyl, and the like.
The term "carbocyclic" means a cyclic ring moiety containing only carbon atoms in the ring(s) without regard to aromaticity. A 3-10 membered carbocyclic means chemically feasible monocyclic and filSGd bicyclic carbocyclics having from 3 to 10 ring atoms. Similarly, a 4-6 membered carbocyclic means monocyclic yclic ring es having 4 to 6 ring s, and a 9-10 membered carbocyclic means fused bicyclic carbocyclic ring moieties having 9 to 10 ring carbons.
The term "cycloalkyl" means a non-aromatic 3-12 carbon yclic, ic, or polycyclic aliphatic ring moiety. Cycloalkyl can be bicycloalkyl, polycycloalkyl, bridged, or spiroalkyl. One or more of the rings may contain one or more double bonds but none of the rings has a completely ated pi-electron system. Examples, without limitation, of cycloalkyl groups are cyclopropane, cyclobutane, cyclopentane, cyclopentene, cyclohexane, exadiene, adamantane, cycloheptane, eptatriene, and the like.
The term "unsaturated carbocyclic" means any cycloalkyl containing at least one double or triple bond. The term "cycloalkenyl" means a cycloalkyl having at least one double bond in the ring moiety.
The terms "bicycloalkyl" and "polycycloalkyl" mean a structure consisting of two or more cycloalkyl moieties that have two or more atoms in common. If the cycloalkyl moieties have exactly two atoms in common they are said to be "fused". Examples include, but are not limited to, bicyclo[3.l .0]hexyl, perhydronaphthyl, and the like. If the cycloalkyl es have more than two atoms in common they are said to be ed". es include, but are not limited to, bicyclo[2.2. l]heptyl ("norbornyl"), bicyclo[2.2.2]octyl, and the like.
The term "spiroalkyl" means a ure consisting of two cycloalkyl moieties that have exactly one atom in common. Examples include, but are not limited to, spiro[4.5]decyl, spiro[2.3]hexyl, and the like.
The term "aromatic" means a planar ring es containing 4n+2 pi electrons, wherein n is an integer.
The term "aryl" means aromatic es containing only carbon atoms in its ring system.
Non-limiting examples include phenyl, naphthyl, and anthracenyl. The terms "aryl-alkyl" or "arylalkyl" or "aralkyl" refer to any alkyl that forms a bridging portion with a terminal aryl.
"Aralkyl" means alkyl that is tuted with an aryl group as defined above; e. g., — CH2 phenyl, —(CH2)2phenyl, —(CH2)3 phenyl, CH3CH(CH3)CH2phenyl, and the like and derivatives thereof.
The term "heterocyclic" means a cyclic ring moiety containing at least one heteroatom (N, O, or S(O)0_2), ing heteroaryl, heterocycloalkyl, including unsaturated heterocyclic rings.
The term "heterocycloalkyl" means a non-aromatic clic, bicyclic, or polycyclic heterocyclic ring moiety of 3 to 12 ring atoms ning at least one ring having one or more heteroatoms. The rings may also have one or more double bonds. However, the rings do not have a completely conjugated pi-electron system. Examples, without limitation, of heterocycloalkyl rings include azetidine, oxetane, tetrahydrofiiran, tetrahydropyran, oxepane, oxocane, thietane, thiazolidine, oxazolidine, oxazetidine, pyrazolidine, olidine, isothiazolidine, tetrahydrothiophene, tetrahydrothiopyran, thiepane, thiocane, azetidine, pyrrolidine, dine, N—methylpiperidine, azepane, azapane, azocane, ioxane, oxazolidine, piperazine, homopiperazine, morpholine, thiomorpholine, l,2,3,6-tetrahydropyridine and the like. Other examples of heterocycloalkyl rings include the oxidized forms of the sulfur—containing rings.
Thus, tetrahydrothiophene- l -oxide, tetrahydrothiophene-l , l-dioxide, thiomorpholine- l -oxide, thiomorpholine- l , 1 -dioxide, tetrahydrothiopyranoxide, ydrothiopyran-l , 1 -dioxide, thiazolidine-l-oxide, and lidine- l , l -dioxide are also considered to be heterocycloalkyl rings. The term "heterocycloalkyl" also es fused ring systems and can include a carbocyclic ring that is partially or fully unsaturated, such as a benzene ring, to form used heterocycloalkyl rings. For e, 3,4-dihydro-1,4-benzodioxine, tetrahydroquinoline, tetrahydroisoquinoline and the like. The term "heterocycloalkyl" also includes heterobicycloalkyl, polycycloalkyl, or heterospiroalkyl, which are bicycloalkyl, polycycloalkyl, or spiroalkyl, in which one or more carbon atom(s) are replaced by one or more heteroatoms selected from O, N, and S. For example, spiro[3.3]heptane, 2,7-diaza- spiro[4.5]decane, 6-oxa-2—thia—spiro[3.4]octane, octahydropyrrolo[l,2-a]pyrazine, 7—azabicyclo [2.2. l]heptane, 2-oxa-bicyclo[2.2.2]octane, and the like, are such heterocycloalkyls.
Examples of saturated heterocyclic groups e, but are not limited to oxiranyl, thiaranyl, aziridinyl, oxetanyl, thiatanyl, azetidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, 1,4-dioxanyl, 1,4-oxathianyl, linyl, l,4-dithianyl, piperazinyl, 1,4-azathianyl, oxepanyl, thiepanyl, azepanyl, 1,4- dioxepanyl, 1,4-oxathiepanyl, 1,4-oxaazepanyl, 1,4—dithiepanyl, 1,4-thieazepanyl, 1,4-diazepanyl Non-aryl heterocyclic groups include saturated and unsaturated systems and can include groups having only 4 atoms in their ring system. The heterocyclic groups include benzo-fused ring systems and ring systems substituted with one or more oxo moieties. Recitation of ring sulfur is tood to include the , sulfoxide or sulfone where feasible. The heterocyclic groups also include partially rated or fully saturated 4—10 membered ring systems, e.g., single rings of 4 to 8 atoms in size and bicyclic ring systems, including aromatic 6-membered aryl or heteroaryl rings fused to a non-aromatic ring. Also included are 4-6 membered ring systems ("4-6 membered heterocyclic"), which e 5-6 membered heteroaryls, and include groups such as inyl and piperidinyl. Heterocyclics can be heteroatom—attached where such is possible. For instance, a group derived from pyrrole can be pyrrol—l-yl (N-attached) or pyrrol- 3-yl (C-attached). Other heterocyclics include imidazo(4,5—b)pyridinyl and benzoimidazol-l- Examples of heterocyclic groups include pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, nyl, piperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, l,2,3,6-tetrahydropyridinyl, 2- pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, oxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, lidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3- yclo[4.1.0]heptanyl, 3H—indolyl, izinyl, and the like.
The term "unsaturated heterocyclic" means a heterocycloalkyl containing at least one unsaturated bond. The term "heterobicycloalkyl" means a bicycloalkyl structure in which at least one carbon atom is replaced with a heteroatom. The term "heterospiroalkyl" means a spiroalkyl structure in which at least one carbon atom is replaced with a heteroatom. es of partially unsaturated heteroalicyclic groups include, but are not limited to: 3,4-dihydro-2H-pyranyl, 5,6-dihydro-2H-pyranyl, ZH-pyranyl, l,2,3,4-tetrahydropyridinyl, and l ,2,5 rahydropyridinyl.
The terms "heteroaryl" or "hetaryl" mean a clic, ic, or polycyclic aromatic heterocyclic ring moiety containing 5-12 atoms. Examples of such heteroaryl rings e, but are not limited to, fiiryl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl. The terms "heteroaryl" also include heteroaryl rings with fused carbocyclic ring systems that are lly or fiJlly unsaturated, such as a e ring, to form a benzofused heteroaryl. For example, benzimidazole, benzoxazole, benzothiazole, benzofuran, quinoline, isoquinoline, quinoxaline, indazole, imidazo[l,2-a]pyridine, 3-methyloxo-2,3- dihydrobenzo[d]oxazol—5-yl, 2—methyl—2H-indazol-5—yl, 3-methylimidazo[l,5—a]pyridine, 2— methyl- 1 o [d]imidazole, lH-pyrrolo [2,3 -b]pyridine, 3,4-Dihydro-2H- benzo[b][1,4]oxazine, 2-oxo-2,3-dihydrobenzo[d]oxazole, 3-oxo-3,4-dihydro-2H- benzo[b] [l ,4]oxazine, 2,3-Dihydrobenzo [b] [l ,4]dioxine, 2-methyl-[l riazolo[ l ,5-a]pyridine, and the like. Furthermore, the terms "heteroaryl" include filSCd 5-6, 5-5, 6-6 ring systems, ally possessing one nitrogen atom at a ring junction. Examples of such hetaryl rings include, but are not limited to, pyrrolopyrimidinyl, imidazo[l,2-a]pyridinyl, imidazo[2,l- b]thiazolyl, imidazo[4,5-b]pyridine, pyrrolo[2,l-f][l,2,4]triazinyl, and the like. Heteroaryl groups may be attached to other groups through their carbon atoms or the heteroatom(s), if applicable. For example, pyrrole may be connected at the nitrogen atom or at any of the carbon atoms.
Heteroaryls include, e.g., 5 and 6 membered monocyclics such as pyrazinyl and pyridinyl, and 9 and 10 membered fused bicyclic ring moieties, such as quinolinyl. Other examples of heteroaryl e quinolinyl, oxy-quinolin-4—yl, pyridinyl, pyridin—3- yl, and pyridinyl. Other examples of heteroaryl include pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furanyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, fiarazanyl, benzofiarazanyl, hiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, furopyridinyl, and the like. Examples of 5-6 membered heteroaryls e, thiophenyl, olyl, triazolyl, oxadiazolyl, 1,2,3-thiadiazolyl, 1,2,4-triazolyl, 1,3,4-oxadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-oxadiazolyl, 1,2,5- thiadiazolyl, pyridyl, zinyl, pyrimidinyl, pyrazinyl, 1,2,4 oxadiazolyl, 1,2,5-triazinyl, 1,3,5-triazinyl, 6-oxo-l,6-dihydropyridine, and the like.
"Heteroaralkyl" group means alkyl, preferably lower alkyl, that is substituted with a aryl group; e.g., —CH2 pyridinyl, —(CH2)2pyrimidinyl, —(CH2)3imidazolyl, and the like, and derivatives thereof.
A pharmaceutically acceptable heteroaryl is one that is sufficiently stable to be attached to a compound of the invention, formulated into a pharmaceutical ition and subsequently administered to a patient in need thereof Examples of monocyclic heteroaryl groups include, but are not limited to: pyrrolyl, l, enyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, 1,2,3— triazolyl, 1,3,4-triazolyl, l-oxa-2,3-diazolyl, 2,4-diazolyl, l-oxa-2,5-diazolyl, l-oxa-3,4- diazolyl, l-thia-2,3-diazolyl, 1-thia-2,4-diazolyl, 1-thia-2,5-diazolyl, l-thia-3,4-diazolyl tetrazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl.
Examples of fused ring heteroaryl groups include, but are not limited to: benzoduranyl, hiophenyl, indolyl, idazolyl, indazolyl, benzotriazolyl, pyrrolo[2,3-b]pyridinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrrolo[3,2-b]pyridinyl, imidazo[4,5- b]pyridinyl, imidazo[4,5-c]pyridinyl, pyrazolo[4,3-d]pyridinyl, pyrazolo[4,3—c]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[3,4-b]pyridinyl, isoindolyl, indazolyl, purinyl, indolinyl, imidazo[l,2-a]pyridinyl, imidazo[l,5—a]pyridinyl, pyrazolo[l ,5-a]pyridinyl, pyrrolo[l ,2- b]pyridazinyl, o[l,2-c]pyrimidinyl, quinolinyl, isoquinolinyl, cinnolinyl, azaquinazoline, quinoxalinyl, phthalazinyl, 1,6—naphthyridinyl, phthyridinyl, 1,8-naphthyridinyl, 1,5- naphthyridinyl, 2,6-naphthyridinyl, 2,7-naphthyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[4,3- d]pyrimidinyl, [3,4-d]pyrimidinyl, pyrido[2,3—d]pyrimidinyl, pyrido[2,3—b]pyrazinyl, pyrido[3,4-b]pyrazinyl, pyrimido[5,4-d]pyrimidinyl, pyrimido[2,3-b]pyrazinyl, pyrimido[4,5- d]pyrirnidinyl.
"Arylthio" means an —S-ary1 or an —S-heteroaryl group, as defined herein.
Representative examples include, but are not limited to, phenylthio, pyridinylthio, furanylthio, thienylthio, pyrimidinylthio, and the like and derivatives thereof.
The term "9-10 membered heterocyclic" means a fused 5,6 or 6,6 bicyclic heterocyclic ring moiety, which can be saturadated, unsaturated or aromatic. The term "9—10 membered fused bicyclic heterocyclic" also means a phenyl fused to one 5 or 6 membered cyclic group. es include benzofuranyl, benzothiophenyl, indolyl, benzoxazolyl, 3H—imidazo[4,5- c]pyridin-yl, dihydrophthazinyl, dazo[4,5-c]pyridinyl, imidazo[4,5-b]pyridyl, 1,3 benzo[1,3]dioxolyl, 2H-chromanyl, isochromanyl, 5-oxo-2,3 dihydro-SH—[l,3]thiazolo[3,2- a]pyrimidyl, 1,3-benzothiazolyl, 1,4,5,6 tetrahydropyridazyl, 1,2,3,4,7,8hexahydropteridinyl, 2- thioxo-2,3,6,9-tetrahydro-1H—purinyl, hydro-1H—purinyl, 3,4-dihydropyrimidin-l-yl, 2,3-dihydro-1,4-benzodioxinyl, 1,3]dioxolyl, omenyl, chromanyl, 3,4— dihydrophthalazinyl, 2,3-ihydro-1H—indolyl, 1,3-dihydro-2H-isoindolyl, 2,4,7-trioxo- 1 ,2,3 ,4,7,8-hexahydropteridin—yl, thieno[3 ,2-d]pyrimidinyl, 4—oxo-4,7-dihydro-3H-pyrrolo[2,3- d]pyrimidin-yl, 1,3-dimethyloxothioxo-2,3,6,9-tetrahydro-1H-purinyl, 1,2- dihydroisoquinolinyl, 2—oxo- 1 ,3-benzoxazolyl, hydro-5H-1,3-thiazolo—[3,2-a]pyrimidinyl, ,6,7,8-tetrahydro-quinazolinyl, 4-oxochromanyl, 1,3-benzothiazolyl, idazolyl, benzotriazolyl, purinyl, fiarylpyridyl, enylpyrimidyl, thiophenylpyridyl, pyrrolylpiridyl, oxazolylpyridyl, thiazolylpiridyl, 3,4—dihydropyrimidinyl imidazolylpyridyl, quinoliyl, isoquinolinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pyrazolyl[3,4]pyridine, 1,2- dihydroisoquinolinyl, cinnolinyl, 2,3—dihydro-benzo[1,4]dioxin4-yl, 4,5,6,7—tetrahydro-benzo[b]— thiophenyl-Z-yl, 1,8-naphthyridinyl, 1,5-napthyridinyl, 1,6-naphthyridinyl, 1,7-napthyridinyl, 3 ,4-dihydro-2H- 1 ,4-benzothiazine, 4,8-dihydroxy-quinolinyl, 1-oxo— 1 ,2-dihydro-isoquinolinyl, 4-phenyl-[1,2,3]thiadiazolyl, and the like.
The term "aryloxy" means an —O-aryl or an —O-heteroaryl group, as defined herein.
Representative es include, but are not limited to, y, nyloxy, furanyloxy, thienyloxy, pyrimidinyloxy, pyrazinyloxy, and the like, and derivatives thereof.
The term "oxo" means a compound containing a carbonyl group. One in the art understands that an "oxo" requires a second bond from the atom to which the oxo is attached.
The term "halo" or "halogen" means fluoro, chloro, bromo, or iodo.
"Acyl" means a —C(O)R group, where R can be selected from the nonlimiting group of hydrogen or optionally substituted lower alkyl, trihalomethyl, unsubstituted cycloalkyl, aryl, or other suitable substituent.
"Thioacyl" or "thiocarbonyl" means a—C(S)R" group, with R as defined above.
The term "protecting group" means a suitable chemical group that can be attached to a functional group and removed at a later stage to reveal the intact functional group. Examples of suitable protecting groups for various onal groups are described in T. W. Greene and P. G.
M. Wuts, tive Groups in Organic Synthesis, 2d Ed, John Wiley and Sons (1991 and later editions); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed. Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995). The term "hydroxy protecting group", as used herein, unless ise indicated, includes Ac, CBZ, and various hydroxy ting groups familiar to those d in the art including the groups referred to in Greene.
The term "linear ure" means a moiety having substituents that do not cyclize to form a ring system. A representative example includes, but is not limited to, a compound including —NRXRY where any atoms of " X" and any atoms of "RY" do not connect to form a ring.
As used herein, the term "pharmaceutically acceptable salt" means those salts which retain the biological effectiveness and properties of the parent compound and do not present safety or toxicity issues. The term aceutically acceptable salt(s)" is known in the art and includes salts of acidic or basic groups which can be present in the compounds and ed or resulting from pharmaceutically acceptable bases or acids.
The term "pharmaceutical composition" means an active compound in any form suitable for effective administration to a subject, e.g., a mixture of the compound and at least one pharmaceutically acceptable carrier.
As used herein, a "physiologically/pharmaceutically acceptable carrier" means a carrier or diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
A "pharmaceutically acceptable excipient" means an inert substance added to a pharmaceutical composition to further facilitate administration of a compound. Examples, without limitation, of excipients include calcium ate, calcium ate, various sugars and types of starch, cellulose derivatives, n, vegetable oils and polyethylene s.
The terms "treat,3, CEtreatment," and "treating" means reversing, alleviating, inhibiting the progress of, or partially or completely preventing the disorder or condition to which such term applies, or one or more symptoms of such er or condition. "Preventing" means treating before an infection occurs.
"Therapeutically effective " means that amount of the compound being administered which will relieve to some extent one or more of the ms of the disorder being treated, or result in inhibition of the progress or at least partial reversal of the ion.
NMR Nuclear magnetic resonance MDP(S) Mass-directed HPLC purification (system) LC/MS Liquid tography mass spectrometry LDA Lithium diisopropylamide tert-BuOH tert-Butanol AcOH Acetic acid CDI 1,1 ’-Carbonyldiimidazole DCE 1,1-Dichloroethane DCM Dichloromethane DMF Dimethylformamide THF Tetrahydrofuran MeOH Methanol EtOH Ethanol EtOAc Ethyl acetate MeCN Acetonitrile DMSO Dimethylsulfoxide Boc utyloxycarbonyl DME 1,2-Dimethoxyethane DMF N,N—Dimethylformamide DIPEA/DIEA ropylethylamine PS—DIEA Polymer—supported diisopropylethylamine PS—PPhg-Pd Polymer-supported Pd(PPh3)4 LAH Lithium aluminum hydride EDC 1-(3-Dimethylaminopropyl)ethylcarbodiimide HATU: l- [bis(dimethylamino)methylene] - l H— l ,2,3—triazolo[4,5 -b]pyridinium—3 -oxide hexafluorophosphate HOBt l-Hydroxybenzotnazole DMAP 4-Dimethylaminopyridine SEM-Cl 2-(Trimethylsilyl)ethoxymethyl chloride TBTU O-(Benzotriazolyl)-N,N,N',N’-tetramethyluronium tetrafluoroborate TEMPO 2,2,6,6-Tetramethylpiperidine- l -oxyl TFA (A) Trifluoroacetic acid (anhydride) TLC Thin layer chromatography TMSCN Trimethylsilyl cyanide Min Minute(s) NMO N—Methylmorpholine N—oxide h Hour(s) d Day(s) RT, R.T., r.t., r.t or rt Room temperature tR Retention time Conc. Concentrated

Claims (10)

1. A compound selected from the group ting of: 6-(3-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; 6-(4-acryloylpiperazinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(4-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; O N N O 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; O NH2 N N 6-(4-acrylamidopiperidinyl)(4-phenoxyphenyl)nicotinamide; O N N N O crylamidopyrrolidinyl)(4-phenoxyphenyl)nicotinamide; HN N N 1-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N N O 1-(1-acryloylazetidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O crylamidophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O 1-(3-acrylamidophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N N N O (S)(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N N N O (R)(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; O N O N N 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)pyrimidinecarboxamide; N NH2 N N (E)(4-(4-(dimethylamino)butenoyl)piperazinyl)(4- phenoxyphenyl)nicotinamide; N N N O cryloylpiperazinyl)(4-(cyclohexyloxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(3-methoxymethylphenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)-6'-phenoxy-2,3'-bipyridinecarboxamide; N N N O cryloylpiperazinyl)(4-(pyridinyloxy)phenyl)nicotinamide; N N O N 1-(1-(4-(dimethylamino)butenoyl)pyrrolidinyl)(4-phenoxyphenyl)-1H-pyrazole- 4-carboxamide; O N N O 6-(4-acryloylpiperazinyl)(4-(3-fluorophenoxy)phenyl)nicotinamide; N N O F 6-(4-acryloylpiperazinyl)(3-fluorophenoxyphenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(4-(4-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(4-(2-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(2-fluorophenoxyphenyl)nicotinamide; N N F O cryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; O S N N 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)thiazolecarboxamide; N N 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; O O N N O 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)oxazolecarboxamide; N N 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)thiazolecarboxamide; S O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; O N N S O cryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; N O 2-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)oxazolecarboxamide; N O O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; S O cryloylpiperidinyl)(4-phenoxyphenyl)thiazolecarboxamide; N O 2-(1-acryloylpiperidinyl)(4-phenoxyphenyl)oxazolecarboxamide; O N N O 5-(1-acryloylpiperidinyl)-4'-phenoxybiphenylcarboxamide; 5-(4-acryloylpiperazinyl)-4'-phenoxybiphenylcarboxamide; cryloylpiperidinyl)(4-(hydroxy(phenyl)methyl)phenyl)nicotinamide; O OH (E)(1-(2-cyanobutenoyl)azetidinyl)(4-phenoxyphenyl)-1H-pyrazole carboxamide; N O 6-(4-acryloylpiperazinyl)(4-((4,4-difluorocyclohexyl)oxy)phenyl)nicotinamide; N N F 6-(1-acryloylpiperidinyl)(4-(phenylcarbamoyl)phenyl)nicotinamide; N N O O 2-(4-phenoxyphenyl)(4-(vinylsulfonyl)piperazinyl)nicotinamide; N N S O 2-(4-phenoxyphenyl)(1-(vinylsulfonyl)piperidinyl)nicotinamide; S O or a pharmaceutically able salt thereof.
2. The compound according to Claim 1, wherein the compound is: 6-(3-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; 6-(4-acryloylpiperazinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(1-acryloylpyrrolidinyl)(4-phenoxyphenyl)nicotinamide; N N 6-(4-acrylamidophenyl)(4-phenoxyphenyl)nicotinamide; O N N O cryloylpiperidinyl)(4-phenoxyphenyl)nicotinamide; O NH2 N N 6-(4-acrylamidopiperidinyl)(4-phenoxyphenyl)nicotinamide; O N N N O 6-(3-acrylamidopyrrolidinyl)(4-phenoxyphenyl)nicotinamide; HN N N (4-(4-(dimethylamino)butenoyl)piperazinyl)(4- phenoxyphenyl)nicotinamide; N N N O 6-(4-acryloylpiperazinyl)(4-(cyclohexyloxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(3-methoxymethylphenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)-6'-phenoxy-2,3'-bipyridinecarboxamide; N N N O 6-(4-acryloylpiperazinyl)(4-(pyridinyloxy)phenyl)nicotinamide; N N O N 6-(4-acryloylpiperazinyl)(4-(3-fluorophenoxy)phenyl)nicotinamide; N N O F cryloylpiperazinyl)(3-fluorophenoxyphenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(4-(4-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(4-(2-fluorophenoxy)phenyl)nicotinamide; N N 6-(4-acryloylpiperazinyl)(2-fluorophenoxyphenyl)nicotinamide; N N F O cryloylpiperidinyl)-4'-phenoxybiphenylcarboxamide; 5-(4-acryloylpiperazinyl)-4'-phenoxybiphenylcarboxamide; 6-(1-acryloylpiperidinyl)(4-(hydroxy(phenyl)methyl)phenyl)nicotinamide; O OH 6-(4-acryloylpiperazinyl)(4-((4,4-difluorocyclohexyl)oxy)phenyl)nicotinamide; N N F 6-(1-acryloylpiperidinyl)(4-(phenylcarbamoyl)phenyl)nicotinamide; N N O O 2-(4-phenoxyphenyl)(4-(vinylsulfonyl)piperazinyl)nicotinamide; N N S O 2-(4-phenoxyphenyl)(1-(vinylsulfonyl)piperidinyl)nicotinamide; S O or a pharmaceutically acceptable salt thereof.
3. The nd according to Claim 2, wherein the compound is N N O .
4. A nd selected from the group consisting of: 2-(4-phenoxyphenyl)(piperidinyl)nicotinamide; 2-(4-phenoxyphenyl)(piperazinyl)nicotinamide; N N 1-(azetidinyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; N O 1-(3-aminophenyl)(4-phenoxyphenyl)-1H-pyrazolecarboxamide; H2N N O 6-(1-acryloylpiperidinyl)(4-phenoxyphenyl)pyridinecarbonitrile; 6-(4-acryloylpiperazinyl)(4-phenoxyphenyl)pyridinecarboxylic acid; N N 2-(4-acryloylpiperazinyl)-N-methyl(4-phenoxyphenyl)thiazolecarboxamide; N N O .
5. A compound having a Formula IId, or a pharmaceutically acceptable salt or e thereof: R3 X Z R2 O O Formula IId wherein X is N , N , N , or ; Z is C or N; G1, G2, and G3 are selected from the group consisting of one or more H, D (deuterium), C1-12 alkyl, halogen, and CF3; R2 and R3 are each independently selected from the group ting of H, D, C1-12 alkyl, halogen, CN, and CF3, wherein C1-12 alkyl is optionally substituted with NR11R12; R4 is selected from the group consisting of H, D, C1-12 alkyl, halogen, CN, and CF3; and R11 and R12 are independently H or C1-6 alkyl.
6. The compound according to Claim 5, wherein X is N or N .
7. The compound according to Claim 5, wherein G1, G2, and G3 are one or more H, or halogen.
8. The nd according to Claim 5, wherein R2 and R3 are H or C1-12 alkyl, wherein C1- 11R12, and R11 and R12 are H or CH 12 alkyl is optionally substituted with NR 3.
9. A pharmaceutical composition for treating a disease mediated by Bruton’s tyrosine Kinase (BTK), comprising the compound of any one of Claims 1-3 and 5-8, and a ceutically acceptable salt thereof.
10. Use of the compound or the pharmaceutically able salt thereof of any one of Claims 1-3 and 5-8, in the cture of a medicament for treating of at least one of cancer, chronic inflammation, and autoimmune disease mediated at least in part by BTK. Guangzhou Innocare Pharma Tech Co., Ltd. By the Attorneys for the Applicant SPRUSON & FERGUSON
NZ718073A 2013-09-30 2014-09-29 Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease NZ718073B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361884958P 2013-09-30 2013-09-30
US61/884,958 2013-09-30
CN201310485048.1 2013-10-16
CN201310485048 2013-10-16
PCT/US2014/058084 WO2015048662A2 (en) 2013-09-30 2014-09-29 Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease

Publications (2)

Publication Number Publication Date
NZ718073A NZ718073A (en) 2021-04-30
NZ718073B2 true NZ718073B2 (en) 2021-08-03

Family

ID=

Similar Documents

Publication Publication Date Title
US11840513B2 (en) Substituted nicotinimide inhibitors of BTK for treating cancer
JP7418395B2 (en) SHP2 phosphatase inhibitors and methods of using them
JP6571215B2 (en) Heteroarylpyridone and azapyridone compounds as inhibitors of BTK activity
JP2022528562A (en) METTL3 inhibitor compound
DK2545045T3 (en) PIPERIDINE-4-YL-azetidine derivatives AS JAK1 INHIBITORS
ES2612503T3 (en) 9H-purine compounds as PI3K-delta inhibitors and methods for their preparation
JP2018150358A (en) Tank-binding kinase inhibitor compounds
CA2857302C (en) Use of inhibitors of the activity or function of pi3k
CN116444515A (en) Di-ring derivative-containing inhibitor, preparation method and application thereof
KR20130002991A (en) 8-methyl-1-phenyl-imidazol[1,5-a]pyrazine compounds
KR20130094710A (en) 5,7-substituted-imidazo[1,2-c]pyrimidines as inhibitors of jak kinases
AU2021309876B2 (en) Rock inhibitor, and preparation method therefor and use thereof
JPWO2014109414A1 (en) Nitrogen-containing heterocyclic compound or salt thereof
NZ718073B2 (en) Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
NZ718073A (en) Substituted nicotinimide inhibitors of btk and their preparation and use in the treatment of cancer, inflammation and autoimmune disease
WO2023185073A1 (en) Parp7 inhibitor and use thereof