NZ621436B2 - Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension - Google Patents

Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension Download PDF

Info

Publication number
NZ621436B2
NZ621436B2 NZ621436A NZ62143612A NZ621436B2 NZ 621436 B2 NZ621436 B2 NZ 621436B2 NZ 621436 A NZ621436 A NZ 621436A NZ 62143612 A NZ62143612 A NZ 62143612A NZ 621436 B2 NZ621436 B2 NZ 621436B2
Authority
NZ
New Zealand
Prior art keywords
alkyl
optionally substituted
alkoxy
imidazo
pyridine
Prior art date
Application number
NZ621436A
Other versions
NZ621436A (en
Inventor
Ian Bruce
Sylvie Chamoin
Stephen Paul Collingwood
Pascal Furet
Vikki Furminger
Diana Janus
Sarah Lewis
Jon Christopher Loren
Valentina Molteni
Alex Michael Saunders
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Priority claimed from PCT/IB2012/054501 external-priority patent/WO2013030802A1/en
Publication of NZ621436A publication Critical patent/NZ621436A/en
Publication of NZ621436B2 publication Critical patent/NZ621436B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Abstract

Provided are imidazopyridine and pyrazolopyridine derivative compounds of the general formula (I), wherein the variables are as defined in the specification. Examples of the compounds include N-(2-Fluoro-5-(2-(4-methylpiperazin-1-yl)benzylcarbamoyl)phenyl)-7-(1-methyl-1H-pyrazol-5-yl)imidazo[1,2-a]pyridine-3-carboxamide and N-(5-((2-(2,2-dimethylpyrrolidin-1-yl)ethyl)carbamoyl)-2-methylpyridin-3-yl)-6-( 1-methyl-1H-pyrazol-5-yl)pyrazolo[1,5-a]pyridine-3-carboxamide. The compounds are inhibitors of PDGFR kinase. The compounds may be useful in the treatment of pulmonary arterial hypertension. yridine-3-carboxamide and N-(5-((2-(2,2-dimethylpyrrolidin-1-yl)ethyl)carbamoyl)-2-methylpyridin-3-yl)-6-( 1-methyl-1H-pyrazol-5-yl)pyrazolo[1,5-a]pyridine-3-carboxamide. The compounds are inhibitors of PDGFR kinase. The compounds may be useful in the treatment of pulmonary arterial hypertension.

Description

ic Heterocycle Derivatives for the Treatment of Pulmonary Arterial Hypertension FIELD OF THE INVENTION The present invention relates to bicyclic heterocycle derivatives, their preparation, their use as pharmaceuticals and ceutical compositions ning them.
More particularly the present invention relates to their use in inhibiting PDGF receptor mediated biological activity.
BACKGROUND Protein kinases (PK) are a large set of urally related phosphoryl transferases having highly conserved structures and catalytic functions. Protein kinases are enzymatic components of the signal transduction pathways which catalyze the transfer of the terminal phosphate from ATP to the hydroxy group of tyrosine, serine and/or threonine residues of proteins, and are ore categorized into families by the substrates they orylate: Protein Tyrosine Kinases (PTK), and Protein Serine/Threonine Kinases. n kinases play a al role in the control of cell growth and differentiation and are responsible for the control of a wide y of cellular signal transduction processes, wherein protein kinases are key ors of cellular signals leading to the production of growth factors and cytokines. The overexpression or inappropriate expression of normal or mutant n kinases plays a significant role in the development of many diseases and disorders including, central nervous system disorders such as mer's, inflammatory disorders such as arthritis, bone diseases such as osteoporosis, metabolic disorders such as diabetes, blood vessel proliferative disorders such as angiogenesis, mune diseases such as rheumatoid arthritis, ocular diseases, cardiovascular disease, atherosclerosis, cancer, thrombosis, psoriasis, restenosis, schizophrenia, pain sensation, transplant rejection and infectious diseases such as viral, and fungal infections.
The agents of the invention act as inhibitors of PDGFR kinase, c-Kit kinase and related receptor and non-receptor tyrosine kinases. PDGFR is activated by binding of the growth factor PDGF to the extracellular portion of the receptor. Upon activation PDGFR phosphorylates many substrate proteins and controls a wide variety of cellular functions including eration and migration. PDGFR mediates these effects on multiple cell types including those of the mesenchymal lineage, fibroblasts, vascular smooth muscles cells and pericytes.
PDGFR kinase inhibition is ed to be a useful target for the treatment of various cardiovascular, ary, tissue remodelling and hypertrophic disorders, many cancers and other indications in which PDGF driven onal responses contribute to pathology, including PAH. PDGFR, PDGFR ligands and activated, phosphorylated PDGFR is found in the proliferating smooth muscle cells that comprise the lesions in the pulmonary vasculature of PAH patients and animal models of PAH. Furthermore, the tyrosine kinase inhibitor Gieevec® has been shown to be ious in the treatment of PAH clinically and in pre-clinical PAH models. Other targets inhibited by the agents of the invention may contribute to the efficacy of the agents in PAH, asthma and other indications. For example, c—kit inhibition contributes to the ion of mast cells and is cial in the treatment of preclinical models of asthma.
SUMMARY OF THE INVENTION In a first aspect of the invention, Embodiment 1, we provide a compound of formula (I); or a pharmaceutically acceptable salt thereof, wherein, A is R3 PCT/IBZOIZIOS4501 R1 is C1—C4 alkyl; C1-C4 alkoxy optionally substituted by one or more halogen atoms; ON; or halogen; R16‘ is H, halogen, C1-C4 alkyl or C1-C4 haloalkyl; X is N or CH; R2 is H; C1—C3 alkyl optionally substituted by one or more OH, -NR9R11 or C1—C4 alkoxy; C1—C3 haloalkyl; C2-Cg alkynyl tuted by one or more halogen, OH, — NRQR11 or C1-C4 alkoxy; C3-C10 cycloalkyl; -(C1-C4 alkyl)~C3-Cg cycloalkyl; 01—03 alkoxy optionally substituted by one or more halogen, -NR9R11 or OH; OH; CN; halogen; -(Co—C4 alkyl)—NR9R“; -(co-<:4 alkyi)—C02R15; -(c:o-c4 alkyl)—C(O)NR9R“; - 1O (Co-C4 —Cs—C14 aryl; or 4 —3 to 14 membered heterocyclyl; wherein the cycioalkyl, -(Co-C4 alkyl)—C6-C14 aryl and -(Co-C4 alkyl)~3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; R3 is H; C1-C8 alkyl optionally substituted by one or more OH, -NR9R11 or C1-C4 alkoxy; C1—Cg haloalkyl; C2-Cg alkynyl tuted by one or more n, OH, ~ NRQR“, or 01-04 alkoxy; 03-010 cycloalkyl; -(c:1—c4 alkyl)—Cs-Cg lkyi; 01-03 alkoxy optionally substituted by one or more halogen, -NR5‘R11 or OH; OH; CN; halogen; -(Co-C4 —NR9R“; -(Co-C4 alkyl)—COZR15; -(Co-C4 alkyi)~C(O)NR9R“; - (Co-C4 —Ca—C14 aryl; or 4 alkyi)—3 to 14 membered heterocyclyl; wherein the cycloalkyl, -(Co-C4 alkyl)-Cs-C14 aryl and -(Co—C4 alkyl)—3 to 14 heterocyclyl are each ally substituted by one or more Za substituents; each Z3 is ndently OH; —(Co~C4 alkyl)—C5 aryl; ~O—Cs aryl; C1-C4 alkyl optionally substituted by one or more OH, ON or —NR193R213; C1-C4 haloalkyl; C1-C4 alkoxy optionally tuted by one or more OH, 98, -NR19"‘R21a or C1-C4 alkoxy; - NR1BaC(O)R21a; —C(O)NR193R21a; _NR18aC(O)NR19aR21a; _NR193R213; —(Co-C4 alkyl)- C(O)OR183; —(Co-C4 alkyl)—C(O)R193; 0x0; CN; N02; halogen or -(oo-c4 alkyl)—4 to 6 membered heterocyclyl; wherein the aryl and heterocyclyl are each optionally substituted by halogen, C1—C4 alkyl, C1—C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens; R4 is H, C1—C4 alkyl or C1-C4 haloalkyl; R5 is H, 01—04 alkyl or C1-C4 haloalkyl; R6 is selected from C1—C3 alkyl optionally substituted by one or more C1-C4 alkoxy; 01-08 haloalkyl; —(Co-C4alkyl)—C3-Cgcycloalkyl; C1-Cgalkoxy optionally substituted by one or more halogen atoms; —NR‘9R21; ~(Co—C4 alkyl)—Ce—C14aryl; —(Co—C4 alkyl)—3 to 14 membered heterocyclyl; and -(Co-C4 alkyl)—COZR15; wherein the —(Co—C4a|kyl)-Cs- Cgcycloaikyl, -(Co-C4 alkyl)—Cs-C14ary| and -(Co-C4 alkyl)-3 to 14 heterocyclyl are each optionally substituted by one or more Z substituents; each Z is independently selected from OH; (Co-C4 alkyi)—Ca aryl; O—Cs aryl; 01-05 alkyl ally substituted by one or more OH, ON or —NR19R21; C1-Ce haloalkyl; C1— 06 alkoxy optionally substituted by one or more OH, —COZR18, -NF<’19R21 or C1-C4 alkoxy; (O)R‘9; -C(O)NR‘3R21; -NR‘BC(O)NR19R2‘; -NR‘9R2‘; (co-c4 - C(O)OR19; (co-c4 alkyI)—C(O)R19; oxo; CN; N02; halogen and (co-c4 aikyl)—4 to 6 membered heterocyclyl; and wherein the aryl and heterocyciyi are each optionally substituted by one or more halogen, 01-05 aikyi, 01—06 haloalkyl and 01—05 alkoxy optionally substituted by one or more halogens; R9 and R" are each ndently selected from H; 01-05 alkyl ally substituted by one or more C1—C4 alkoxy or OH; (31—05 haloalkyl; 1aikyl)~Cg—C5 cycloalkyl; (Co-C4 alkyl)- (36-014aryl optionally substituted by one or more groups selected from C1—C6 alkyl, C1~Ca alkoxy and halogen; and (Co-C4 alkyl)— 3- to 14-membered heterocyclyl optionally substituted by one or more groups seiected from halogen, oxo, C1—Ce alkyl and —Ca alkyl; or R9 and R11 together with the nitrogen atom to which they are attached form a 5- to -membered heterocyclyl, which heterocyclyl inciudes O to 3 further heteroatoms selected from N, O and S, the heterocyclyl being ally substituted by one or more substituents selected from OH; n; phenyl, 5— to iO-membered heterocyclyl; C1-Cs aikyi; C1-C5 haloalkyl; 01—05 alkoxy optionally substituted by one or more OH or 01-04 alkoxy; and C(O)OC1-Csaikyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by 01—05 alkyl, C1-C5 haloalkyl or 01—05 alkoxy; R15 is selected from H; 01—08 alkyl; C1—Cs haloalkyl; Cg-Cm lkyl; (—C1—C4alkyl)- Cg-Ca cycloalkyl; —(Co—C4 alkyi)~Cs-C14aryi and -(Co-C4 aikyl)-3 to 14 membered heterocyclyl group; wherein the lkyl, aryl and heterocyclyl groups are each optionally substituted by one or more Z substituents; R183 is independently H or 61-05 alkyl; R193 and R218 are each independently H; (31-05 alkyl optionally substituted by one or more C1—C4 alkoxy or OH; C1-C6 haloalkyl; —(CO-C1alkyl)—C3~Cacycloa|kyl; (Co-C4 alkyl)- 05-014aryl optionally substituted by one or more groups seiected from 01-06 alkyi, C1- 06 alkoxy and halogen; or (Co-C4 alkyl)— 3— to 14—membered heterocyclyl optionally substituted by one or more groups seiected from halogen, oxo, 01-06 alkyl and C(O)C1—CG alkyl; or R198‘ and R213 together with the nitrogen atom to which they attached form a 5- to 10- membered heterocyclyl, which cyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more tuents selected from OH; halogen; ; 5— to ‘lO-membered heterocyclyl; C1-Cs alkyl; C1-C5 haloalkyl; C1—C5 alkoxy optionally substituted by one or more OH or C1—C4 alkoxy; and C(O)OC1-C5alkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by C1-C6 alkyl, 01-05 haloalkyl or C1—Ce alkoxy; R18 is independently H or C1—C5 alkyl; R19 and R21 are each independently H; 01-05 alkyl optionally substituted by one or more C1—C4 alkoxy or OH; C1-Ce kyl; ~(Co-C1alkyl)—C3—Cscycloalkyl; (C0—C4 alkyl)— Ce-Cmaryl optionally substituted by one or more groups selected from 01—05 alkyl, C1- Ce alkoxy and halogen; or (Co—C4 alkyl)- 3- to 14—membered heterocyclyl, optionally 1O substituted by one or more groups selected from halogen, oxo. C1-Cs alkyl and C(O)C1—Cs alkyl; or R19 and R21 together with the nitrogen atom to which they attached form a 5— to 10— membered cyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from OH; halogen; ; 5— to 10-membered heterocyclyl; C1—C5 alkyl; 01-06 haloalkyl; 01-05 alkoxy optionally substituted by one or more OH or C1-C4 alkoxy and C(O)OC1-Csalkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by a substituent selected from C1-Cs alkyl, 01-05 kyl and C1—05 .
DEFINITIONS For purposes of reting this specification, the following definitions will apply and whenever appropriate, terms used in the singular will also include the plural and vice versa.
As used herein, the term “alkyl” refers to a fully saturated ed or unbranched hydrocarbon moiety having up to 20 carbon atoms. Unless othenNise provided, alkyl refers to hydrocarbon moieties having 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. Representative examples of alkyl include, but are not limited to, , ethyl, n-propyl, opyl, l, sec—butyl, iso-butyl, tent-butyl, n-pentyl, isopentyl, neopentyl, n—hexyl, 3-methylhexyl, 2,2— dimethylpentyl, methylpentyl, n—heptyl, or n—octyl.
As used herein, the term “alkoxy” refers to alkyl-O-, wherein alkyl is defined herein above. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, , tert—butoxy, pentyloxy, hexyloxy, cyclopropyloxy— , cyclohexyloxy— and the like. Typically, alkoxy groups have about 1~6, more preferably about 1—4 carbons.
As used herein, the term ”haloalkyl” refers to an alkyl as defined herein that is substituted by one or more halo groups as defined . The haloalkyl can be monohaloalkyl, dihaloalkyl or polyhaloalkyl including oalkyl. A loalkyl can have one iodo, bromo, chloro or fluoro within the alkyl group. Dihaloalky and polyhaloalkyl groups can have two or more of the same halo atoms or a combination of different halo groups within the alkyl. Typically the polyhaloalkyl contains up to 12, or 10, or 8, or 6, or 5, or 4, or 3, or 2 halo groups. Non—limiting examples of haloalkyl e fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, roethyl, difluoropropyl, dichloroethyl and dichloropropyl. A perhaloalkyl refers to an alkyl having all hydrogen atoms replaced with halo atoms.
As used , the term "cycloalkyl" refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-10 carbon atoms. Unless othen/vise provided, cycloalkyl refers to cyclic hydrocarbon groups having between 3 and 10 ring carbon atoms or between 3 and 8 ring carbon atoms. Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, exyl or cyclohexenyl. Exemplary bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.‘l.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 6,6—dimethylbicyclo[3.1.1]heptyl, trimethylbicyclo[3.i .1]heptyl, or bicyclo[2.2.2]octyl.
The term “02-8 alkynyl” as used herein refers to a linear or branched saturated hydrocarbon group containing from 2 to 8 carbon atoms that contains at least one carbon to carbon triple bond. Examples of such groups include l, propynyl, butynyl and yl.
The term "aryl" refers to an aromatic hydrocarbon group having 6-14 carbon atoms in the ring portion. Typically, aryl is monocyclic or bicyclic aryl having 6—14 carbon atoms and includes one aromatic ring fused to one non-aromatic hydrocarbon ring. miting examples include phenyl, indane, naphthyl or tetrahydronaphthyl.
As used herein, the term “aryloxy” refers to both an --O-aryl and an --O—heteroaryl group, wherein aryl and heteroaryl are defined herein.
As used herein, the term “4- to 8-Membered cyclyl”, “5— to 6- membered heterocyclyl”, “3— to 10-membered heterocyclyl”, “3— to 14-membered heterocyclyl”, “4- to 14—membered heterocyclyl” and “5- to 14-membered heterocyclyl”, refers, respectively, to 4- to 8—membered, 5- to 6—membered, 3- to lO—membered, 3— to 14— membered, 4- to 14—membered and 5- to 14-membered heterocyclic rings containing at least one ring heteroatom selected from the group consisting of nitrogen, oxygen ’IO and sulphur, which may be saturated, partially saturated or unsaturated (aromatic).
The heterocyclyl es single ring groups, fused ring groups and bridged groups. es of such heterocyclyl include, but are not limited to, furan, pyrrole, pyrrolidine, pyrazole, imidazole, triazole, isotriazole, tetrazole, thiadiazole, azole, oxadiazole, pyridine, piperidine, pyrazine, oxazole, isoxazole, pyrazine., pyridazine, pyrimidine, piperazine, pyrrolidine, pyrrolidinone, morpholine, triazine, oxazine, yrofuran, ydrothiophene, tetrahydrothiopyran, tetrahydropyran, 1,4-dioxane, athiane, indazole, ine, indazole, indole, 8~aza- bicyclo[3.2.1]octane, 2,3-dihydrobenzofuran or thiazole.
"Heteroaryl" is a subset of heterocyclyl, wherein "heteroaryl" are completely unsaturated (aromatic). Examples of such groups are pyridine and pyrazine.
As used herein, the term “halogen” or “halo” refers to fluoro, chloro, bromo, and iodo. s ments of the invention are described herein. lt will be ized that features specified in each embodiment may be combined with other specified features to provide further embodiments.
Embodiment 2: A compound of formula (I); WO 30802 PCT/182012/054501 or a pharmaceutically acceptable salt f, wherein, A is R3 R1 is C1-C4 alkyl; C1-C4 alkoxy optionally substituted by one or more halogen atoms; ON; or halogen; R1a is H, halogen, C1-C4 alkyl or C1-C4 haloalkyl; X is N or CH; R2 is H; C1-Cg alkyl optionally substituted by one or more OH, ~NR9R“ or C1—C4 alkoxy; C1~Cg haloalkyl; Cz'Cg alkynyl substituted by one or more halogen, OH, — NRS’R11 or C1-C4 alkoxy; 03-010 cycloalkyl; -(C1—C4 aikyl)—Cg—Cg cycioalkyl; C1-Cg alkoxy optionally substituted by one or more halogen, —NR9R11 or OH; OH; CN; halogen; ~(Co-C4 alkyl)—N RQR”; -(co-c4 awn-0021215; «co-c:4 alkyl)-C(O)NR9R“; - (Co-C4 alkyl)-Ca-C14 aryl; or —(Co—C4 alkyl)—3 to 14 membered cyclyl; wherein the cycloalkyl, 4 -CG-C14 aryl and -(Co-C4 alkyl)~3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; R3 is H; 01-08 alkyl optionally substituted by one or more OH, -NR9R11 or C1-C4 alkoxy; C1-C3 haloalkyl; C2-03 l substituted by one or more halogen, OH, - NRQR“, or C1-C4 alkoxy; C3-C10 cycloalkyl; ~(C1-C4 alkyi)-C3-Ca cycloalkyl; C1-C8 alkoxy optionally substituted by one or more halogen, 1 or OH; OH; CN; n; -(Co-C4 alkyl)—NR9R”; ~(Co—C4 alkyl)—COZR15; -(CO-C4 alkyl)-C(O)NR9R“; - (Co—C4 alkyl)—Cs—C14 aryl; or -(Co—C4 alkyl)—3 to 14 membered heterocyolyl; wherein the cycloalkyl, -(Co—C4 —Ca-C14 aryl and -(Co—C4 alkyl)~3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; each Z3 is independently OH; (Co—C4 alkyl)—C5 aryl; O-Ca aryl; C1—C4 alkyl optionally substituted by one or more OH, ON or —NR193R213; C1-C4 haloalkyl; 01-04 alkoxy optionally substituted by one or more OH, 'COQR193, -NR19E‘R21a or C1—C4 ; — NR18aC(O)R21a; —C(O)NR198R213; _NR18aC(O)NR1QaR21a; _NR193R21a; —(Co—C4 alkyl)- 13‘“; -(co—c4 alkyl)-C(0)R‘9a; oxo; CN; N02; halogen; «co—cl, alkyl)-4 to 6 membered cyclyl; or —O-(4 to 6 membered heterocyclyl); wherein the (Co-C4 —Co aryl, O-Cs aryl, -(Co—C4 alkyl)—4 to 6 membered heterocyclyl and —O—(4 to 6 membered heterocyclyl) are each optionally substituted by OH, halogen, C1-C4 alkyl, C1—C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens; R4 is H; R5 is H, C1-C4 alkyl or C1—C4 haloalkyl; R6 is selected from C1-C8 alkyl optionally substituted by one or more C1—C4 alkoxy or —NR19R21; c1-c8 haloalkyl; ~(Co-C4alkyl)—C3-CecycloaIkyl; C1-Cgalkoxy optionally tuted by one or more halogen atoms; 21; —(Co—C4 alkyt)—Ce-C14aryl; and — (Co-C4 alkyI)-3 to 14 membered cyclyl; wherein the —(Co-C4a|kyl)-C3- Cgcycloalkyl, —(Co-C4 alkyl)—Ce-C14ary| and -(Co—C4 alkyl)—3 to 14 heterocyclyl are each optionally substituted by one or more Z substituents; 2O each Z is independently ed from (Co—C4 -Cs aryl; O-Cs aryl; Cl-C5 alkyl optionally substituted by one or more 01-05 alkoxy, ON or -NR19R21; 01—05 kyl; C1~C5 alkoxy optionally substituted by one or more -NR19R21 or C1-C4 alkoxy; - NR‘QRZ‘; (co-c4 alkyl)—C(O)R19; CN; halogen and (co-c:4 alkyl)—4 to 6 membered cyclyl; and wherein the aryl and heterocyclyl are each optionally substituted by one or more halogen, 01-05 alkyl, 01—06 haloalkyl and 01-05 alkoxy optionally substituted by one or more ns; R9 and R" are each independently selected from H; 01—05 alkyl optionally substituted by one or more C1-C4 alkoxy or OH; 01-06 haloalkyl; -(Co-C1a|kyl)—C3-Cs lkyl; (C0-C4 alkyl)- C6-C14aryl optionally substituted by one or more groups selected from C1-C6 alkyl, C1-Cs alkoxy and halogen; and (Co-C4 alkyl)- 3- to 14—membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, C1-C5 alkyl and C(O)Cl~C5 alkyl; or R9 and R“ together with the nitrogen atom to which they are attached form a 5- to -membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from OH; halogen; phenyl, 5- to 10—membered heterocyclyl; C1~Cs alkyl; C1—C5 haloalkyl; 01-05 alkoxy optionally substituted by one or more OH or C1-C4 alkoxy; and C(O)OC1-Caalkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by 01-05 alkyl, C1-Cs haloalkyl or C1-C5 alkoxy; R15 is selected from H; C1-Cs alkyl; C1-Cs haloalkyl; 03-010 cycloalkyl; (C4alkyl)- Cg'Cg cycloalkyl; -(Co-C4 alkyl)-C5-C14aryl and -(Cg-C4 alkyl)—3 to 14 membered heterocyclyl group; wherein the C3—C10 cycloalkyl, (-C1-C4alkyl)—Ca~Cg cycloalkyl, -(Co- C4 alkyl)—Ca—C14aryl and —(Co—C4 alkyl)—3 to 14 membered heterocyclyl groups are each optionally substituted by one or more Z substituents; R183 is independently H or 01—05 alkyl; R193 and R21a are each independently H; C1-C5 alkyl optionally substituted by one or more C1-C4 alkoxy,— NR22R23‘, or OH; c1-c6 haloalkyl; —(Co-C1alkyl)-Cg~Cscycloalkyl; - (Co-C4 alkyl)— C5—C14aryl optionally substituted by one or more groups selected from C1-C5 alkyl, 01—05 alkoxy and halogen; or -(Co-C4 alkyl)~ 3- to 14-membered heterocyclyl optionally substituted by one or more groups ed from halogen, oxo, 01-06 alkyl and C(O)C1~Cs alkyl; or R19‘1 and R213 together with the nitrogen atom to which they attached form a 5- to 10- membered cyclyl, which heterocyclyl es 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from OH; halogen; phenyl; 5- to 10-membered heterocyclyl; 01-05 alkyl; C1—Ce haloalkyl; C1-C6 alkoxy optionally tuted by one or more OH or C1—C4 alkoxy; and C(O)OC1-Cealkyl; wherein the phenyl and heterocyclyl tuent groups are themselves optionally substituted by C1—Ce alkyl, C1-Cs haloalkyl or C1-C5 ; R18 is ndently H or C1-C5 alkyl; R19 and R21 are each independently C1—Ce alkyl ally substituted by one or more C1-C4 alkoxy; C1—Cs haloalkyl; ~(Co-C1alkyI)-Cg-Cscycloalkyl;- (Co-C4 —CG-C14aryl optionally substituted by one or more groups selected from C1—C6 alkyl, 01—05 alkoxy and halogen; or -(Co-C4 — 3- to 14—membered heterocyclyl, ally substituted by one or more groups selected from halogen, 01-06 alkyl and -C(O)C1—C6 alkyl; or R19 and R21 together with the nitrogen atom to which they attached form a 5- to 10— membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally tuted by one or more substituents ed from halogen; phenyl; 5— to 10—membered heterocyclyl; 01-05 alkyl; 01-06 haloalkyl; 01-05 alkoxy optionally substituted by one or more C1-C4 alkoxy and C(O)OC1-Cealkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by a substituent selected from C1-C5 alkyl, C1— Cs haloalkyl and C1-Ce alkoxy; and R22 and R23 are each independently H or C1—C6 alkyl.
Embodiment 3: A compound of formula (I), according to Embodiment 1 or Embodiment2, n R1 is C1-C4 alkyl, C1-C4 alkoxy, ON or n.
Embodiment 4: A compound of formula (1), according to any preceding Embodiment, wherein R1 is C1-C4 alkyl or halogen. ment 5: A compound of formula (l), ing to any preceding Embodiment. 1O wherein R1 is methyl or halogen.
Embodiment 6: A compound of formula (l), according to any preceding Embodiment, wherein R1 is methyl or F.
Embodiment 7: A compound of formula (I), according to any preceding Embodiment, wherein R13 is H, methyl or F; particularly R1a is H. ment 8: A compound of formula (I), according to any preceding Embodiment, wherein X is N.
Embodiment 9: A compound of formula (I), according to any one of Embodiments 1 to 7, wherein X is CH.
Embodiment 10: A compound of formula (I), according to any preceding Embodiment, n R2 is H; C1~Cs alkyl optionally substituted by one or more OH, ~ NR9R1‘or C1-C4 alkoxy; 01—06 haloalkyl; (32—06 alkynyl tuted by one or more halogen, OH, -NRQR11 or C1-C4 alkoxy; Cg'CB cycloalkyl; -(C1—C4 alkyl)—Ca-Cs cycloalkyl; C1-C5 alkoxy optionally substituted by one or more halogen, ~NR9R‘1 or OH; OH; CN; halogen; -(Co-C4 -NR9R“; 4 alkyl}COzR15; -(C0—C4 alkyl)- C(O)NR19R21; -(co-c4 alkyl)—Cs~C14aryl; or -(cO—c4 alkyl)—3 to 14 membered heterocyclyl; wherein the Cs-Cecycloalkyl, -(CO-C4 alkyl)—C6-C14aryl and -(Co-C4 alkyl)- 3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents. ment 11: A compound of formula (I), according to any ing Embodiment, wherein R2 is H; C1-C4 alkyl optionally substituted by one or more OH, - NR9R11or C1-C4 alkoxy; C1-C4 haloalkyl; C2-C5 alkynyl tuted by one or more halogen, OH, -NR9R“, or c1-c4 alkoxy; 03-05 cycloalkyl; —(c1-c4 alkyl)—Cg-Ce cycioalkyi; 01—05 alkoxy optionally substituted by one or more halogen, -NRQR11 or OH; OH; CN; halogen; —(c0—c4 alkyl)—NR9R“; -(co-c4 alkyi}COzR15; —(Co—C4 alkyl)— C(O)NR19R“; -(Co-C4 alkyl)-C5—C14aryi; or —(Co-C4 aikyi)-3 to 14 membered heterocyclyi; wherein the C3-Cscycloalkyi, -(Co-C4 -Ce-C14aryl and —(Co-C4 alkyl} 3 to 14 cyclyi are each optionally substituted by one or more Za substituents. ment 12: A compound of formula (l ), according to any preceding Embodiment, wherein R2 is H; C1—C4 alkyi ally tuted by one or more OH or ~NR9R“; Cl—C4 haioalkyl; C1-C4 alkoxy optionally substituted by one or more halogen, -NR9R11 or OH; OH; CN; halogen; —(c0-c4 alkyI)-NR9R”; ~(Co—C4 alkyl)— C(O)NR9R"; phenyl; or -(Co—C4 alkyl)—5 to 6 membered heterocyclyi; wherein the phenyl and -(Co—C4 aikyl)—5 to 6 heterocyclyi are each optionally substituted by one or more Za substituents.
Embodiment 13: A nd of formula (1), according to any preceding ment,wherein R2 is H; C1—C4 alkyl optionally substituted by one or more OH or -NH2; C1-C4 alkoxy ally substituted by one or more -NR9RH or OH; F; Br; —(C1- CZ alkyl)—NR9R“; —C(O)NR9R“; phenyl; or —(Co—C4 aikyi)—5 to 6 ed heterocyclyl; wherein the phenyl and —(Co-C4 alkyl)—5 to 6 heterocyclyl are each optionally substituted by one or more 2" substituents.
Embodiment 14: A compound of formula (I), according to any preceding Embodiment,wherein R2 is H; C1—C4 alkyl optionally substituted by one or more OH or -NH2; C1—C4 alkoxy optionally substituted by -NR9R”; F; Br; 2 aikyl)—NR9R"; — C(O)NR9R“; phenyl; or -(Co—C4 aiky|)-5 to 6 membered heterocyclyi; wherein the phenyl and —(Co—C4 alkyI)—5 to 6 heterocyclyi are each optionally substituted by one or more Za substituents.
Embodiment 15: A compound of formula (1), according to any preceding Embodiment,wherein R2 is H; C1-C4 alkyl optionally substituted by one or more OH; C1-C4 alkoxy optionally substituted by -NR9R11; F; Br; HR“; phenyl; or -(Co-C4 —5 to 6 membered heterocyclyi; wherein the phenyl and -(Co-C4 —5 to 6 membered heterocyclyi are each optionally substituted by one or more Za substituents.
Embodiment 16: A compound of formula (I), according to any preceding Embodiment, n R2 is phenyl or -(Co—C4 alkyl)—5 to 6 membered heterocyclyl, each optionally substituted by one, two or three Za substituents.
Embodiment 17: A compound of a (I), according to any preceding Embodiment, wherein R2 is phenyl or 5- or 6-membered heterocyclyl, each ally substituted by one, two or three Za substituents.
Embodiment ‘18: A compound of formula (I), according to any one of Embodiments 1 to 15, wherein R2 is H, F, Br, /‘<\/Q\\ OH 1‘ F C F O l [Elm/OH ;t\\ , gtifiYkH/k/DH“a.
H 6*“H ;‘\\\ /“\“ / /——/ urn Na—N/J N\N Vt kl. ' 5? g2/ /‘ Embodiment 19: A compound of formula (l), according to any preceding Embodiment, wherein R3 is H; C1—C4 alkyl optionally substituted by one or more OH, - NRgRflor C1-C4alkoxy; C1—C4 haloalkyl; C2-C5 alkynyl substituted by one or more halogen, OH, -NRQR11 or lkoxy; 03-06 cycloalkyl; -(C1-C4 alkyl)-Cg-Cg cycloalkyl; -C1—C4alkoxy optionally substituted by one or more halogen, -NR9R11 or OH; OH; CN; halogen; —(Co-C4 alkyl)-NR9R“; -(cQ-c4 alkyl)—C02R15;-(Co-C4 alkyl)- C(0)NR9R“; —(co-c4 -Cearyl; or {co-ca, alkyl)-5 to 6 ed heterocyclyl; wherein the 03-05 cycloalkyi, -(Co—C4 alkyl)-Caaryl and ~(Co-C4 alkyl)—5 to 6 heterocyclyl are each optionally tuted by one or more Za substituents.
Embodiment 20: A compound of formula (I), according to any preceding Embodiment, wherein R3 is H; C1—C4 alkyl optionally substituted by one or more OH or —NR9R“; C1-C4 haloalkyl; 03-05 cycioalkyl; C1-C4alkoxy optionally substituted by one or more halogen or ~NR9R“; OH; CN; halogen; —(Co-C4 alkyl)—C(O)NR19R21; phenyl; or -5 to 6 ed cyclyl, wherein the Cst lkyl, phenyl and 5 to 6 cyclyl are each optionally substituted by one or more Za substituents.
Embodiment 21: A compound of formula (I), according to any preceding Embodiment, wherein R3 is H; C1-C4 alkyl; -C1—C4alkoxy; OH; CN; halogen; - C(O)NR9R“; phenyl or -5 to 6 ed heterocyclyl; wherein the phenyl and 5 to 6 heterocyclyl are each optionally substituted by one or more 22‘1 substituents.
Embodiment 22: A compound of formula (l), according to any preceding Embodiment, n R3 is H; C1—C4 alkyl; -C1-C4 alkoxy; halogen or —C(O)NR9R”.
PCT/IBZOIZ/054501 Embodiment 23: A compound of a (I), according to any preceding ment, wherein R3 is H.
Embodiment 24: A compound of formula (I), according to any preceding Embodiment, wherein R4 is H.
Embodiment 25: A nd of a (l), according to any ing Embodiment, wherein R5 is H, C1—C4 alkyl or C1-C4 haloalkyl.
Embodiment 26: A compound of formula (1), according to any preceding Embodiment, wherein R5 is H or methyl.
Embodiment 27: A compound of a (l), according to any preceding Embodiment, wherein R5 is H.
Embodiment 28: A compound of formula (I), according to any preceding Embodiment, wherein R4 is H and R5 is H.
Embodiment 29: A compound of formula (l), according to any preceding Embodiment, wherein R6 is 01-05 alkyl ally substituted by C1—C4 alkoxy; C1-Ce haloalkyl; ~(Co—C4alkyl)—Cg—Cs cycloalkyl; Ci-C4 alkoxy optionally substituted by one or more halogen atoms; —NR19R21; -(Co—C4 alkyl)—Cs-C14aryl or —(Co~C4 alkyl)-3 to 14 membered heterocyclyl; wherein the cycloalkyl, -(Co-C4 alkyl)—Ce—C14 aryl and -(Co-C4 alkyl)—3 to 14 heterocyclyl are each optionally substituted by one or more Z substituents.
Embodiment 30: A compound of formula (l), according to any preceding Embodiment, wherein R6 is 01-05 alkyl optionally substituted by one or more C1-C4 3O alkoxy; C1-C6 haloalkyl; alkyl)—Ca-Cscycloalkyl; C1—C4 alkoxy optionally substituted by one or more halogen atoms; —NR19R21; 4 alkyl)—Cs—C1oaryl or -(Co- C4 alkyl)-5 to 6 membered heterocyclyl; wherein the —(Co-C1alkyl)-Cs-Cecycloalkyl, - (CO-C4 alkyl)-C5—C14aryl and ~(Co—C4 alkyl)—5 to 6 membered heterocyclyl are each optionally tuted by one or more Z tuents.
Embodiment 31: A nd of formula (I), according to any preceding Embodiment, wherein R6 is 01—05 alkyl optionally substituted by C1—C4 alkoxy; -(Co— Czalkyl)—Cg-Cecycloalkyl; C1-C4alkoxy; -Ca~C1oaryl or -(CO-Cz alkyl)—5 to 6 membered heterocyclyl; wherein the —(Co—Czalkyl)-C3—Cecycloalkyl, —(CO-02 alkyl)-Ce—C1oaryl and — (Co-Czalkyl)-5 to 6 membered heterocyclyl are each optionally substituted by one or more Z substituents.
Embodiment 32: A nd of formula (l), according to any preceding Embodiment, wherein R6 is C1-Ca alkyl optionally substituted by C1-C4 ; —(Co— C1alkyl)-cyclohexyl; phenyl or 1 alkyl)-5 to 6 membered heterocyclyl; wherein the -(Co~C1alkyl)—cyclohexyl, phenyl and -(Co-C1 ~5 to 6 membered heterocyclyl are each optionally substituted by one or more Z substituents.
Embodiment 33: A compound of formula (I), according to any one of Embodiments 1 to 30,wherein R6 is C1-C4 alkyl ally substituted by one or more C1-C4 alkoxy; phenyl; C1-C4 haloalkyl; tetrahydrofuran; pyrrolidine, yrrolidine or -CH2- piperidine; wherein phenyl, tetrahydrofuran, pyrrolidine, -CH2—pyrrolidine and -CH2- piperidine are each optionally substituted by one or more Z substituents.
Embodiment 34: A compound of formula (l), according to any one of Embodiments 1 to 30, wherein R5 is -(Co-C2 alkyl)—5 to 6 membered heterocyclyl, optionally substituted by one, two or three Z substituents.
Embodiment 35: A compound of formula (l), according to any one of Embodiments 1 to 30, wherein R6 is ZOlZ/054501 " F -..I-_ N >,. o I I ! " \—0 or \<.
Embodiment 36: A compound of formula (1), according to any one of Embodiments ‘l to 30, wherein R6 is Embodiment 37: A nd of formula (l), according to any preceding ment, wherein each Z3 is independently OH; ~(Co-C4 alkyl)—C5 aryl; -O—Ca aryl; C1—C4 alkyl optionally substituted by one or more OH, ON or ~NR193R213; Ci-C4 haloalkyl; C1—C4 alkoxy optionally substituted by one or more OH, —COZR193, - NR1QaR21a or 01-04 alkoxy; -NR1BQC(O)R213; ~C(O)NR193R213; -NR18aC(O)NR‘QaR21a; — NR193R213; -(c0-c4 alkyl)—C(O)OR188; -(co—c4 —C(O)R1Qa; oxo; CN; N02; halogen or ~(Co—C4 alkyl)-4 to 6 membered cyclyl; wherein the -(Co-C4 alkyl)—Ce aryl, -O- 06 aryl and -(Co-C4 alkyl)—4 to 6 membered heterocyclyl are each optionally substituted by halogen, C1-C4 alkyl, C1-C4 haloalkyl or C1—C4 alkoxy optionally substituted by one or more halogens. 2O Embodiment 38: A compound of formula (I), according to any preceding Embodiment, n each Z8 is independently OH; C1—C4 alkyl optionally substituted by one or more OH or ~NR‘93R213; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more OH, C1—C4 alkoxy or —NR‘93R213; —C(O)NR‘93R213; CN; halogen or —(Co~ C4 alkyl)—4 to 6 membered cyclyl; wherein the -(Co-C4 alkyl)—4 to 6 membered heterocyclyl are each optionally substituted by halogen, C1—C4 haloalkyl, or C1—C4 alkoxy optionally substituted by one or more halogens.
Embodiment 39: A compound of formula (I), according to any preceding Embodiment, wherein each Z3 is independently C1~C4 alkyl optionally substituted by one or more OH or ~NR193R213; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more OH, C1—C4 alkoxy or -NR193R213; -C(O)N R193R21a; halogen or ~(Co—C4 alkyl)~4 to 6 membered heterocyclyl; n the —(Co-C4 alkyl)—4 to 6 ed heterocyclyl is optionally substituted by halogen, C1-C4 alkyl or C1-C4 haloalkyl.
Embodiment 40: A compound of formula (l), according to any preceding Embodiment, wherein each 2a is independently C1—C4 alkyl ally substituted by one or more OH or -NR193R21a.
Embodiment 41: A compound of formula (l), according to any one of Embodiments 1 to 39, wherein each Z3 is independently fluorine, bromine, chlorine, , y, N ., -CH2NH2, ‘ H '. V , , , 1 or |\/O.
Embodiment 42: A compound of formula (I), according to any ing Embodiment, wherein each Z is independently -(Co-C4 alkyl)-Cs aryl; -O-Ce aryl; C1- 04 alkyl optionally substituted by one or more ON or —NR19R21; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more ~NR19R21 or C1-C4 ; ~NR‘9R21; - (Co-C4 —C(O)R19; CN; n or -(Co—C4 alkyl)-4 to 6 membered heterocyclyl; wherein the -(C0—C4 alkyl)—Cs aryl, —O—Ce aryl and —(Co—C4 alky|)-4 to 6 membered cyclyl are each optionally substituted by halogen, C1—C4 alkyl, C1-C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens.
Embodiment 43: A compound of formula (l), according to any preceding Embodiment, wherein each 2 is independently C1-C4 alkyl optionally substituted by one or more NHZ; C1-C4 kyl; C1-C4 alkoxy optionally substituted by one or more C1-C4 alkoxy or —NR19R2‘; CN; halogen or -(Co-C4 alkyl)—4 to 6 membered heterocyclyl; wherein the ~(Co-C4 alkyl)—4 to 6 membered heterocyclyl is optionally substituted by n, C1-C4 alkyl, C1-C4 haloalkyl, or C1—C4 alkoxy optionally substituted by one or more halogens.
Embodiment 44: A compound of formula (I), according to any preceding Embodiment n each Z is independently C1—C4 alkoxy, halogen, C1—C4 alkyl or C1-C4 kyl.
Embodiment 45: A compound of formula (l), ing to any preceding Embodiment, wherein R9 and R11 are each independently H; 01-05 alkyl optionally substituted by C1-C4 alkoxy or OH; 01—05 haloalkyl; -(Co-C1alkyl)-Ca-Cscycloalkyl; (Co- C4 alkyl)- Ca—C14aryl optionally substituted by one or more groups selected from 01-06 alkyl, C1—Ce alkoxy and halogen; or ~(Co—C4 alkyl)— 5- to 6-membered cyclyl optionally substituted by one or more groups selected from halogen, 0x0, C1—C5 alkyl and C(O)C1-Ce alkyl.
Embodiment 46: A compound of formula (I), according to any one of ments 1 to 44, wherein R9 and R“ together with the nitrogen atom to which they attached form a 5- to 6-membered heterocyclyl, the heterocyclyl including 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from OH; n; phenyl; 5- to 10-membered heterocyclyl; C1—C5 alkyl; 01—06 haloalkyl; 01—05 alkoxy optionally substituted by one or more OH or C1-C4 alkoxy; and C(O)OC1—Cfia|kyl; wherein the phenyl and heterocyclyl tuent groups are themselves optionally substituted by C1—C6 alkyl, 01—05 kyl or C1-C5 alkoxy.
Embodiment 47: A compound of formula (I), according to any preceding Embodiment, wherein R15 is H; C1-C4 alkyl; c1-c4 haloalkyl; cs-c10 cycloalkyl; +01— C4alkyl)-Cg-C8 cycloalkyi; -(Co~C4 alkyl)—Ce—C14aryi or 4 alkyl)—3 to 14 membered heterocyclyl group; wherein the 03—010 cycioalkyl, —(—C1—C4alkyI)-C3-Ca cycloalkyl, - (Co—C4 alkyl)—C6—C14aryl and -(Co-C4 -3 to 14 membered heterocyclyl groups are each optionally substituted by one or more Z substituents.
Embodiment 48: A compound of formula (i), according to any preceding Embodiment, wherein R15 is H or C1-C4 alkyl.
Embodiment 49: A nd of a (l), according to any preceding Embodiment, wherein R18 is independently H or C1-C4 alkyl. ment 50: A compound of formula (l), according to any preceding Embodiment, wherein R19 and Ft21 are each independently C1-Cs alkyl optionally substituted by one or more C1—C4 alkoxy; C1—Ce haloalkyl; -(Co-C1a|kyl)-Cg- Cscycloalkyl; (C0-C4 alkyl)-aryl optionally substituted by one or more groups selected from 01-06 alkyl, C1-05 alkoxy and halogen; or (Co—C4 alkyl)— 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, C1- C6 alkyl and -Cs alkyl.
Embodiment 51: A compound of formula (I), according to any one of Embodiments 1 to 49, wherein R19 and R21 together with the nitrogen atom to which they are attached form a 5— to 6—membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from halogen; phenyl; 5- to 10~membered heterocyclyl; C1—C4 alkyl; C1-C4 haloalkyl; C1-Cs alkoxy optionally tuted by one or more OH or C1-C4 alkoxy and C(O)OC1-C6alkyl; wherein the phenyi and heterocyclyl tuent groups are themselves optionally tuted by 01-06 alkyl, C1~Cs haloaikyl or 01-05 alkoxy. 3O Embodiment 52: A compound of formula (i), according to any preceding Embodiment, wherein R18a is independently H or C1-C4 alkyl.
Embodiment 53: A compound of formula (l), according to any ing Embodiment, n R193 and R213 are each independently H; 01-05 alkyl optionally substituted by C1-C4 alkoxy or OH; C1-C5 haloalkyl; -(Co-C1alkyl)-Cg-Cscycloalkyl; (C0- C4 alkyl)~aryl optionally substituted by one or more groups selected from 01—05 alkyl, 01-05 alkoxy and halogen; or (Co—C4 alkyl)— 5- to ered cyclyl optionally PCT/IBZOIZ/054501 substituted by one or more groups ed from halogen, oxo, C1-C6 alkyl and C(O)C1-Ce alkyl. ment 54: A compound of formula (I), according to any one of Embodiments 1 to 52, wherein R1961 and R215‘ together with the nitrogen atom to which they attached form a 5- to 6—membered cyclyl which heterocyclyl includes 0 to 3 further atoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from OH; halogen; phenyl; 5— to 10—membered heterocyclyl; C1-C4 alkyl; C1-C4 haloalkyl; C1-C6 alkoxy optionally substituted by one or more OH or C1—C4 ; and C(O)OC1-Cealkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by C1—C5 alkyl, 01-06 haloalkyl or 01-05 alkoxy.
Embodiment 55: A compound of formula (l), according to any preceding Embodiment, wherein the compounds are represented by a II: 35qu "H. [3“].([1 R13? . LE... )5ng Jf, W RS ,- (ll).
Embodiment 56: A compound of formula (II), or a pharmaceutically acceptable salt thereof: (II) wherein R1 is C1-C4 alkyl, C1-C4 , ON or halogen; R12‘ is H, halogen, C1—C4 alkyl or C1-C4 haloalkyl; X is N or CH; R2 is H; C1-C4 alkyl optionally substituted by one or more OH, -NR$’R11 or C1-C4 alkoxy; C1—C4 haloalkyl; 02—05 alkynyl substituted by one or more halogen, OH, - NRQR“ or C1—C4 alkoxy; 03-05 cycloalkyl; —(C1-C4 alkyl)—Cg-Cs cycloalkyl; 01-06 alkoxy optionally substituted by one or more halogen, —NR9R11 or OH; OH; CN; halogen; - (co-c4 alkyl)—NR9R”; —(Cg—C4 alky|)—COQR15; -(Co—C4 alkyl)—C(O)NR9R”; -(c:o-c4 alkyl)- Cs-C14aryl; or 4 alkyl)—3 to 14 membered heterocyclyl; wherein the Cg- Cecycloalkyl, -(Co-C4 alkyl)—C5—Ci4aryl and -(Co—C4 —3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; R6 is 01-06 alkyl optionally substituted by one or more C1-C4 alkoxy; C1—C5 haloalkyl; - (Co-C1alkyl)-Cg~Cecycloalkyl; C1-Ca alkoxy optionally substituted by one or more halogen atoms; ~NR‘9R21; {Co-C4 alkyl)—C5-C1oaryl; or ~(C0-C4 alkyi)—5 to 6 membered heterocyclyl; wherein the ~(Co—C1alky|)—Cg-Cecycloalkyl, ~(Co-C4 alkyl)-Ce—C14aryl and — (Co-C4 alkyl)-3 to 14 heterocyclyl are each optionally tuted by one or more Z substituents. each Z21 is independently OH; C1—C4 alkyl optionally tuted by one or more OH or N H2; C1-C4 haloalkyl; C1-C4 alkoxy ally substituted by one or more OH, 01-04 alkoxy or -NR193R213; -C(O)NR193R213; CN; halogen or 4 alkyl)-4 to 6 membered heterocyclyl; wherein the — heterocyclyl is optionally substituted by halogen, C1—C4 alkyl optionally substituted by one or more halogen atoms, or C1—C4 alkoxy optionally substituted by one or more halogens; each Z is independently C1—C4 alkyl optionally substituted by one or more NH2; C1-C4 haloalkyl; C1-C4 alkoxy ally substituted by one or more C1-C4 alkoxy or — NR19R2‘; CN; halogen or 4 alkyl)—4 to 6 membered heterocyclyl; wherein the heterocyclyl contains at least one heteroatom selected from N, O and S, wherein the heterocyclyl is optionally substituted by n, C1—C4 alkyl, C1-C4 haloalkyl or 01—04 alkoxy optionally substituted by one or more halogens; R9 and R“ are each ndently H; 01-05 alkyl optionally substituted by one or more C1-C4 alkoxy or OH; 01-05 haloalkyl; -(Co—C1alkyl)—C3-Csoycioalkyl; -(Co-C4 alky|)- 06—014aryl optionally substituted by one or more groups selected from 01-06 alkyl, 01—06 alkoxy and halogen; or -(CO—C4 — 5- to 6—membered cyclyl optionally substituted by one or more groups ed from halogen, oxo, 01-05 alkyl and -C(O)C1-Ce alkyl; R15 is H or C1—C4 alkyl; PCT/IBZOIZ/054501 R19 and R21 are each independently 01-06 alkyl optionally substituted by one or more C1—C4 alkoxy; 01-05 haloalkyi; -(C0—C1aIkyl)—Cg—Cscycloalkyl; —(C0-C4 — aryl optionally substituted by one or more groups selected from 01-05 alkyl, C1-Cs alkoxy and halogen; or —(Co-C4 alkyl)— 5- to 6—membered heterocyclyl optionally substituted by one or more groups selected from halogen, C1-C5 alkyl and —C(O)C1—Ce alkyl; R193 and R21:11 are each ndently H; C1-Ce alkyl optionally substituted by one or more C1—C4 alkoxy or OH; 01—05 haloalkyl; -(Co-C1alkyl)—03~Cecycloaikyl; (Co—C4 alkyl)— aryl optionally substituted by one or more groups selected from 01-05 alkyl, C1-Ce alkoxy and halogen; or (Co-C4 alkyl)— 5- to 6—membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, C1'Cs alkyl and C(O)C1—Cs alkyl.
Embodiment 57: A compound of formula (1), according to any one of Embodiments 1 to 54, wherein the compounds are represented by formula (Ht): (ill). ment 58: A compound of a (til), or a pharmaceuticaily acceptable salt thereof: H [gN‘t N Ex. ’N Fiat-ilk;”r' HI-fl/ Ill] D L R2 (Ill) wherein R1 is C1-C4 alkyl, C1-C4 alkoxy, ON or halogen; R16 is H, halogen, Ci—C4 alkyl or C1-C4 kyl; X is N or CH; R2 is H; C1-C4 alkyl optionally substituted by one or more OH, -N RQR11 or C1-C4 alkoxy; C1—C4 haloalkyl; Cz-Cs l substituted by one or more halogen, OH, - NRQR11 or C1—C4 alkoxy; Cs—Ce cycloalkyl; -(C1-C4 alkyl)—Cs-Ca cycloalkyl; C1—C4 alkoxy optionally substituted by one or more halogen, -N RQR11 or OH; OH; CN; halogen; - (co-c4 alkyI)-NR9R“; ~(co-c4 alkyl)—COZR15; -(c0-c4 alkyl)—C(O)NR9R“; 4 — aryl; or —(Co—C4 alkyl)-3 to 14 membered heterocyclyl; wherein the Cg- Cecycloalkyl, 4 —C6-C14aryl and ~(Co-C4 alkyl)—3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; R6 is 01-06 alkyl ally substituted by one or more 01-04 alkoxy; C1-C4 haloalkyl; — (C0—C1alkyl)—Cg-Cecycloalkyl; C1-C4 alkoxy optionally substituted by one or more halogen atoms; -N R19R21; -(C0-C4 alkyl)-Ce-C10aryl or -(Co-C4 alkyl)—5 to 6 membered heterocyclyl; wherein the —(Co—C1alkyl)—C3—Cacycloalkyl, —(Co—C4 alkyl)—Cs-C1oaryl and - (Co-C4 alkyl)—5 to 6 ed cyclyl are each optionally substituted by one or more Z substituents; each Z6‘ is independently OH; C1—C4 alkyl optionally substituted by one or more OH or NH2; C1-C4 alkyl; C1-C4 haloalkyl; C1—C4 alkoxy optionally substituted by one or more OH, c,-c4 alkoxy or -NR193R213; —C(0)NR193R213; CN; halogen or -(Co-C4 alkyl)—4 to 6 membered heterocyclyl; n the heterocyclyl is optionally substituted by halogen, C1—C4 alkyl, C1—C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens; each Z is independently C1-C4 alkyl optionally substituted by one or more NHZ; C1—C4 alkyl; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more C1—C4 alkoxy or —NR’9R21; CN, halogen or 4 alkyl)-4 to 6 membered heterocyclyl; wherein the heterocyclyl is optionally substituted by halogen, C1—C4 alkyl, C1~C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens; R9 and R11 are each independently H; C1-Cs alkyl optionally substituted by one or more C1—C4 alkoxy or OH; C1-C5 haloalkyl; -(CO—C1alkyl)-C3-Cecycloalkyl; -(Co—C4 alkyl)— Ce-C14aryl optionally substituted by one or more groups selected from C1-C5 alkyl, (31-06 alkoxy and halogen; or -(Co-C4 alkyl)— 5— to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, C1-C6 alkyl and 1-Cb- alkyl; R15 is H or C1—C4 alkyl; R19 and R21 are each independently 01-05 alkyl optionally substituted by one or more C1—C4 alkoxy; C1—Ce haloalkyl; —(Co-C1alkyl)—Cg-Cacycloalkyl; —(Co-C4 alkyl)— Ce~C14aryl optionally substituted by one or more groups selected from 01—05 alkyl, (31—05 alkoxy and halogen; or ~(Co-C4 alkyl)— 5- to ered cyclyl optionally substituted by one or more groups selected from halogen, C1—Ce alkyl and —C(O)C1—06 alkyl; R193 and R218 are each independently H; C1-C5 alkyl ally substituted by one or more C1—C4 alkoxy or OH; 01-05 haloalkyl; -(Co-C1atkyl)—Cg-Cacycloalkyl; (Co-C4 alkyl)— aryl optionally substituted by one or more groups selected from C1—Ce alkyl, 01—05 alkoxy and halogen; or (Co-C4 alkyl)— 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, (31—05 alkyl and C(O)Ci-C5 alkyl. 1O Embodiment 59: A compound of formula (I), (I) or (III), according to any preceding Embodiment, wherein x, R1, R13, R2, R3, R4, R5, R6, 2, 2a R15, R18, R19 and R19 are those defined by the Embodiments above or by the Examples section below.
Embodiment 60: A compound of formuia (l), which is ed from: N—(2-Fluoro(2—(4-methylpiperazin~1-yl)benzylcarbamoyl)phenyI)—7-(1-methyl-1H— pyrazolyl)imidazo[1,2—a]pyridinecarboxamide; 7-(3-Fluoro(2-hydroxyethylcarbamoyl)phenyl)-N-(2-fluoro(2-(4-methylpiperazin- 1—y|)benzylcarbamoyi)phenyi)imidazo[1,2-alpyridine-S—carboxamide; o-N-(2-methyl(2—(4-methylpiperazin-1— y|)benzylcarbamoyl)phenyi)imidazo[1,2-a]pyridinecarboxamide; 7-(1 ~MethyI-1 H—pyrazoI—S—yl)—N-(2-methyl~5-(2—(4—methylpiperazin-1 ~ yl)benzylcarbamoyl) phenyl)imidazo[1,2—a]pyridine—3-carboxamide; N-(2-Fluoro(2-(4—methylpiperazin-t -yl)benzylcarbamoyl)phenyl)-7—(pyridine~3- yl)imidazo[1,2—a]pyridinecarboxamide; N-(2—Flu0r0(2-(4—methyipiperazinyl)benzylcarbamoyl)phenyl)~7-(1~methyl-1 H— pyrazolyl)imidazo[1,2-a]pyridine—3-carboxamide; N-(5-(3,4—difluorobenzylcarbamoyl)—2—tluorophenyl)(1-methyl-1H-pyrazolyl) imidazo [1,2—a]pyridine -3—carboxamide; N—(5-(benzylcarba moyi)—2-fluoropheny|)—6—(1~methyI-1H-pyrazoly|)imid azo[1,2- a]pyridinecarboxamide; N-(4-fluoro—2—methyi—5-(2—(4-methyl piperazinyl) benzylcarbamoyl)phenyl)—7— (pyridineyl)imidazo[1,2-a]pyr idinecarboxamide; N-(5-(3,4—difluorobenzylcarbamoyl)—2—f|uorophenyI)—7—(6-(3-(dimethyl amino) propoxy) pyridineyi)imid 2-a]pyridinecarboxamide; N-(5-(2-(2.6-cis-dimethylpiperidin—t-yl)ethyicarbamoyl)—2-fluorophenyl)—7-(1-methyl- 1 H-pyrazol-S—yl)imidazo[1,2-a]pyridine-3—carboxamide; WO 30802 7-(4—(aminomethyl) )-N—(2-fluoro—5—(2—(4-methylpiperazin-1 - yl)benzy|carbamoyl) phenyl)imidazo[1,2-a]pyridine-3—carboxamide; N—(5-(2-tert—butoxyethyloarbamoyl)—2—fluorophenyl)—7—(1—(2-morpholinoethyl)—1 H— pyrazolyl)imidazo[1,2-a]pyridine—3—carboxamide; N-(5-((5,5—dimethyltetrahyd rofuran—Z—yl)methyl ca rbamoy!)~2—fluorophenyl )—7—(1 -(2~ morpholinoethyI)-1H-pyrazol-4—yl)imidazo [1 ,2-a]pyridineca rboxamide; N—(2—fl uoro— -(2~(4-methy}piperazinyl)benzylcarbamoyl)phenyl)(6—methoxy ne yl)imidazo [1,2-a]pyridine—3-carboxamide; N—(2~fluoro—5—(2-(4-methylpiperazinyl)benzylcarbamoy!)pheny!)-6~(1—methyl—1 H— pyrazol—5—yl)pyrazolo[1,5-8]pyridinecarb0xamide; 1-(2—(4-fluoro(7~(pyridine~3-yl)imidazo [1,2—a]pyridine~3— carboxamido)benzamido)ethyi)-2,6-cis-dimethyl piperidine; N-(5-(2-tert-butoxyethylcarbamoyl)—2—fluorophenyl)—6-(6—(3—(dimethylamino) propoxy)pyridine—3-y|)pyrazolo[1,5-a]pyridine-3—carboxamide; N-(2~methyl(2—(4-methylpiperazin-1—yl)benzylcarbamoyl)phenyl)-7—(pyridine yl)imidazo[1 yridine—3-carboxamide; 1-methyI(2-((6-methyl(7-(1-methyl -1 H-pyrazolyl)imidazo[1,2—a]pyridine—3- carbox amido)nicotina mido)methy|)pheny!)piperazine; 7—(1—Methyl-1 H-pyrazol-4—yl)—imidazo[1,2—a]pyridine—3-carboxylic acid {5-[2—(2,6—ois- dimethyI-piperidin-1 thy|carbam oyl ]—2-fluoro—phenyl}-amid e; N—(5-(2—tert-Butoxyethylcarbamoyl)—2—fiuorophenyl)—7—(6-(3-(dimethylamino)propoxy) pyridin—S—yl)imidazo[1,2-a]pyridine-3—carboxamide; N-(5—(3,4—DifluorobenzylcarbamoyI)—2-fl uorophenyl)~7-(6-(2—(pyrrolidin~1—yl)ethoxy) pyridin-S-yl)imidazo[1,2-a]pyridine—S-carboxamide; 6—(1—MethyI-1 H-pyrazol-5—yl)—N-(2—methyl-5—(2—(4—methyipiperazin-1— yl)benzylcarbamoy!) phenyi)pyrazolo[1,5-a]pyridine—3—carboxamide; N-(2-Bromo—5-(2~(4—methylpiperazinyl)benzylcarbamoyl)pheny|)imidazo[1,2- a]pyridine—3—carboxamide; N—(2-Bromo(2-(4-methylpiperazin-1~yl)benzylcarbamoyl)phenyl)pyrazolo[1,5- a]pyridinecarboxamide; N-(5-(3,4-Difluorobenzylcarbamoyl)—2—fluor0phenyl )(3—hyd roxy—S-methyl butyl)imidazo[1,2—a]pyridine—3—carboxamide; N~(5-(3,4—Difluorobenzylcarbamoyl)—2—fl uorophenyl)—7—(3—f|uoro—4—(2—(piperidin—1— ylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide; N-(5-(3,4-Difluorobenzylcarbamoy!)fluorophenyl)(3-f|uoro—4-(2-(tetrahyd ro-2H— pyran-4—yl)ethy|carbamoyl )phenyl)imidazo[1 ,2—a]pyridine—3—carboxamid e; WO 30802 N-(5-(3,4-Diflu0robenzylcarbamoyI)-2~fluorophenyl)—7—(3-fluoro-4—(3-morpholino propylcarbamoyl)phenyl)imidazo[1,2-a}pyridinecarboxamide; 6-(1—MethyI-1H—pyrazoIy|)-N-(2-methyl(2—(4—methylpiperazin y|)benzy|carbamoyl)pyridinyl)pyrazolo[1,5-a]pyridine—3-carboxamide; N—(5—((5,5—Dimethyltetrahydrofuran-Z—yl )methylca rbamoyl)fluorophenyE)-7—(6-(4— methylpiperazin-1—y|)pyridinyl)imidazo[1,2—a]pyridine—3-carboxamide; 3,4-Difluorobenzylcarbamoyl)—2—fluorophenyl)—7-(1-(3—(dimethy| amino)propyl)- 1 H-pyrazol-4—yl)imidazo[1,2-a]pyridine—3-carboxamide; N-(5—((5,5—Dimethyltetra hyd rofuran—Z—yl )methylcarbamoyl)f|uor0pheny|)—7—(5- 1O ((tetrahydro-2H—pyranylamin0)methyl)pyridinyl)imidazo[1,2—a]pyridine—3— carboxamide; (S)—N-(5-(((5,5-Dimethyltetrahydrofuranyl)methyl) carbamoyl)—2~fluorophenyl)—7-(5— (((2-fluoroethyl) amino)methy!)pyridin—3-yl)imidazo[1,2-a]pyridine-3—carboxamide ; (R)—N~(5—(((5,5—dimethyltetrahydrofura n-2—yl)methyl) oyl)f|uorophenyl)—7-(5— ethoxyethyl) (methyl)amino)methyl)pyridinyl) imidazo[1,2—a] pyridine carboxamide ; (R)—7-(5—((tert—butylamino)methy| )pyridin—S-yl)-N—(5-(((5, 5-dimethyltetrahydrofuran yl)methyl)carbamoyI)—2~fluorophenyl) imidazoh,2—a]pyridine-3—carboxamide ; N-(5-(3,4-Difluorobenzylcarbamoyl)fluorophenyl)—7-(6-(2—(pyrrolidiny!)ethoxy) pyridinyl)imidazo[1,2-a]pyridine—3-carboxamide; N~(5-((2—(tert—Butoxy) ethyl)carbamoyl)—2—fluoro phenyl)—7-(8-(2—(pyrrolidin~1—yl) ethoxy)pyridin~3-yl) imidazo[1,2-a] ne-S—carboxamide ; N-(5—(((5,5-dimethyl tetrahydrofuran—Z—yl)methyi)carbamoyI)—2~fluorophenyl)(6-((1 - methyl piperidin-4—yl)oxy) pyridinyl)imidazo[1,2-a]pyridine—3-carboxamide ; 6-(1-Methyl~1 H-pyrazol-S-yl)-N-(2—methyl(2—(4—methyipiperazin-1 - yl)benzylcarbamoyl) )pyrazolo[1,5—a]pyridine—3-carboxamide; N—(Z-Bromo—5—(2—(4-methylpiperazin—1~yl)benzylcarbamoyl)phenyl)imidazo[1 ,2- a]pyridinecarboxamide; N-(Z-Bromo-S-(Z-(4—m ethylpiperazin-1 -yl)benzylcarbamoyl)phenyl)pyrazolo[1 ,5- a]pyridinecarboxamide; N—(5~(3,4-Difluorobenzylcarbamoyl)—2—fluorophenyl )—7—(3-hyd roxymethyl butyl)imidazo[1,2—a]pyridine—3-carboxamide; N—(5—(3,4—DifluorobenzylcarbamoyI)—2—fl uorophenyl)—7~(3-fluoro—4-(2—(piperidin—1— yl)ethylcarbamoyl)phenyl)imidazo[1,2-a1pyridine—3-carboxamide; N-(5-(3,4-DifluorobenzylcarbamoyI)fl uorophenyl)(3—fluoro—4-(2-(tetrahyd ro-2H- pyran-4—yl)ethyioarbamoyl)pheny|)imidazo[1,2—a]pyridine—3-carboxamide; N-(S-((2-(2,2—Dimethylpyrrolidin-1~yl)ethy1)carbamoyl)f|uorophenyl)—7-(1~methyl— 1 H-pyrazol-4—yl)imidazo{1,2-3]pyridine-S—carboxamide; N-(5-(2-(2,2-Dimethylpyrrolidiny|)ethylcarbamoyl)—2—fluorophenyl)—7-(3-fluoro ((1 R,2R)—2—hyd roxycyclohexylcarbamoyl)phenyl)imidazo[1 ,2—a]pyridine—3- carboxamide; N—(5—(2-(2,2-Dimethylpyrrolidin-1~yl)ethylcarbamoyl)—2-fl uorophenyl)—7—(3-fluoro(1 - hydroxy—Z-methylpropanylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3— carboxamide; N—(5-(2—(2,2—Dimethylpyrrolidiny|)ethyicarbamoyl)-2~methylpyridinyl)—6-(3—fluoro~ 1O 4—(1»hydroxy~2—methylpropan—Z—ylcarbamoyl)phenyl)pyrazolo[1,5—a]pyridine~3— carboxamide; N—(5—(3,4-Difluorobenzylcarbamoyi)-2—fluorophenyl)(3~fluoro—4-(3-morpholino propylcarbamoyl)phenyl)imidazo[1,2—3]pyridine—3-carboxamide; 2—(2,6-cis-Dimethylpiperidin-1—y!)ethylcarbamoyl)—2-fluoropheny|)—7-(3-fluoro (1 —hydroxy~2—methylpropa n—2-ylca rbamoyl)ph enyl)imidazo[1 ,2-a]pyridine carboxamide; 7—(3—F1uoro—4-(2-fluoroethylcarbamoyi )phenyl)—N-(2-f|uoro—5—(2—(4—methylpiperazin-1 — zylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3—carboxamide; N-(2-Fluoro(2-(4-methylpiperazin—1-yl)benzylcarbamoyl)phenyl)(3-fluoro(2- hydroxy ethylcarbamoyl )ph enyl)imid azo[1 ,2—a]pyridine—3-carboxamide; N-(5-(2—(2.2-dimethylpyrrolidin-1—y1)ethylcarbamoyl)methylpyridin—3-yl)—6-(1 - methyl-1 H-pyrazoI—4-yl)pyrazolo[1 ,5-a]pyridine—3~carboxamid e; (S)~N-(2—Fluoro(2-(2—(methoxy methyl) pyrrolidin~1~yl)ethyl oyl)phenyI)—7— (1-methyl—1H—pyrazolyl)imidazo[1,2—a]pyridine—S-Carboxamide; N—(2—Fluor0((2—(3-propylpyrrolidinyl)ethyl)carbamoyl)phenyI)—7-(1-methyl—1 H— pyrazolyl)imidazo [1 yridinecarboxamide; (R)-N-(2—F|uoro-5—((2—(2-(methoxymethyl) pyrrolidin—1-yl)ethyl) carbamoyl)phenyl)-7— (1-methyI-1 zol—4-yl)imidazo[1,2-a]pyridine—3-carboxamide; N~(5-((2-(3,5-Dimethylpiperidin—1-y1)ethyl)carbamoyl)—2-fluoropheny|)—7-(1—methyl—1 H- pyrazolyl)imidazo[1,2—a]pyridinecarboxamide; N-(2—fiuoro((2-(2,2,6,6-tetra methylpiperidin—1-yl)ethyl)carba moyl)phenyl)—7-(1 - methyl-1 H-pyrazoIy1)imidazo[1,2—3]pyridinecarboxamide; N—(5—((2—(tert—buty1(methyl)amino)ethy1 )carbamoy1)a2-fluorophenyl)(1~methyl—1 H— pyrazol-S-yl)imidazo[1,2-a]pyridinecarboxamide; (2-(2,2-dimethylpyrrolidin—1 -y1)ethyl)carba moyl)-2—methy|pyridinyl)(1 - methyl-1 H—pyrazoI-S-y|)pyrazo|o[1,5—a]pyridine—3-carboxamide; N—(5-((2—(butyl (ethyl)amino)ethyl)carbamoyl)~2-fluoro )—7-(3-fluoro-4~((2— hydroxy ethyl)carb amoyl)phenyl)imidazo[1,2—a]pyridine—S—carboxamide; uoro-4—((1-hydroxymethylpropanyl)oarbamoyl)phenyl)-N-(2-fluoro-5—((2— (3-propylpyrrolidinyl)ethy!)oarbamoyl)phenyl)imidazo[1,2—a]pyridine carboxamide; N-(5-((2—(3,3—dimethylmorpholino)ethyl)carbamoyl)fluorophenyl)—7-(3—fluoro-4—((1— hydroxy-Z—methylpropanyl)oarbamoyl)phenyl)imidazo[1 ,2—a]pyridine carboxamide; (R)—7-(3—fluoro—4—((1-hydroxy-2—methylpropan-Z-yl)carbamoyl)phenyl)—N—(2—fluoro-5— 1O ((2—(2—(methoxymethyl)pyrrolidin—1—yl)ethyl) carbamoyl)phenyl)imidazo[1,2—a]pyridine— 3—carboxamide; N-(5-((3,4-Difluorobenzyl)oarbamoyl)fluorophenyl)—7—(6—((2— (dimethylamino)ethyl)carbamoyl)pyridin-3—yl)imidazo[1,2—a]pyridine—B-carboxamide; N—(5-((2-(2,2-Dimethylpiperidin-1~y!)ethyl)oarbamoy|)-2—methylpyridinyl)—6—(1- methyl-1 H~pyrazo|—4—yl)pyrazolo[1,5—a]pyridine—3-carboxamide; N—(5—((2—(2,6—cis-Dimethyipiperidinyl)ethyl)carbamoyl}2—methylpyridinyl)—6—(1- methyl-1 H-pyrazoI—4—y|)pyrazolo{1,5-a]pyridine-S-oarboxamide; (2—((28,3R)—2,3—diethylazetidiny|)ethyl) oyl)—2-fluorophenyl)—7—(1— methyl-1 H—pyrazoIyl)imidazo[1,2—a]pyridine-S-oarboxamide; N-(5-(3,4-Difluorobenzylcarbamoyl)-2—f|uorophenyl)(6-(((2-hydroxyethyl) (methyl)amino)methyl)pyridin—3—y!)imidazo[1,2-a]pyridineoarboxamide; N—(5—((3,4—difluorobenzyl)carbamoyI)—2-fluorophenyl)—7—(6—((methyl (phenethyl)amino)methy|)pyridinyl)imidazo[1,2—a]pyridinecarboxamide; N—(5-((3,4—Difluoro benzyl)carbamoy!)—2—fluorophenyl)—7-(6— yl(phenethyl)amino)methyl)pyridinyl)imidazo[1,2—a]pyridine—3-carboxamide; N—(5~((3,4-Difluoro benzyl)carbamoyi)—2—fluorophenyl)(5-((methylamino) methyl)pyridin—3-yl)imidazo[1,2—a]pyridinecarboxamide; 7~(5-((Cyclohexyl amino)methyl)pyridin-B-yl)-N-(5~((3,4-difiuoroben2yl) carba moyl)—2- fluoro ) imidazo[1,2-a]pyrid inecarboxamide; and 3O N-(5-((3,4-Difluoro benzyl)carbamoyl)fluorophenyl)(5—(((2-methoxyethyl) (methyl)amino)methyl)pyridinyl)imid azo[1,2—a]pyridine-3—oarboxamide; or a pharmaceutically acceptable salt thereof.
Embodiment 61: A compound of formula (I), which is selected from: N—(5-((2-(2,2— Dimethylpyrrolidinyl)ethyl)carbamoyl)—2—fluorophenyl)—7-(1-methyl-1H-pyrazoI-4— yl)imidazo[1,2—a]pyridine~3-carboxamide; N-(5-(2-(2,6-cis-Dimethylpiperidinyl)ethylcarbamoyl)fluorophenyl)—7-(1-methyl- 1 H—pyrazol—5-y|)imidazo[1,2-a]pyridinecarboxamide; N-(5-(2—(2,2-dimethylpyrrolidin—1-yl)ethylcarbamoyl)-2—methylpyridinyl)(1 - methyl-1 H—pyrazol—4—yl)pyrazolo[1,5-a]pyridinecarboxamide; 7-(1-Methyl-1 H—pyrazol-4—yl)-imidazo[1,2—aipyridinecarboxylic acid {5—[2—(2,6—cisdimethyl —piperidin-1—yl)-ethylcarbamoyl ]-2—fluoro—phenyl}—amide; and N—(5-(2—(2.6—cis-Dimethylpiperidin—1-yl)ethylcarbamoyl)fluorophenyl)~7-(3—tluoro-4— (1-hydroxy—2—methylpropan—2—ylcarbamoyl)phenyl)imidazo[1,2—alpyridine-3— carboxamide; 1O or a ceutically acceptable salt f. in another embodiment, the individual nds according to the invention are those listed in the es section below, as the free base or as a pharmaceutically acceptable salt thereof.
As used herein, the term “an optical isomer” or “a stereoisomer” refers to any of the various stereo isomeric configurations which may exist for a given compound of the present invention and includes geometric isomers. It is understood that a substituent may be attached at a chiral center of a carbon atom. The term "chiral“ refers to molecules which have the property of non-superimposability on their mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner. Therefore, the invention includes enantiomers, reomers or racemates of the compound. iomers” are a pair of stereoisomers that are non-superimposable mirror images of each other. A 1:1 mixture of a pair of enantiomers is a "racemic” mixture. The term is used to designate a racemic mixture where appropriate. "Diastereoisomers” are stereoisomers that have at least two asymmetric atoms, but which are not mirror- images of each other. The absolute chemistry is specified according to the Cahn~ lngold- Prelog R-S system. When a compound is a pure enantiomer the chemistry at each chiral carbon may be ied by either R or 8. Resolved nds whose absolute configuration is unknown can be designated (+) or (-) depending on the direction (dextro— or levorotatory) which they rotate plane polarized light at the wavelength of the sodium D line. Certain compounds described herein contain one or more asymmetric centers or axes and may thus give rise to enantiomers, reomers, and other stereoisomerlc forms that may be defined, in terms of absolute stereochemistry, as (R)— or (S)-.
Depending on the choice of the starting materials and procedures, the compounds can be present in the form of one of the possible isomers or as mixtures thereof, for example as pure optical isomers, or as isomer mixtures, such as racemates and diastereoisomer es, depending on the number of asymmetric carbon atoms.
The present invention is meant to include all such possible isomers, including racemic es, diasteriomeric mixtures and optically pure forms. Optically active (R)— and (S)— isomers may be prepared using chiral synthons or chiral ts, or ed using conventional techniques. if the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis— or trans— configuration. All tautomeric forms are also intended to be included.
As used herein, the terms “salt” or “salts” refers to an acid on or base addition salt of a compound of the ion. “Salts” include in particular “pharmaceutical acceptable salts”. The term aceutically acceptable salts” refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable. In many cases, the compounds of the present invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, isulfonate, fumarate, tate, gluconate, onate, ate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, , maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalacturonate, propionate, stearate, succinate, sulfosalicylate, te, tosylate and trifluoroacetate salts. lnorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived inciude, for example, acetic acid, propionic acid, giycolic acid, oxalic acid, malic acid, maleic acid, malonic acid, succinic acid, fumaric acid, L-tartaric acid, citric acid, c acid, 4—hydroxybenzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p—toluenesulfonic acid, sulfosalicylio acid, L—glutamic acid, hippuric acid, nicotinic acid, adipic acid, saccharin and the like. Pharmaceutically acceptable base on salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns l to XII of the periodic table. In certain embodiments, the salts are d from sodium, potassium, ammonium, m, magnesium, iron, silver, zinc, and copper; ularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be d include, for example, primary, ary, and tertiary amines, substituted amines including naturally occurring substituted , cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
The pharmaceutically acceptable salts of the present invention can be sized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these nds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are lly carried out in water or in an organic solvent. or in a mixture of the two.
Generally, use of non—aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where cable. Lists of additional le salts can be found, e.g., in ”Remington's Pharmaceutical Sciences”, 20th ed., Mack Publishing Company, Easton, Pa., ; and in “Handbook of Pharmaceutical Salts: Properties, Selection, and Use” by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. lsotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be orated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H, 2012/054501 3H, “C, 13C, 14C, 15N, 18F ”Cl, 1251 tively. The invention includes various isotopically d compounds as defined herein, for e those into which radioactive isotopes, such as 3H and 14C, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically labelled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single—photon emission computed tomography (SPECT) including drug or ate tissue distribution assays, or in radioactive treatment of patients. in particular, an 18F or labeled compound may be particularly desirable for PET or SPECT studies. isotopically-labeled compounds of formula (i) can generally be ed by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled t previously employed.
Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain eutic advantages resulting from greater metabolic stability, for example sed in vivo ife or reduced dosage requirements or an improvement in therapeutic index. it is understood that deuterium in this t is ed as a substituent of a compound of the formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by the isotopic enrichment factor. The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each ated deuterium atom of at least 3500 (52.5% deuterium incorporation at each ated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium oration), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium oration), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, d5- acetone, de-DMSO.
Compounds of the invention, i.e. compounds of formula (i), (ll) or (III) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of g co—crystals with suitable co—crystal formers. These co-crystals may be ed from compounds of formula (i), (ll) or (III) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co- melting, or contacting in solution compounds of a (l), (ll) or (Ill) with the cocrystal former under crystallization conditions and ing co-crystals thereby formed. Suitable co-crystal formers include those described in .
Hence the invention further provides co-crystals comprising a compound of formula 1O (I), (ll) or (in).
As used herein, the term "pharmaceutically able r" includes any and all solvents, dispersion media, coatings, tants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, ng agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289- 1329).
Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
The term "a therapeutically effective amount" of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the ical or medical response of a subject, for example, reduction or tion of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease ssion, or prevent a disease, etc. in one non—limiting embodiment, the term “a therapeutically effective amount” refers to the amount of the compound of the present ion that, when administered to a t, is effective to (1) at least partially alleviating, ting, preventing and/or ameliorating a condition, or a disorder or a disease (i) mediated by PDGFR or (ii) associated with PDGFR activity, or (iii) characterized by activity (normal or abnormal) of PDGFR; or (2) ng or inhibiting the activity of PDGFR. in another non-limiting embodiment, the term “a therapeutically ive amount” refers to the amount of the compound of the present invention that, when stered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of PDGFR.
As used herein, the term “subject” refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. in certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
As used herein, the term it”, “inhibition" or “inhibiting” refers to the reduction or suppression of a given ion, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
As used herein, the term ”treat”, “treating" or "treatment" of any disease or er refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms f). In another embodiment “treat", "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter ing those which may not be nible by the patient. in yet another embodiment, “treat", "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), logically, (e.g., stabilization of a physical parameter), or both. in yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the e or disorder.
As used herein, a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
As used herein, the term "a,” "an,” "the” and similar terms used in the context of the present invention (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly dicted by the t.
All methods described herein can be performed in any suitable order unless otherwise ted herein or ise y contradicted by context. The use of any and all examples, or exemplary language (e.g. "such as”) provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed.
Any asymmetric atom (e.g., carbon or the like) of the compound(s) of the present invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)— or (R,S)— configuration. In certain embodiments, each asymmetric atom has at least 50 % enantiomeric excess, at least 60 % enantiomeric excess, at least 70 % omeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % omeric excess, or at least 99 % enantiomeric excess in the (R)— or (S)— configuration. Substituents at atoms with unsaturated double bonds may, if possible, be present in cis- (Z)— or trans— (E)— form.
Accordingly, as used herein a compound of the t invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, l s (antipodes), racemates or mixtures thereof.
Any resulting mixtures of s can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or onal crystallization. 2O Any resulting racemates of final products or intermediates can be resolved into the l antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound. in particular, a basic moiety may thus be ed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, 6.9., tartaric acid, dibenzoyl tartaric acid, diacetyl ic acid, di-0,0’—p-toluoy| tartaric acid, mandelic acid, malic acid or camphor—1 O-sulfonic acid. Racemic ts can also be resolved by chiral chromatography, e.g., high pressure liquid chromotography (HPLC) using a chiral adsorbent.
Furthermore, the compounds of the present invention, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization. The compounds of the present ion may ntly or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms. The term "solvate" refers to a molecular x of a compound of the present invention (including pharmaceutically acceptable salts thereof) with one or more solvent WO 30802 molecules. Such solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like. The term "hydrate" refers to the complex where the solvent molecule is water.
General Synthetic Schemes The nds of the invention may be synthesized by the general synthetic routes below, specific examples of which are described in more detail in the Examples.
Scheme 1. Method A and A1 F21 F5“ Arm-i2 )ng >1 \ Rilaff R15 lirleth [ti Al Method A/ HOAO\Ef Cl) KO \x / HM" “*0 1O Ra/EOR‘} In scheme, the formation of the amide bond is shown using either Method A or Method A1. Method A is an amide coupling. Methedo A1 is a TBD coupling. This amide ion uces the R5 moeity. X, R1, R13, R4, R5 and R6 are as defined herein.
Scheme 2. lmidazopyridines Method 81 Method BE 1 W - “ R” \f -1’11~11 \ R" FY a 1/ M‘KN a“ NH Cl) 0 , LE? 11111111111110 Kl x“ 1’2 Riakf HM” weo R41” ‘95 In scheme 2, the formation of the amide bond at the aniline nitrogen is shown using Method 81 and Method 82. Method Bl and Method B2 are e amide couplings using acid chlorides. ln on, scheme 2 depicts the formation of an amide bond to introduce the R6 moiety using Method A2, which is an amide coupling.
One on and Y2 is a halogen, such as bromine, and one on and Y2 is hydrogen. X, R1, R13, R4, R5 and R6 are as defined herein.
Scheme 3. '3 “y R1 N\_}‘Y “(IN H XI \ Ear-'2 —ll‘~l 1 Fit.
Q5 .5 Method D / ‘\ Method 0 F!1 is not Br \Q‘ B rict Br Nit fill R 1 if)» R2 C L \1, 11—; R1 ‘ 't HL‘X‘RZ-__ R1 3 /k“4/" HM 0 Wk] HM 0 P‘ 7k4 ”"“K F:E R5 FEB In scheme 3, the introduciton of R2 and/or R3 is depicted as a substitution reaction at Y or Y2 using Method C or Method D. Method C is a Suzuki reaction to couple aryl group at R2 or R3. Method D is a Negishi reaction to couple an alkyl group at R2 or R3. R1 cannot be bromine for this reaction. One on and Y2 is a halogen, such as e, and one on and Y2 is hydrogen. X, R1, R13, R2, R3, R4, R5 and R6 are as defined herein.
Scheme 4. Pyrazolopyridines 2012/054501 NH 1 2 R l NH2 R13 xl \ O O HN O R6A\H Method 81 Method 82 O \ \ R1 Y X \ y2 /N\ l N / O \ R13 \ R1 Y (I) 0 Method A2 >i \ Y2 HN O H R6 In scheme 4, the formation of the amide bond at the aniline nitrogen is shown using Method Bi and Method 82. Method Bi and Method BZ are aniline amide couplings using acid chlorides. In addition, scheme 2 depicts the formation of an amide bond to introduce the R6 moiety using Method A2, which is an amide coupling.
One of Y and Y2 is a halogen, such as bromine, and one on and Y2 is hydrogen. X, R1, R”, R4, R5 and R6 are as defined herein. ’10 Scheme 5.
/N\N O \ \ R1 Y XI \ Y2 HN 0 R49\ R5 R6 Method D Memo" C R1 is not Br R1 is not Br / \N N / \N O \ \ R1 R2 O \ \ \_ R1 R2 NH \ XI \ R3 NH / X] \ R13 / R41 0 HN o R >j\4 R5 R6 R5 R6 ln scheme 5, the introduciton of R2 and/or R3 is depicted as a substitution reaction at Y or Y2 using Method C or Method D. Method C is a Suzuki reaction to couple aryl group at R2 or R3. Method D is a Negishi reaction to couple an alkyl group at R2 or R3. R1 cannot be bromine for this reaction. One of Y and Y2 is a n, such as bromine, and one on and Y2 is hydrogen. X, R1, R“, R2, R3, R4, R5 and R6 are as defined herein.
Scheme 6 | N\ N N \ l R1 0 o MeMgl N \ x R1 NH ———> - /O Method E HN O kRG HN O in scheme 6, the introduciton of alkyi group alpha to an ester group is ed using Method E, a Grignard addition, to form tertiary alcohol. R1 and R6 are as defined herein.
Scheme 7 l \ F N N \ 0 NH R1 / N —————> HN 0 Method F kR6 TBD or h droiysis ”N 0 then HATSIJ k in scheme 7, the introduciton of amide bond from an ester group is depicted using Method F. Method F is a TBD reaction. R1, R6, and R21 are as defined herein.
Scheme 8 I N\ N o “N \ N \ I F O N K CI N \ \ \ / F NH - \ / OR' Method G HN 0 K NaH HN O In scheme 8, the introduciton of alkoxy moiety is depicted using methog G. Method G is a nucleophilic cement of a halide. R’ is C1—C4 alkoxy optionally tuted by one or more OH, -COZR1B, -NR19F€21 or C1—C4 alkoxy. R1, R6, R18, R1g and R21 are as defined herein.
The invention further es any variant of the present processes, in which an intermediate product obtainable at any stage thereof is used as starting material and the remaining steps are carried out, or in which the starting materials are formed in situ under the reaction conditions, or in which the reaction components are used in 1O the form of their salts or iiy pure material.
Compounds of the invention and intermediates can also be converted into each other according to methods generally known to those skilled in the art.
Within the scope of this text. only a readily removable group that is not a constituent of the particular desired end product of the compounds of the present invention is designated a "protecting group", unless the context indicates othen/vise. The tion of functional groups by such protecting groups, the protecting groups themselves, and their cleavage reactions are described for example in standard reference works, such as J. F. W. McOmie, "Protective Groups in Organic Chemistry", Plenum Press, London and New York 1973, in T. W. Greene and P. G.
M. Wuts, "Protective Groups in c Synthesis", Third edition, Wiley, New York 1999, in "The Peptides"; Voiume 3 (editors: E. Gross and J. Meienhofer), Academic Press, London and New York 1981, in "Methoden der organischen Chemie" (Methods of Organic Chemistry), Houben Weyi, 4th edition, Volume 15/l. Georg Thieme Verlag, art 1974, in H.-D. Jakubke and H. eit. "Aminosauren, Peptide, Proteine” (Amino acids, Peptides, Proteins), Verlag Chemie, Weinheim, Deerfield Beach, and Basel 1982, and in Jochen Lehmann, "Chemie der hydrate: Monosaccharide und Derivate" (Chemistry of Carbohydrates: Monosaccharides and Derivatives), Georg Thieme Verlag, Stuttgart 1974. A characteristic of protecting groups is that they can be removed readily (Le. without the occurrence of undesired secondary ons) for example by ysis, reduction, photolysis or alternatively under physiological ions (9.9. by enzymatic cleavage).
Salts of compounds of the t invention having at least one salt—forming group may be prepared in a manner known to those skilled in the art. For example, salts of compounds of the present invention having acid groups may be formed, for example, by ng the compounds with metal compounds, such as alkali metal salts of suitable organic carboxylic acids, e.g. the sodium salt of 2-ethylhexanoic acid, with organic alkali metal or alkaline earth metal compounds, such as the corresponding hydroxides, carbonates or hydrogen carbonates, such as sodium or potassium hydroxide, carbonate or hydrogen ate, with corresponding calcium compounds or with ammonia or a suitable organic amine, stoichiometric amounts or only a small excess of the salt-forming agent ably being used. Acid addition salts of compounds of the present ion are obtained in customary manner, e.g. by treating the compounds with an acid or a suitable anion ge reagent. Internal salts of compounds of the present ion containing acid and basic salt-forming groups, e.g. a free carboxy group and a free amino group, may be formed, e.g. by the neutralisation of salts, such as acid addition salts, to the isoelectric point, e.g. with weak bases, or by treatment with ion exchangers.
Salts can be converted into the free compounds in accordance with methods known to those skilled in the art. Metal and ammonium salts can be converted, for example, by treatment with suitable acids, and acid addition salts, for example, by treatment with a suitable basic agent.
Mixtures of isomers obtainabie according to the invention can be separated in a manner known to those skilled in the art into the individuai isomers; reoisomers can be separated, for example, by partitioning between polyphasic solvent mixtures, recrystallisation and/or chromatographic separation, for example over silica gel or by e.g. medium pressure liquid chromatography over a reversed phase column, and racemates can be separated, for example, by the formation of salts with optically pure salt-forming reagents and tion of the mixture of diastereoisomers so obtainable, for example by means of fractionai crystallisation, or by chromatography over optically active column ais. ediates and final products can be worked up and/or purified ing to standard methods, e.g. using chromatographic methods, bution methods, (re- )crystallization, and the iike.
The following applies in general to all processes mentioned herein before and hereinafter.
All the above-mentioned process steps can be d out under reaction ions that are known to those skilled in the art, including those mentioned specifically, in the e or, customarily, in the presence of solvents or diluents, including, for e, solvents or diluents that are inert s the reagents used and dissolve them, in the e or ce of catalysts, condensation or neutralizing , for example ion exchangers, such as cation exchangers, 9.9. in the H+ form, depending on the nature of the reaction and/or of the reactants at reduced, normal or elevated temperature, for example in a temperature range of from about —100 °C to about 190 °C, ing, for example, from imately -80 °C to approximately ‘IO 150 °C, for example at from -80 to -60 °C, at room temperature, at from -20 to 40 °C or at reflux temperature, under atmospheric pressure or in a closed vessel, where appropriate under pressure, and/or in an inert atmosphere, for example under an argon or nitrogen atmosphere.
At all stages of the reactions, mixtures of isomers that are formed can be ted into the dual isomers, for example diastereoisomers or enantiomers, or into desired mixtures of isomers, for example racemates or mixtures of diastereoisomers, for example ously to the methods described under "Additional process steps".
The solvents from which those solvents that are suitable for any particular reaction may be selected include those mentioned specifically or, for example, water, esters, such as lower alkyl—lower alkanoates, for example ethyl acetate, ethers, such as aliphatic ethers, for example diethyl ether, or cyclic ethers, for example tetrahydrofuran or dioxane, liquid aromatic hydrocarbons, such as benzene or toluene, alcohols, such as methanol, ethanol or i- or 2—propanol, nitriles, such as acetonitrile, halogenated hydrocarbons, such as methylene chloride or chloroform, acid amides, such as dimethylformamide or dimethyl acetamide, bases, such as heterocyclic nitrogen bases, for example pyridine or N-methylpyrrolidin-2—one, carboxylic acid anhydrides, such as lower alkanoic acid anhydrides, for example acetic anhydride, cyclic, linear or branched hydrocarbons, such as cyclohexane, hexane or isopentane, methycyclohexane, or mixtures of those solvents, for example aqueous solutions, unless othenivise indicated in the ption of the processes.
Such solvent mixtures may also be used in working up, for e by chromatography or partitioning.
The compounds, including their salts, may also be obtained in the form of hydrates, or their crystals may, for example, include the solvent used for crystallization.
Different crystalline forms may be present.
The invention relates also to those forms of the process in which a nd obtainable as an intermediate at any stage of the process is used as ng material and the remaining process steps are d out, or in which a starting material is formed under the reaction conditions or is used in the form of a derivative, for example in a protected form or in the form of a salt, or a compound obtainable by the process according to the invention is produced under the process conditions and processed further in situ.
All starting als, building blocks, reagents, acids, bases, dehydrating agents, solvents and catalysts utilized to synthesize the compounds of the present invention are either commercially available or can be produced by organic synthesis methods known to one of ordinary skill in the art (Houben-Weyl 4th Ed. 1952, Methods of Organic Synthesis, Thieme, Volume 21).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically able r. The pharmaceutical composition can be formulated for particular routes of stration such as oral administration, parenterai administration, and rectal administration, etc. in addition, the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation ons, suspensions or emulsions). The pharmaceutical itions can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as nts, such as preservatives, stabilizers, wetting agents, emulsifers and buffers, etc.
Typically, the pharmaceutical compositions are tablets or n capsules comprising the active ingredient together with a) diluents, e,g., lactose, dextrose, e, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, c acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) s, 6.9., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent es; and/or e) absorbents, colorants, flavors and sweeteners.
Tablets may be either film coated or enteric coated ing to methods known in the art.
Suitable compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Compositions ed for oral use are prepared according to any method known in the art for the manufacture of ceutical itions and such compositions can contain one or more agents ed from the group consisting of ning agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
Tablets may contain the active ingredient in a mixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for e, corn , or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer . For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. ations for oral use can be presented as hard gelatin capsules wherein the 3O active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or , or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
Certain able compositions are s isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
Said compositions may be sterilized and/or contain adiuvants, such as preserving, stabilizing, g or fying agents, solution promoters, salts for regulating the osmotic re and/or buffers. in addition, they may also contain other therapeutically valuable substances. Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 01—75%, or contain about 1-50%, of the active ingredient.
Suitable compositions for transdermal application include an effective amount of a compound of the invention with a suitabie r. Carriers suitable for transdermai delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, ally a rate controlling barrier to deiiver the compound of the skin of the host at a controlled and ermined rate over a prolonged period of time, and means to secure the device to the skin.
Suitable compositions for topical application, e.g., to the skin and eyes, e aqueous solutions, suspensions, ointments, creams, gels or sprayabie formulations, e.g., for delivery by l or the like. Such topical delivery systems will in particuiar be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for 2O prophylactic use in sun creams, lotions, sprays and the like. They are thus particularly suited for use in topical, including ic, formulations well—known in the art. Such may contain soiubiiizers, izers, tonicity enhancing agents, buffers and preservatives.
As used herein a topical application may also n to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either atone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phosphoiipids) from a dry powder r or an aerosol spray presentation from a pressurised container, pump, spray, atomizer or nebuliser, with or without the use of a suitable propellant.
Where the inhalable form of the active ingredient is an aerosol ition, the tion device may be an aerosol vial provided with a valve adapted to deliver a metered dose, such as 10 to 100 at, e.g. 25 to 50 pl, of the composition, Le. a device known as a metered dose inhaler. Suitable such l vials and procedures for containing within them l compositions under pressure are well known to those skilled in the art of inhalation therapy. For example, an aerosol composition may be administered from a coated can, for example as described in EP-A—0642992. Where the inhalable form of the active ingredient is a nebulizabie aqueous, c or aqueous/organic dispersion, the inhalation device may be a known nebulizer, for example a conventional tic nebulizer such as an airjet nebulizer, or an ultrasonic nebulizer, which may contain, for example, from 1 to 50 ml, commonly 1 to ml, of the dispersion; or a hand—held nebulizer, sometimes referred to as a soft mist or soft spray inhaler, for example an electronically controlled device such as an AERX (Aradigm, US) or Aerodose (Aerogen), or a mechanical device such as a AT (Boehringer ingelheim) nebulizer which allows much smaller nebulized volumes, e.g. 10 to 100 pl, than conventional nebulizers. Where the inhalable form of the active ingredient is the finely d particulate form, the inhalation device be, for example, a dry powder inhalation device adapted to deliver dry powder from a capsule or blister containing a dry powder comprising a dosage unit of (A) and/or (B) or a multidose dry powder inhalation (MDPl) device adapted to deliver, for example, 3—25 mg of dry powder comprising a dosage unit of (A) andlor (B) per actuation. The dry powder composition ably contains a diluent or r, such as lactose, and a compound that helps to protect against product performance deterioration due to moisture e.g. ium stearate. Suitable such dry powder inhalation devices include s sed in US 3991761 (including the AEROLIZERTM device), WO 042, WO 97/20589 ding the CERTIHALERTM device), WO 97/30743 (including the ALERTM device) and WO 05/37353 (including the LERTM device).
The invention also includes (A) an agent of the invention in free form, or a pharmaceutically acceptable sait or soivate thereof, in ble form; (B) an inhalable medicament comprising such a compound in inhatable form together with a pharmaceutically acceptable carrier in inhalable form; (C) a pharmaceutical product comprising such a compound in inhalable form in association with an inhalation device; and (D) an inhalation device containing such a compound in inhalable form.
Dosages of agents of the invention ed in practising the present invention wilt of course vary depending, for example, on the particular condition to be treated, the effect desired and the mode of administration. tn general, suitable daily dosages for administration by inhalation are of the order of 0.0001 to 30 mg/kg, typically 0.01 to 10 mg per patient, while for oral administration suitable daily doses are of the order of 0.01 to 100 mg/kg.
PCT/1B2012/054501 The present invention further provides anhydrous ceutical compositions and dosage forms comprising the compounds of the present invention as active ingredients, since water may tate the degradation of certain compounds.
Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture ning ingredients and low moisture or low humidity conditions. An anhydrous pharmaceutical ition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to ‘IO water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e. g., vials), blister packs, and strip packs.
The invention further provides pharmaceutical compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the t invention as an active ingredient will ose. Such agents, which are referred to herein as "stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
The compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent. The compound of the present ion may be administered separately, by the same or different route of administration, or together in the same ceutical composition as the other agents.
Another aspect of this invention relates to the fact that the compounds of formula (I), (II) or (III) and their pharmaceutically acceptable salts have beneficial pharmacological activity and, therefore, are useful as pharmaceuticals. ore, according to a further aspect of the invention a compound of formula (I), (ll) or (III) is before described as a medicament. The use of compounds of formula (I), (II) or (III) in inhibiting PDGF receptor mediated biological ty is novel per se. Therefore, a nd of formula (l), (II) or (III) or pharmaceuticall able salt thereof is an inhibitor of PDGF receptor mediated ical activity.
We particularly provide a compound of formula (I), (II) or (III) or a pharmaceuticallly acceptable salt thereof for the treatment of a respiratory disorder.
According to a further aspect of the invention the use of a compound of formula (I) is hereinbefore described in the manufacture of a medicament. More ularly, the use is hereinbefore described in the manufacture of a medicament for inhibiting PDGF receptor mediated biological activity. Another aspect of the invention is the use of a compound of formula (I) in the cture of a medicament for the treatment of a respiratory disorder.
Another aspect provided herein are pharmaceutical itions that include a therapeutically effective amount of a compound of Formula (I), (II), or (III), or a pharmaceutical salt thereof and a pharmaceutically acceptable r. in certain embodiments of such pharmaceutical compositions, the pharmaceutical ition is formulated for intravenous administration, intravitrial administration, intramuscular administration, oral administration, rectal administration, transdermal administration, ary administration, inhalation administration, nasal administration, topical administration, ophthalmic administration or otic administration. In other embodiments, the pharmaceutical compositions are in the form of a tablet, a pill, a capsule, a liquid, an inhalant, a nasal spray solution, a suppository, a solution, an emulsion, an ointment, eye drop or ear drop. In other embodiments, such pharmaceutical compositions further include one or more additional therapeutic agents.
Use and Method of ng Another aspect provided herein is the use of the compound of Formula (I), (II) or (III) or a ceutically acceptable salt thereof for the treatment of a disorder or disease in a subject by inhibiting c—kit and/or PDGFR kinase activity, and such suc use e a therapeutically effective amount of the compound of a (I), (II) or (III). r aspect provided herein is the use of the compound of Formula (I), (II) or (Ill) 3O or a pharmaceutically able salt thereof for the treatment of a disorder or disease in a subject by inhibiting c-kit and/or PDGFR kinase activity, and such suc use include a therapeutically effective amount of the compound of Formula (I), (II) or (III), wherein the disease or disorder is a mast-cell associated disease, a respiratory disease, an inflammatory disorder, irritable bowel syndrome (IBS), inflammatory bowel disease (I BD), an autoimmune er, a metabolic disease, a fibrosis disease, a dermatological disease, cariac hypertrophy, cancers of the lung or other tissues in which a PDGFR isoform is mutated, pressed or activiated, 012/054501 pulmonary arterial ension (PAH) or primary pulmonary hypertension (PPH). In other embodiments of this aspect, the disease is asthma, allergic rhinitis, pulmonary al hypertension (PAH), pulmonary fibrosis, hepatic fibrosis, cardiac fibrosis, derma, irritable bowel syndrome (IBS), inflammatory bowel disease (lBD), ia, dermatosis, type I diabetes or type II diabetes. r aspect provided herein are uses for treating a disease mediated by a kinase in a patient in need therof, and such uses include a therapeutically effective amount of a compound of Formula (l), (II) or (III), the kinase is selected from c-kit, c, ’10 PDGFRB, p38, BCR—abl and c-FMS and the disease is a mast-cell associated disease, a respiratory disease, an inflammatory disorder, irritable bowel syndrome (IBS), inflammatory bowel disease (lBD), an autoimmune disorder, a metabolic disease, a fibrosis disease, a dermatological disease, pulmonary arterial hypertension (PAH) or primary pulmonary hypertension (PPH).
In certain embodiments of this aspect, the disease is asthma, allergic rhinitis, pulmonary arterial hypertension (PAH), pulmonary fibrosis, c fibrosis, cardiac fibrosis, cardiac hypertrophy, scleroderma, irritable bowel syndrome (IBS). inflammatory bowel disease (lBD), uticaria, dermatosis, type I diabetes or type II diabetes.
Another aspect ed herein is the use ofa compound of a (I), (II) or (III), in the manufacture of a medicament for treating a disease or disorder in a t where modulation of a c—kit and/or PDGFR kinase is implicated.
Another aspect provided herein includes methods for treating a disease or disorder where modulation of c—kit and/or PDGFR kinase is implicated, wherein the method includes administering to a system or subject in need of such treatment an effective amount of a compound of Formula (I), (II) or (III), or pharmaceutically acceptable salts or pharmaceutical compositions thereof, y treating the disease or er. In such s, the compound of Formula (I), (II) or (III), is an inhibitor of c-kit and/or PDGFR kinases. In certain embodiments of such s, the methods e administering the compound to a cell or tissue system or to a human or animal subject. In certain embodiments of such methods, the disease or condition is a metabolic disease, a fibrotic e, cardiac hypertrophy, a respiratory disease, an inflammatory disease or disorder, a dermatological disease or an autoimmune disease. In certain embodiments of such s, the disease or condition is asthma, allergic rhinitis, irritable bowel syndrome (188), inflammatory bowel disease (lBD), pulmonary arterial hypertension (PAH), pulmonary fibrosis, liver fibrosis, cardiac fibrosis, scleroderma, urticaria, dermatoses, atopic itis, type I diabetes or type II diabetes.
In another embodiment the disease is selected from a a respiratory disease, an inflammatory er, irritable bowel syndrome (188), matory bowel disease (IBD), aa fibrosis disease, pulmonary arterial ension (PAH) and y pulmonary hypertension (PPH). in other embodiments the es is asthma, pulmonary arterial hypertension (PAH), pulmonary fibrosis, hepatic fibrosis, cardiac fibrosis, or cardiac hypertrophy.
Pharmaceutical Composition and Combinations The pharmaceutical composition or combination of the present invention can be in unit dosage of about 1 -1000 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1—500 mg or about 1-250 mg or about 1-150 mg or about 0.5-100 mg, or about 1-50 mg of active ingredients. The therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof. is dependent on the species of the t, the body weight, age and individual condition, the er or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the er or disease.
The above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, 9.9., mice, rats, dogs, monkeys or isolated organs, tissues and ations thereof. The compounds of the present invention can be applied in vitro in the form of solutions, e.g., aqueous ons, and in vivo either 3O enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution. The dosage in vitro may range between about 10‘3 molar and 10'9 molar concentrations. A therapeutically ive amount in vivo may range depending on the route of administration, between about 01—500 mg/kg, or between about 1-100 mg/kg.
The activity of a compound ing to the present invention can be assessed by the following in vitro & in vivo methods.
Pharmaceutical Use and Assay Compounds of a (l), (ll) or (III) provided herein were assayed to measure their capacity to t PDGFR kinases using the appropriate assay described below: PDGFR inhibition was evaluated using the Rat A10 cell proliferation.
Rat A10 cell proliferation assay Rat A10 cells (ATCC) were resuspended in DMEM mented with 1% FBS and ng/mL recombinant rat PDGF-BB at 20,000 cells/mL. The cells were ted into 384 well plates at 50 pL/well and incubated for 4 hours at 37°C. 0.5 uL of test compound 3-fold serially diluted in DMSO was added to each well. The plates were returned to the incubator for a further 68 hours. 25 pL of CellTiter—Glo (Promega) was added to each well and the plates were incubated on the bench for 15 minutes.
Luminescence was then read using a CLIPR CCD camera (Molecular Devices).
Data were analysed using non—linear regression fitted to sigmoidal ble slope) curves using a four parameter logistic equation to generate |C50 values for each compound.
Compounds of the examples, herein below, generally have PDGFR Kb values in the Rat A10 cell based assay below 10pM. Table A provides a list of representative compounds with their |C50 values.
Table A.
PDGFR RAT(A10) leo luM 0.009 0.040 —'*0.010 40.004 0.011 PDGFR inhibtors, including the compounds of formula (l), (ll) or (ill) are also useful as co-therapeutic agents for use in ation with second agents, such as organic nitrates and NO-donors, such as sodium nitroprusside, nitroglycerin, isosorbide mononitrate, isosorbide dinitrate, molsidomine or SlN—i, and lnhalational NO; compounds that inhibit the ation of cyclic guanosine monophosphate (cGMP) and/or cyclic adenosine monophosphate , such as inhibitors of phosphodiesterases (PDE) 1, 2, 3, 4 and/or 5, especially PDE 5 inhibitors such as sildenafil, vardenafil and tadalafil; NO—independent, but haem-dependent stimulators of guanylate cyclas'e, such as in particular the compounds described in WO 00/06568, WO 00/06569, WO 02/42301 and WO 03/095451; NO- and haem- independent activators of guanylate cyclase, such as in particular the nds described in WO 01/19355, WO 01/19776, WO 01/19778, WO 01/19780, WO 02/070462 and WO 02/070510; compounds which inhibit human philic se, such as sivelestat or DX-890 (Reltran); compounds inhibiting the signal uction cascade, such as tyrosine kinase and/or serine/threonine kinase inhibitors, in particular imatinib, ib, erlotinib, sorafenib and sunitinib; compounds influencing the energy metabolism of the heart, for example and preferably etomoxir, dichloroacetate, ranolazine or trimetazidine; antithrombotic agents, for example and preferably from the group comprising platelet aggregation inhibitors, anticoagulants or profibrinolytic substances; active nces for lowering blood pressure, for example and ably from the group comprising calcium antagonists, angiotensin ll nists, ACE inhibitors, endothelin nists, renin inhibitors, aldosterone synthase inhibitors, alpha receptor blockers, beta receptor blockers, locorticoid or antagonists, Rho-kinase inhibitors and diuretics; and/or active nces that modify lipid metabolism, for example and preferably from the group comprising thyroid receptor ts, inhibitors of cholesterol synthesis, for example and preferably HMG-CoA—reductase inhibitors or inhibitors of ne synthesis, ACAT inhibitors, CETP inhibitors, MTP inhibitors, PPAR—alpha, PPAR-gamma and/or PPAR—delta agonists, cholesterol absorption inhibitors, lipase inhibitors, polymeric bile acid adsorbers, bile acid reabsorption inhibitors and lipoprotein(a) antagonists, particulariy in the treatment of PAH or diseases and ers such as those mentioned hereinbefore, e.g., as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs.
In particular, an embodiment of this invention is a pharmaceutical combination comprising the compounds of formula (i), (ll) or (Ill) and a second agent wherein the second agent is a PDEV inhibitor or neutral endopeptidase inhibitor.
The compounds of formula (I), (ll) or (III) may be mixed with a second agent in a fixed pharmaceutical composition or it may be administered tely, , simultaneously with or after the other drug substance.
Other useful combinations of PDGFR inhibitorwith anti—inflammatory drugs are those with antagonists of ine receptors, e.g., OCR-1, OCR-2, OCR-3, CCR-4, COR— , COR-6, OCR-7, OCR-8, COR-9 and CCR10, CXCR1, CXCRZ, CXCR3, CXCR4, CXCRS, particularly OCR-5 antagonists, such as Schering-Plough antagonists SC— 351125, SCH-55700 and SCH-D; Takeda antagonists, such as N-[[4-[[[6,7-dihydro hyl-phenyl)-5H—benzo-cycloheptenyl]carbonyl]amino]phenyi}- methyl]tetrahydro—N,N-dimethyl—2H—pyran-4—amin—ium chloride (TAK—770); and COR— antagonists described in USP 6,166,037 (particularly claims 18 and 19), WO 00/66558 (particularly claim 8), WO 00/66559 (particularly claim 9), W0 04/018425 and W0 04/026873.
Suitable anti-inflammatory drugs include steroids, in ular, glucocorticosteroids, such as budesonide, beclamethasone dipropionate, fluticasone propionate, onide or mometasone e, or steroids described in W0 02/88167, W0 02/12266, WO 02/100879, WO 02/00679 (especially those of Examples 3, 11, 14, 17, 19, 26, 34, 37, 39, 51, 60, 67, 72, 73, 90, 99 and 101), WO 03/35668, WO 81, W0 03/62259, W0 03/64445, W0 03/72592, WO 27 and WO 04/66920; non—steroidal glucocorticoid or agonists, such as those described in DE 10261874, WO 00/00531, WO 02/10143, W0 03/82280, WO 03/82787, WO 03/86294, WO 03/104195, W0 03/101932, WO 04/05229, W0 04/18429, WO 1O 04/19935 and WO 04/26248; LTD4 antagonists, such as montelukast and zafirlukast; PDE4 inhibitors, such as cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden),V—11294A (Napp), BAY19—8004 (Bayer), SCH—351591 (Schering-Plough), Arofylline (Almirall Prodesfarma), PD189659/PD168787 (Parke-Davis), AWD—12—281 (Asta ), CDC-801 (Celgene), SelClD(TM) CC—10004 (Celgene), VM554/UM565 (Vernalis), T—440 e), KW-4490 (Kyowa Hakko Kogyo), and those disclosed in WO 92/19594, WO 93/19749, WO 93/19750, WO 93/19751, WO 98/18796, WO 99/16766, WO 01/13953, WO 03/104204, W0 03/104205, WO 03/39544, W0 04/000814, W0 04/000839, W0 04/005258, W0 450, W0 04/018451, W0 04/018457, W0 04/018465, W0 04/018431, W0 04/018449, W0 2O 04/018450, W0 04/018451, WO 04/018457, W0 04/018465, WO 04/019944, W0 04/019945, W0 04/045607 and W0 04/037805; adenosine A28 receptor antagonists such as those described in W0 02/42298; and beta—2 ceptor agonists, such as albuterol tamol), metaproterenol, terbutaline, salmeterol fenoterol, procaterol, and especially, formoterol, carmoterol and pharmaceutically able salts f, and compounds (in free or salt or solvate form) of formula (I) of WO 4, which document is incorporated herein by nce, preferably indacaterol and pharmaceutically acceptable salts thereof, as well as compounds (in free or salt or solvate form) of formula (I) of WO 04116601, and also compounds of EP 6, JP 05025045, WO 93/18007, WO 99/64035, USP 2002/0055651, WO 01/42193, W0 01/83462, WO 02/66422, W0 02/70490, W0 02/76933, WO 03/24439, W0 60, WO 64, WO 03/72539, WO 03/91204, WO 03/99764, WO 04/16578, WO 04/22547, WO 04/32921, WO 04/33412, WO 04/37768, WO 04/37773, W0 04/37807, WO 04/39762, WO 04/39766, WO 04/45618, W0 04/46083, WO 04/80964, W0 04/108765 and W0 04/108676.
Suitable bronchodilatory drugs include anticholinergic or antimuscarinic agents, in particular, ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate, but also those described in EP 424021, USP 3,714,357, USP 5,171,744, WO 01/04118, WO 02/00652, WO 02/51841, WO 02/53564, WO 40, WO 03/33495, WO 03/53966, WO 03/87094, WO 04/018422 and WO 04/05285.
Suitable dual anti-inflammatory and bronchodilatory drugs include dual beta—2 adrenoceptor t/muscarinic antagonists such as those disclosed in USP 200410167167, WO 46 and WO 04/74812.
Suitable antihistamine drug nces include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hloride, activastine, astemizole, azelastine, ne, epinastine, mizolastine and tefenadine, as well as those disclosed in JP 7299, WO 03/099807 and WO 04/026841.
Accordingly, the invention includes as a further aspect a combination of PDGFR inhibitor with agents that inhibit ALK5 and/or ALK4 phosphorylation of Smad2 and Smad3.
Accordingly, the invention includes as a further aspect a combination of PDGFR inhibitor with second agents that are Rho—kinase inhibitors. Accordingly, the invention includes as a r aspect a combination of PDGFR inhibitor with second agents that are TPH’i antagonists.
Accordingly, the invention includes as a further aspect a combination of PDGFR inhibitor with second agents that are [P receptor agonist.
Accordingly, the invention includes as a further aspect a combination of PDGFR tor with second agents that are multi-kinase inhibitors, such as imatinib mysilate( Gleevec®) or nilotinib. lmatinib functions as a c inhibitor of a number of tyrosine kinase enzymes. it occupies the TK active site, leading to a decrease in activity. TK enzymes in the body, include the insulin receptor. lmatinib is specific for the TK domain in the Abelson proto—oncogene, c—kit and PDGF-R (platelet—derived growth factor or).
Accordingly, an embodiment of this invention provides a pharmaceutical combination comprising the compounds of a (I), (ll) or (ill) ,or pharmaceutical salts thereof, and a second agent wherein the second agent is selected from phosphodiesterase V (PDEV) tors, such as sildenafil or tadalafil; neutral ptidase inhibitors such as neutral endopeptidase 1 inhibitors; anti—inflammatory drugs including nists of chemokine ors; steroids including corticosteroids such as long— acting corticosteroids; Bz—agonists including ultra—long-acting Bg-agonists; bronchodilatory drugs including anticholinergic or antimuscarinic , such as long—acting muscarinic antagonists; dual anti-inflammatory and bronchodilatory drugs including dual beta-2 adrenoceptor agonist/muscarinic antagonists; antihistamine drug substances; lP receptor agonists, such as as those disclosed in W02012/007539; agents that induce pulmonary vascular vasodilation; agents that are tryptophan hydroylase i (TPHi) tors; multi-kinase inhibitors such as c—Kit inhibitors; tyrosine kinase inhibitors such as ib (Gleevec®) or nilotinib; MAPK (e.g. p38) inhibitors; mTOR inhibitors (alone or in combination with P13K inhibitors); LPA-i tors; endothelin antagonists; diuretics; aldosterone receptor blockers; and endothelin receptor blockers.
Formulation and Administration The nds of the invention may be administered by any appropriate route, e.g., orally, e.g., in the form ofa tablet or capsule; parenterally, e.g., intravenously; by inhalation, e.g., in the treatment of inflammatory or obstructive airways disease; intranasally, e.g., in the treatment of allergic rhinitis; topically to the skin, e.g., in the treatment of atopic dermatitis; topically to the eye, e.g., in the treatment of glaucoma; or rectally, e.g., in the treatment of atory bowel disease.
In a further aspect, the invention also provides a pharmaceutical composition comprising a compound of a (l), (ll), or (ill) in free form or in the form of a pharmaceutically able salt, optionally together with a pharmaceutically acceptable nt, diluent or carrier.
The composition may contain a co—therapeutic agent such as an anti-inflammatory, broncho-dilatory, antihistamine or anti-tussive drug as hereinbefore described. Such compositions may be prepared using conventional diluents or excipients and techniques known in the galenic art. Thus oral dosage forms may include tablets and capsules. Formulations for topical administration may take the form of creams, nts, gels or transdermal delivery s, e.g., patches. Compositions for inhalation may comprise aerosol or other atomisable formulations or dry powder formulations. 2012/054501 When the composition comprises an aerosol formulation, it preferably contains, e.g., a hydro-fluoro—alkane (HFA) propellant, such as HFA134a or HFA227 or a mixture of these, and may contain one or more co-solvents known in the art such as ethanol (up to 20% by weight), and/or one or more surfactants such as oleic acid or sorbitan trioleate, and/or one or more bulking agents, such as lactose. When the composition ses a dry powder formulation, it preferably contains, e.g., the compound of formula (I)—(Ill) having a particle diameter up to 10 microns, optionally together with a diluent or carrier, such as lactose, of the desired particle size distribution and a compound that helps to protect against product performance deterioration due to moisture, e.g., magnesium stearate. When the composition comprises a nebulised formulation, it preferably contains, e.g., the compound of formula (I), (II) or (III) either dissolved, or suspended, in a vehicle containing water, a co-solvent, such as ethanol or propylene glycol and a stabiliser, which may be a surfactant.
Thus, the present invention further es: (a) a compound of a II) in inhalable form, e.g., in an aerosol or other atomisable composition or in inhalable particulate, e.g., micronised, form; (b) an inhalable medicament sing a compound of formula (I), (II) or (III) in inhalable form; (c) a pharmaceutical product comprising a compound of formula (I), (II) or (III)) in inhalable form in association with an inhalation device; and (d) an tion device containing a compound of formula (I), (II) or (III) in inhalable form.
Dosages of nds of a (I), (II) or (III) employed in practising the present invention will of course vary ing, e.g., on the particular condition to be treated, the effect desired and the mode of administration. In general, suitable daily dosages for stration by inhalation are of the order of 0005-10 mg, while for oral 3O administration suitable daily doses are of the order of 0.05-100 mg.
The present invention includes all pharmaceutically acceptable isotopicalIy-Iabelled compounds of formula (I), (II) or (III) wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. 2012/054501 Examples of es suitable for inclusion in the compounds of the invention e isotopes of hydrogen, such as 2H and 3H, , such as 110, 130 and 14C, ne, such as 360i, fluorine, such as 18F, iodine, such as 123| and 125i, nitrogen, such as 13M and 15N, oxygen, such as 150, 170 and 180, phosphorus, such as 32F, and sulphur, such as 358.
Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo ife or reduced dosage requirements, and hence may be preferred in some circumstances. lsotopically~labeled compounds of formula (I), (ll) or (lll) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate icalIy-labeled reagents in place of the non-labeled reagent usly employed.
Compounds of formula (1), (H) or (III) may be prepared by the general reactions shown in the examples herein.
Referring to the examples that follow, compounds of the preferred embodiments are synthesized using the methods bed herein, or other methods, which are known in the art.
It should be understood that the organic compounds according to the preferred embodiments may exhibit the phenomenon of tautomerism. As the chemical structures within this specification can only represent one of the possible tautomeric forms, it should be understood that the preferred embodiments encompasses any tautomeric form of the drawn structure.
It is understood that the invention is not limited to the embodiments set forth herein for illustration, but embraces all such forms thereof as come within the scope of the above disclosure.
The ion is illustrated by the following Examples.
Examples General Conditions: Mass spectra were run on LCMS systems using electrospray ionization. These were either t 1100 HPLC/Micromass Platform Mass Spectrometer combinations or Waters Acquity UPLC with SQD Mass Spectrometer. [M+H]+ refers to sotopic molecular weights.
NMR spectra were run on open access Bruker AVANCE 400 NMR spectrometers using ICON—NM R. a were measured at 298K and were referenced using the t peak.
The following es are intended to illustrate the invention and are not to be construed as being limitations thereon. Temperatures are given in degrees centigrade. If not mentioned otherwise, all ations are performed under reduced pressure, preferably between about 15 mm Hg and 100 mm Hg (= 20-133 mbar). The structure of final products, intermediates and starting materials is confirmed by standard analytical methods, e.g., microanalysis and spectroscopic characteristics, e.g., MS, IR, NMR. Abbreviations used are those conventional in the art. if not defined, the terms have their generally accepted meanings.
Abbreviations: aq. aqueous br broad d doublet DCM dichloromethane DMF methylformamide DMAC dimethylacetamide DMSO dimethylsulfoxide EtOAc ethyl acetate hr hour HOBt hydroxybenzotriazole HPLC high pressure liquid chromatography LC-MS liquid chromatography and mass spectrometry MeOH methanol MS mass spectrometry m multiplet min minutes ml milliliter(s) m/z mass to charge ratio 2012/054501 NMR> nuclear magnetic resonance ppm parts per million PEAX PE-anion exchange (e.g. e® PE-AX columns from Biotage) Rt retention time RT room temperature 8 t SCX—2 strong cation exchange (e.g. lsolute® SCX~2 columns from Biotage) t triplet TEA triethylamine TBD 2,3,4,6,7,8—hexahydro-1H-pyrimido[1,2-a]pyrimidine THF tetrahydrofuran Referring to the examples that follow, compounds of the preferred embodiments were synthesized using the methods described herein, or other methods. which are known in the art.
The various starting materials, intermediates, and compounds of the red ments may be isolated and purified, where appropriate, using tional ques such as precipitation, filtration, crystallization, evaporation, distillation, 2O and chromatography. Unless ise stated, all starting materials are obtained from commercial suppliers and used without further purification. Salts may be ed from compounds by known salt-forming procedures.
It should be understood that the organic compounds according to the preferred embodiments may exhibit the phenomenon of tautomerism. As the chemical structures within this specification can only represent one of the possible tautomeric forms, it should be understood that the preferred embodiments encompasses any tautomeric form of the drawn structure. 3O if not indicated otherwise, the analytical HPLC conditions are as follows: Method 2minLC_v001 Column Waters BEH 018 100x21 mm, 1.7 pm Column Temp. 50 °C Eluents A: H20, B: acetonitrile, both containing 0.1% TFA Flow Rate 0.7 ml/min nt 0.25 min 5% B; 5% to 95% B in 1.00 min, 0.25 min 95% Method 2minLC_v002 Column Waters BEH C18 50x2.1 mm, 1.7 pm Column Temperature 50 °C Eluents A: H20, B: MeOH, both containing 0.1% TFA Flow Rate 0.8 ml/min Gradient 0.20 min 5% B; 5% to 95% B in 1.30 min, 0.25 min 95% B Method 2minLC_30_v003 Column Waters BEH C18 50x2.1 mm, 1.7 pm Column Temperature 50 °C Eluents A: H20, B: acetonitrile, both containing 0.1% TFA Flow Rate 0.8 ml/min Gradient 0.25 min 30% B; 30% to 95% B in 1.00 min, 0.25 min 95% B Method 10minLC_v003 Column Waters BEH C18 50x2.1 mm, 1.7 pm Column Temperature 50 °C Eluents A: H20, B: itrile, both containing 0.1% TFA Flow Rate 0.8 ml/min Gradient 0.20 min 5% B; 5% to 95% B in 7.80 min, 1.00 min 95% Method Lopr_v002 Column Phenomenex Gemini C18 50X4.6 mm, 3.0 mm Column Temperature 50 °C Eluents A: H20, B: MeOH, both containing 0.1% TFA Flow Rate 1.0 ml/min Gradient 5% to 95% B in 2.0 min, 0.2 min 95% B Method A Column: Aoquity HSS T31.8um 2.1x 50mm at 50°C WO 30802 Eluent A: water + 0.05 % formic acid + 3.75 mM ammonium acetate Eluent B: acetonitrile + 0.04 % formic acid Gradient: 2% to 98 % B in 1.4 min — flow 1.2 ml/min Method 10 min LC Column Aglient, Poroshell 120 88- C18 2.7 um 3.0x 50mm Column Temp. 30 °C Eluents B: H20, C: acetonitrile, both containing 0.1% 1O Formic acid Flow Rate min Gradient 0.50 min 5% C; 5% to 95% C in 6.50 min, 95% to % C in 3 min Example compounds of the present invention include: Preparation of Final Compounds Example 1.1 N -(2-F|uoro(2-(4-methyl pi perazin-1 -yl)benzylcarbamoyl)phenyl)(1 -methy| - 1H-pyrazoly|)imidazo[1,2-a]pyridinecarboxamide / N HN O Step 1: 7-Bromo-N-(2—fluoro(2—(4-methylpiperazinyl)benzylcarbamoyl) phenyl)imidazo[1,2—a]pyridinecarboxamide Methyl 3-(7-bromoimidazoi1,2-a]pyridinecarboxamido)—4-fluorobenzoate (Intermediate 08 g, 2.75 mmol), TBD (0.383 g, 2.75 mmol) and (2—(4- methylpiperaziny|)phenyl)methanamine (0.565 g, 2.75 mmol) in toluene (35 ml) were heated to 80 °C for 6 hrs. A further portion of TBD (0.06 g, 0.431 mmol) and (2- (4-methylpiperazin—1-y|)phenyl)methanamine (0.100 g, 0.487 mmol) were added and the mixture was heated at 80 °C overnight. TBD (0.06 g, 0.431 mmol) was added and heating continued at 110 °C overnight. e was d in vacuo and the ing solid was partitioned between s sodium bicarbonate solution and EtOAc. The organic portion was washed with sodium bicarbonate solution (2 x 50 ml) 1O and concentrated in vacuo. The resulting oil was dissolved in MeOH and purified by chromatography on silica eluting with 0 — 10% MeOH in DCM. The fractions were combined and the solvent removed in vacuo. The resulting solid was recrystallised from EtOAc (50 ml) to afford the title compound; LC-MS: Rt 0.85 mins; MS m/z 567.4 {M+H}+; Method 2minLC_v003 1H NMR (400MHz, d6-DMSO) 5 10.3 (1H, s), 9.4 (1 H, d), 8.9 (1H, t), 8.6 (1H, s),8.2 (1H, d), 8.1 (1H, s), 7.8 (1H, m), 7.4 (1H, d), 7.35 (1H, d), 7.2 (2H, m), 7.1 (1H, d), 7.0 (1H, t), 4.6 (1H, d), 2.9 (4H, m), 2.4 (3H, t), 2.2 (3H, s). _St_ep_2: N—(2—Fluoro(2-(4-methylpiperazin-1—yl)benzylcarbamoyl)phenyl)—7-(1- methyl-1 H—pyrazol-S-yl)imidazo[1,2-a]pyridinecarboxamide A mixture sing 7—bromo—N-(2-fluoro(2-(4-methylpiperazinyl)benzyl carbamoyl)phenyl)imidazo[1,2-a]pyridine—3—carboxamide (step 1) (120 mg, 0.212 mmol), 1-methyl(4,4,5,5-tetramethy|—1,3,2-dioxaborolanyI)—1H-pyrazole (44.2 mg, 0.212 mmol), cesium carbonate (69.1 mg, 0.212 mmol) and PdCl2(dpp0.CHzclg adduct (17.33 mg, 0.021 mmol) in DMF (5 ml) under N2 was heated at 85 °C for 2 hrs.
After cooling to RT, the reaction mixture was partitioned between with EtOAc and water. A precipiate formed which was removed by filtration and discuarded. The organic portion was separated and washed with , water, dried ) and concentrated in vacuo. The crude product was purified by chromatography on silica eluting with 020% MeOH/DCM and the product fractions were combined and 3O concentrated in vacuo. The product was dissolved in MeOH and passed through a 1g 2,4,6-trimercaptotriazine silica column under gravity. The column was washed with MeOH and the ed eluents were concentrated in vacuo. The product was triturated with THF (1% MeOH) foilowed by ether to afford the title compound; LC-MS: Rt 0.80 mins; MS m/z 567 {M+H}+; Method 2minLC_v003 1H NMR (400MHz, MeOD) 6 9.6 (1H, s), 8.6 (1H, s), 8.4 (1H, m), 7.9 (1H, s), 7.8 (1H, m), 7.6 (1H, s), 7.45-7.3 (5H, m), 7.2 (1H, m), 6.7 (1H, s), 4.75 (2H, s), 4.1 (3H, s), 3.4 (4H, s broad), 3.2 (4H, s broad), 2.95 (3H, s).
WO 30802 Example 1.2 7-(3-Fluoro(2-hydroxyethylcarbamoyl)phenyl)-N—(2-fluoro(2-(4- methylpiperazinyl)benzyicarbamoyl)phenyl)imidazo[1,2-a]pyridine carboxamide / /N HN O A mixture comprising 7-bromo—N-(2-fluoro(2-(4-methylpiperazin yl)benzylcarbamoyl) phenyl)imidazo[1,2-aipyridinecarboxamide (Ex 1.1, step 1) 1O (120 mg, 0.212 mmol), 3-fluoro—4—(2—hydroxyethylcarbamoyl) phenylboronic acid (48.2 mg, 0.212 mmol), cesium carbonate (69.1 mg, 0.212 mmol) and PdC|2(dppf).CHZCl2 adduct (17.33 mg, 0.021 mmol) in DMF (3 ml) under N2 was heated at 85 °C for 2 hrs. Further portions of 3—fluoro~4—(2-hyd roxyethylcarbamoyl) boronic acid (48.2 mg, 0.212 mmol), cesium carbonate (69.1 mg, 0.212 mmol) and PdCl2(dppf).CH2Cl2 adduct (17.33 mg, 0.021 mmol) were added and heating continued at 100 °C overnight. The reaction mixture was diluted with EtOAc and water and the biphasic mixture was filtered. The filtercake was washed with MeOH and the filtrate was trated under vacuum. Purification of the crude residue by chromatography on silica eluting with 0—20% NH3 MeOH/DCM afforded a beige solid.
The solid was triturated with THF/ether and dried in vacuo at 45 °C overnight to afford the title nd; LC-MS: Rt 0.81 mins; MS m/z 668/669 {M+H}+; Method _v003 av55463 1H NMR (400MHz, CD3OD) 6 9.6 (1H, s), 8.6 (1H, s), 8.4 (1H, d), 8.1 (1H, s), 8.0 (1H, t), 7.8 (3H, m), 7.6 (1H, d). 7.5-7.2 (4H, m), 7.2 (1H, t), 4.7 (2H, s), 3.75 (2H, t), 3.6 (2H, t), 3.1 (4H, s broad), 3.0 (4H, s broad), 2.6 (3H, s).
Example 1.3(i) and 1.3(ii) Step1: Example 1.3 (i): 7-Bromo—N-(2—methyl-5—(2-(4—methylpiperazin yl)benzylcarbamoy|)phenyl)imidazo[1,2-a]pyridine—3—carboxamide Methyl 3—(7-bromoimidazo[1,2-a]pyridinecarboxamido)methylbenzoate (Intermediate 1B)(4OO mg, 1.030 mmol) and TBD (143 mg, 1.030mmol) in toluene (10 ml) was treated with (2-(4-methylpiperazinyl)phenyl)methanamine (212 mg, 1.030 mmol) and heated at 100 °C under nitrogen overnight. Further TBD (106 mg. 0.82 mmol) was added the mixture was heated at 100 °C for a further 24hrs.The t was removed in vacuo and the residue was partitioned between EtOAc (50 ml) and sodium onate solution (50 ml).The organic portion was separated and washed with sodium bicarbonate (2 x 25 ml), dried (MgSO4), filtered and evaporated to s to give a yellow solid. Purification by chromatography on silica eluting with O — 20 % 2M NH3 in MeOH / DCM ed a colourless oil. The oil was dissolved in minimum volume of EtOAc and titurated iso—hexane to afford the title nd as a white solid; LC—MS: Rt 0.85 mins; MS m/z 561.4 {M+H}+; Method 2minLC_vOO3 1H NMR (400MHz, DMSO-d6)510.1 (s, 1H), 9.4 (d, 1H), 8.9 (t, 1H), 8.5 (s, 1H), 8.1 (s, 1H), 7.9 (s, 1H), 7.7 (d, 1H), 7.4 (d, 1H), 7.3 (d, 1H), 7.2 (m, 2H), 7.1 (d, 1H), 7.0 (m, 1H), 4.6 (d, 2H), 3.1 (t, 4H), 2.4 (s, 3H), 2.3 (s, 3H) Step 2 Example 1.3 (ii): 7-(1-MethyI-1H-pyrazol—S-yl)—N-(2—methyl—5—(2-(4- methylpiperaziny|)benzylcarbamoyl) phenyl)imidazo[1,2-a]pyridinecarboxamide Under nitrogen, a mixture comprising 7-bromo—N—(2—methyl—5—(2—(4—methylpiperazin- 1—yl)benzylcarbamoyl) phenyl)imidazo[1,2-a1pyridinecarboxamide (step 1) (77.8 mg, 0.125 mmol), 1~methyl—5-(4,4,5,5—tetramethyl-1,3,2-dioxaborolanyl)—1 H- pyrazole (51.9 mg, 0.249 mmol) and cesium carbonate (40.6 mg, 0.125 mmol) in DMF (1 ml) was treated with PdCl2(dppf).CH2Cl2 adduct (50.9 mg, 0.062 mmol) and heated at 110 °C for 1 hr. The solvent was removed in vacuo and purification by chromatography on silica eluting with 0 — 20% 2M NH3 in MeOH / DCM afforded a black solid. The solid was dissolved in 7M NH3 in MeOH (12 ml) and passed through a 1 g Pd scavenger dge. The solvent was removed in vacuo and the ing solid was dried to yield the title compound; LC-MS: Rt 0.98 mins; MS m/z 563.5 {M+H}+; Method 2minLC__v003 av55760 1H NMR z, MeOD) 6 9.5 (1H, d), 8.55 (1 H, s), 8.0 (1H, s), 7.8 (1H, s), 7.7 (1H, dd), 7.55 (1 H, s), 7.4 (2H, m), 7.2 (3H, m), 7.1 (1H, t), 6.6 (1H, d), 4.7 (2H, t), 4.0 (3H, s), 3.1 (7H, b), 2.7 (4H, s), 2.4 (3H, 8) Example 1.4 N -(2-Fluoro (2-(4-me1:hylpiperazin-1 -yl)benzylcarbamoyl)phenyl)—7-(pyridine yl)imidazo[1,2-a]pyridinecarboxamide W0 2013/030802 PCT/IBZOIZ/054501 \ / /N \ N / HN O Under nitrogen, 7-bromo-N-(2-fluoro(2-(4—methylpiperazin—1-yl)benzylcarbamoyl) phenyl)imidazo[1,2-a]pyridine-3—carboxamide (Ex 1.1, step 1) (120 mg, 0.212 mmol), pyridineylboronic acid (33.8 mg, 0.212 mmol), triethylamine (0.030 ml, 0.212 mmol), cesium carbonate (69.1 mg, 0.212 mmol) and PdCl2(dppf).CHQC|2 adduct (17.33 mg, 0.021 mmol) in DMF (3 mi) were heated at 85 °C overnight. A r portion of cesium carbonate (24.34 mg, 0.075 mmol) and PdCl2(dppf).CH2Cl2 adduct (6.10 mg, 7.47 pmol) were added along with 3-(1,3,2-dioxaborinanyl)pyridine (34.6 mg, 0.212 mmol) and heated continued at 85 °C for 26 hrs. A further n of 1O cesium carbonate (24.34 mg, 0.075 mmol) and PdCl2(dppf).CH2Cl2 adduct (6.10 7.47 pmol) and 3—(1,3,2—dioxaborinan-2—yl)pyridine (34.6 mg, 0.212 mmol) were added. The reaction was heated at 100 °C for 2hrs. The reaction mixture was diluted with EtOAc and water and the resulting product which iated out was collected by filtration. Further product was contained in the organic phase, which was washed with set. NaHCOs, brine, dried (MgSO4) and concentrated in vacuo. The crude residue was combined with the precipitated product and purified by chromatography on silica eluting with 0-20% CM. The ing solid was purified by ative LC-MS eluting with 0.1% diethylamine 30-70% acetonitrile/water. The product fractions were partitioned between EtOAc and NaHCOS. The organic portion was separated, dried (MgSO4) and concentrated in vacuo to afford the title compound; LC-MS: Rt 1.79/1.81 mins; MS m/z 564/565 {M+H}+; Method 10minLC_v003 1H NMR (400MHz, CD30D) 5 9.7 (1H, d), 9.1 (1 H, d), 8.7 (1H, m), 8.6 (1H, s), 8.4 (1H, m), 8.3 (1H, m), 8.2 (1H, s), 8.1 (1H, s), 7.8 (1H, m), 7.7-7.5 (2H, m), 1 (5H, m), 4.3 (2H, s), 3.1 (4H, m), 2.8 (4H, s broad), 2.5 (3H, t).
Example 1.5 N-(2-Fluoro(2-(4-methylpiperazinyl)benzylcarbamoyl)phenyI)(1-methyl- 1H-pyrazolyl)imidazo[1,2-a]pyridinecarboxamide hydrochloride 83:11; N-(2-Fluoro-5—(2—(4-methylpiperazinyl)benzylcarbamoyl)phenyl)(1- methyle1 H-pyrazolyl)imidazo[1,2~a]pyridinecarboxamide A e comprising 7-bromo-N-(2—fluoro—5—(2-(4—methylpiperazin-1— y|)benzy|carbamoyl) phenyl)imidazo[1,2—a]pyridinecarboxamide (Ex 1.1, step 1) (100 mg, 0.177 mmol) and cesium carbonate (230 mg, 0.707 mmol) in DME (2.5 ml) and water (1 ml) under nitrogen was treated with 1-methyl-4—(4,4,5,5-tetramethyl- 1,3,2—dioxaborolan-2—yl)-1H-pyrazole followed by PdCl2(dppf).CH2Ci2 adduct (7.22 mg, 8.84 pmol) and heated using microwave radiation at 100 °C for 1 hr. After cooling to RT, the on mixture was partitioned between water (4 ml) and EtOAc (10 ml)/ MeOH (1ml). The organic portion was separated, dried (M9804) and concentrated in vacuo. Puriifcation by chromatography on silica g with 0—20% MeOH in DCM ed by trituration of the resulting solid with EtOAc afforded the title compound; LC-MS: Rt 0.89 mins; MS m/z 567 {M+H}+; Method 2minLC_v003 mg; N-(2—Fluoro(2—(4—methylpiperazin-1~yl)benzylcarbamoyl)phenyl)—7~(1- methyl—1H-pyrazol—4-yl)imidazo[1,2-a]pyridinecarboxamide hydrochloride N-(2—Fluoro—5-(2-(4-methylpiperazin-1—yl)benzyicarbamoyl)phenyl)-7—(1—methyl—1H- pyrazol-4—yl)imidazo[1,2-a]pyridine-S-carboxamide (step 1) (30 mg, 0.053 mmol) in MeOH (2 ml) was d with HCl in ether (0.053 ml, 0.053 mmol) and the solution was evaporated to dryness and dried in vacuo overnight to afford the title compound; LC-MS: Rt 0.82 mins; MS m/z 56, [M+H]+; Method 2minLC_v003 2012/054501 The compounds of the following tabulated Examples (Table 1) were prepared by a similar methods to that of Example 1.1—1.5 from the appropriate starting compounds, the preparations of which are detailed in the ‘Preparation of intermediates’ section.
INMR 505.4 [M+H]+; Method 10m in LC_v003 1H NMR (400 MHz, DMSO) 10.35 (1H, s), 9.50 (1H, d), 9.17 (1H, t), 8.68 (1H, s), 8.19 (1H, d), 8.00 (1H, s), 7.75 (1H, m), 7.58 (1H, s), 7.42-7.50 (3H, m), 7.18 (1H. m), 4.48 (2H, d), 4.01 l (3H, s). l 5N—(5-(3,4-difluoro benzyicarbamoyl)—2- l fluorophenyl)—7—(1—methyl—1H—pyrazol—S—yl) 1.6 ' imidazo [1 ,2-a]pyridine—3—carboxamide l Rt 0.93 mins; MS m/z 489/470/471 {M+H}+; Method 2minLC_v003 1H NMR (400MHz, DMSO- d6) 510.3 (1H, s), 10.0 (1H, s), 9.1 (1H. t), 8.6 (1H. s), 8.2 (1H, m). 7.9 (1H, d), 7.8 (3H. m), 7.5 (1H, t), 7.35 (4H, m), 7.3 (1H, m), 5.7 (1H, s). 4.5 (2H, d), 3.9 (3H, N—(5—(benzylcarbamoyl)—2-f|uorophenyl)—6—(1 — methyl-1 zoI—3-yl)imidazo[1,2—a]pyrldine—3— “|.7 Ecarboxamide Rt =0.78 mins; MS mlz 578.5 [M+H]+; Method 2minLC_v003 ; 1H NMR (400 MHz, DMSO) 9.60 (1H, s), 8.93 (1H, s), 8.62 (1H, d), 8.49 (1H. s), 8.18 (1H, d), 7.98 (1H, s), 7.87 (1 H, d), 7.57 (3H. m), 7.42 (1H, d). 7.38 (1H, d), 7.30 (1H, dd), 7.22 (1H, d), N-(4—fluoro—2-me’thyl(2‘("r‘meth3"l piperazin-l- 7.13 (2H, m), 4.73 (2H, d), yl) benzylcarbamoyl)phenyl)-7*(Dyridin9'3' 3.05 (4H. m), 2.90-2.75 yl)imidazo[1,2—a]pyridine—3—carboxamide (4H, m). 2.48 (3H, s), 2.39 (3H, s).
F 0.92 mins; MS m/z n |:\ 2 603.6 {M+H}+; Method _vOO3 , / 1H NMR(400MH2, MeOD) \ 6 9.6 (1H, s), 8.6 (1H, d), 8.5 (1 H, s), 8.4 (1H, dd), F 8.2 (1 H, dd), 8.0 (1H, s), F 7.8 (1H, m), 7.5 (1H, d), 7.4 N—(5-(3,4-dif!uorobenzyloarbamoyl)—2- (1H, d), 4.3 (3H, rn), 7.0 fluorophenyl)—7—(6~(3—(dimethyl amino) propoxy) (1H, d), 4.6 (2H, s), 4.5 (2H, pyridine—3—yl)imid azo[1,2~a]pyridine—3— d), 2.6 (2H, t), 2.3 (6H, s), 1.9 carboxamide 2.0 (2H, m) Rt 1.91mins; MS m/z 518.40 [M+H]+; Method 10min LC_v003 1H NMR z, DMSO— d6) 6 10.7 (1H, s), 10.0 (1/2H,3 m), 9.5 (1H, d), 9 (1H, t), 8.8 (1H, s), 8.2 (1H, m) 8.1 (1H, s), 7.6 (1H, s), 7.5 (2H, d), 6.6 (1H, s) 4 (3H, s), 3.6 (2H, m), 3.4 (2H, m), 3.3 (2H, m), 3.1 N-(5—(2-(2,S-cis-dimethylpiperidin (1H, m), 1.9 (1H, m), 1.6 (2H, yl)ethylcarbamoyl)—2—fluorophenyI)—7-(1methyl- m), 1.5 (2H, m), 1.4 (4H, d), 1H—pyrazoI—5—y|)imidazo[1,2—a]pyridine~3- 1.3(2H,d) 31.10 carboxamide Rt 0.88 mins; MS m/z 592 ; Method 2minLC_v003 1H NMR (400MHz, DMSO— d6) 510.3 (broad 1H, s), 9.52 (1H, d), 9.0 (1H, t), 8.7 (1H, s), 8.25 (1H, d), 8.1 (1H, s), 7.9 (2H, d), 7.6 (1H, d), 7.5-: 7.4 (3H, m), 7.25—72 (2H, m), 7.15 (1H, d), 7.1 (1H, t), 4.6 (2H, d), 3.8 (2H, s), 2.9 (4H, s), 2.5 (4H, s), 2.25 U\ (3H, s), 2.1 (2H, s broad). :7-(4-(aminomethyl) phenyl)—N—(2-fluoro-5—(2—(4- methylpiperaziny|)benzylcarbamoyl) 1.11 phe:yl)imidazo[1,2—-a]pyridine—3—carboxamide Rt= 0.83 mins; MS m/z 578.5, [M+H]+; Method _v003 1H NMR (400 MHz, DMSO) 10.25 (1H, s), 9.35 (1H, d), 8.60—8.43 (2H, m), 8.15 (2H, s), 7.97 (1H, s), 7.77 2 N—(5-(2-tert—butoxy ethylcarbamoyI)-2— (1H, m), 7.60-7.35 (3H, m), fluorophenyI)(1-(2—morphoIInoethyI)—1 H— 4.28 (2H, m), 3.55 (4H, m), pyrazoly1)imidazo[1,2-a]pyridine—3- 3.40 (2H, m), 2.50—2.80 :1.12 carboxamide (8H, m), 1.004 .21 (9H, m).
Rt = 0.96 mins; MS m/z 590.5, [M+H]+; Method (FEES?!F H I "1‘ 2minLC_v003 1H NMR (400 MHz, DMSO) \ \ [E 1 o 5 10.20 (1H, s), 9.38 (1H, d), 8.81 (2H, m), 8.45 (2H, s), 8.18 (2H, m), 7.98 (1H, O s), 7.79 (1H, m), 7.48-7.37 (2H, m), 4.28 (2H, t), 4.07 (5,5-dimethyltetrahydrofuran—2-yl)methyl EcarbamoyI)—2—fluorophenyIH—(l -(2- (1H, m), 3.57 (4H, m), 2.78 (2H, t), 2.52 (2H, m), 2.45 morpholinoethyl)—1H—pyrazol-4—yl)imidazo [1 ,2- Ea]pyridine-3~carboxamide (4H, m), 2.02 (1H, m), 1.70 (3H, m), 1.21 (3H, s), 1.17 (3H, s).
Rt =1.03 mins; MS mlz Fm /\ 594.4, [M+H]+; Method N\/N 2minLC_v003 o \/ 1H NMR (400 MHz, DMSO) 10.30(1H, s), 9.50 (1H, d), 8.99 (1H, t), 8.78 (1H, s), 8.65 (1H. s), 8.30 (1H, m d), 8.23 (1H, d), 8.17 (1H, N\ s), 7.85 (1H, m), 7.83 (1H, N—(2—fluoro—5—(2~(4—methylpiperazin-1 - d), 7.52—7.36 (3H, m), 7.27 Eyl)benzlearbamoyl)phenyl)—7-(6-methoxy (1H, m), 7.05 (2H, m), 4.58 pyridineyl)imidazo [1,2-a]pyridine—3- (2H, d), 3.32 (3H, s), 2.87 Ecarboxamide (4H, m), 2.52 (4H, m), 2.23 (3H, s).
Rt 0.89 mins; MS m/z 567/568/569 {M+H}+; Method 2minLC__vOO3 1H NMR (400MHz, DMSO- d6) 5 10.0 (1H, s). 9.9 (1H, m), 9.1 (1H, s), 9.0 (1H, t), 8.9 (1H, s), 8.3 (2H, m), 7.8 1 (1H. m), 7.7 (1H, d), 7.55 / (1H, s), 7.4 (1H, t), 7.3 (2H, N-(2—fluoro—5-(2-(4-methylpiperazin m), 7.2 (2H, m), 6.6 (1H, s), y1)benzylcarbamoyl)pheny1)—6—(1 ~methy1-1 H- 4.6 (2H, d), 4.0 (3H, s), 3.6— pyrazoI-S—yl)pyrazolo[1 ,5—a]pyridine~3— 3.0 (8H, m), 2.9 (3H, d). 1.1 5 carboxamide Rt 0.70 mins; MS m/z 515{M+H}+; Method 2minLC_v003 \ NE. \ [/7474 N1 I 1H NMR z, DMSO) / 0 \ ,4} 510.71 (1H, 8), 10.01 (1H, t), 9.72 (1H, d), 9.33 (1H, 0’; \3H s), 9.05 (1H, m), 8.90 (1H, 1/ CL s), 8.85 (1H, d), 8.70 (1H, M8111 m), 8.47 (1H, s), 8.23 (1H, m), 7.89 (3H, m), 7.51 (1H, m), 3.55 (2H, m), .33 1-(2-(4—fluoro—3-(7-(pyridine—S-ymmidazo [1’2— (2H. m), 1.83 (1H, m), 1.70 Ia]pyridine—3—carboxamido)benzamido)ethyl)—2,6~ (2H, m), 1.50 (2H, m), 1.40 :1.1 6 cis—dimethylpiperidinium Chloride End 1.30 (6H, 2 X d).
Rt 0.92 mins; MS m/z 577{M+H}+; Method 2mlnl.C_v003 1H NMR(4OOMHz, MeOD) 9.0 (1H, s), 837 (1H, s), 8.5 (1H, d), 8.4 (1H, d), 8.3 (1H, d), 8.1 (1H, dd), 7.85 (1H, d), 7.75 (1H, m), 7.35 (1H, t), 5.95 (1H, d), 4.4 (2H, 1), 3.6 (2H, m), 3.5 (2H, m), 2.6 (2H, t), 2.3 (6H, s), 2.0 (2H, m), 1.25 (9H, s) 2-tert-butoxyethylcarbamoyl) Efluorophenyl)—6—(6—(3-(dimethylamino) 1'17; propoxy)pyridine—3—yl)pyrazolo[1,5—a]pyridine—3- 3 carboxamide Rt 0.75 mins; MS m/z 550.5 {M+H}+; Method 2minLC_vOO3 1H NMR z, MeOD), 9.6 (1H, d), 9.0 (1H, d), 8.6 (1H, dd), 8.5 (1H, s), 8.3 (1 H, d), 8.05 (1H. s), 7.95 (1H, d), 7.7 (1H, dd). 7.5 (1 H. s), 7.55 (1H, dd), 7.4 (1 H. d), 7.35 (1H, d). 7.25 (2H, m), 7.1 (1H, t), 4.7 (2H, s), 3.0 (4H, b), 2.8 (4H, b), 2.5 (3H, s), 2.4 (3H, N-(2—methyl(2—(4—methylpiperazin-1 — yI)benzylcarbamoyl)pheny|)—7—(pyridine—3- 1.1 8 yl)imidazo[1 ,2—a]pyridine—3~carboxamide PCT/IBZOIZ/054501 Rt 0.76 mlns; MS mlz 564.5% {M+H}+; Method 2minLC_vOO3 , 1H NMR (400MHz, MeOD) 6 9.80 (1H. d), 8.85 (1H, d), 8.59 (1H, \ s), 8.40 (1H, d), 7.91 (1H. s), 7.59 (1H. d).
N \ SW l O 7.39 (2H, m), 7.30 (2H, m), 7.16 (1H, t), 8.85 (1H. d). 4.76 (2H, s). 4.04 (3H, s), 3.10 (4H, m), 2.98 (4H, b), 2.88 (3H, s), 2.59 (3H, s) /OCIH 1—methyl-4—(2-((6-methyl-5—(7-(1-methyl -1H- pyrazoI-S-yl)imld azo[1,2—a]pyrldlnecarbox nicotina mido)methyl)phenyl)piperazin chloride Example 1.10 N-(5-(2-(2,6-cis-Di methyl piperidin-1 -yl)ethylcarbamoyl)f|uorophenyl)(1 - methyl-1H-pyrazol-S-yl)imidazo[1,2-a]pyridinecarboxamide PdCl2(dppf).CHZCl2 adduct (39.5 mg, 0.048 mmol) was added to a mixture comprising 1-methy|(4,4,5,5-tetramethyl-1,3,2—dioxaborolanyl)—1H-pyrazole (commercially available) (212 mg, 1.017 mmol), 7-bromo-N-(5—(2—(2,6—cis— dimethylpiperidinyl)ethylcarbamoyl)—2-fluorophenyl)imidazo[1,2-a]pyridine-3— carboxamide (lntermediate 4C) (500 mg, 0.968 mmol), Cs2C03 (1262 mg, 3.87 mmol) in 1,2-dimethoxyethane (10 ml) and water (4.29 ml). The mixture was degassed thoroughly refilling with nitrogen (x3). The mixture was heated using microwave radiation at 100°C for 1 hour. The water was removed by pipette and the organic portion was concentrated in vacuo. The residue was dissolved in MeOH and dry loaded onto . The crude t was purified by chromatography on silica eluting with 0—20% MeOH in DCM to afford the title compound. (See Table 1 for characterising data).
The citrate salt of 2-(2,6-cis—dimethylpiperidin—1-yi)ethylcarbamoyl)—2- 1O fluoropheny|)(1-methyl-1 H-pyrazoi—5—yl)imidazo[1,2-a]pyridinecarboxamide was prepared according to the ing procedure: $9.1; 3-[(7~Bromo-imidazo[1,2-a]pyridlne—3-carbony|)-amino]-4—fluoro—benzoic acid methyl ester To the solution of compound 7-bromoimidazo[1,2-a]pyridinecarboxylic acid (intermediate A step 3) (1.26 Kg, 5.23 mol) in DMAC (15 L) was added dropwise SOClz (1.86 kg, 15.6 mol) at 10 °C in 30 min. To the resulting mixture warmed to °C was added compound methyl 3-amino—4—fluorobenzoate (884 g, 5.23 mol) in DMAC (3.0 L) over 30 min. After addition, the reaction temperature went up to 30 °C.
HPLC showed the reaction went to completion within 5 min. To the reaction mixture was added water (20 L) over 20 min. The mixture was filtered and dried under vacuum to afford the title compound as a white solid; 1H NMR (400 MHZ, DMSO-de) 8 3.87 (S, 3 H) 7.57 (dd, J=7.28, 2.01 Hz, 1 H) 7.51 (dd, J=10.16, 8.66Hz, 1 H) 7.90 (td, J=4.33, 2.38 Hz, 1 H) 8.29 (m, 2H) 8.90 (s, 1 H)9.43 (d, J=7.53 Hz,1 H) 10.78 (s, 1 H) Rt 6.90 mins; MS m/z 394.0 ; Method 10 min LC flag ro-3—{[7-(2-methyl-2H-pyrazol—3-yl)-imidazo[1,2-a]pyridinecarbonyl]~ amino}-benzoic acid 3-[(7-Bromo~imidazo[1,2—a]pyridine—3—carbonyl)—amino]—4-fluoro-benzoic acid methyl ester (step 1)(1200 g, 3.060 mol), 1-methyl(4,4,5,5-tetramethyI-1,3,2- dioxaborolan-Z-yl)-1H-pyrazole (commercially available) (764 g, 3.67 mol), PdC12(dppf)-CH2C3|2 (75.0 g, 91.8 mmol) in dioxane (10 L) and aqueous N82C03 (2 N, 4.6 L) were heated to reflux for 6 hr. The reaction e was cooled to 50 °C and filtered. The filtrate was heated to reflux, to which was added AcOH (600 g, 10.0 mol) was added dropwise. During the course of on solids came out of solution to give pale pink slurry. After addition the mixture was slowly cooled to RT and filtered. To PCT/132012/054501 the filter cake was added dioxane (20 L) followed by heating to reflux to obtain a on. The solution was cooled to RTand filtered to provide the title compound as a white solid; 1H NMR (400 MHz, DMSO-ds) 54.00 (s, 3 H) 6.67 (s, 1 H) 7.46 (t, J=9.41 Hz, 1 H) 7.40 (d, J=7.03 Hz,1 H) 7.54 (s, 1 H)7.85 (d, J=2.26 Hz, 1 H) 7.99 (s, 1 H) 8.28 (d, J=6.27 Hz, 1 H) 8.67 (s, 1 H) 9.47 (d, J=7.03Hz, 1 H) 10.35 (s, 1 H).
Rt 5.40 mins; MS m/z 380.1 {M+H}+; Method 10 min LC _S_tep__3_: ethyl—2H-pyrazol-3—yl)—imldazo[1,2-a]pyridine—3-carboxylic acid {5-{2— 1O (2,6-dimethyl—piperidinyl)-ethylcarbamoyl]—2—fluoro~phenyl}-amide 4—F|uoro—3—{[7-(2-methyl-2H—pyrazoiyl)—imidazo[1,2-a]pyridine—3-carbonyl]—amino}— benzoic acid (step 2) (450 g, 1.19 mol), EDC°HCl (454.8 g, 2.372 mol) and HOBt (181.6 g, 1.186mol) in DMF (3.2 L) at 25 °C were stirred for 1.5 hr. The reaction was monitored by HPLC. To the reaction mixture was se added cis 2-(2,6—dimethyl- piperidinyl)-ethylamine (222.5 g, 1.423 mol) over 10 min and stirring continued for min. To the on mixture was se an aqueous solution of Na2C03 (5%, 6 L) over 120 min and the ing solid was collected by filtration and washed with water (5 L). To the solid was added ethanol (5 L) foliowed by heating to 70 °C to obtain a clear solution. Water (1. 5 L) was se added at 70 °C and stirred for 30 min. The clear solution was slowly cooled to 25 °C over 2hr. The solid was filtered. washed with ethanol (500 mL) and dried under vacuum at 50°C overnight to afford the title compound as a white solid ; 1H NMR (400 MHZ, DMSO-d6)51.03 — 1.19 (m, 3 H) 1.10 (d, J=6.02 HZ, 6 H) 1.55 (br. s., 1 H) 1.50(d, J=12.30 Hz, 2 H) 2.42 (br. s., 2 H) 2.71 (br. 5., 2 H) 3.27 (d, J=5.77 Hz, 2 H) 4.00(s, 3 H) 6.60 (s, 1H) 7.41 (d, J=6.02 Hz, 2 H) 7.54 (s, 1 H) 7.77(s, 1 H) 8.00 (s, 1 H) 8.14 (d, 1 H) 8.54 (s, 1 H) 8.67(s, 1 H) 9.48 (d, 1 H) 10.35 (s, 1 H).
Rt 4.80 mins; MS m/z 518.2 {M+H}+; Method 10 min LC 8395; N-(5—((2-(2,6-cis-Dimethylpiperidiny|)ethyl)carbamoyl)—2—fluorophenyl)—7- (1-methyl-1H-pyrazolyl)imidazo[1,2-a]pyridineS—carboxamide citric acid (1:1) 7-(2-Methyl-2H-pyrazol—B-yl)-imidazo[1,2-a]pyridinecarboxylic acid {5-[2-(2,6- dimethyl-piperidin-1—yl)-ethylcarbamoyl]—2—fluoro—phenyl}-amide (step 3)(480 g) was suspended in ethanol (2300 mL) in a 5000 mL four-necked flask equipped with thermometer, reflux condenser and a nitrogen inlet. The mixture was heated to 55 °C and the suspension gradually became clear. A solution of citric acid (180 g) in PCT/IBZOIZ/054501 acetone (2.4 L) was added over 1h and the internal temperature was controlled at 45—50°C. The clear solution was stirred at 50°C for 2h. A crystal seed (1 g) was added to the reactor and the internal temperature was cooled to 20°C at a speed of 8°C/h.
The mixture was stirred at 20°C for 60 h. The resulting solid was filtered and the filter cake was washed with e (1 L) and dried in vacuum (under 4 mbar at 55°C) for 24 h to afford the title compound; 1H NMR (400 MHz, DMSO—ds) 8 1.46 (m, 3 H) 1.29 (d, J=6.02 Hz, 6 H) 1.65 (3., 1 H) 1.73(d, 2 H) 2.59 (m, 4 H) 3.18 (m. 4 H) 3.53 (d, 2 H) 4.00(s, 3 H) 6.68 (s, 1H) 7.41 (d, 1 H)7.48 (d, 1 H) 7.54 (s, 1 H)7.80(s, 1 H) 8.00 (s, 1 H)8.20 (d, 1 H)8.68 (s,1 H) 8.90 (s, 1 H) 9.48 (d, 1 H) 10.38 (s, 1 H).
Example 1.20 7-(1-Methyl-1 H-pyrazolyl)-imidazo[1,2-a]pyridinecarboxylic acid {5-[2-(2,6- cis-dimethyl-piperidinyl)-ethylcarbamoyl ]fluoro-phenyl}-amide hydrochloride o-N—(5-(2-(2,6-cis—dlmethylpiperidinyl)ethylcarbamoyl)—2-fluorophenyl) imidazo[1,2—a]pyridine-B—carboxamide (intermediate 4C) (50 mg, 0.097 mmol), 1- methyl-1H—pyrazol~4—ylboronic acid (24.38 mg, 0.194 mmol) and cesium carbonate (126 mg, 0.387 mmol) was dissolved in DME (215 pl)/water (108 pl) to form a solution. Nitrogen was bubbled though the reaction mixture for 2 minutes.
PdCl2(dppf).CH20l2 adduct (3.95 mg, 4.84 umol) was added and the mixture was heated using microwave radiation at 100°C for 15 mins. The water was removed and the organic portion was dry loaded onto .
. The crude product was purified by chromatography on silica g with O-20°/o 2M NH3 in MeOH and the product fractions were ed and concentrated in vacuo. The product was dissolved in MeOH and passed through a 1g trimercaptotriazine silica. The solvent was 2012/054501 removed in vacuo and the residue was triturated with ether. The resulting precipitate was filtered and dried in the oven to afford the title compound; LC—MS: Rt 0.30 mins; MS m/z H}+; Method 2minLC_v003 1H NMR (400MHz, DMSO)610.4(1H, s), 10 (1H, d), 9.4 (1H, d), 8.9 (1H, t), 8.7 (1H, s), 8.55 (1 H, s), 8.2 (2H, d), 8 (1H, s), 7.85 (1H, m), 7.6 (1H, d), 7.5 (1H, t), 4.2 (2H, q), 3.55 (2H, m), 3.4 (4H, m), 3.3 (2H, m), 3.1 (1H, m) 1.9 (1H, d), 1.7 (2H, m), 1.5 (2H, m), 1.4 (4H, m), 1.3 (2H, d), 1.1 (1H t).
Example 1.21: 1O 6-(1-Methyl-1H-pyrazoly|)-N-(2-methy|(2-(4-methylpiperazin yl)benzylcarbamoyl)pyridinyl)pyrazolo[1,5-a]pyridinecarboxamide The title compound was prepared from intermediate 1D and (2—(4—methylpiperazin-1— yl)phenyl)methanamine analogously to Example 1.1 step 1; LC-MS: Rt 0.66 mins; MS m/z 564.7{M+H}+; Method 2minLopr Example 2.1 N-(5-(2-tert-ButoxyethylcarbamoyI)fluorophenyl)(6-(3- (dimethylamino)propoxy) pyridinyl)imidazo{1,2-a]pyridinecarboxamide PCT/IBZOIZ/054501 \\\\N/ 7-Bromo—N-(5-(2—tert—butoxyethylcarbamoyI)—2-fluorophenyl)imidazo[1,2—a]pyridine—3— carboxamide (Intermediate 2A) (300 mg, 0.629 mmol), N,N-dimethyl—3—(5—(4,4,5,5— tetramethyl-1,3,2-dioxaborolan—2—yl)pyridinyloxy)propan—1~amine (202 mg, 0.660 mmol) and cesium carbonate (819 mg, 2.51 mmol) were combined in 1,2- dimethoxyethane (7 ml) and water (3 ml). The mixture was degassed thoroughly refilling with en and treated with PdCI2(dppf).CH20l2 adduct (25.7 mg, 0.031 mmol). The mixture was once again degassed thoroughly refilling with nitrogen and heated using microwave radiation at 100 0C for 1 hr. The aqueous was removed by 1O pipette and the organic portion was absorbed onto silica and purified by chromatography eluting with 0-20% MeOH in DCM. The resulting solid was tallised from EtOAc to afford the title nd; LC-MS: Rt 2.55 mins; MS m/z 577.5, [M+H]+; Method C_v003 1H NMR (400 MHz, DMSO) 6 10.25 (1H, s), 9.46 (1H, d), 8.73 (1H, s), 8.65 (1H, s), 8.55 (1H, t), 8.24 (1H, d), 8.16 (2H, m), 7.80 (1H, m), 7.63 (1H, d), 7.45 (1H, t), 6.95 (1H, d), 4.36 (2H, t), 3.45 (2H, m), 3.35 (2H, m), 2.35 (2H, m), 2.15 (6H,s), 1.87 (2H, m), 1.16 (9H, 3).
Example 2.2 N~(5-((5,5-Dimethyltetrahydrofuran-Z-yl)methylcarbamoyl)-2—fl uorophenyl)(6- (4-methylpiperaziny|)pyridinyl)imidazo{1,2-a]pyridinecarboxamide The title nd was prepared from commercially available 1-methyl(5—(4,4,5,5- tetramethyl-1,3,2—dioxaborolan—2—yl) pyrldin—Z-yl)piperazine and lntermediate 4E analogously to Example 2.1; LC-MS: Rt 0.71 mins; MS m/Z 586/587 {M+H}+; Method 2mlnLC_v003 Example 2.3: N-(5-(3,4-Difluorobenzylcarbamoyl)fluorophenyl)—7-(1-(3-(dimethyl amino)propyl)-1H-pyrazolyl)imidazo[1,2-a]pyridinecarboxamide F N The title compound was prepared from commercially available N,N—dimethyl—3—(4— (4,4,5,5—tetramethyI—1,3,2—dioxaborolan—2—yl)4 H—pyrazoIy|)propan—1—amine and lntermediate 3A analogously to Example 2.1; LC-MS: Rt 0.69 mins; MS m/z 576/577/578 {M+H}+; Method 2minLC_vOO3 Example 2.4 N -(5-((5,5-Dimethyltetrahyd rofu ranyl)methylcarbamoyl)fluorophenyl)(5- ahydro-2H-pyranylamino)methyl)pyridinyl)imidazo[1,2-a]pyridine amide Step 1: N-(5—(((5,5—Dimethyltetrahydrofuran—2-yl)methyl)carbamoyl)—2-fluoropheny|)- 7-(5—formylpyridinyl)imidazo[l ,2—a]pyridine—3-carboxamide The title compound was prepared from 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolanyl)nicotinaldehyde and Intermediate 4E analogously to Example 2.1; LC-MS: Rt 0.87 mins; MS m/z 7/518 {M+H}+; Method 2minLC_v003 flap—2: N-(5—((5,5-Dlmethyltetrahydrofuranyl)methy|carbamoyl)-2—fluorophenyl)—7- (5-((tetrahydro-2H—pyran-4—ylamino)methyl)pyridin—3-yl)imidazo[1,2-a]pyridine—3— carboxamide A suspension of N-(5-(((5,5-dimethyltetrahydrofuran-2—yl)methyl)carbamoyl)—2— fluorophenyl)(5-formylpyridin-S—yl)imidazo[1,2—a]pyridine—3—carboxamide (step 1)(98 mg, 0.19 mmol), tetrahydro—2H-pyranamine (38.4 mg, 0.380 mmol), acetic 1O acid (0.2 ml, 3.49 mmol) and 2—picoline borane (24.16 mg, 0.228 mmol) in MeOH (2 ml) was heated at 50°C for 3 hrs. The on mixture was concentrated under vacuum and the residue was redissolved in 10% MeOH/EtOAc. The mixture was washed with sat. NaHC03 and H20. The organic portion was dried MgSO4 and concentrated in vacuo. The crude product was purified by chromatography on silica eluting with 0-20% 2M NH3 in MeOH/DCM. The product ons were combined and concentrated in vacuo. The residue was triturated with EtOAc/iso—hexane to afford the title compound; LC-MS: Rt 0.69 mins; MS m/z 601/602 {M+H}+; Method 2minLC_v003 The compounds of the following ted Examples (Table 2) were prepared by a similar method to that of e 2.4 from the appropriate starting compounds, the preparations of which are ed in the ‘Preparation of intermediates’ section.
Table 2 Stru‘ct—r m«HA 7" - LC-MS/NMR Name LCMS: Rt 0.66 mins; MS m/z 563/564 {M+H}+; Method 2minLC__v003 (S)-N-(5—(((5,5—Dimethyltetrahydrofuran—2—yl)methyl) carbamoyl)—2-fluorophenyl)—7-(5—(((2-fluoroethyl) amino)methyl)pyridin—3-yi)imidazo[1,2-a]pyridine LCMS: Rt 0.68 mins; MS m/z 589 ; Method 2min LC_v003 (R)-N-(5-(((5,5—dimethyltetrahydrofuran-2—yl)methyl) carbamoyl)—2—fluorophenyl)—7-(5-(((2-methoxyethyl) E(methyl)amino)methyl)pyridinyl) imidazo[1,2~a] 2.6 pyridine—3-carboxamide _*E (R)(5-((tert—butyiamino)methy|)pyridin—3—yi)—N—(5- LCMS: Rt 0.69 mins; (((5,5~dimethylt etrahydrofuran-2— MS m/z 573 {M+H}+; yi)methyi)carbamoyi)-2—fluorophenyl) imidazo[1,2— Method 2min LC_v003 32.7 :;a]pyridine~3-carboxamide Example 3.1 N-(5-(3,4—Difluorobenzylcarbamoyl)~2-fluoropheny|)(6-(2-(pyrroiidin yl)ethoxy) pyridinyl)imidazo[1,2-a]pyridinecarboxamide hydrochloride N \ \N O \/o F m1; 7-(6—Chioropyridin—3-yl)—N-(5-(3,4—difluorobenzyicarbamoyi)—2— fluorophenyl)imidazo[1,2—a]pyridine—3-carboxamide A e comprising 7-bromo-N-(5-(3,4—difluorobenzyicarbamoyi)—2— phenyl)imidazo[1,2—a]pyridine—3-carboxamide (intermediate 3A)100 mg, 0.199 mmol), 6-chloropyridinyiboronic acid (31.3 mg, 0.199 mmoi) and cesium carbonate (259 mg, 0.795 mmol) in DME (631 uL)/water (31.5 (L) was purged with nitrogen and d with PdClg(dppf).CH2C|2 adduct (8.11 mg, 9.93 umol) The resulting mixture was heated using microwave radiation at 100°C for 15 mins. The water was removed PCT/IBZOIZ/054501 by pipette and the reaction mixture was d with MeOH. Purification of the mixture by tography on silica eluting with 0—20% 2M NH3 in MeOH/DCM afforded the title nd; LC-MS: Rt 0.75 mins; MS m/z 536 {M+H}+; Method _30_v003 mN-(5-(3,4-Difluorobenzylcarbamoyl)—2-fluorophenyI)—7-(6—(2-(pyrrolidin~1- yl)ethoxy) pyridin-3—yl)imidazo[1,2—a]pyridine-3~carboxamide hydrochloride A solution of 2-(pyrrolidin—1-y|)ethanol (30.5 mg, 0.265 mmol) and sodium hydride (6.36 mg, 0.265 mmol) in toluene (442 pl) was stirred at RT for 30 mins and treated with 7-(6-chloropyridinyl)—N-(5-(3,4—difluorobenzylcarbamoyl)—2- 1O fluorophenyl)imidazo[1,2—ajpyridinecarboxamide (step 1). The reaction mixture was heated at 60°C for 1 hour and the reaction was ed with wet MeOH. cation of the mixture by chromatography on silica eluting with 0-20% (2M NH3 in MeOH)/DCM afforded a yellow solid. The solid was dissolved in MeOH and treated with 2M HCI in diethyl ether (1eq). The solvent was removed in vacuo and purification of the crude product by chromatography on silica eluting with 2M NH3 in MeOHlTBME afforded a solid. The solid was treated again with 2M HCI in diethyl ether and concentrated in vacuo to afford the title nd; LC—MS: Rt 0.52mins; MS m/z 615 [M+H]+; Method 2minLC_30_v003 1H NMR (400MHz, DMSO-d6) 610.5 (1H, s), 9.6 (1H, s), 8.8 (2H, d) 8.4 (1H, d), 8.2 (1H, d), 7.9 (1H, m), 7.7 (1H, d), 7.45 (1H, t), 7.4 (1H, m), 7.3 (1 H, s), 7.2 (1H, m), 7.15 (1H, s), 7.1 (1H, d), 7(1H, s), 4.7(2H, d), (4.5 (2H, d), 3.3 (4H, m), 3.1 (2H, m), 2.05 (2H, m), 1.95 (2H, m).
The compounds of the following tabulated Examples (Table 3) were prepared by a similar method to that of Example 3.1 from the appropriate starting compounds, the preparations of which are detailed in the ‘Preparation of Intermediates’ section.
Table 3 - LC-MSINMR Name LC-MS: Rt 0.87 mins; MS m/z 589 [M+H]+; Method 2minLC_v003 N—(5-((2-(tert—Butoxy)ethyl)carbamoyl)—2— fluorophenyl)~7—(6—(2-(pyrrolidin—1—yl) )pyridin— 13.2 3—yl) imidazo[1,2—a] pyridinecarboxamide LC—MS: Rt 0.71 mins; MS m/z 601/602 {M+H}+; Method 2minLC_v003.
:N—(5—(((5,5-dimethyl tetrahydrofuran—Z— yl)methyl)carbamoyl)—2—fluorophenyl)—7-(6-((1—methy| piperidinyl)oxy) pyridin—S—yl)imidazo[1 ,2— 3.3 a]pyridine—3-carboxamide Example 4.1 6-(1 -Methyl-1 zol-S-yI)-N-(2-methyl(2-(4-methy|piperazin yl)benzylcarbamoyl) phenyl)pyrazolo[1,5-a]pyridinecarboxamide hydrochloride 6-Bromo—N—(Z-methyl~5~(2—(4~methyipiperazin-t—yl)benzylcarbamoyl)phenyl)pyrazolo [1 ,5—a]pyridine—3—carboxamide (169 mg, 0.301 mmol), -dimethyl-2,4-dioxa borabicyclo[3.1.0]hexan-3—yl)-1~methyl—1H-pyrazole (75 mg, 0.391 mmol) and cesium carbonate (392 mg, 1.204 mmol) in DME (3209 1.1L) and water (1284ul) were combined to give a yellow solution. PdCl2(dppf).CH20l2 adduct (6.15 mg, 7.52 umol) was added and the mixture was heated using microwave radiation at 100°C for 1hr A further portion of PdCl2(dppf).CHQCl2 adduct (6.15 mg, 7.52 umol) was added and heating continued using microwave radiation at 100°C for 1hr. 5-(1,5-Dimethyl-2,4- dioxaborabicyclo[3.1.0]hexanyl)—1-methyl-1H-pyrazole (75 mg, 0.391 mmol), PdCl2(dppf).CH20|2 adduct (6.15 mg, 7.52 pmol) and cesium carbonate (392 mg, 1.204 mmol) were added. The mixture was heated using micriowave radiation at 100°C for 2hrs.The product was purified by tography on silica eluting with 0— % 2M NH3 in MeOH/DCM followed by a second column using 0—15% 2M NH3 in MeOH/DCM. The resulting e was ved in CM, filtered through a glass fibre filter paper and purified by preparative chromatography eluting with 20- 50% MeCN/water (0.1% TFA). The appropriate fractions were partitioned between with NaHC03 and EtOAc and the c portion was dried (M9804) and concentrated to afford a colourless oil. HCl (1 equiv.) in dioxane was added and trituration with EtOAc/EtOH afforded the title compound as a solid; LC-MS: Rt 2.66 mins; MS m/z 563/564/565 {M+H}+; Method C__v003 1H NMR (400MHz, CDgOD) 6 8.9 (1 H, s), 8.7 (1 H, s), 8.4 (1H, d), 7.95 (1H, s), 7.7 (1H, d), 7.65 (1h, d), 7.6 (1H, s), 7.4 (1H, d), 7.35 (1H, d), 7.3 (1H, m), 7.2 (1H, m), 7.1 (1 H, t), 6.6 (1 H, s), 4.7 (2H, s), 4.0 (3H, s), 3.0 (4H, m), 2.7 (4H, s broad), 2.45 (6H, d).
Example 5.1 N-(2-Bromo-S-(Z-(4-methylpiperazinyl)benzylcarbamoyl)phenyl)imidazo[1,2- a]pyridinecarboxamide WO 30802 @111; 3—Amino—4—bromo—N—(2~(4—methylpiperazin—1~yl)benzyl)benzamide N1,N1—dimethyl—N2—((propylimino)methylene)ethane~1,2—diamine (0.70 ml, 3.99 mmol) was added to a stirred solution / sion of o—4—bromobenzoic acid (719 mg, 3.33 mmol), (2—(4-methylpiperazinyl)phenyl)methanamine (820 mg, 3.99 mmol) and HOBt (140 mg, 1.0 mmol) in dry DCM (20 ml) under argon. After 24 hrs the reaction mixture was diluted with DCM and washed several times with water.
The solvent was removed in vacuo and the resulting yellow oil was triturated with DCM/ diethyl to give a yellow crystalline powder; LCMS: Rt 0.85 min; MS m/z 403.2 [M+H]+; Method 2minLC_v003 1H NMR (400 MHz, CDCI3) 5 6.48 (1H, d), 6.42 (1H, br s), 7.30 (3H, m), 7.20 (1H, d), 7.14 (1H, t), 6.92 (1H, d), 4.72 (2H, d), 4.23 (2H, br s), 3.02 (4H, br s(, 2.62 (4H, br s), 2.35 (3H, s) mN-(2—Bromo—S—(Z—(4-methylpiperazin—1-y|)benzylcarbamoyl)phenyl)imidazo [1 ,2-a]pyridine—3-carboxamide lmidazo[1,2-a]pyridine—3~carbonyl chloride hydrochloride (0.97 mg, 4.46 mmol) was slowly added in portions to a stirred on of 3-amino—4-bromo-N-(2—(4— methylpiperazin-1~yl)benzyl)benzamide (step 1)(900 mg, 2.23 mmol) in dry pyridine (15 ml). The reaction was stirred at RT for 18 hrs and quenched with water. The solvent removed in vacuo. The residue was treated with a small amount of MeOH to dissolve insoluble material then partitioned between aq. NaHC03 and DCM. The organic t was washed with brine, dried over M9804 and concentrated in vacuo.
The crude product mixture was crystallised from MeOH to give a white solid; LCMS: Rt 0.78 min; MS m/z 549.2 [M+H]+; Method 2minLC_v003 1H NMR (400 MHz, MeOD) 5 9.52 (1H, s), 8.53 (1 H, s), 8.25 (1H, s), 7.85 (1H, d), 7.77 (1H, d), 7.68 (1H, d), 7.60 (1H, t), 7.35 (1H, d), 7.22 (3H, m), 7.10 (1H, t), 4.72 (2H, d), 3.0 (4H, br s), 2.67 (4H, br s), 2.37 (3H, s).
Example 5.2 N-(2-Bromo(2-(4-methylpiperazinyl)benzy|carbamoyl)phenyl)pyrazolo[1,5- a]pyridinecarboxamide /N The title compound was prepared from obromo-N-(2—(4-methylpiperazin yl)benzyl)benzamide (Ex 5.1, step 1) and pyrazolo[1,5-a]pyridine-3~carbonyl chloride (commercially available) ously to Example 5.1; LCMS: Rt 2.12 min; MS m/z 547.3, 549.3 [M+H]+; Method Lopr_v002. 1O 1H NMR (500MHz, dB-DMSO) 6 9.90 (1H, s), 9.05 (1H, t), 8.85 (1H , d), 8.79 (1H,s), 8.22 (1H,d), 8.14 (1 H, d), 7.84 (1 H, d), 7.74 (1H, dd), 7.54 (1 H, m), 7.23 (2H, m), 7.13 (2H, m), 7.05 (1H, m), 4.57 (2H, d), 2.87 (4H, m), 2.50 (4H, m), 2.23 (3H, s).
Example 6.1 N —(5-(3,4-Difluorobenzylcarbamoyl)fl uorophenyl)(3-hydroxymethyl butyl)imidazo[1,2-a]pyridinecarboxamide WO 30802 HN O mEthyl 3—(3-(5-(3,4-difluorobenzylcarbamoyl)fluorophenylcarbamoyl) imidazo[1 yridin-7—yl)propanoate 7-Bromo-N-(5—(3,4-difluorobenzylcarbamoyl)fluorophenyl)imidazo[1,2-a]pyridine carboxamide (intermediate 3A)(1g, 1.987 mmol), Pd2(dba)3 (0.091 g, 0.099 mmol) and tritertbutylphosphonium tetrafluoroborate (0.058 g, 0.199 mmol) were combined in THF(60 ml) under nitrogen and the mixture was degassed thoroughly refilling with nitrogen (X3). (3-Ethoxy—3—oxopropyl)zinc(ll) bromide (19.87 ml, 9.93 mmol) was added and the mixture was heated at 60 °C for 100 mins. After cooling to RT, the 1O mixture was absorbed onto silica and purification by chromatography eluting with 0— 6% MeOH in DCM afforded a yellow oil which was triturated with EtOAc to give the title compound; LCMS: Rt 0.99 min; MS m/z 525 [M+H]+; Method _v003 SEQ; N—(5-(3,4—Difluorobenzylcarbamoyl)flu0rophenyl)—7—(3—hydroxymethyl butyl)imidazo[1,2-a]pyridinecarboxamide Ethyl 3—(3-(5-(3,4-dif|uorobenzylcarbamoyl)~2—fluorophenylcarbamoyl)imidazo[1 ,2- a]pyridinyl)propanoate (step 1)(50 mg, 0.095 mmol) was suspended in dry ether and cooled to 0 °C. Methylmagnesium bromide (0.318 ml, 3.0M in ether, 0.953 mmol) was added and the e was stirred and allowed to warm to RT over 16 hrs. The mixture was partitioned between 10% aq. Citric acid and EtOAc. The organics were ted, dried (M9804), filtered and evaporated to dryness. The residue was purified by chromatography on silica eluting with 0-10% MeOH in DCM to afford the title product; LCMS: Rt 0.97 min; MS m/z 511.4 [M+H]+; Method 2minLC_v003 1H NMR (400 MHz, MeOD) 5 9.45 (2H, d), 9.08 (1H, t), 8.45 (1H, s), 8.35 (1H, d), 7.75 (1H, m), 7.55 (1H, s), .38 (3H, m), 7.10 (1H, d), 4.58 (2H, s), 2.88 (2H, m), 1.85 (2H, m), 1.30 (6H, 3).
Example 7.1 N-(5-(3,4-Difluorobenzyicarbamoyl)fluorophenyl)(3-fluoro(2-(piperidin 1O ylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide hydrochloride (P“#5 F . %13 §t_ep_1_: Methyl 4-(3-(5-(3,4-ditiuorobenzylcarbamoyl)—2-fluorophenylcarbamoyl) imidazo[1,2—a]pyridinyl)fluorobenzoate 7~Bromo-N~(5-(3,4—difluorobenzylcarbamoyl)—2—fluorophenyl)imidazo[1,2—a]pyridine carboxamide (intermediate 3A)(500 mg, 0.993 mmol), methyl 2—fluoro—4—(4,4,5,5- tetramethyI—1,3,2-dioxaborolan—2-yl)benzoate (306 mg, 1.093 mmol) and cesium carbonate (1295 mg, 3.97 mmol) in water (2 ml) /DME (5 ml) were mixed to give a suspension. The mixture was degassed with nitrogen and PdCl2(dppt).CH2Clz adduct (40.6 mg, 0.050 mmol) was added. The reaction mixture was heated using microwave radiation at 100°C for 1hr and partitioned between EtOAc and water. The organic portion was separated, dried (M9804) and concentrated in vacuo. The crude product was triturated with MeOH/diethyl ether to afford the title compound; LCMS: Rt 1.09 min; MS m/z 567.[M+H]+; Method _v003 fig; N-(5-(3,4-Difluorobenzylcarbamoyl)-2—fluorophenyl)(3-fluoro—4-(2- (piperidinyl)ethylcarbamoyl)pheny|)imidazo[1,2-a]pyridinecarb0xamide Methyl 4-(3-(5-(3,4-difluorobenzylcarbamoyl)fiuorophenylcarbamoyl)imidazo[1,2- a]pyridin-7—yl)fluorobenzoate (step 1)(50 mg, 0.087 mmol), ridin-t—yl- ethylamine ( 0.260 mmol), and TBD (12.07 mg, 0.087 mmol) in THF (289 pl) were heated to 70°C for 2 days. The mixture was diluted with water (4 ml) and extracted with DCM using a phase separator. The c portion was concentrated in vacuo and the residue was dissolved in DMSO. Puriication of the crude product was carried out by preparative LC-MS. The resulting ons were loaded onto a tted (MeOH) Isolute® SCX-Z cartridge and washed with MeOH. The product was eluted with 7M ammonia in MeOH. The resulting residue was d with 2M HCI (in EtOH, 1 equiv) and the concentrated in vacuo to afford the title compound as a hloride salt; LS-MS: Rt 3.79 mins; MS m/z 673 [M+H]+; Method 10minLC_v003 1O 1H NMR (400MHz. DMSO—d6) 6 9.1(1H, s), 9(1H, bs). 8.6(1H. d). 8.4(1H, s), 8.2(2H, m), 7.9-7.7 (3H, m), 7.6(1H, s), 7.4(2H. m), 7.2(1H, m), 4.5(2H, d), 3.4(2H, m), 3.2(2H, s), 2.4(4H, m), 1.5 (4H, m) 1.4(2H, m).
Example 7.2 N-(5-(3,4-Difluorobenzylcarbamoyl)fluorophenyl)(3-fluoro(2-(tetrahyd ro- 2H-pyranyl)ethylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide hydrochloride H I N\ The title compound was prepared analogously to Example 7.1 using the appropriate amine in step 2. The hydrochloride salt was obtained by ng N—(5—(3,4— difluorobenzylcarbamoyl)—2—fluorophenyl)—7—(3—fluoro~4—(2—(tetrahydro—2H-pyran-4— yl)ethylcarbamoyl)phenyl)imidazo[1,2—a]pyridine~3—carboxamide with 2M HCI (in diethyl ether) and removing the solvent in vacuo; LC-MS: Rt 3.49 mins; MS m/z 674 [M+H]+; Method 10minLC_v003 1H NMR (400MHz. DMSO-dG) 6 9.1 (1H. s) 8.4(2H, m), H, m), 7.9(1H, d), 7.8(1H, d), 7.7(1H, t), 7.65(2H, d), 7.4(3H, m), 7.2(1H, m), 4.5 (2H, d), 3.8 (2H, M). 3.3 (3H, m) 3.15 (1H, s), 1.6 (3H, m), 1.5 (2H. m). 1.1(2H, m).
Example 7.3 (2~(2,2—Dimethylpyrroiidin—1—yi)ethyi)carbamoyl)—2~fluorophenyi)—7—(1—methyi— 1 H-pyrazol-4—y|)imidazo[1,2—a]pyridine—3—carboxamide HN O m4-FIuoro—3-(7-(1-methyI—1H-pyrazoIy|)imidazo[1,2—a]pyridine carboxamido)benzoic acid A mixture comprising methyl 3—(7-bromoimidazo[1,2-a]pyridine-3~carboxamido)—4— benzoate (Intermediate 1A)(5.41 g, 13.79 mmol), 1-methyi—1H—pyrazol yiboronic acid (1.911 g, 15.17 mmol), Pd(dppf)Cl2.DCM (1.127 g, 1.379 mmol) and cesium carbonate (13.48 g, 41.4 mmol) in DME (100 mi) and water (10ml) was heated at 100°C for 4 hrs. 3 equivalents of sodium carbonate were added and the mixture was heated to 100°C for 6 hrs. A further 3 equivalents of sodium carbonate were added and the reaction was heated at 100°C overnight.
The reaction e was cooled to 50°C and filtered through glass fiber filter paper.
The filtrate was acidified to pHS and allowed to cool to room temperature. A brown precipitate formed which was collected by vacuum filtration and dried at 45°C to afford the title nd; LC—MS: Rt 0.68 mins; MS m/z 380/381 {M+H}+; Method 2minLC_v003.
MN-(5-((2—(2,2—Dimethylpyrrolidin-1—yl)ethyl)carbamoyl)-2—fluorophenyi)—7-(1 1H-pyrazoi-4—yi)imidazo[1,2—a]pyridinecarboxamide A solution of4-fiuoro—3-(7-(1-methyi—1H—pyrazolyl)imidazo[1,2-a]pyridine carboxamido) benzoic acid (step 1)(6.31 g, 16.63 mmol), 2—(2,2—dimethyipyrroiidin yl)ethanamine (2.84 g, 19.96 mmol) and triethyiamine (6.96 mi, 49.9 mmol) in DMF PCT/1B2012/054501 (36.9 ml) and EtOAc (2 ml) was treated dropwise with ®T3P (propylphosphonic anhydride) (50% w/w in EtOAc) (15.88 g, 24.95 mmol). The resulting mixture was stirred at RT for 16 hrs and diluted with 10% MeOH/EtOAc (62.7 ml). The mixture was washed with 0.5 M LiCl, H20 and sat. NaHC03. The aqueous layer was back- ted with 10% MeOH EtOAc (62.7 ml) (3 X 100 ml). The combined organic extracts was dried M9804, filtered and trated in vacuo. The resulting solid was triturated with EtOAc and dried at 45°C to afford the title nd; LC-MS: Rt 0.59 mins; MS m/z 504/505/506 {M+H}+; Method 2mlnLC_v003 Further purification was carried out by loading the product onto 2 pre-washed 109 1O lsolute SCX—2 columns and washing through with MeOH. The product was eluted with 2N NH3 in MeOH to afford a brown solution which was triturated with EtOAc to the title compound; 1H NMR (400MHz), CDgOD) 6 9.4 (1H, d), 8.45 (1H, s), 8.3 (1 H, m), 8.2 (1H, s), 8.0 (1H, s), 7.85 (1H, s), 7.8 (1H, m), 7.4 (1H, m), 7.3 (1H, t), 4.0 (3H, s), 3.5 (2H, m), 2.9 (2H, t), 2.7 (2H, t), 1.8 (2H, m), 1.7 (2H, m), 1.0 (6H, 8).
Example 7.4 N -(5-(2-(2,2-Dimethylpyrrolidinyl)ethylcarbamoyl)fluorophenyl)(3-fluoro- 4-((1R,2R)hydroxycyclohexylcarbamoyl)phenyl)imidazo[1,2-a]pyridine amide )1 \ F \ “MI,HYLVN l/ b CNN/«Km 0 SEE 7-Bromo—N-(5-(2—(2,2—dimethylpyrrolidinyl)ethylcarbamoyI)-2— fluorophenyl)imidazo[1,2-a]pyridine-3—carboxamide 3—(7-Bromoimidazo[1,2-a]pyridine-3—carboxamido)—4—fluorobenzoic acid (prepared by hydrolysis of methyl 3-(7—bromoimidazo[1,2—a]pyridine—3-carboxamido)—4- fluorobenzoate (Intermediate 1A) using NaOH) (700 mg, 1.851 mmol), 2-(2,2— dimethylpyrrolidin-‘l-yl)ethanamine (290 mg, 2.036 mmol) and triethylamine (0.774 ml, 5.55 mmol) were dissolved in EtOAc (7.284 ml) and DMF (1.28mi) to give a yellow solution. ®T3P (propylphosphonic anhydride)(1.620 ml, 2.036 mmol) was added and the mixture was stirred at room temperature for 90 mins. The reaction 1O mixture was diluted with EtOAc and washed with 0.5M LiCI in H20 and sat NaHCO3.
The organic portion was dried M9804, ed and concentrated in vacuo. The residue was triturated with EtOAc/iso-hexane to afford the title compound as a white solid; LC—MS: Rt 0.67 mins; MS m/z 503/504/505 {M+H}+; Method 2minLC_v003 mMethyl 4-(3—(5—(2—(2,2—dimethylpyrrolidin—1~yl)ethylcarbamoyl)—2- fluorophenylcarbamoyl)imidazo[1,2-a]pyridin-7—yl)—2-fiuorobenzoate The title compound was prepared from 4-(methoxycarbonyl)phenylboronic acid and 7—bromo—N—(5—(2—(2,2-dimethylpyrrolidin—1—yl)ethylcarbamoyl)-Z-fluorophenyl) imidazo[1,2—a]pyridine—3-carboxamide (step 1) analogously to Example 7.3 step 1; LC—MS: Rt 0.75 mins; MS m/z 576/577 [M+H]+; Method 2minLC_v003 : 4-(3—(5~(2-(2,2—Dimethylpyrrolidinyl)ethylcarbamoyl)fluorophenyl carbamoyl)imidazo[1,2—a]pyridinyl)fiuorobenzoic acid A mixture comprising methyl 4—(3—(5—(2—(2,2—dimethylpyrrolidinyl)ethylcarbamoyl} 2—fluorophenylca rbamoyl) imidazo[1,2—a]pyridinyl)~2-fluorobenzoate (step 2)(409 mg, 0.711 mmol) and sodium hydroxide (142 mg, 3.55 mmol) in MeOH (5 ml) was heated at 60°C overnight. The reaction mixture was concentrated under vacuum and the resulting residue was ved in water and adjusted to pH 5 using 1M HCl. The solid precipitate was collected by filtration and dried at 45°C. The aqueous portion was concentrated in vacuo and the residue was ted in 5% MeOH inDCM. The resulting suspension was filtered and the filtrate was ed with the solid precipitate from the first filtration. The combined t were evaporated to dryness and dried at 45°C in the vacuum oven.
LC-MS: Rt 0.68 mins; MS m/z 562/563 {M+H}+; Method _v003 mN-(5—(2-(2,2-Dimethylpyrrolidiny|)ethylcarbamoyl)f|uorophenyl)-7—(3- fluoro((1 2—hydroxycyclohexylcarbamoyl)phenyl)imidazo[1,2-a]pyridine carboxamide The title compound was prepared from (1R,2R)-2—aminocyclohexanol and 4-(3-(5~(2— (2,2—dimethylpyrrolidin—1—yl)ethylcarbamoyl)—2—fluorophenylcarbamoyl)imidazo[1,2- a]pyridin—7—yl)—2—fluorobenzoic acid (step 3) analogously to Example 7.3 step 2; LC—MS: Rt 0.70 mins; MS m/z 659/660 {M+H}+; Method 2minLC_v003 Example 7.5 N-(5-(2-(2,2-Dimethylpyrrolidinyl)ethylcarbamoyl)fluorophenyl)—7-(3-fluoro- 4-(1-hydroxymethylpropanylcarbamoyl)phenyl)imidazo[1,2-a]pyridine amide The title compound was prepared analogously to Example 7.4 from 4—(3—(5—(2—(2,2— dimethylpyrrolidin—1—y|)ethylcarbamoyl)—2—fluorophenylcarbamoyl)imldazo[1 ,2~ a]pyridin—7—yl)-2~fluorobenzoic acid (step 3) and 2—amino—2—methylpropan—1—ol; LC-MS: Rt 0.69 mins; MS m/z 633/634/635 ; Method 2minLC_v003.
Example 7.6 N-(5-(2-(2,2-Dimethylpyrrolidinyl)ethylcarbamoyl)methylpyridinyl)(3- fluoro(1-hydroxy-Z-methylpropan-2~ylcarbamoyl)phenyl)pyrazolo[1,5- a]pyridlnecarboxamide V”WK T.
““WDH Steps 1, 2 and 3: 4—(3—(5-(2-(2,2~Dimethylpyrrolidinyl)ethylcarbamoyl)—2— methylpyridin-B—ylcarbamoyl)pyrazolo[1,5—a]pyridin-6—yI)—2-fluorobenzoic acid The title compound was prepared from intermediate 1E analogously to Example 7.4 steps 1, 2 and 3; LC-MS: Rt 0.72 mins; MS m/z 559 {M+H}+; Method 2minLoprv01. flew—4: N—(5-(2—(2,2-Dimethylpyrrolidin-1—yl)ethylcarbamoyl)-2—methylpyridin-B-yl)-6— (3-fluoro-4—(1—hydroxymethylpropan-Z—ylcarbamoyl)phenyl)pyrazolo[1,5—a]pyridine- 3-carboxamide 1O A solution of 2-aminomethylpropanol (12.64 mg, 0.142 mmol), 4—(3-(5-(2—(2,2- dimethylpyrrolidiny|)ethylcarbamoyl)methylpyridinylcarbamoyl)pyrazolo[1,5— a]pyridinyl)fluorobenzoic acid (72 mg, 0.129 mmol) and ylamine (0.054 ml, 0.387 mmol) in DCM (5 ml) was treated with HATU (53.9 mg, 0.142 mmol) and DMF (1 ml). After stirring at RT for 3 hrs, the reaction e was diluted with DCM and washed with sat NaHCOs and H20. The organic portion was separated, dried MgSO4 and trated in vacuo. The product was purified by flash column chromatography on silica eluting with 0-25% 2M NH3 in MeOH/DCM. The product fractions were combined and concentrated to afford an orange oil which was triturated with EtOAc/hexane to afford a pale brown sold.
LC-MS: Rt 0.74 mins; MS m/z 630/631/632 {M+H}+; Method prvO1 Example 8.1 N-(5-(3,4-Difluorobenzylcarbamoyl)fluorophenyl)(3-fluoro(3-morpholino propylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide hydrochloride H ImN\ N N N \ HN\/\/ O NH SELL: 4—(3-(5—(3,4-Difluorobenzylcarbamoyl)-2—f|uorophenylcarbamoyl)imidazo[1 ,2- a]pyridin-7—yl)—2-f|uorobenzoic acid A solution of methyl 4—(3—(5—(3,4—difluorobenzylcarbamoyl)—2—fluorophenylcarbamoyl) imidazo ]pyridin—7-yl)fluorobenzoate (Example 7.1, step 1) (497 mg, 0.862 mmol) and 2M NaOH (4311 uL, 8.62 mmol) in MeOH (2874 pt.) was stirred at RT overnight. The solvent was removed in vacuo and the residue was acidified with 2M HCI to pH 2 and extracted with DCM. The organic extracts were dried over MgSO4, filtered and concentrated in vacuo to afford the title compound; 1O LCMS: Rt 0.66 min; MS m/z 563 ; Method 2minLC_30_v003 mN—(5-(8,4-Difluorobenzylcarbamoyl)-2—fluoropheny|)—7—(3-f|u0ro~4—(3- morpholino propylcarbamoyl)phenyl)imidazo[1,2—a1pyridinecarboxamide hydrochloride A mixture comprising 4-(3—(5—(3,4-difluorobenzylcarbamoyl)—2—fluorophenyl ‘15 carbamoyl)imidazo[ 1,2-a]pyridin-7—yl)—2—fluorobenzoic acid (100 mg, 0.178 mmol), 3- morpholinopropanamine (51.3 mg, 0.356 mmol), HATU (74.4 mg, 0.196 mmol) and s base (34.2 uL, 0.196 mmol) in THF (593 uL) was stirred at room temperature overnight. The solvent was removed in vacuo and the residue was partitioned between water and DCM. The organic portion was dried over M9804, filitered and concentrated in vacuo. The residue was purified by chromatography on silica eluting with 0-20% 2M NH3 in Methanol in TBME. The resulting solid was treated with 2M HCl in diethyl ether and concentrated in vacuo to afford the title LCMS: Rt 3.09mins; MS m/z 688 ; Method 10minLC_v003 1H NMR (400MHz, DMSO-d6)610.5(1H, s), 10.4(1 H, bs), 9.5(1H, d), 9.2(1H, t), 8.8(1 H, s), 8.6(1 H, bs), , s), 8.2(1H, d), 7.9(1H, d) 7.8(1H, m), 7.7(1H, d), 7.5(1H, m)m 7.4(1H, m) 7.2(1H, m) 4.5(2H, d), 4(2H, d), 3.75(2H, t), 3.45(4H, m), 3.1(4H, m) 2(2H, m).
Example 8.2 N-(5-(2-(2,6-cis-Dimethylpiperidinyl)ethy|carbamoyl)f|uorophenyI)(3- fluoro(‘l -hydroxymethylpropanylcarbamoyl)phenyl)imidazo[1,2- a]pyridinecarboxamide M4—(3—(5-(2-(2,6—cis—Dimethylpiperidin-1 ~yl)ethylcarbamoyl)—2—fluorophenyl 1O oyl)imldazo[1,2—a]pyridin—7~yl)—2—fluorobenzoic acid The title compound was prepared from 7-bromo—N—(5-(2—(2,6—cis~dimethylpiperidin —1— yl)ethylcarbamoyl)—2-fluorophenyl)imidazo[1,2—a]pyridine-3—carboxamide (Intermediate 4C) and 3-fluoro—4—(methoxycarbonyl)phenylboronic acid analogously to Example 1.20; LCMS: Rt 0.50 mins; MS m/z 294 [M+H]+; Method pr MN-(5-(2-(2,6~cis—Dimethylpiperidin-t—yl)ethylcarbamoyl)—2-fluorophenyl)-7—(3— fluoro~4—(1-hydroxy-Z-methylpropan—Z-ylcarbamoyl)phenyl)imidazo[1,2-a]pyridine—3- carboxamide The title nd was ed from 4-(3—(5-(2—(2,6-cis-dimethylpiperidin yl)ethylcarbamoyl)—2—fluorophenyl carbamoyl)imidazo[1,2-a]pyridinyl) fluorobenzoic acid (step 1) and 2-aminomethylpropanol analogously to Example 8.1, step 2; LCMS: Rt 0.73 mins; MS m/z 647 [M+H]+; Method 2minLopr Example 8.3 7-(3-Fluoro(2-fluoroethylcarbamoyl)phenyI)-N-(2-fluoro(2-(4- methylpiperaziny|)benzylcarbamoyl)phenyl)imidazo[1,2-a]pyridine carboxamide F O I HWF NR \\ \ HM C! K‘VN \ mMethyl 2-fiuoro—4-(3-(2-fluoro(2—(4-methylpiperaziny|)benzylcarbamoy|) phenylcarbamoy|)imidazo[1,2—a]pyrldinyl)benzoate The title compound is prepared from 7-Bromo-N~(2—fluoro—5-(2-(4-methylpiperazin yl) benzylcarbamoyl)phenyl)imidazo[1,2—a]pyridinecarboxamide (Example 1.1 step 1O 1) and ro-4—(methoxycarbonyl)phenylboronic acid analogously to Example 1.1 step 2; LC-MS: Rt 0.79 mins; MS m/z 639/640 {M+H}+; Method 2minLC_v003 Step 2 and 3: 7—(3—Fluoro—4-(2—fluoroethylcarbamoyl)phenyl)—N-(2—fluoro—5-(2~(4— methylpiperazin—1-yl)benzylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3—carboxamide The title compound was prepared from methyl 2—fluoro-4—(3—(2—fluoro—5—(2-(4- methylpiperaziny|)benzylcarbamoyl)phenylcarbamoyl)imldazo[1,2—a]pyridin-7— yl)benzoate (step) and oethanamine analogously Example 8.1 steps 1 and 2; LC—MS: Rt 0.74 mins; MS m/z 670/671 {M+H}+; Method 2minLC_v003 e 8.4 N-(2-Fluoro(2-(4-methy|piperazinyl)benzylcarbamoyl)phenyl)(3-fluoro (2-hydroxy ethylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide The title compound was prepared from 7-bromo-N-(2-fluoro(2—(4-methylpiperazin- 1-yl)benzylcarbamoyl) phenyl)imidazo{1,2—a]pyridine-3—carboxamide (Example 1.1 step1 and 2—amlnoethanol analogously to Example 8.2 steps 1 and 2; LC—MS: Rt 0.71 mins; MS m/z 668/669/670 ; Method 2minLC_v003.
Example 9.0 N -(5-(2-(2,2-dimethylpyrrolidinyl)ethylcarbamoyl)methy|pyridinyl)(1- methyl-1 H-pyrazolyl)pyrazolo[1 ,5-a]pyridinecarboxamide §tap_1: Methyl 5-(6-bromopyrazolo[1,5-a]pyridine-3—carboxamido)methylnicotinate opyrazolo[1,5-a]pyridinecarboxylic acid (7.71 g, 32.0 mmol) in toluene (80 ml) was treated with thionyl chloride (18.67 ml, 258 mmol) and was heated to 110 °C for 6 hr. The solvent was removed in vacuo and the residue was treated with pyridine (80 ml), methyl 5-aminomethylnicotinate (4.25 g, 25.6 mmol) and oven dried molecular sieves. The reaction mixture was stirred at RT overnight and then treated with MeOH (250 ml). The resulting suspension was removed by filtration. The filtrate was triturated with ol and the solid produced was ed to afford the title compound; LCMS: Rt 0.91 mins; MS m/z 391.4 [M+H]+; Method 2minLopr SE92; 6—Methyl—5—(6—(1-methyI-1H—pyrazolyl)pyrazolo[1,5-a]pyridine carboxamido)nicotinic acid Methyl 5—(6-bromopyrazolo[1.5-a]pyridine—3—carboxamido)—6-methylnicotinate (step 1) (7 g, 17.99 mmol), 1~methyl(4,4,5,5—tetramethyI-1,3,2—dioxaborolan-Z-yl)-1 H- pyrazole (4.49 g, 21.58 mmol) and cesium carbonate (23.44 g, 71.9 mmol) were stirred in methoxyethane (60 ml) and water (25.00 ml). The mixture was degassed thoroughly ing with nitrogen. PdCl2(dppf).CHZCl2 adduct (0.350 g, 0.429 mmol) was added and the mixture was degassed thoroughly refilling with nitrogen, The mixture was stirred at 100 °C for 7 hrs and then cooled to 50°C and filtered through glass-fiber paper. The filtrate was acidified to pH 5 by the addition of 2M HO! and filtered. The foam residue was dissolved in OH (1:1) and azeotroped with toluene (x2). The resulting solid was dried in a vacuum oven to afford the title compound; WO 30802 LCMS: Rt 0.69 mins; MS m/z 377.5 [M+H]+; Method 2minLopr §_t_ep_3: N—(5—(2—(2,2—Dimethylpyrrolidin—1—yl)ethylcarbamoyi)methylpyridin—3—y|)—6— (1-methyl-1H-pyrazoIyl)pyrazolo[1 yridine—3-carboxamide 6-Methyl(6-(1—methyl-1H-pyrazoiyl)pyrazolo[1,5-a]pyridine carboxamido)nicotinic acid (6.6 g, 1.1 equiv) and 2-(2,2-dimethylpyrrolidin—1- yl)ethanamine (2.397 g, 11.14 mmol) were combined in DMF (100 ml) and treated with DlPEA (8.34 mi, 47.7 mmol) followed by HATU (4.44 g, 11.67 mmol). After strring at RT for 90 mins, the mixture was partitioned between water (1 L) and EtOAc (750 ml).The resulting suspension was removed by filtration and the organic portion 1O was washed with aqueous sodium bicarbonate, 0.5M lithium chloride, brine, dried M9804, filtered and evaporated to s. cation by chromatography on silica eluting with 0-20% 2M NH3 in MeOH/TBME afforded residue which was recrystallised from acetone to afford the title compound; LCMS: Rt 0.61 mins; MS m/z 501 ; Method 2minLopr 1H NMR (400 MHz, DMSO) 5 9.75 (1H, s), 9.15 (1 H, s), 8.75 (2H, m), 8.58 (1H, t), 8.32 (1H, s), 8.25 (1 H, s), 8.21 (1H, d), 8.07 (1 H, s), 7.82 (1H, d), 3.89 (3H, s), 3.34 (4H, m), 2.76 (2H, t), 2.56 (3H, s), 1.69 (2H, m), 1.53 (2H, m) 0.92 (6H, s) The compounds of the following tabulated Examples (Table 4) were prepared by a similar methods to that of Example 9 from the appropriate starting compounds, the preparations of which are detailed herein and in the ‘Preparation of intermediates’ [M+H]+/NMR WO 30802 Rt 0.59 mins; MS m/z 520 [M+H]+; Method 2minLoprv01 (S)—N-(2-Fluoro—5-(2—(2—(methoxy ) pyrrolidin-l -yl)ethyl carbamoyl)phenyl)(1— methyl-1 H-pyrazolyl)imldazo[1,2—a]pyridine—3- carboxamide Rt 2.40mins: MS m/z 516.5[M+H]+ Method 10minLoprv0‘l N-(2-Fluoro((2-(3-propylpyrrolidin yl)ethyl)carba moyl)phenyl )(1 methyl-1 H- pyrazolyl)imidazo [1 ,2-a]pyrldine 9.2 carboxamide ’l’lO PCT/132012/054501 Rt 0.57 mins; MS m/z F N R [\ 520/521 [M+Hr; Method N \ / N/ 2minLoprvO1 (R)—N—(2~Fluoro—5—((2-(2—(methoxymethyl) idin—1~y|)ethyl) carbamoyl)pheny1)—7—(1~ methyl—1 H—pyrazoI—4—yl)imidazo[1,2—a]pyridine~3— 9.3 carboxamide Rt 2.36mins: MS m/z 516 F N a I \ [M+H]+ Method N \ \ N 10minLoprv01 o \ \ [‘3 o N; I LA N-(5—((2—(3,5-Dimethylpiperidin-1~ yl)ethyl)carbamoyl)fluorophenyl)—7—(1~methyl- 1 H-pyrazo!-4—yl)imidazo[1,2-a]pyridine—3- 9.4 carboxamide LCMS: Rt 0.81 mins; MS m/z 546.4 [M+H]+; Method 2minLopr N-(2—fluoro—5-((2—(2,2,6,6-tetra methylpiperidln-l- y$1)ethy1)carba moy|)phenyI)—7-(1-methyl-‘l H- ; pyrazol—5—yl)imidazo{1,2—a]pyridine~3— carboxamide hydrochloride salt [Eon/18: Rt 0.75 mins; MS m/z 492.3 [M+H]+; Method 2minLC_v003 N—(5-((2-(tert- butyl(methyl)amino)ethyl)carbamoyl)—2- fluorophenyl)—7-(1-methyl-1H-pyrazol—S— yl)imidazo[1,2—a]pyridine—3-carboxamide $9.6 chloride salt LCMS: Rt 0.59 mins; MS m/z 501/502/503{M+H}*; Method _v003 l N-(5-((2-(2,2-dimethylpyrrolidin-1 - yl)ethyl)carbamoyl)methylpyrldin-3—y|)(‘l - methyl-1 H-pyrazolyl)pyrazolo[1,5-a]pyridine- 3—carboxamide hydrochloride salt LCMS: Rt 0.66 mins; MS m/z 607/608 {M+H}*; Method 2minLC_v003 fluoro )(3-fluoro—4—((2—hyd roxy 35myl)0arb amoy|)phenyl)imidazo[1,2—a]pyri dine- 3-carboxamide LCMS: Rt 0.79 mins; MS m/z 648.6 [M+H]+; Method 2minLopr Int BB and Int 8A 7-(3—fluoro—4—((1-hydroxy—2—methylpropan—Z— yl)carbamoyl)phenyl)—N—(2—fluor0—5—((2~(3— propylpyrrolidin yl)ethyl)carbamoyl)phenyl)imidazo[1,2- a]pyridine-3~carboxamide LCMS: Rt 0.74 mins; MS m/z 650.6 [M+H]+; Method 2minLopr oA‘K \\/N\/\m O N—(5-((2-<3,3- dimethylmorpholino)ethyl)carba moyl)—2— fluorophenyl)(3-fiuoro—4-((1-hyd roxy-Z- methylpropan-Z- ; bamoyl)phenyl)imidazo[1,2-a]pyridine-3— 19.10 carboxamide LCMS: Rt 0.75 mins; MS m/z 648.7 ; Method 2minLopr (R)—7-(3-fluoro-4~((1-hydroxy—Z-methylpropan—Z— yl)carbamoyl)ph enyl)-N-(2-fluoro-5—((2—(2— (methoxymethyl)pyrrolidinyl)ethyl) carbamoyl)phenyl)imidazo[1,2-a1pyridine 9.11 carboxamide LCMS: Rt 0.74 mins; MS m/z 616.6 [M+H]+; Method 2minLopr N-(5~((3,4-Difluorobenzyl)carbamoyl)—2— fluorophenyI)(6-((2~ (dimethylamino)ethyl)Carbamoyl)pyridin—3— LCMS: Rt 0.66 mins; M8 m/z 515.7 ; Method 2minLopr N-(5-((2—(2,2—Dimethylpiperidin—1- yl)ethy|)oarba moyl)-2—methy|pyridin—S—yl)—6—(1 — methyl-1H-pyrazoIyl)pyrazolo[1,5-a]pyridine— 9.13 3-carboxamide LC—MS: Rt 0.62 mins; MS m/Z 515.7 {M+H}+; Method 2minLC_v003 N—(5—((2-(2,6—dimethyl piperidin-1—yl)ethyl) carbamoyl)—2~methylpyridinyl)—6-(1~methyl— 1 H-pyrazol-4—yl)pyrazolo[1 ,5-a]pyridine—3- LC-MS: Rt 0.66 mins; MS E \ m/z 518.5 M+H +; F H \ [ 1 N N \ \r}! \ Method 2minLoprv01 _ N\ L O ‘ NH ‘ N-(5—((2—((28,3R)~2,3-diethylazetidin-1~yl)ethyl) carbamoyl)fluoropheny!)~7-(1~methyl—1 H- pyrazol-4—y8)imidazo[1,2—a]pyridine~3— 9.1 5 amide Example 10.1: N-(5-(3,4-Difluorobenzylcarbamoyl)—2-fluorophenyi)(6-(((2-hydroxyethyl) (methyl)amino)methyl)pyridinyl)imidazo[1,2-a]pyridine—3-carboxamide PCT/IBZOlZ/054501 (3,4-Difluorobenzylcarbamoyl)fluorophenyl)(6-formylpyridin yl)imidazo[1,2—a]pyridine—3-carboxamide A e comprising 7—bromo—N—(5—(3,4—difluorobenzylcarbamoyl)—2~fluorophenyl) imidazo[1,2—a]pyridine—3—carboxamide (Intermediate 3A)(2.7 g, 5.36 mmol), 5» (4,4,5,5—tetramethyl-1,3,2-dioxaborolanyl)picolinaldehyde (1.375 g, 5.90 mmol) and cesium carbonate (6.99 g, 21.46 mmol) in DME (30 ml) and water (3 ml) was d with PdCl2(dppf).CHQCl2 adduct (0.219 g, 0.268 mmol). The mixture was placed under nitrogen and heated at 100°C for 1hr. The resulting mixture was concentrated in vacuo and the residue was dissolved in 10% trifluoroethanol/CHClg.
The organics were washed with water and NaHCOa and concentrated in vacuo.
Purification by chromatography on silica eluting with 0-30% 2M NH3 in CM afforded the title product; LC-MS: Rt 0.67 mins; MS m/z 530/531 {M+H}+; Method 2minLC_v003 grep—2: N-(5-(3,4-Difluorobenzylcarbamoyl)-2—fluorophenyl)—7-(6—(((2—hydroxy ethyl)(methyl)amino)methyl)pyridin-S-yl)imidazo[1,2-alpyridinecarboxamide A suspension of N-(5-(3,4-difluorobenzylcarbamoy|)fluorophenyl)(6- formylpyridin—B-yl)imidazo[1,2-a]pyridinecarboxamide (step 1)(1OO mg, 0.189 mmol), 2-(methylamino)ethanol (70.9 mg, 0.944 mmol) and molceular sieves in EtOH (2 ml) was heated at 70°C overnight. The mixture was cooled to 0°C and treated with sodium borohydride (7.15 mg, 0.189 mmoi). The mixture was allowed to warm to RT and was stirred overnight. The resulting suspension was removed by filtration and the filtrate was concentrated in vacuo. The residue was partitioned between EtOAc and water. The organic portion was separated, washed with Na H003, dried (MgSO4) and concentrated in vacuo. Purification of the residue by preparative chromatography eluting with 25—50% 0.1% TFA acetonitrile/water ed fractions that were combined and diluted with NaHCOa and 5% trifluoroethanol/DCM. The organics were separated, dried and trated in vacuo to afford a white solid. The solid was 1O triturated with EtOH/Ether to afford the title nd as a white solid; LC—MS: Rt 0.74 mins; MS m/z 589/590/591 {M+H}+; Method 2minLC__v003 Example 10.2 N-(5-((3,4-difluorobenzyl)carbamoyl)fluorophenyl)(6-((methyl (phenethyl)amino)methyl)pyridinyl)imidazo[1,2-a]pyridine-B-carboxamide N\ VD The title compound was prepared analogously to Example 10.1 from the appropriate amine in step 2; LC-MS: Rt 0.83 mins; MS m/z 649/650 {M+H}+; Method 2minLC_v003.
The compounds of the following tabulated Examples (Table 5) were prepared by a r methods to that of Example 10 from the appropriate ng compounds, the preparations of which are detailed herein and in the ‘Preparation of lntermediates’ section.
Table 5 Structure [M+H]*/NMR m/z 649/650 {M+H}+; Method 2minLC_v003. ((3,4-Dif|uoro benzyl)carbamoyl)—2— Efluorophenyl)—7~(6— ((methyl(phenethy|)amino)methyl)pyridin-3— yl)imidazo[1,2—a]pyridine—3—carboxamide EN-(5-((3,4-Difluoro benzyl)carbamoyl)-2— LCMS: Rt 0.71 mins; MS Efluorophenyl)-7—(5—((methylamino) m/z 544.5 [M+H]+; Method methyl)pyridinyl)lmidazo[1 ,2—a]pyridine—3~ 110.3 carbox amide LCMS: Rt 0.80 mins; MS m/z 612.6 ; Method 7—(5—((Cyclohexyl amino)methyl)pyridin—B-yl)—N— E(5-((3,4-dlfluorobenzyl) carba moyl)—2-fluoro A E 10.4 i phenyl) imidazo[1,2-a]pyrid inecarboxamide WO 30802 LCMS: Rt 0.74 mins; MS N—(5—((3,4—Difluoro benzyl)carbamoyl)—2— m/z 602.6 [M+H]+; Method fluorophenyl)—7—(5-(((2-methoxyethyi) (menu/[)3min0)m8thy1)pyridinyl)imid azo[1,2— .5 a]pyridine~3-carboxamide Preparation of Intermediates Intermediate 1A Methyl 3-(7-bromoimidazo[1,2-a]pyridinecarboxamido) fluorobenzoate N\ \ Br O O §t§p_1: Potassium (Z)—2—chloro—1 -ethoxy—3~oxoprop-1~enolate A cooled (0°C) sion of ethyl 2-chloroacetate (17.47 ml, 163 mmol) and ethyl formate (13.18 ml, 163 mmol) in ether (250 ml) was treated slowly (over 3 hrs) with ium 2-methylpropan-2—olate (18.31 g, 163 mmol) keeping the temperature below 5°C. The mixture was concentrated in vacuo and the resulting solid was washed with ether and dried (47 °C in a vacuum oven) to afford the title compound; 1H NMR (400MHz, d6—DMSO) 6 8.95 (1H, s), 3.9 (2H, q), 1.1 (3H, t).
E92; Ethyl 7-bromoimidazoi1,2-a]pyridine—S—carboxylate WO 30802 A solution of 4-bromopyridin—2-amine (10 g, 57.8 mmol) and potassium (Z)chloro- 1-ethoxy—3—oxoprop—1—en—1—olate (step 1)(23.4 g, 124 mmol) in ethanol (200 ml) was cooled to 5°C. Sulfuric acid (7.70 ml, 144 mmol) was added dropwise and the reaction heated to reflux at 90°C for 3 hrs. The mixture was cooled to RT and TEA (20.03 ml, 144 mmol) was slowly added and heating continued at 90°C for 18 hrs.
After cooling to RT, the mixture was filtered and the solid was partitioned between EtOAc and aqueous 2M HCl. The aqueous layer was d (NaOH, solid pellets) and extracted using EtOAc. The combined organic extracts were dried (M9804) and concentrated in vacuo to afford the title compound; 1O 1H NMR (400MHz, d6-DMSO) 5 9.1 (1H, d), 8.3 (1H, s), 8.2 (1H, s), 7.4 (1H, d), 4.4 (2H, q), 1.4 (3H, t) $93: 7—Bromoimidazo[1,2—a]pyridine—3-carboxylic Ethyl 7—bromoimidazo[1,2~a]pyridine-3—carboxylate (step 2)(30.81 g, 114 mmol) in MeOH (172 ml) was treated with 2M NaOH (172 ml, 343 mmol) and the mixture was heated to 60°C for 40 minutes.The volatile solvent was removed in vacuo and the crude material was treated with 2M sodium bisulfate solution to adjust the pH to 6—7.
The resulting solid was collected by filtrationand added to water (400 ml). The mixture was stirred and heated to 90°C for 1h. After cooling to RT, the suspension was filtered and dried in a vacuum over at 40°C to afford the title product; LC-MS: Rt 0.59 mins; MS m/z 243.1 {M+H}+; Method 2minLC_v003 flap—4: Methyl 3-(7—bromoimidazo[1,2—a]pyridine—3-carboxamido)—4-fluorobenzoate A e comprising 7-bromoimidazo[1,2-a]pyridinecarboxylic acid (step 3)(1.8 approximately 7.47 mmol) and thionyl chloride (10 ml, 137 mmol) under N2 was heated at reflux for 1.5 hrs. The reaction e was concentrated in vacuo and azeotroped with toluene. Methyl 3—aminofluorobenzoate (1.263 g, 7.47 mmol) (pre- dried at 45°C) was added followed by pyridine and the mixture was stirred at room temperature under N2 overnight. The reaction mixture was d with EtOAc and washed with H20. The resulting solid was collected by tion. The filtrate was dried (M9804) and concentrated in vacuo and triturated with ether to afford cream solid.
The solids were combined and dried at 450C to afford the title compound; LC—MS: Rt 0.97 mins; MS m/z 392 {M+H}+; Method 2minLC_v003 1H NMR (400MHz, DMSO-d6) 5 10.3 (1H, s), 9.4 (1 H, d), 8.6 (1H, s), 8.3 (1H, m), 8.2 (1H, s), 7.9 (1 H, m), 7.5 (1H, t), 7.4 (1H, d), 3.9 (3H, 8).
Intermediate 1B Methyl romoimidazo[1,2-a]pyridinecarboxamido) methylbenzoate ([3 O A mixture comprising 7-bromoimidazo[1,2—a]pyridine-3—carboxylic acid (intermediate 1A step 3) (750 mg, 3.11 mmol) and thionyi chloride (5 mi, 68.5 mmol) under N2 was heated at reflux for 2hrs. The mixture was concentrated in vacuo and azeotroped with e. Methyl 3-amino-4—methylbenzoate (514 mg, 3.11 mmol) (pre—dried at 45°C) was added foiiowed by ne (5 mi) and the mixture was d at room temperature under N2 overnight. The reaction mixture was diluted with EtOAc and washed with sat. NaHC03, brine, H20, dried (MgSO4) and concentrated in vacuo.
Purification by chromatography on sitica eiuting with 50-100% EtOAc in iso-hexane 1O afforded the title compound as an orange solid; LC—MS: Rt 0.94 mins; MS m/z 390/391/392 {M+H}+; Method 2minLC_v003 1H NMR (400MHz, DMSO-d6) 6 10.0 (1H, s), 9.4 (1H, d), 8.6 (1H, s), 8.2 (1H, d), 8.0 (1H, d), 7.8 (1H, d), 7.5 (1 H, d), 7.3 (1H. d), 3.9 (3H, s), 2.4 (3H, s). ediate 10 Methyl 5-(7-bromoimidazo[1,2-a1pyridinecarboxamido)methylnicotinate N \N l o O 0\ Step 1: Methyl 2-chloromethyinitronicotinate To a suspension of 6-methyl—5-nitro—2—oxo—1,2-dihydropyridine—B—carboxyiic acid (commercially available)(12.5 g, 63.1 mmol) in chlorobenzene (210 ml) was added DMF (2.442 mi, 31.5 mmol) followed by POCls (23.52 mi, 252 mmol). The mixture was heated at 133 °C for 1 hr. After cooling to RT, the mixture was concentrated in vacuo. The residue was cooled in an ice bath, treated with MeOH (200 ml, 4944 mmol) and stirred at RT for 16hrs. The mixture was concentrated in vacuo and the residue was ioned between water (300 ml) and EtOAc (300 ml). The organics were dried (M9804) and concentrated in vacuo to afford the title compound as a red crystalline solid; LC-MS: Rt 1.10 mins; MS m/z 230.9 {M+H}+; Method 2minLC_vOO3 §teg_2_: Methyl 5-aminomethylnicotinate Methyl 2—chloro~6—methyl-5—nitronicotinate (step 1)(6.9 g. 29.9 mmol) was added to a suspension of ammonium formate (18.87 g. 299 mmol) and 10% Pd(Carbon)(0.522 g, 0.491 mmol) in MeOH (330 ml) and the e was heated at reflux for 3 hrs.
After cooling to RT, the mixture was filtered through Celite® (filter material) and washed through with MeOH. The t was removed in vacuo and the crude ’15 product was ated with EtOAc to give an orange solid. Purification by chromatography on silica eluting with O—100% EtOAc in iso-hexane afforded the title product. $93; Methyl 5-(7—bromoimidazo[1,2—a]pyridine—3—carboxamido)—6—methylnicotinate The title compound was prepared from 7-bromoimidazo[1,2—a]pyridinecarboxylic 2O acid (Intermediate 1A, step 3)and methyl 5—aminomethylnicotinate (step 2) analogously to intermediate 1A; 1H NMR(4OOMHZ,d6-DMSO)610.21 (1H, s), 9.40 (1H, J=7.4, d), 8.83 (1H, s), 8.55 (1H, s), 8.39 (1H, s), 8.13 (1H, J=1.6, d), 7.35 (1H, J=2.0,7.4, dd), 3.89 (3H, s), 2.58 (3H, 3) Intermediate 1D Methyl yl(6-(1-methyI-1H-pyrazolyl)pyrazolo[1,5-a]pyridine carboxamido)nicotinate 0 OMe mEthyl 6-(1—methyl-1H—pyrazol—S-yl)pyrazolo[1,5-a]pyridine-3—carboxylate A mixture comprising ethyl 6-bromopyrazolo[1,5-a]pyridinecarboxylate (1.5 g, 5.57 mmol), 1-methyl-5—(4,4,5,5-tetramethyl-1,3,2-dioxaborolan~2-yl)—1H-pyrazole (1.218 g, 5.85 mmol), cesium carbonate (7.26 g, 22.30 mmol) and PdCl2(dppf).CH2012 adduct (91 mg, 0.111 mmol) in DME (10 ml) and water (4.00 ml) was heated using microwave ion at 70 °C for 1 hr. r dppf).CH20l2 adduct (91 mg, 0.111 mmol) was added and the mixture was heated at 80 °C for 1 hr. 1-Methyl (4,4,5,5-tetramethyl—1,3,2-dioxaborolan-2—y|)-1H-pyrazole (1.218 g, 5.85 mmol) and PdCl2(dppf).Cchl2 adduct (91 mg, 0.111 mmol) were added and heating continued at 100 °C for 3 hrs. The e was diluted with 10% MeOH in EtOAc (200 ml) and washed with sat. NaHCOB. The organic solvent was removed under vacuum and azeotroped with toluene. The resulting solid was loaded onto silica and purified by chromatography eluting with 0 ~ 100% EtOAc in iso~hexane to afford the title compound; LC—MS: Rt 0.92 mins; MS m/z 271.4 {M+H}+; Method 2minLopr m6-(1-Methyl—1H—pyrazol—S-yl)pyrazolo[1,5-a]pyridine-3~carboxylic acid The title compound was prepared from ethyl 6—(1—methyl-1H—pyrazol—S- yl)pyrazolo[1,5—a]pyridine—3-carboxylate analogously to Intermediate 1A step 3; LC—MS: Rt 0.72 mins; MS m/z 243.3 {M+H}+; Method 2minLopr l 6—methyl—5-(6—(1-methyl—1H-pyrazol-5—yl)pyrazolo[1,5-a]pyridine~3— carboxamido)nicotinate 6-(1-Methyl—1H-pyrazolyl)pyrazolo[1,5-a]pyridine-3—carboxylic acid (step 2) was dissolved in pyridine (5 ml, 61.8 mmol) and 6-methylnicotinate (247 mg, 1.486 mmol) was added. The reaction mixture was stirred under nitrogen overnight. The mixture was diluted with EtOAc (200 ml) and washed with sat. aq. NaH003 (200 ml). The 2012/054501 s portion was back-extracted with EtOAc (100 ml) and the combined organic extracts were, dried MgSO4, filtered and concentrated under vacuum to give yellow solid. Purification of the solid by tography on silica eluting with 0 - 20% 2M NH3 in MeOH / TBME afforded the title compound; LC—MS: Rt 0.83 mins; MS m/z 391.3 {M+H}+; Method 2minLopr Intermediate 1E -(6-Bromopyrazolo[1,5-a]pyridinecarboxamido)methylnicotinic acid NEH KM... 6%!VJ .2 “EM.
N xxx- ”Cb O O H A solution of methyl 5-(7-bromoimidazo[1,2-a1pyridinecarboxamido) methylnicotinate (Intermediate 1C)(1 g, 2.57 mmol) and sodium hydroxide (1.028 g, .7 mmol) in MeOH (20 ml) was heated at 50°C ght. The mixture was concentrated in vacuo and the residue was dissolved in water. The pH was adjusted to pH4 by addition of 1N HCI and the resulting solid was collected by filtration and dried at 45°C to afford the title compound; LC-MS: Rt 0.82 mins; MS m/z 376/377 {M+H}+; Method 2minLoprv01 Intermediate 2A 7—Bromo-N-(5-(2-tert-butoxyethylcarbamoyl)fluorophenyl)imidazo[1,2- aIpyridinecarboxamide F N E. l \ N \ 0 \ o ”No Step 1: 3-Amino—N-(Z—tert—butoxyethyl)—4-fluorobenzamide WO 30802 A mixture comprising 2—tert~butoxyethanamine (1.2 g, 5.12 mmol, 50%w/w) and methyl ofluorobenzoate (0.866 g, 5.12 mmol) in THF (10 ml) was treated with TBD (0.713 g, 5.12 mmol) and heated at 90 °C for 16 hrs. After cooling to RT, the solvent was removed in vacuo and the residue was partitioned between water and EtOAc. The organic portion was separated and washed with 10% aq citric acid (x 2), NaHC03(sat. aq), brine, dried (MgSO4) and concentrated in vacuo. Purification of the crude product by chromatography on silica eluting with O-100% EtOAc in iso- hexane afforded the title compound as a clear oil; LC—MS: Rt 1.09 mins; MS m/z 255 [M+H]+; Method 2minLC_v003 1O §E_Q_2_: 7—Bromo—N-(5—(2-tert—butoxyethylcarbamoyl)fluoropheny|)imidazo[1 ,2- dine—3—carboxamide 6-Bromoimidazo[1,2-a]pyridine—3-carboxylic acid (commercially available)(1024 mg, 4.25 mmol) was ded in toluene (30 ml) and thionyl chloride (1.550 ml, 21.23 mmol) was added. The mixture was heated at 110 °C for 3 hrs. The solvent was removed in vacuo and the resulting residue was treated with a solution of 3-amino—N- (2-tert-butoxyethyl)-4—fluorobenzamide (step 1) (900 mg, 3.54 mmol) in pyridine (10 ml). Molecular sieves were added the mixture was stirred at RT for 16 hrs. In a separate flask, 6-bromoimidazo[1,2-a]pyridine-3—carboxylic acid 640 mg, 2.65 mmol) in e (30 ml) was treated with l chloride (0.969 ml, 13.27 mmol) at 110 °C for 3 e solvent was removed in vacuo and the solid e was added to the reaction mixture in the original flask. Stirring continued for 3 days. The mixture was poured into MeOH and the resulting suspension was removed by filtration.The filtrate was azeotroped with toluene to give a solid, which was triturated with MeOH to afford the product. The filtrate was concentrated in vacuo and the residue was dissolved in DCM (2% MeOH) and washed with water. The organic portion was dried (M9804) and concentrated in vacuo to afford a solid that was triturated with EtOAc to give the title compound; LC-MS: Rt 0.96 mins; MS m/z 477(479); 1) [M+H]+; Method 2minLC_v003 Intermediate ZC 7-Bromo-N-(4-f|uoromethyl(2-(4-methylpiperazinyl)benzylcarbamoyl) phenyl) imidazo[1,2-a]pyridinecarboxamide YES.
O NH [NJ ml; 2-FIuoro-4—methyl—5-nitrobenzoic acid 2-FIuoro—4-methyibenzoic acid (1 g, 6.49 mmol) in H2804 (19 ml, 356 mmol) was cooled to 0 °C in an ice salt water bath and treated dropwise with mixture of H2804 (0.763 ml, 14.31 mmol) and nitric acid (0.65 ml, 14.54 mmol) over 10 min. The reaction mixture was stirred at 0 °C for 3 hrs and poured into ice/water (200 ml) and stirred for a further hour. The resulting sion was ted by filtration, dried in vacuo and collected in EtOH, azeotroping to dryness to afford the title compound. mg5—Aminofluoromethylbenzoic acid 2-Fluoro-4—methyI—5-nitrobenzoic acid (900 mg, 4.52 mmol) in MeOH (70 ml) was treated with ammonium formate 1(425 mg, 22.60 mmol) and Pd (Carbon) (144 1.356 mmol). The mixture was degassed ghly refilling with nitrogen and heated to 60 °C for 2 hrs. The mixture was filtered through silica and washed with MeOH.
The filtrate was passed through SCX—Z resin (309 0.67 mmoi lg) eluting with MeOH (250 ml) followed by 2M ammonia in MeOH (250 ml). The ammonia/MeOH washings were evaporated to dryness and the resulting crude residue was purified by recrystallisation from MeOH to afford the title compound; LC~MSz Rt 0.53 mins; MS m/z 170 {Mi-HF; Method 10minLC_v003 nofluoro-4—methyl-N-(2-(4-methylpiperazinyl)benzyl)benzamide A mixture sing (2—(4-methylpiperaziny|)phenyl)methanamine (413 mg, 2.010 mmol) and 5-aminofluoromethylbenzoic acid (step 2)(340 mg, 2.010 mmol) in DMF (3 ml) was treated with DIPEA (0.351 ml, 2.010 mmol) followed by HATU (764 mg, 2.010 mmol) and stirred at 25 °C for 24 hrs. The mixture was partitioned between water and EtOAc. The organic portion was washed with sat. aq. NaHCOa, 0.5 M LiCI and brine (each back extracted with EtOAc). The combined c layers were dried (M9804), ed and evaporated to dryness to give a pink oil. Purification by chromatography on silica eluting with O- 20% MeOH in DCM afforded the title compound; LC—MS: Rt 0.73 mins; MS m/z 357 {M+H}+; Method 2minLC_v003 _S_tep_4: o—N-(4—fluoromethyl—5-(2-(4—methylpiperazinyl)benzylcarbamoyl) phenyl)imidazo[1,2-a]pyridine—3—carboxamide The title compound was prepared from 5-amino-2—fluoro—4~methyI-N—(2-(4- methylpiperazin-1~yl)benzyl)benzamide (step 3) analogously to 7-bromo—N—(5—(2—tert— butoxyethylcarbamoyl)—2-fluorophenyl)imidazo[1,2-a]pyridine-S-carboxamide mediate 2a step 2); 1O LC-MS: Rt 0.91 mins; MS m/z 5794/5824 {M+H}+; Method 2minLC_v003 Intermediate 2D 6-Bromo-N-(5-(2-tert-butoxyethylcarbamoyl)fluorophenyl)pyrazolo[1,5- a]pyridi necarboxamide N\ Br / N O NH The title compound was prepared analogously to Intermediate 2A by replacing 6- bromoimidazo[1,2-a]pyridine-S-carboxylic acid (step 2) with 6—bromopyrazolo[1,5— a]pyridine—3—carboxylic acid (commercially available); LC-MS: Rt 1.13 mins; MS m/z 477.1 {M+H}+; Method _v003.
Intermediate 3A o-N~(5-(3,4-difluorobenzylcarbamoyl)fluorophenyl)imidazo[1,2- a]pyridinecarboxamide 955.F H l N\ no-N-(3,4~difluorobenzyl)-4—fluorobenzamide A mixture comprising methyl 3—aminofiuorobenzoate (2 g, 11.82 mmol), (3,4- difiuorophenyl)methanamine (2.54 g, 17.74 mmol) and TBD (1.646 g, 11.82 mmol) in THF (39.4 ml) was heated at 80°C overnight. After cooling to RT, the mixture was ed by chromatography on silica eluting with 0—20% 2M NH3 in MeOH/DCM to afford the title compound. grip—2: 7-Bromo—N-(5-(3,4-difiuorobenzylcarbamoyl)fiuorophenyl)imidazo[1,2— a]pyridine-3~carboxamide 1O A mixture comprising 7-bromoimidazo[1,2—a]pyridine—3-carboxylic acid (intermediate 1A step 3) (1.4 g, 5.81 mmol) and thionyl chloride (8.48 ml, 116 mmol) was heated at 100°C for 1.5 hrs. The mixture was concentrated in vacuo. 3-Amin0-N—(3,4- difluorobenzyl)—4—fluorobenzamide (step 1) (1.4 g, 5.00 mmol) and pyridine (16.65 ml) was added and the ing suspension was stirred at RT for 1 hour. EtOAc and MeOH were added and the mixture was filtered. The white solid was dried to afford the title compound; LC—MS: Rt 0.74 mins; MS m/z 503 {M+H}+; Method 2minLC_30_v003 Intermediate 4A 6-Bromo-N-(2-methyl(2-(4-methylpiperazin yl)benzylcarbamoyl)phenyl)pyrazolo [1 ,5-a]pyridinecarboxamide m3-Amino4—“methyl—N-(22-(—4~methyipiperazin——1—yl)benzyl)benzamide A on of methyl 3—amin04-methylbenzoatec(commercially available) (1.609 9.74 mmol), TBD (2,3,4,6,7,8-hexahydro-1H-pyrimido[1,2-a]pyrimidine) (0.678 4.87 mmol) and methylpiperazinyl)phenyl)methanamine (commercially available) (2 g, 9.74 mmol) in toluene (30 ml) was heated at reflux overnight. The reaction mixture was diluted with EtOAc and washed with sat.NaH003 and water.
The organic portion was separated, dried ) and concentrated in vacuo. The product was purified by chromagraphy on silica eluting with a gradient of 0-20% 2M NH3 in MeOH/DCM to afford the title compound; LC-MS: Rt 0.64-0.8 mins; MS m/z 339{M+H}+; Method 2minLC_v003 1H NMR (400MHz, DMSO-d6) d 8.6 (1H, t), 7.25-6.95 (7H, m), 5.0 (2H, s), 4.5 (2H, d), 2.9 (4H, m), (4H, m), 2.2 (3H, s), 2.1 (3H, s). m6—Bromo-N-(2—methyl-5—(2—(4-methylpiperazin—1 - yl)benzylcarbamoyl)phenyl)pyrazolo [1 yridine-3—carboxamide A mixture comprising 6-bromopyrazolo[1,5-a]pyridinecarboxylic acid (commercially available)(196 mg, 0.813 mmol) and thionyl chloride (2 mi, 27.4 mmol) was heated at 60°C for 1 hr and concentrated in vacuo. To this was added 3—amino—4-methyl-N—(2- (4-methylpiperazinyl)benzyl)benzamide (step 1) (220 mg, 0.651 mmol) and pyridine (5 ml). The reaction was stirred under nitrogen at room temperature for 2 hrs. The mixture was diluted with 10% MeOH in EtOAc and washed with water, sat NaHCOs, brine and concentrated in vacuo. The crude product was purified by chromatography on silica eluting with 0—20% 2M NHS in MeOH/DCM to afford the title compound; LC-MS: Rt 0.95 mins; MS m/z 561/563/564 {M+H}+; Method 2minLC_v003 Intermediate 4B N enzylcarbamoyl)f|uorophenyl)iodoimidazo[1,2-a]pyridine carboxamide $9.1; 6—lodoimidazo[1,2—a]pyridine—B—carboxyIic acid The title compound was prepared from 5—iodopyridin—2—amine analogously to 7— bromoimidazo[1,2-a]pyridine~3-carboxylic acid (Intermediate 1A step 2 and step 3); LC-MS: Rt 1.07 mins; MS m/z 317 [M+H]+; Method 2minLC_v003 m3-Amino-N-benzylfluorobenzamide The title compound was prepared from methyl o-4—fiuorobenzoate, 1O benzylamine and 2,3,4,6,7,8-hexahydro-1H-pyrimido[1,2-a]pyrimidine analogously to 3-amin0—4-methyl-N—(Z-(4—methylpiperazin—1-yI)benzyI)benzamide (Intermediate 4A, step 1); 1H NIVIR (400MHz, DMSO-dS) 6 8.9 (1H, t), 7.4-7.2 (6H, m), 7.0 (2H, d), 5.3 (2H, s), 4.5 (2H, d). _S_jt_e_g_3_: N—(5—(Benzylcarbamoyl)fluorophenyi)—6—iodoimidazo[1,2—a]pyridine—3— carboxamide The title compound was prepared from 6—iodoimidazo[1,2-a]pyridine~3~carb0xyIic acid (step 1) and 3-3mino-N-benzyI—4~fluorobenzamide (step 2) anangously 6—bromo-N- (2—methyI-5—(2—(4—methylpiperazin—1—yI)benzylcarbamoy|)phenyI)pyrazolo [1 ,5- a]pyridine—3—carboxamide (Intermediate 4A); LC-MS: Rt 0.99 mins; MS m/z 515/516/517 ; Method 2minLC_v003 Intermediate 4C 7-Bromo-N-(5—(2-(2,6-cis-dimethylpiperidinyl)ethylcarbamoyl) fluorophenyl)imidazo[1,2-a]pyridine-S-carboxamide 8439;]; 3—Amino—N-(2—(2,6-cis—dimethylpiperidinyi)ethyi)—4—fluorobenzamide A mixture comprising 2-(2,6—cis-dimethylpiperidin—1-yl)ethanaminium chloride (4 .75 mmol) and methyl 3—amino—4-fluorobenzoate (3.51 g, 20.75 mmol) in THF (50 ml) was treated with TBD (2.89 g, 20.75 mmol) and stirred at 80°C for 16 hrs. A further portion of methyl o-4~fluoro benzoate (1 g) and TBD (0.5 g) were added and heating continued for 24 hrs. The resulting mixture was partitioned between ethyl acetate and aqueous sodium bicarbonate. The mixture was extracted once with ethyl acetate and once with chloroform. The ed organic layers were 1O dried (M9804), filtered and ated to dryness. Purification of the e by chromatography on silica eluting with 0-20% MeOH in DCM afforded the title compound; LC-MS: Rt 0.71 mins; MS m/z 294 {M+H}+; Method 2minLC_v003.
Step 2: 7—Bromo-N-(5-(2-(2,6—cis-dimethylpiperidinyl)ethylcarbamoyl)—2- fluorophenyl)imidazo[1,2—a]pyridine—3—carboxamide 7—Bromoimidazo[1,2-a]pyridine—3-carboxylic acid (intermediate 1A step 3) (1.150 g, 4.77 mmol) was suspended in toluene (10 ml) and treated with thionyl chloride (1.045 ml, 14.32 mmoi). The mixture was at 100°C for 2hrs. The solvent was removed in vacuo and the solid was added to a stirred solution of 3-amino—N—(2-(2,6-cis—dimethyl piperidinyl)ethyl)fluorobenzamide (1.4 g, 4.77 mmol) in dry pyridine (5 ml) containing oven dried molecular sieves. The mixture was stirred at RT under nitrogen here overnight. The t was removed in vacuo and purification of the crude t by chromatography on silica eluting with 0-20% MeOH in DCM afforded the title compound; LC—MS: Rt 0.82 mins; MS m/z 516{M+H}+; Method 2minLC_v003 Intermediate 4D 6-Bromo-N-(2-fluoro(2-(4-methylpiperazinyl)benzylcarbamoyl)phenyi) pyrazolo[1,5-a]pyridinecarboxamide The title compound was prepared analogously to lnteremdiate 4A from the appropriate ng compounds; LC—MS: Rt 0.95 mins; MS m/z 565/568/569 {M+H}+; Method 2minLC__v003 Intermediate 4E 7-Bromo-N-(5-((5,5-dimethyltetrahydrofuranyl)methylcarbamoyl) phenyl) imidazo[1,2-a]pyridinecarboxamide rjlj/ TEEa—jfiafirr: tn“? 1O Sim; 5—(Azidomethyl)-2,2-dimethyltetrahydrofuran momethyl)—2,2-dimethyltetrahydrofuran (36 g, 186 mmol) in DMF (300 ml) was treated with solid sodium azide (12.73 g, 196 mmol) and heated at 90 °C for 4 hrs.
The mixture was allowed to cool to RT and partitioned between water(1.5 l) and ether (2 x 500 ml). The ether layer was separated and washed with (0.5M ) LiCl (500 ml), dried M9804, filtered and ated to afford the title compound; Slap; (5,5-Dimethyltetrahydrofuran—2-yl)methanamine -(Azidomethyl)-2,2-dimethyltetrahydrofuran (22 g, 142 mmol) in THF (500 ml) was treated with Triphenylphosphine (39.0 g, 149 mmol) and t stirred for 5 mins. Water (50.0 ml) was added and the reaction mixture was heated at 80 °C for4 hrs. The mixture was passed through lsolute® SCX—2 resin (2009 0.67 mmol Kg) eluting with MeOH(500 ml), DMSO (100 ml), 20% MeOH2DCM (500 ml), MeOH (500 ml) followed 7M ammonia in MeOH (500 ml). The ammonia layer was evaporated to dryness to afford the title compound.
LC—MS: Rt 0.63 mins; MS m/z 243 [M+H]+; Method 2minLC_v002_low mass Step 3—4: 7-Bromo-N-(5-((5,5-dimethyltetrahydrofuranyl)methylcarbamoyl)—2— fluorophenyl)imidazo[1,2-a]pyridine-3—carboxamide The title compound was ed from (5,5-dimethyltetrahydrofuran—Z-yl)methan amine (step 2) and methyl 3—amino-4—fluorobenzoate analogously to lntermediate 4A steps 1 and 2; LC—MS: Rt 0.96 mins; MS m/z 491 [M+H]+; Method 2minLC_v003 intermediate 4F 7-Bromo-imidazo[1,2-a]pyridinecarboxylic acid {5-[((S)-5,5-dimethyl -tetrahydro-furanylmethyl)wcarbamoyfl-Z-fluoro-phenyl}-amide The title compound was ed analogously to Intermediate 4E by replacing (5,5- dimethyltetrahydrofuran-2—yl)methanamine (step 3) with (S)—(5,5~dimethyltetra hydrofuran-Z—yl)methanamine; LCMS Rt 0.87 mins; MS m/z 491/492 [M+H]+; Method __v003 Intermediate 5A 4-Fluoro(imidazo[1,2-a]pyridinecarboxamido)benzoic acid FH]N\ O OH Step 1: |midazo[1,2-a]pyridinecarbonyl chloride A suspension of imidazo[1,2-a]pyridine—3—carboxylic acid (5.270 g, 32.5 mmol) in DCM (200 ml) was treated with oxalyl chloride (3.13 ml, 35.8 mmol) followed by the addition of DMF (0.252 ml, 3.25 mmol). The reaction mixture was stirred at RT overnight. The solvent was removed in vacuo to afford the title compound as a hydrochloride salt; 1H NMR (400 MHz, DMSO-d6) 6 9.48 (1H, d), 8.77 (1H, s), 7.99 (2H, m), 7.56 (1 H, §t_e_p_2: Methyl o-3—(imidazo[1,2—a]pyridine—3-carboxamido)benzoate A solution of methyl 3—amino—4—fluorobenzoate (5 g, 29.6 mmol) in pyridine (200 ml) 1O was treated with imidazo[1,2—a]pyridine-3—carbonyl chloride.HCl (step 1)(6.43 g, 29.6 mmol) and the mixture was stirred at RT for 2 days. The e was poured into water (30 ml) and a small exotherm was observed. After cooling to RT, the resulting itate was filtered and dried in a vacuum oven to afford the title compound; LC-MS: Rt 0.81 mins; MS m/z 314.2 {M+H}+; Method 2minLC_v003 1H NMR (400 MHz, DMSO-d6) 6 10.30 (1H, s), 9.44 (1H, d), 8.63 (1H s), 8.34 (1H, dd), 7.83 (1H, m), 7.78 (1H, d), 7.54 (1H, m), 7.48 (1H, m), 7.20 (1H, t), 3.90 (3H, s). gm4—Fluoro(imidazo[1,2-a]pyridine—3-carboxamido)benzoic acid A suspension of methyl 4-fluoro—3-(imidazo[1,2-a]pyridinecarboxamido)benzoate (step 1) (7.2 g, 22.98 mmol) in water (30 ml), THF (45.0 ml) and MeOH (15.00 ml) 2O was treated with lithium hydroxide monohydrate (4.82 g, 115 mmol). The reaction mixture was stirred at RT ght and concentrated in vacuo to remove THF and MeOH. The resulting mixture was acidified with 2M HCl to yield a solid that was collected by filtration and washed with ether (3 x). The white solid was dried in the vacuum oven at 50°C to afford the title compound as a hloride salt; LC-MS: Rt 0.71 mins; MS m/z 300.2 {M+H}+; Method 2minLC_v003 1H NMR (400 MHz, 6) 5 13.21 (1H, br 8), 10.68 (1 H, s), 9.60 (1 H, s), 8.94 (1H, s), 8.29 (1 H, dd), 8.00 (1H, d), 7.86 (2H, m), 7.47 (2H, m).
Intermediate 6A (S)(2-(Methoxymethyl)pyrrolidinyl)ethanamine §_t_e_L1_: (S)—tert—Butyl 2-(2-(methoxymethyl)pyrrolidin—1-yl)ethylcarbamate A suspension comprising (S)—2—(methoxymethyl)pyrrolidine (1 g, 8.68 mmol), tert- butyl 2—bromoethylcarbamate (1.946 g, 8.68 mmol), triethylamine (1.210 ml, 8.68 mmol) and potassium carbonate (1.200 g, 8.68 mmol) in MeCN (10 ml) was heated at 85°C ght. The resulting mixture was filtered and washed with MeCN.
Purification of the crude product by chromatography on silica eluting with 0—5% MeOH in DCM afforded the title compound as a colourless oil; 1H NMR (400MHz), DMSO-d6) 6 3.3 (1H, s), 3.25 (3H, s), 3.15 (1H, m), 3.0 (2H, m), 2.9 (1H, m), 2.8 (1H, m), 2.55 (1H, m), 2.3 (1H, m), 2.15 (1H, q), 1.8 (1H, m), 1.65 (2H, m), 1.45 (1H, m), 1.4 (9H, s). _: (S)—2-(2-(Methoxymethyl)pyrrolidlnyl)ethanamine A solution of (S)-tert-buty| 2—(2-(methoxymethyl)pyrrolidin-1—yl)ethylcarbamate (step 1)(1.89 g, 7.32 mmol) and in MeOH (5 ml) and treated with 2M HCl in MeOH (10 equivalents) at room temperature over the weekend. The solvent was d in vacuo to afford the title compound; 1H NMR (400MHz, CDCl3) 5 3.45 (1H, m), 3.25 (1 H, m), 3.1 (1H, m), 2.9 (1H, m), 2.8 (2H, m), 2.6 (1H, m), 2.5 (3H, s), 2.4 (1H, m), 2.2 (1H, m), 0.8 (1H, m), 1.7 (2H, m), 1.6 (1H, m).
The intermediates of the following table (Table 6) were ed by a similar method to that of intermediate 6A from the appropriate commercially available ng compounds.
MeOD)5 3.5 (2H, t), 2.9 2-(3-propylpyrrolidin- (1H, t): 2-75 (3H, m), 1-yl)ethanamine 2-55 (1H, m), 2 (3H, m), 1.4 (4H, m), 1 (3H, t) PC17IB2012/054501 1H NMR (400MHz, CDCI3)53.4 (1H, m), 3.38 (3H, s), 3.5 (1H, m),5 (S)-2—(2—(methoxy 2.9 (1H, m), 2.8 (2H, m), methyl) pyrrolidin-‘l- 2.6 (1H, m), 2.4 (1H, m), yl)ethanamine 2.2 (1H, q), 1.9 (1H, m), 1.7 (2H, m), 1.6 (1H, m). 1H NMR (400MHz, 00013) 5 3.45 (1H, m), 3.25 (1H, m), 3.1 (1H, (2-(methoxy m), 2.9 (1H, m), 2.8 (2H, methyl) pyrrolidin m), 2.6 (1H, m), 2.5 (3H, yl)ethanamine s), 2.4 (1H, m), 2.2 (1H, m), 0.8 (1H, m), 1.7 (2H, m), 1.6 (1H, m). 0 _.
H NMR z), (2-(tert-butyl(methyl) DMSO) 5 8.15 (3H, br), amino)ethanamine 3.60 (1H, m), 3.30 (2H, trifluoro acetate m), 3.07 (1H, m), 2.76 Mun—“mg“ (3H, s), 1.34 (9H, s), 1H NMR (400MHz), CDSOD) 5 3.75—3.42 (4H, m), 3.17 (2H, m), 2—(2,2-dimethyl piper 1.87 (4H, m), 1.70 (2H, idin—1-yl) ethanamine m), 1.55-0.82 (6H, m), Intermediate 7A 3-(5-Carboxyfluorophenylcarbamoyl)(1-methy|-1H-pyrazoI yl)imidazo[1,2-a]pyridinium chloride Lula JIM tax ( I/ ' 0"" Kim The title compound was prepared analogously to yl—5—(6—(1—methy|—1 H~ pyrazol~4—yl)pyrazolo[1,5-a]pyridine—3—carboxamido)nicotinic acid (Example 9.0 step 2) from methyl romoimidazo[1,2—a}pyridine3—carboxamido)—4-f|uorobenzoate (Intermediate 1A) and 1-methyl(4,4,5,5—tetramethyl-[1,3,2]dloxaborolan-2—yl)—1 H- pyrazole followed hydrolysis of the resulting methyl ester using sodium ide; LC-MS: Rt 0.77 mins; MS m/z 380 [M+H]+; Method 2minLC_v003 Intermediate TB 6-MethyI(6-(1-methyl-1H-pyrazolyl)pyrazolo[1,5-a]pyridinecarboxamido) nicotinic acid The title compound was prepared analogously to 6-methyl—5-(6-(1-methyl-1H- pyrazolyl)pyrazolo[1,5-a]pyridine—3—carboxamldo)nicotinic acid (Example 9.0 step 2) from Intermediate 10 and 1-methy|—5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2— yl)—’l H—pyrazole followed ysis of the resulting methyl ester using sodium hydroxide; LC—MS: Rt 0.69 mins; MS m/z 377/378 [M+H]+; Method 2minLC_vOO3 Intermediate 8A 4-Fluoro(7-(3-fluoro(1-hydroxymethylpropan ylcarbamoyl)phenyl)imidazo[1 ,2-a]pyridinecarboxamido)benzoic acid “‘ J t/ \\ F {4“-N’Z : H N K/\mRa HO who Steps 1 and 2: 7-(3-Fluoro-4~(1-hydroxy—2-methylpropan-2—ylcarbamoyl)phenyl)—N—(2- fluoro(1-hydroxymethylpropanylcarbamoyl)phenyl)imidazo[1,2-a]pyridine—3- carboxamide The title compound was analogously to Example 9.0 steps 2 and 3 from (1) ediate 1A and 4—borono—2—fluorobenzoic acid analogously to Example 9 step 2; (2) 4-(3-(5-Carboxy—2-fluorophenylcarbamoyl)imidazo[1,2-a]pyridin-7—yI) fluorobenzoic acid (step 1) and omethylpropanol; LC-MS: Rt 0.91 mins; MS m/z 580.4 [M+H]+; Method prvO1 SE12; 4—Fluoro—3-(7-(3-fluoro—4-(1-hydroxy-2—methy|propan—2- yloarbamoyl)phenyl)imidazo[1,2—a]pyridine—3—carboxamido)benzoic acid 7-(3—FIuoro—4—(1—hydroxy—2-methylpropan—2—ylcarbamoyl)pheny|)-N—(2~fluoro~5—(1— hydroxy-2—methylpropan-2—ylcarbamoyl)phenyl)lmldazo[1,2-a]pyridine—3-carboxamide (step 1)(590 mg, 1.129 mmol) in 1.2-dimethoxyethane (10 ml) and water (10.00 ml) was treated with sodium hydroxide (181 mg, 4.52 mmol) and warmed to 60°C for 1hr.
After cooling to RT, the pH of the mixture was adjusted to pH5 using 2M HCl (2.2 ml).
The solvent was removed in vacuo and the resulting crude product was triturated with water to afford the title compound; LCMS: Rt 0.92 mins; MS m/z 509.4 [M+H]+; Method 2minLopr intermediate 9A (S)-(5,5-Dimethyltetrahydrofuranyl)methanamine . '1. ”fit/1 ”f To a stirred solution of (S)—(5,5~dimethyltetrahydrofuran—2-yl)methanol (14 g, 108 mmol) and PPh3 (33.8 g, 129 mmol) in THF (140 ml) under N2 was added phthalimide (17.40 g, 118 mmol) to give a suspension. The mixture was cooled to 8°C and DlAD (27.2 ml, 140 mmol) was added dropwise over 30 mins keeping 1O internal T <10 °C. The resulting white slurry was diluted with water (100 ml) and ted with EtOAc (100 ml). The organics were washed with sat. NaH003 (100 ml), brine (100 ml) and dried (M9804) and concentrated under reduced pressure.
Purification by chromatography on silica g with 0—20% EtOAc/iso—hexane ed the title compound as a white crystalline solid; LC—MS: Rt 1.02 mins; MS m/z 260 [M+H]+; Method 2minLopr Step; (S)—(5,5-Dimethyltetrahydrofuran—Z-yl)methanamine hydrochloride A solution of (S)—2-((5,5-dimethyltetrahydrofuran-2—yl)methyl)isoindoline—1,3-dione (step 1) (21.9 g, 84 mmol) in EtOH (440 ml) was stirred at 60 °C under N2 and hydrazine e (4.51 ml, 93 mmol) was added. The mixture was stirred at 60 °C overnight and then allowed to cool to RT. 2M HCl (60 ml) was added dropwise to adjust pH to pH 1. The slurry was filtered g with EtOH (50 ml) and the filtrate was concentrated in vacuo to a volume of approximately 50 ml.
The mixture was filtered and washed through with TBME (20 ml) and water (20 ml).
The te was washed with TBME (100 ml) and the organic phase was extracted with 2M HCl (50 ml). The acidic aqueous layers were combined and basified to pH 10 with 2M NaOH (~70 ml). This mixture was extracted with TBME (3 x 200 ml) and the combined organic layers were washed with brine (200 ml) and dried (M9804) and filtered. 4M HCl in dioxane (21 ml, 84 mmol) was added slowly and the resulting solution was then concentrated in vacuo to yield a yellow oil. The oil was triturated with diethyl ether to afford the title compound as a pale yellow solid; LC-MS: Rt 0.57 mins; MS m/z 164.1 [M+H]+; Method 2minLopr Intermediate 9B ,5-Dimethyltetrahydrofuranyl)methanaminium chloride “sf /””.1.\‘I /NH3+ ..--‘-1’5..- _S_tep_£ (R)—(5,5—Dimethyltetrahydrofuran-Z-yl)methyl 4-methylbenzenesulfonate 1O (R)-(5,5-dimethyltetrahydrofuran—2—yl)methanol [refer Bull chem Soc Japan Vol. 45, No.3, pp. 916-921. 1972 J Yoshimura et.al p921) (6.7 g, 51.5 mmol) in pyridine (50 ml) was treated with Tosyl-Cl (9.81 g, 51.5 mmol) at room temperature and stirred for 72hrs.The solvent was removed by evaporation and azeotroping with toluene. The mixture was then partitioned between ethyl acetate and 10% aqueous citric acid. The organics were washed with brine and the s was back extracted with ethyl acetate. The combined c layers were dried M9804, filtered and evaporated to dryness to give a dark oil of (R)—(5,5-dimethyltetrahydrofuranyl)methyl 4- methylbenzenesulfonate; 1H NMR (400 MHz, CDClg) 8 7.85 (2H, d), 7.36 (2H, d), 4.17 (1 H, m), 3.98 (2H, d), 2.47 (3H, s), 2.07 (1 H, m), 1.85-1.70 (3H, m), 1.18 (6H, s). flap—2: (R)—5—(Azidomethyl)-2,2-dimethyltetrahydrofuran (R)—(5,5~Dimethyltetrahydrofuran~2—yl)methyl 4—methylbenzenesulfonate (step 1) (12.8 g, 45.0 mmol) in DMF (50 ml) was d with sodium azide (3.80 g, 58.5 mmol) at RT overnight then warmed to 70°C for 3hrs. Sodium azide (3.80 g, 58.5 mmol) was added and the mixture was warmed to 100°C for 3hrs. allowed to cool to RT. The e was diluted with water and ethyl acetate. The ethyl acetate layer was washed with 0.5 M lithium de in water and the organic layers were dried M9804, ed and evaporated to dryness to afford the title compound; 1H NMR (400 MHz,CDC13)5 4.17 (1H, m), 3.49 (1H, dd), 3.20 (1H, dd), 2.05 (1H, m), 1.85~1.75 (3H, m), 1.30 (3H, s) 1.23 (3H, s) m(R)—(5,5-Dimethyltetrahydrofuran-2—yl)methanaminium chloride (R)(Azidomethyl)—2,2-dimethyltetrahydrofuran (step 2)(6.98 g, 45 mmol) in tetrahydrofuran (175 ml) and Water (35.0 ml) was treated with triphenylphosphine (12.98 g, 49.5 mmol) and stirred at RT for 20 mins then warmed to 80 °C for 4 hrs.
Solid Isolute® SCX resin was added and stirred at RT for 1hr. The SCX—2 resin was washed with 7M ammonia in MeOH (1 L). The ammonia layer was ated to dryness with a cool water bath and vacuum > 80 mbar. The oily residue was treated with 2N HCI (aq) and the solid precipitate was removed by filtration. The aqueous portion was washed with ethyl e and DCM (containing 10% trifluoroethanol.) The aqueous was basified by the addition of 2N NaOH (aq) and was extracted with ethyl acetate (x3). The combined organic layers were dried M9804, d and treated with excess HCl in dioxane before being evaporated to dryness to afford the title compound; 1H NMR (400 MHz, MeOD) 5 4.17 (1 H, m), 3.09 (1H, dd), 2.87 (1 H, dd), 2.19 (1 H, m), 1.95—1.70 (3H, m), 1.31 (3H, s) 1.27 (3H, 3).
Intermediate 9C (S)Bromo-N-(5-((5,5-dimethyltetrahydrofuranyl)methylcarbamoyl) fluorophenyl)imidazo[1,2-a]pyridinecarboxamide l 'i C' ”“0 4‘. WEN The title compound was prepared from (R)~(5,5—dimethyltetrahydrofuran-Z- yl)methanaminium chloride (intermediate QB) and 3-(7—Br0m0imidazo[1,2-a]pyridine- 3—carboxamido)—4—fluoro c acid (prepared by hydrolysis of methyl 3-(7- bromoimidazo[1,2-a]pyridinecarboxamido)fluorobenzoate (Intermediate 1A) using NaOH) ously to Example 7.4 step 1; LC-MS: Rt 0.97 mins; MS m/z 489/4912 {M+H}+; Method 2minLopr Intermediate 9!): 2-((2RS,3SR)-2,3-Diethylazetidinyl)ethanamine H EH -.‘vflx‘ Diffs)r l3 xi” \H o t] . [2furl? Chlorosulfonyiisocyanate (5.17 ml, 59.4 mmol) in DCM (12 ml) was stirred at 25 °C and treated dropwise with (E)—hexene (7.39 mi, 59.4 mmoi) in DCM (6 ml). The mixture was stirred at RT for 72 hrs. The reaction mixture was heated over 6 hrs at 40°C, before being poured onto ice. The mixture was extracted with DCM (3 x 100 ml) and the ed organics were washed with water (x1),dried M9804 and evaporated to dryness to afford the title compound; 1H NMR (400 MHz, CDCI3) 6 3.95 (1H, dt), 3.09 (1H, cit), 2.21 (1H, m), 1.85 (3H, m), 1.11 (3H, t), 1.05 (3H, t). $5522.; (3RS,4RS)—3,4~Diethyiazetidin—2-one (2RS,3RS)—2,3-Diethyi—4—oxoazetidine~1—sulfonyl chloride (step 1) (3.2 g, 14.18 mmoi) in e (7 ml) was treated with enol (2.92 mi, 28.4 mmoi) and cooled to -30 °C. ne (1.376 ml, 17.01 mmoi) in acetone (2.55 ml) was added dropwise over 30 mins, maintaining the temperature around —30 °C. After stirring for 30 minutes, water (10 ml) was added slowly and the mixture was filtered. The e was extracted with diethyl ether (5 X 25 ml) and the combined organic layers were dried M9804, filtered and evaporated to dryness (colorless oil 2.39). Purification by chromatography on silica eluting with iso-hexane followed by diethyl ether afforded the title compound; 1H NMR (400 MHZ, CDCI3) d 3.36 (1H, dt), 2.71 (1H, cit), 1.81 (1 H, m), 1.72-1.55 (3H, m), 1.03 (3H, t), 0.95 (3H, t). m:2-((2RS,3RS)—2,3-Diethyl—4-oxoazetidinyl)acetonitrile (3RS,4RS)—3,4-diethylazetidin-Z-one (step 2)(100 mg, 0.786 mmol) in dry THF (5 ml) was cooled to —78 °C and was treated with lithium bis(trimethylsilyl)amide (0.786 ml, 0.786 mmol) [1 M in THF]. The solution was allowed to warm to 0 °C then re—cooled to 0°C before adding bromoacetonitrile (0.060 ml, 0.865 mmol). The e allowed to warm to RT overnight. 10% Aqueous citric acid (30 ml) was added and the e was extracted with ether (4 x 40 ml). The ed organic layers were dried M9804, filtered and evaporated to dryness. Purification by chromatography on silica eluting with 0-100% Et20 in iso-hexane ed the title compound; 1H NMR (400 MHz, CDCIg) 8 4.21 (1H, d), 3.99 (1H, d), 3.32 (1H, dt), 2.71 (1H, dt), 1.81 (1H, m), 1.72-1.55 (3H, m), 1.03 (3H, t), 0.95 (3H, t). §t§_p_4_: 2—((2RS,38R)—2,3-Diethylazetidinyl)ethanamine hydrochloride AlCls (3.37 g, 25.3 mmol) in dry ether (140 ml) was added to a stirred suspension of 1M LAlH4 in ether (25.3 mi, 25.3 mmol) in ether (140 ml). The mixture was heated at reflux for 30 mins and after g to RT the mixture was transferred by cannula into a solution of 2-((2RS,3RS)~2,3—diethyl—4—oxoazetidin~1—yl)acetonitrile (step 3) (1.4 8.42 mmol) in dry ether (50 ml). Stirring was continued at RT for 16 hrs. The reaction mixture was cooled to 0°C and Rochelle salt (aq 50 ml) was added. The mixture allowed to stir at RT for 24hr. The aqueous was separated and the ether layer retained. The aqueous was stirred with 10% trifluoroethanol/DCM (250 ml) for 3hrs then the layers were separated and the aqueous was further stirred with 10% trifluoroethanol/DCM {250 ml} for a further 2hrs. The remaining aqueous was extracted with 10% trifluoroethanol/DCM (x3). The ed organic layers were treated with 1 M HCl in methanol and concentrated in vacuo to afford the title compound; 1H NMR (400 MHz, MeOD) 8 4.55 (1H, dd), 4.33 (1 H, t), 4.12 (1H, dd), 3.87 (1H, m), 3.72-3.46 (3H, m), 2.56 (1H, m), .91 (2H, m), 1.71 (2H, m),1.05 (3H, t), 0.90 (3H, t).
From the foregoing it will be appreciated that, although specific embodiments of the ion have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not d except as by the appended claims.

Claims (24)

Claims
1. A compound represented by formula (1) 5 or a pharmaceutically acceptable salt f, wherein, A is R3 R1 is C1—C4 alkyl; 01-04 alkoxy optionally substituted by one or more halogen atoms; 10 CN; or halogen; R1:11 is H, halogen, C1—C4 alkyl or C1-C4 haloalkyl; X is N or OH; R2 is H; C1—C8 alkyl optionally substituted by one or more OH, ~NR9R11 or C1—C4 alkoxy; C1—Cg haloalkyl; C2-C3 alkynyl tuted by one or more halogen, OH, - 15 NRQR“ or 01-04 alkoxy; 03-010 cycloalkyl; -(C1-C4 alkyl)-Cg-Cg cycloalkyl; 01-08 alkoxy optionally substituted by one or more halogen, -NR9R“ or OH; OH; CN; halogen; -(C0-C4 alkyl)—NR9R“; -(Co—C4 alkyl)—COZR15; -(C0~C4 alkyl)-C(O)NR9R“; - (Co-C4 alkyl)-C6-C14 aryl; or -(Co-C4 alkyl)—3 to 14 membered heterocyolyl; n the cycloalkyl, -(Co-C4 alkyl)—CG-Ci4 aryi and ~(Co-C4 alkyl)—3 to 14 heterocyclyl are each 20 optionally substituted by one or more 2" substituents; R3 is H; C1-C8 alkyl optionally substituted by one or more OH, -N R9R11 or C1-C4 alkoxy; C1-C8 haloalkyl; C2—C3 alkynyl tuted by one or more halogen, OH, — NRQR", or C1-C4 alkoxy; 03-010 cycloalkyl; —(C1—C4 —Cs-Ca oycloalkyl; C1-C8 alkoxy ally substituted by one or more halogen, —NR9R11 or OH; OH; CN; halogen; -(Co-C4 alkyl)—NR9R”; ~(Co-C4 alkyl)—COzR15; -(Co-C4 alkyl)-C(O)NR9R”; - (Co-C4 alkyl)-C5—C14 aryl; or ~(C0—C4 alkyl)-3 to 14 ed heterocyclyl; wherein the cycloalkyl, -(Co-C4 alkyl)—Ce-C14 aryl and -(Co-C4 alkyl)—3 to 14 heterocyclyl are each optionally substituted by one or more Za substituents; each Z3 is independently OH; (Co—C4 alkyl)—Cs aryl; O-Cs aryl; C1—C4 alkyl optionally substituted by one or more OH, ON or -NR193R213; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more OH, —COZR193, —NR1QE‘R21a or C1—C4 alkoxy; — (O)R21a; —C(O)NR19aR21a; _NR18aC(O)NR1QaR21a; _NR193R21a; 4 alkyl)- C(O)OR188; ~(Co-C4 alkyl)-C(O)R1ga; oxo; CN; N02; halogen; —(Co-C4 alkyl)—4 to 6 10 membered heterocyclyl; or ~O-(4 to 6 membered heterocyclyl); wherein the (Co—C4 alkyl)—Cs aryl, O—Cs aryl, -(Co-C4 alkyl)—4 to 6 membered heterocyclyl and —O-(4 to 6 membered heterocyclyl) are each optionally substituted by OH, halogen, C1-C4 alkyl, C1-C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more ns; R4 is H; 15 R5 is H, C1—C4 alkyl or 01-04 haloalkyl; R6 is selected from 01-08 alkyl optionally tuted by one or more C1-C4 alkoxy -NR‘9R2‘; 01-08 haloalkyl; -(Co-C4alkyl)-Cg-Cscycloalkyl; C1—Caalkoxy optionally substituted by one or more halogen atoms; -NR19R21; —(Co—C4 alkyl)—Cs-C14aryl; and — (Co—C4 alkyl)—3 to 14 membered heterocyclyl; wherein the ~(Co-C4alkyl)-Cs- 20 Cgcycloalkyl, -(Co-C4 alkyl)—C5-C14aryl and -(Co-C4 alkyl)-3 to 14 heterocyclyl are each optionally substituted by one or more Z substituents; each Z is independently ed from (Co—C4 alkyl)—Cs aryl; O—Cs aryl; C1—Ce alkyl optionally substituted by one or more C1-Ce alkoxy, CN or —NR19R21; C1—Cs haloalkyl; C1-Cs alkoxy optionally substituted by one or more 21 or C1-C4 alkoxy; — 25 NR19R21; (Co~C4 alkyl)—C(O)R19; CN; n and (co-c4 alkyl)~4 to 6 membered heterocyclyl; and wherein the aryl and cyclyl are each optionally substituted by one or more halogen, C1-C5 alkyl, (31-06 haloalkyl and 01-06 alkoxy optionally substituted by one or more halogens; R9 and R11 are each independently selected from H; C1—C5 alkyl optionally substituted 30 by one or more C1-C4 alkoxy or OH; 01—06 haloalkyl; —(Co-C1alkyl)—Cs-Ce cycloalkyl; (Co-C4 - 06-014aryl optionally substituted by one or more groups selected from 01—05 alkyl, Ci-Cs alkoxy and halogen; and (Co-C4 alkyl)- 3- to 14-membered heterocyclyl ally substituted by one or more groups selected from halogen, oxo, 01-05 alkyl and C(O)C1—Cs alkyl; or 35 R9 and R11 together with the nitrogen atom to which they are ed form a 5- to 1 O-membered heterocyclyl, which heterocyclyl es 0 to 3 r heteroatoms selected from N, O and S, the heterocyclyl being ally substituted by one or more substituents selected from OH; halogen; phenyl, 5- to 10-membered heterocyclyl; C1—C6 alkyl; 01—06 haloalkyl; C1—C5 alkoxy optionally substituted by one or more OH or C1—C4 alkoxy; and C(O)OC1-C5alkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally tuted by 01-06 alkyl, 01—05 haloalkyl or 01-05 alkoxy; R15 is selected from H; c1-c8 alkyl; 01—08 haloalkyl; 03-c10 cycloalkyl; (-01—C4alkyl)- Cg-Cg cycloalkyl; -(Co—C4 alkyl)—CG~C14aryl and -(Co-C4 alkyl)—3 to 14 membered cyclyl group; wherein the C3-C1o lkyl, (-C1—C4alkyl)—C3-Cg cycloalkyl, —(Co— C4 alkyl)—Cs—C14aryl and —(Co-C4 alkyl)—3 to 14 membered heterocyclyl groups are 10 each optionally substituted by one or more Z substituents; R183 is independently H or (31-06 alkyl; R193 and R213 are each ndently H; C1-Cs alkyl optionally substituted by one more C1—C4 alkoxy,~ 3', or OH; C1—C5 haloalkyl; ~(Co-C1alkyl)—C3-Cecycloalkyl; - (Co—C4 alkyl)— C5-C14aryl optionally substituted by one or more groups selected from 15 01-05 alkyl, 01-05 alkoxy and halogen; or -(Co—C4 alkyl)- 3- to 14—membered heterocyclyl optionally substituted by one or more groups ed from halogen, oxo, C1-Cs alkyl and C(O)C1-Cs alkyl; or R193 and R21"1 together with the nitrogen atom to which they attached form a 5— to 10— membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms 2O selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents ed from OH; n; phenyl; 5- to 10—membered heterocyclyl; 01—05 alkyl; 01—05 haloalkyl; 01-05 alkoxy optionally tuted by one or more OH or C1-C4 alkoxy; and C(O)OC1-C5alkyl; wherein the phenyl and heterocyclyl substituent groups are themselves optionally substituted by 01-05 alkyl, 25 C1-Cs haloalkyl or 01—05 alkoxy; R18 is independently H or (31-06 alkyl; R19 and R21 are each independently C1—C5 alkyl optionally substituted by one or more C1—C4 alkoxy; C1-Ce kyl; —(Co—C1alkyl)~Cg-Cscycloa|kyl;- (Co—C4 alkyl)-C5-C14aryl optionally substituted by one or more groups selected from C1-C5 alkyl, C1-Cs alkoxy 30 and halogen; or -(Co-C4 alkyl)— 3— to 14—membered cyclyl, optionally substituted by one or more groups selected from n, 01—05 alkyl and -C(O)C1-CB alkyl; or R19 and R21 together with the nitrogen atom to which they attached form a 5- to 10— membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or 35 more substituents selected from halogen; phenyl; 5- to 10—membered heterocyclyl; C1-C5 alkyl; C1—Ce haloalkyl; C1-C5 alkoxy optionally substituted by one or more C1-C4 alkoxy and C(O)OCq-Cealkyl; n the phenyl and heterocyclyl substituent groups are themselves optionally substituted by a substituent ed from C1-C6 alkyl, C1- CG haloalkyl and C1—C5 alkoxy; and R22 and R23 are each ndently H or 01—06 alkyl.
2. A compound ing to claim 1, wherein R5 is H; R6 is 01—05 alkyl ally substituted by C1—C4 alkoxy; C1-Ce haloalkyl; —(Co—C4aikyl)- Cg-Cgcycloalkyl; C1-Cs alkoxy; ~Ce-C1oaryl; or -(Co-C2 alkyl)—5 to 6 membered heterocyclyl; wherein the -(Co-C4alkyl)-Cg—Cgcycloalkyl, -Ce—C1oaryl and 2 - 10 5 to 6 heterocycylyl are each optionally substituted by one or more Z substituents; each Z is independently selected from -(Co-C4 alkyl)—Cs aryl; ~O-Ce aryl; C1-C4 alkyl optionally substituted by one or more CN or -NR’9R21; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more -NR19R21 or C1—C4 alkoxy; -NR19R21; -(Co~C4 alkyl)—C(O)R19; CN; halogen or -(Co—C4 alkyl)—4 to 6 membered heterocyclyl; wherein 15 the —(Co-C4 alkyl)—Ce aryl, —O-Cs aryi and or -(CO~C4 alkyl)—4 to 6 ed heterocyclyl are each optionally substituted by n, 01-05 alkyl; 01-05 haloalkyl or C1—C6 alkoxy optionally substituted by one or more halogens; R18 is independently H or 01—05 alkyl; R19 and R21 are each ndently C1-Ce alkyl ally substituted by one or more 20 C1-C4 alkoxy; C1-C5 haloalkyl; -(Co-C1alkyl)-Cg—Cscycloaikyl; (Co-C4 alkyl)-ary| optionally substituted by one or more groups selected from 01-06 alkyl, C1—Cs alkoxy and halogen; or (Co—C4 alkyl)— 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, C1435 alkyl and C(O)C1-Cs alkyl; R19 and R21 together with the nitrogen atom to which they attached form a 5— to 10— 25 membered heterocyclyl, which heterocyclyl includes 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally substituted by one or more substituents selected from n; ; 5- to 10-membered heterocyclyl; 01-06 alkyl; C1—C5 haloalkyl; C1—C5 alkoxy optionally substituted by one or more C1-C4 alkoxy; or C(O)OC1-Cealkyl; wherein the phenyl and heterocyclyl substituent groups 3O are themselves optionally substituted by (31—06 alkyl, 01—06 haloalkyl or C1-Ce alkoxy.
3. A compound ing to claim 1 or 2, wherein R5 is H; R6 is 01-05 alkyl optionally substituted by C1—C4 alkoxy; —(Co-Cza|kyl)-Cg—Cacycloalkyl; 35 C1-C4 alkoxy; -Ce-C1oaryl or '(Co-Cz alkyl)—5 to 6 membered heterocyclyl; wherein the -(Co—Czalkyi)—C3—Cscycloaikyl, '(Cg-Cz alkyl)—Ce—C1oaryl and ~(Co-CgalkyI)-5 to 6 membered heterocyclyl are each optionally substituted by one or more Z substituents; each Z is independently C1-C4 alkyl ally substituted by one or more NHZ; C1-C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more C1-C4 alkoxy or - NngRZ‘; CN; halogen or 4 —4 to 6 membered heterocyclyl; wherein the - (Co—C4 alkyl)—4 to 6 membered heterocyclyl is optionally substituted by halogen, 01-05 alkyl, C1—C6 haloalkyl or 01-05 alkoxy optionally substituted by one or more halogens; R19 and R21 are each independently H; 01-05 alkyl optionally substituted by one or more C1-C4 alkoxy; C1—Cs haloalkyl; ~(Co-C1alkyl)—Cg-Cacycl0alkyl; (Co-C4 alkyl)—aryl 1O optionally substituted by one or more groups selected from C1-C5 alkyl, C1—Cs alkoxy and n; or (Co-C4 alkyl)— 5— to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, C1-Cs alkyl and C(O)C1—C5 alkyl.
4. A compound according to any one of claims 1 to 3, wherein 15 R5 is H; R6 is C1-C4 alkyl optionally substituted by one or more C1-C4 alkoxy; phenyl; C1-C4 haloalkyl; ydrofuran; pyrrolidine, yrrolidine or —CH2—piperidine; wherein phenyl, tetrahydrofuran, pyrrolidine, —CH2-pyrrolidine and iperidine are each optionally substituted by one or more Z substituents; 20 each Z is independently C1-C4 alkoxy, halogen, C1-C4 alkyl or C1-C4 haloalkyl.
5. A compound ing to any one of claims 1 to 4, wherein R2 is H; C1-Cg alkyl optionally substituted by one or more OH, -NR9R“or C1-C4 alkoxy; C1—Ce haloalkyl; 02—08 alkynyl substituted by one or more halogen, OH, - 25 NRQR“ or c1-c4 alkoxy; 03-010 cycloalkyi; -(c1-c4 alkyl)-C3-Cg cycloalkyl; 01-08 alkoxy optionally substituted by one or more halogen, -NR9R11 or OH; OH; CN; halogen; -(co-c4 alkyl)—N RQR”; -(c0-c4 —COZR15; -(cO-c4 alkyl)-C(O)NR9R”; - (C0-C4 alkyl)—C5—C14 aryl; or {Co—C4 atkyl)—3 to 14 membered cyclyl; wherein the cycloalkyl, -(Co-C4 alkyl)-Cs-C14 aryl and -(C0-C4 alkyl)~3 to 14 heterocyclyl are each 3O optionally substituted by one or more Za substituents; R3 is H; R9 and R11 are each independently H; 01-05 alkyl ally substituted by one or more C1—C4 alkoxy or OH; 01-05 haloalkyl; 1alkyl)—C3—Cacycloalkyl; (Co-C4 alkyl)- Ce-C14 aryl optionally substituted by one or more groups selected from 01-05 alkyl, 35 C1-C5 alkoxy and halogen; or (Co-C4 — 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, 0X0, C1—Ca alkyl and C(O)C1-Cs alkyl; WO 30802 2a is independently OH; (Co-C4 allow-Cs aryl; O-Cs aryl; C1-C4 alkyl optionally substituted by one or more OH, ON or —NR198R21a; C1—C4 haloalkyl; C1—C4 alkoxy optionally substituted by one or more OH, 93, —NR193‘R21a or C1—C4 alkoxy; - NR1saC(O)R2‘a; —C(O)NR193R213; -NR18“C(O)NR193R213; —NR193R213; (Co-C4 - C(O)OR18a; (co-c4 alkyl)—C(O)R193; oxo; CN; N02; halogen; or (co-c4 alkyl)—4 to 6 membered heterocyclyl; wherein the aryl and heterocyclyl are each optionally substituted by halogen, C1-C4 alkyl, C1-C4 haloalkyl or C1-C4 alkoxy optionally substituted by one or more halogens; R18a is independently H or Cl-Cs alkyl; 1O R193 and R216‘ are each independently H; Ci-Ce alkyl optionally substituted by one or more C1-C4 alkoxy or OH; 01-06 haloalkyl; 1alkyl)—Cg-Cscycloalkyl; -(Co-C4 alkyl)- 06-014 optionally substituted by one or more groups selected from (21-05 alkyl, (31-05 alkoxy and halogen; or (C0—C4 — 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, C1-Ce alkyl and 15 C(O)C1-C.~, alkyl; or R19a and R218 er with the nitrogen atom to which they attached form a 5- to 10- membered heterocyclyl, which heterocyclyl es 0 to 3 further heteroatoms selected from N, O and S, the heterocyclyl being optionally tuted by one or more substituents selected from OH; halogen; phenyl; 5- to 10-membered 20 cyclyl; C1-Cs alkyl; C1-C6 haloalkyl; C1-Ce alkoxy optionally substituted by one or more OH or C1-C4 alkoxy; or C(O)OC1—Csalkyl; wherein the phenyl and heterocyclyl substituent groups are lves optionally substituted by 01—05 alkyl, C1-C5 haloalkyl or C1-Ce . 25
6. A compound according to any one of claims 1 to 5, wherein R2 is H; C1—C4 alkyl optionally substituted by one or more OH or ~NR9R“; C1—C4 haloalkyl; C1-C4 alkoxy optionally substituted by one or more halogen, -NR9R11 or OH; OH; CN; halogen; -(Co-C4 alkyl)-NR9R“; ~(Co—C4 alkyl}C(O)NR9R“; phenyl; or - (Co-C4 —5 to 6 membered heterocyclyl; wherein the phenyl and —(Co-C4 alkyl)—5 30 to 6 heterocyclyl are each ally substituted by one or more Za substituents; R3 is H; R9 and R“ are each independently H; 01-05 alkyl optionally substituted by one or more C1-C4 alkoxy or OH; 01-06 haloalkyl; —(CO-C1alkyl)—C3—Cecycloalkyl; (Co—C4 alkyl)- 05-014 optionally substituted by one or more groups selected from 01-06 alkyl, C1-C5 35 alkoxy and halogen; or (Co-C4 alkyl)- 5- to 6—membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, 01-06 alkyl and C(O)C1—Cs alkyl; each Za is ndently OH; C1-C4 alkyl optionally substituted by one or more one or more OH or -NR19aR21a; C1—C4 haloalkyl; C1-C4 alkoxy optionally tuted by one or more OH, 01—04 alkoxy or -NR193R213; -C(O)NR193R213; CN; halogen or -(<:0-C4 alkyl)- 4 to 6 membered heterocyclyl; wherein the heterocyclyl is optionally substituted by n, C1—C4 alkyl, C1-C4 haloalkyl or C1—C4 alkoxy optionally substituted by one or more halogens; R1981 and R21a are each independently H; C1—C4 alkyl optionally substituted by one or more C1—C4 alkoxy or OH; C1—C4 haloalkyl; -(Co—C1alkyl)—Cg-Cecycloalkyl;— (Co—C4 alkyl)~ Ce-C14 optionally substituted by one or more groups selected from C1—C5 alkyl, 10 C1-C6 alkoxy and n; or ~(Co-C4 alkyl)— 5— to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, C1-Cs alkyl and -Ce alkyl.
7. A compound according to any one of claims 1 to 6, wherein 15 R2 is H; C1-C4 alkyl optionally tuted by one or more OH or —NH2; C1-C4 alkoxy optionally substituted by -NR9R11; F; Br; -(C1—Cz alkyl)-NR9R“; —C(O)NR9R“; phenyl; or -(Co-C4 alkyl)—5 to 6 membered heterocyclyl; wherein the phenyl and -(Co-C4 alkyl)- 5 to 6 membered heterocyclyl are each optionally substituted by one or more 23 substituents; 20 R3 is H; Z3 is independently C1-C4 alkyl optionally substituted by one or more OH or — NR193R213; C1-C4 haloalkyl; C1-C4 alkoxy optionally tuted by one or more OH, C1-C4 alkoxy or -NR19R21; -C(O)N ; halogen; or (Co-C4 alkyl)—4 to 6 membered heterocyclyl; wherein the heterocyclyl is optionally substituted by halogen, C1-C4 alkyl 25 or C1—C4 haloalkyl; R9 and R“ are each independently H; C1-Cs alkyl ally substituted by one or more C1-C4 alkoxy or OH; (31-06 haloalkyl; 1alkyl)—C3-Cecycloalkyl; (Co-C4 alkyl)- aryl optionally substituted by one or more groups selected from Ci-Ce alkyl, C1-Ce alkoxy and halogen; or (CO-C4 - 5- to 6-membered heterocyclyl optionally 30 substituted by one or more groups selected from halogen, oxo, 01—06 alkyl and C(O)C1-05 alkyl; R1921 and R21:3 are each independently H; C1-C4 alkyl optionally substituted by one or more C1-C4 alkoxy or OH; C1-C4 haloalkyl; -(Co—C1alkyl)—Cs—C60ycloalkyl; (Co—C4 alkyl)— aryl optionally substituted by one or more groups selected from 01-05 alkyl, 01-05 35 alkoxy and n; or (Co-C4 alkyl)— 5- to 6-membered heterocyclyl optionally substituted by one or more groups selected from halogen, oxo, 01-05 alkyl and C(O)C1—Cs alkyl.
8. A nd according to any one of claims 1 to 7, wherein R1 is fluorine or methyl; R121 is H; 5 R2 is H, F, Br, /‘\\\ /‘ F O H N/fi \ \ \ K/O I I /\\ ‘ ,\\ / ;\ / O \ N O \ R6 is F I \ —--N N F \——/ , or 5
9. A compound according to Ciaim 1, which is selected from: N—(2—Fluor0(2-(4—methylpiperazinyl)benzylcarbamoy!)phenyl)—7—(1-methyl-1H- pyrazol-S—yl)imidazo[1,2-a]pyridine—3—carboxamide; 7—(3—FIuoro(2-hydroxyethylcarbamoy!)pheny!)-N-(2—fluoro(2-(4-methylpiperazin- 1-y|)benzylcarbamoyl)phenyl)imidazo[1,2—a]pyridinecarboxamide; 10 7—Bromo—N—(2-methyI(2-(4-methylpiperazin-1— yl)benzylcarbamoyl)phenyl)imidazo[1,2-a]pyridine—3—carboxamide; 7-(1-MethyI-1 H-pyrazol—S-yl)—N—(2-methyl-5—(2—(4—methylpiperazin~1 - yl)benzylcarba moyl) phenyl)imidazo[1,2—a1pyridinecarboxamide; N—(2-Fluoro—5-(2-(4—methylpiperazin—1-yl)benzylcarbamoyl)phenyi)—7—(pyridinc 15 dazo[1,2-a]pyridine—3-carboxamide; N—(2—Fluoro—5-(2—(4—methylpiperazin—1-y1)benzyicarbamoyl)phenyi)-7~(1-methyI-1 H— pyrazol-4—yl)imidazo[1,2—a]pyridine—3-carboxamide; N-(5—(3,4-difluorobenzyicarbamoyl)-2—fluorophenyl)—7—(1methyl-1H-pyrazol-5—yl) imidazo [1,2-a]pyridine -3~carboxamide; 20 N-(5-(benzylcarba moyl)—2-fluorophenyl)—6—(1—methyl-1H-pyrazol—S-yl)imid 2— a]pyridine-3—carboxamide; uoromethyl(2-(4-methy! piperazin—fiyl) benzylcarbamoyl)phenyl)—7— (pyridineyl)imidazo[1,2—a]pyr idine—3—carboxamide; N~(5-(3,4-difluorobenzylcarbamoyl)fluorophenyl)—7-(6—(3-(dimethyl amino) propoxy) 25 pyridine—3~yl)imid azo[1,2-a]pyridine—3—carboxamide; N-(5-(2—(2,6—cis~dimethylpiperidinyl)ethy!carbamoyl)fluorophenyl)—7—(1-methyl- 1 H-pyrazol-S-yl)imidazo[1,2-a]pyridine-3—carboxamide; WO 30802 7~(4-(aminomethyl) phenyl)-N-(2-fluoro(2—(4-methylpiperazin—1 - yI)benzylcarbamoyl) phenyl)imidazo[1,2-a]pyridine—3—carboxamide; N—(5-(2-tert—butoxyethylcarbamoy1)-2—fluorophenyl)—7-(1-(2-morpholinoethyI)-1 H— pyrazolyl)imidazo[1,2-a]pyridine—S-carboxamide; N—(5—((5,5-dimethy1tetrahyd rofu yl)methy1 ca rbamoyl)—2—fluorophenyl )(1 -(2— morpholinoethy1)-1H—pyrazol—4-yl)imidazo [1 ,2—a]pyridine~3-carboxamide; N—(Z—fluoro—5-(2—(4—methylpiperaziny|)benzylcarbamoyl)phenyl)(6~methoxy pyridine—3—yl)imidazo [1 yridine—S—carboxamide; N-(Z—fluoro—5-(2—(4—methylpiperazin—1-yl)benzy10arbamoyl)phenyl)—6—(1-methyl—1 H— 1O pyrazol-5—yl)pyrazol0[1,5-a]pyridine-3—carboxamide; 1~(2-(4-fluoro(7—(pyridine—S-yl)imidazo [1 ,2—ajpyridine carboxamido)benzamido)ethy1)—2,6-cis-dimethyl dine; N—(5-(2—tert-butoxyethylcarbamoyl)—2—fluorophenyl)(6—(3-(dimethylamino) y)pyridine—3-y|)pyrazolo[1,5-a]pyridine-3—carboxamide; 15 N—(2—methyl(2~(4-methylpiperazinyl)benzylcarbamoyl)phenyl)-7—(pyridine yl)imidazo[1,2-a]pyridine—3—carboxamide; 1—methyl(2-((6—methyI(7-(1-methyl -1H—pyrazol-S—y1)imidazo[1,2-a]pyridine carbox amido)nicotina mido)methy1)phenyl)piperazine; 7-(1-Methyl-1 H-pyrazol—4—yl)—imidazo[1,2—a]pyridinecarboxylic acid {5-[2—(2,6-cis— 20 dimethyI-piperidinyI)-ethylcarbam oyl ]fluoro-phenyl}-amide; N-(5-(2—tert-Butoxyethylcarbamoyl)—2-fl uorophenyl)—7—(6—(3—(dimethylamino)propoxy) pyridin-S—yl)imidazo[1,2—a]pyridine—3—carboxamide; N—(5-(3,4—Difluorobenzylcarbam0y1)—2-fluorophenyl)—7-(6-(2~(pyrrolidin—1 —yl)ethoxy) pyridinyl)imidazo[1,2-a]pyridine-S-carboxamide; 25 6-(1-Methyl—1 H-pyrazol—S—y1)—N—(2—methyl(2—(4-methylpiperazin-1 — yl)benzylcarbamoyl) phenyl)pyrazolo[1,5-a]pyridine-3—carboxamide; N-(2-Bromo—5-(2—(4-methy1piperazin-1~yl)benzylcarbamoyl)pheny|)imidazo[1 ,2— a]pyridine—3-carboxamide; N-(2—Bromo—5-(2—(4—methylpiperazin-1 —yl)benzyicarbamoyl)phenyl)pyrazolo[1,5- 3O a]pyridinecarboxamide; N-(5—(3,4-Difluorobenzylcarbamoy1)-2—fluoroph enyl )—7-(3-hyd roxymethyl imidazo[1,2-a]pyridinecarboxamide; N~(5-(3,4—Difluorobenzylcarba moy1)-2—fl uorophenyl)—7—(3-fluoro—4~(2—(piperidin—1~ yl)ethylcarbamoy|)phenyl)imidazo[1,2-a]pyridine—3—carboxamide; 35 N—(5-(3,4-Difluoro benzylcarbamoyl)—2—fluorophenyl)(3-fluoro(2-(tetrahyd ro-2H- pyran-4~yl)ethylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3-carboxamide; PCT/182012/054501 N-(5-(3,4-Difluorobenzylcarbamoyl)-2—fluorophenyl)(3-f|uoro—4-(3-morpholino propylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3—carboxamide; 6-(1-Methyl-1 H-pyrazoI-S-yl)—N-(2-methyl—5—(2-(4—methylpiperazin yl)benzylcarbamoyl)pyridinyl)pyrazolo[1,5-a]pyridine-S—carboxamide; N-(5-((5,5~Dimethyitetrahyd rofuran—Z-yl )methylcarbamoyl)—2—fluorophenyl)—7—(6~(4- methylpiperaziny|)pyridin-3—yl)imidazo[1,2—a]pyridine-3—carboxamide; N—(5—(3,4—DifluorobenzylcarbamoyI)—2—fl uoropheny!)—7-(1~(3-(dimethyl amino)pr0pyl)— 1 H-pyrazoI—4-yl)imidazo[1 ,2—a]pyridine—3-carboxamid e; N~(5-((5,5-Dimethyltetrahyd rofuran-Z-yl lcarbamoyI)—2-fluorophenyl)—7—(5- 1O ((tetrahydro—2H-pyranylamino)methyl)pyridin-B-yl)imidazo[1,2-a]pyridine—3- carboxamide ; (S)-N-(5-(((5,5-Dimethyltetrahydrofuran-Z—yl)methyl) carbamoyl)—2-flu0r0phenyI)—7—(5- (((Z—fiuoroethyl) amino)methyl)pyridin—3-yl)imidazo[1,2-a}pyridine—S—carboxamide ; (R)—N—(5—(((5,5-dimethyltetrahydrofuran-2—yl)methyl) carbamoyl)fluorophenyl)—7-(5- 15 (((2-methoxyethyl) (methyl)amino)methyl)pyridinyl) imidazo[1,2—a] pyridine carboxamide ; (R)—7—(5-((tert—butylamino)methyl )pyridin-3—yl)-N-(5—(((5, 5-dimethyltetrahydrofura n~2- yl l)carbamoyl)-2—fluorophenyl) imidazofl ,2—a1pyridinecarboxamide ; N-(5-(3,4-DifluorobenzylcarbamoyI)-2—fluorophenyl)(6-(2—(pyrrolidinyl)ethoxy) 20 pyridin-B-yl)imidazo[1,2-a]pyridine—3—carboxamide ; N—(5-((2~(tert—Butoxy) ethyl)oarbamoyl)—2—fluoro phenyl)—7—(6-(2—(pyrrolidin—1 -yl) ethoxy)pyridin—3-yl) imidazo[1,2—a] pyridine~3—carb0xamide ; N—(5~(((5,5-dimethyl tetra hyd rofuran-Z-y|)methyl)ca rba moyl)—2~fluorophenyl)—7~(6—((1 - methyl din—4—yl)oxy) pyridin-3—yl)imidazo[1,2—a]pyridine—S-carboxamide ; 25 6-(1—Methyl—1 H—pyrazoI-S—yI)—N-(2—m ethyl—5-(2—(4—methyl piperazin—1 — yl)benzylcarbam0yl) phenyl)pyrazolo{1,5-a]pyridine—3—carboxamide ; N~(2-Bromo—5-(2—(4~methylpiperazin—1-yl)benzylcarbamoyl)phenyl)imidazo[1 ,2— a]pyridinecarboxamide; N-(2-Bromo—5-(2—(4—m ethylpiperazin-1 -yl)benzylcarbam oyl)phenyl ol0[1 ,5- 30 dine—3-carboxamide; N-(5-(3,4-Difluorobenzylcarbamoyl)-2—fluor0phenyl )(3—hyd roxy—3-methyl butyl)imidazo[1,2-a]pyridinecarboxamide; 3,4—Difluorobenzylcarba moyl)—2—fluorophenyl)-7—(3-f|uoro—4-(2—(piperidin—1— yl)ethylcarbamoyl)phenyl)imidazo{1,2—a]pyridine—3~oarboxamide; 35 N-(5-(3,4—Difluor0benzylcarbamoyl)fl enyl)—7-(3-fluoro(2-(tetrahyd ro-2H— pyran—4-yl)ethylcarbamoyl)pheny|)imidazo[1,2-a]pyridine—3—carboxamide; N-(5-((2-(2,2-Dimethylpyrro|idin-1—yl)ethyl)carbamoy|)—2—f|uorophenyl)(1-methy|— 1 H—pyrazol~4—yl)imidazo[1,2—21]pyridine—3—carboxamide; 2-(2.2—Dimethylpyrroiidiny1)ethylcarbamoyl)fluorophenyl)—7-(3—fluoro—4— ((1 R,2R)—2—hydroxycyclohexylcarbamoyl)phenyl)imidazo[1 ,2—a]pyridine carboxamide; N-(5-(2—(2,2—Dimethylpyrrolidin—1—yl)ethylcarbamoy1)—2—fl uorophenyl)—7—(3—f|uoro—4-(1 - y—Z-methylpropanylcarbamoyl)phenyl)imidazo[1,2—a]pyridine—3— carboxamide; N-(5—(2-(2,2-Dimethylpyrrolidin—1-yl)ethylcarbamoyl)—2-methylpyridinyl)~6—(3—fluoro— 1O 4-(1—hydroxy—Z—methylpropan-Z-ylcarbamoyl)phenyl)pyrazolo[1,5-a]pyridine—3- carboxamide; N—(5-(3,4-Difluorobenzylcarbamoyl)—2—fluoropheny!)—7-(3-f|uoro—4-(3-morpholino propylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide; N-(5-(2-(2,6-cis-Dimethylpiperidin—1-yl)ethylcarbamoyl)fluorophenyl)—7-(3-fluor0 15 (1-hydroxy-2~methy|propan-Z—ylcarbamoyl)pheny|)imidazo[1,2-a]pyridine-3— carboxamide; 7—(3—FIuoro(2-fluoroethylca rbamoyl )phenyl)—N-(2-fluoro(2—(4-methylpiperazin yl)benzylcarbamoyl)phenyl)imidazo[1,2—a]pyridine-3—carboxamide; N-(2-FIuoro—5-(2—(4-methylpiperazin—1-yl)benzylcarbamoyl)phenyl)(3-fluoro—5-(2— 20 hydroxy ethylcarbamoyl)phenyl)imidazo[1,2-a]pyridinecarboxamide; N-(5—(2—(2,2—dimethy|pyrrolidin—1~yl)ethylcarbamoyl)~2—methylpyridin-3—yl)—6—(1 — methyl—1 H—pyrazoI—4—y1)pyrazolo[1,5-a]pyridine—3—carboxamide; (S)-N—(Z—Fluoro—5-(2-(2-(methoxy methyl) pyrrolidinyl)ethyl carbamoyl)phenyi)—7— (1-me’thyl—1H-pyrazol—4-yl)imidazo[1,2~a]pyridine—3~carboxamide; 25 N—(2-F1uor0-5—((2—(3-propylpyrrolidin-1 -y|)ethy1)carbamoyl)phenyl)—7~(1-methyl—1 H- I-4—yl)imidazo [1 ,2—a}pyridinecarboxamide; (R)—N-(2-Fluoro-S-((2~(2—(methoxymethyl) pyrrolidinyl)ethyl) carbamoyl)phenyI) (1-methyl-1H-pyrazol—4—y1)imidazo[1,2-a]pyridine—3-carboxamide; N-(5-((2-(3,5-Dimethylpiperidin-1~yl)ethyl)carbamoyl)—2—fluorophenyI)-7—(1-methy|—1 H— 30 pyrazolyl)imidazo[1,2-a]pyridine-S-carboxamide; uoro—5-((2-(2,2,6,6—tetra methylpiperidin—1-yl)ethy!)carbamoyl)phenyl)—7-(1 - methyl—1 H-pyrazolyl)imidazo[1,2—a]pyridine—3-carboxamide; N—(5—((2—(tert—butyl(methyl)amino)ethy| )carbamoyl)—2-fluorophenyl)—7-(1—methyl—1 H- pyrazol-5—yl)imidazo[1,2-a]pyridine—3-carboxamide; 35 (2-(2,2-dimethylpyrrolidin—1-y|)ethyl)carbamoyl)—2-methylpyridinyl)(1- methyl—1 H-pyrazoi~5-yl)pyrazolo[1 ,5-a]pyridine—S—carboxamide; N-(5-((2-(butyl (ethyl)amino)ethyl)carbamoyI)fluor0 phenyt)(3-fluoro-4—((2- hydroxy carbamoyl)phenyl)imidazo{1,2-a1pyridinecarboxamide; 7-(3-fluoro-4—((1-hydroxy-2—methylpropan—2-yl)carbamoyl)phenyl)-N¥(2-fluoro-5—((2- (3-propylpyrrolidiny|)ethyl)carbamoyl)phenyl)imidazo[1,2-a]pyridine carboxamide; (2—(3,3-dimethylmorpholino)ethyl)carbamoyt)-2—fluorophenyl)-7—(3-fluoro((1 - hydroxy-2—methylpropan-Z-yl)carbamoyt)phenyl)imidazo[1 ,2—a]pyridine-3— carboxamide; (R)(3-fluoro—4-((1—hydroxy—2—methytpr0panyt)carbamoyt)phenyl)-N—(2-tluoro-5— 1O ((2-(2—(methoxymethyl)pyrrolidiny|)ethyl) carbamoyl)phenyl)imidazo[1,2—a]pyridine- 3—carboxamide; N-(5-((3,4—Difluorobenzyt)carbamoyt)f|uorophenyl)—7-(6—((2— (dimethylamino)ethyl)carbamoyl)pyridin-B-yl)imidazo[1,2-a}pyridinecarboxamide; N-(5—((2-(2,2-Dimethylpiperidin-1 -y|)ethyl)carbamoyl)methylpyridinyl)(1 - 15 methyl-1 H-pyrazol-4—yl)pyrazolo[1 ,5-a]pyridinecarboxamide; N-(5-((2-(2,6-cis-Dimethylpiperidin~1 -y|)ethyl)carbamoyt)-2—methylpyridin—3-yl)—6—( 1 - methyl-1H-pyrazo|y|)pyrazo|o[1 ,5-a]pyridine-3—carboxamide; N-(5-((2-(v(28,3R)-2,3-diethylazetidinyt)ethyl) carbamoyl)-2—fluorophenyl)—7-(1 - methyl-1H-pyrazol-4—yl)imidazo[1,2-a]pyridinecarboxamide; 20 N-(5-(3,4-Difluorobenzylcarbamoyl)f!uorophenyl)—7-(6-(((2—hydroxyethyl) (methyl)amino)methy|)pyridin—B—yl)imidazo[1,2—a]pyridinecarboxamide ; N-(5—((3,4—difluorobenzyl)carbamoyl)fluorophenyl)—7-(6-((methyl (phenethyl)amino)methyl)pyridin-B-yl)imidazo[1,2-a]pyridinecarboxamide; N-(5—((3,4-Difluoro )carbamoyl)f|uorophenyt)—7-(6- 25 ((methyl(phenethyl)amino)methyl)pyridin-3—yl)imidazo[1,2—a]pyridinecarboxamide; N-(5—((3,4—Difluoro benzyl)carbamoyl)—2—fluorophenyl)—7-(5-((methylamino) methyl)pyridin—3-yl)imidazo[1,2-a]pyridine-S-carboxamide; 7-(5-((Cyclohexyl amino)methy|)pyridin-3—yl)-N—(5-((3,4—difluorcbenzyl) carba moyl)~2- fluoro phenyl) o[1,2-a]pyridine—3—carboxamtde; and 30 N-(5—((3,4-Difluoro benzyl)carbamoyl)-2—fluorophenyl)—7~(5-(((2—methoxyethyt) (methyl)amino)methyl)pyridinyl)imidazo[1,2—a]pyridinecarboxamide; or a pharmaceutically acceptable salt thereof.
10. The compound of claim 1, which is 35 N—(5—((2—(2,2—Dimethylpyrrolidin-1—y|)ethyl)carbamoyl)-2—fluorophenyl)—7-(1-methyl- azoI-4—yt)imidazo[1,2—a]pyridine-3—carboxamide or a pharmaceutically acceptable salt thereof.
11. The compound of claim 1, which is N-(5-(2-(2,6-cis-Dimethylpiperidinyl)ethylcarbamoyl)-2—fluorophenyl)(1-methyl— azolyl)imidazo[1,2-a]pyridinecarboxamide or a pharmaceutically acceptable salt thereof.
12. The compound of claim 1, which is N-(5-(2—(2,2—dimethylpyrrolidin-1—yl)ethy|carbamoyl)methylpyridinyl)—6—(1- methyl-1H-pyrazol—4—yl)pyrazolo[1,5~a]pyridine—3-carboxamide or a pharmaceutically 1O acceptable salt thereof.\
13. The compound of claim 1, which is 7-(1-Methyl-1H-pyrazolyl)—imidazo[1,2—a]pyridine-3—carboxylic acid {5—[2-(2,6-cis- dimethyl-piperidinyl)-ethylcarbamoyl uoro-phenyl}-amide or a 15 pharmaceutically acceptable salt thereof.
14. The compound of claim 1, which is N-(5-((2-(2,2-dimethylpyrrolidin—1-y|)ethyl)carbamoyl)—2-methy|pyridinyl)(1— methyl-1H-pyrazol-S-yl)pyrazolo[1,5-a]pyridine—3-carboxamide or a pharmaceutically 20 acceptable salt thereof.
15. The compound of claim 1, which is N—(5-(2-(2,6-cis—Dimethylpiperidin—1-yl)ethylcarbamoyl)—2—fluorophenyl)-7—(3—fluoro—4- (1—hydroxy—2—methylpropanylcarbamoyl)phenyl)imidazo[1,2—a]pyridine-3— 25 carboxamide or a pharmaceutically acceptable salt thereof.
16. A pharmaceutical composition, sing: a therapeutically effective amount of the compound ing to any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, and 30 one or more pharmaceutically acceptable carriers.
17. A pharmaceutical combination, comprising: a therapeutically effective amount of the compound according to any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, and 35 a second active agent.
18. Use of a nd ing to any one of claims 1 to 15, or a pharmaceutically able salt thereof, in the manufacture of a medicament for the treatment of a disorder or disease mediated by the PDGF receptor.
19. Use of a compound according to any one of claims 1 to 15, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of pulmonary arterial hypertension.
20. The compound according to claim 1, substantially as herein described with 1O reference to any one of the Examples thereof.
21. The compound according to any one of claims 1 to 15, substantially as herein described. 15
22. The pharmaceutical composition according to claim 16, substantially as herein described.
23. The ceutical combination according to claim 17, substantially as herein described.
24. The use according to claim 18 or 19, substantially as herein described.
NZ621436A 2011-09-01 2012-08-31 Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension NZ621436B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161530049P 2011-09-01 2011-09-01
US61/530,049 2011-09-01
US201261680119P 2012-08-06 2012-08-06
US61/680,119 2012-08-06
PCT/IB2012/054501 WO2013030802A1 (en) 2011-09-01 2012-08-31 Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension

Publications (2)

Publication Number Publication Date
NZ621436A NZ621436A (en) 2015-10-30
NZ621436B2 true NZ621436B2 (en) 2016-02-02

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012303674B2 (en) Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
JP7208142B2 (en) Tyrosinamide derivatives as RHO kinase inhibitors
JP6976953B2 (en) Spiroheptane salicylamide as a ROCK inhibitor and related compounds
JP5369302B2 (en) Pyridopyrimidinediones as PDE4 inhibitors
AU2014356583B2 (en) New octahydro-cyclobuta (1,2-c;3,4-c&#39;)dipyrrol-2-yl
MX2015004151A (en) Gdf-8 inhibitors.
AU2016352813A1 (en) Novel pyrazolo pyrimidine derivatives
BR112020000284A2 (en) 5-membered heterocyclic and bicyclic amides as rock inhibitors
KR20150092173A (en) Phenylethylpyridine derivatives as pde4-inhibitors
WO2014146493A1 (en) Acyclic cyanoethylpyrazolo pyridones as janus kinase inhibitors
TW201811766A (en) N-(pyridin-2-yl)pyridine-sulfonamide derivatives and their use in the treatment of disease
CN112752757B (en) Tyrosine amide derivatives as RHO-kinase inhibitors
KR20230157432A (en) Heterocyclic Derivatives as Janus Kinase Inhibitors
JP6353899B2 (en) Inhibitors of leukotriene production
KR101803866B1 (en) 5-benzylaminomethyl-6-aminopyrazolo[3,4-b]pyridine derivatives as cholesteryl ester-transfer protein(cetp) inhibitors useful for the treatment of atherosclerosis
JP2020502252A (en) Derivatives of quinoline as inhibitors of DYRK1A and / or DYRK1B kinase
KR102611539B1 (en) GSK-3 inhibitor
TW202334167A (en) Fused tetracyclic quinazoline derivatives as inhibitors of erbb2
NZ621436B2 (en) Bicyclic heterocycle derivatives for the treatment of pulmonary arterial hypertension
CA3212872A1 (en) Indoline derivatives as ddrs inhibitors
CA3202134A1 (en) Dihydrofuropyridine derivatives as rho- kinase inhibitors
JP2024510849A (en) Tetrahydrothienopyridine derivatives as DDR inhibitors