NZ619776B2 - Heteroaryl compounds as 5-ht4 receptor ligands - Google Patents

Heteroaryl compounds as 5-ht4 receptor ligands Download PDF

Info

Publication number
NZ619776B2
NZ619776B2 NZ619776A NZ61977612A NZ619776B2 NZ 619776 B2 NZ619776 B2 NZ 619776B2 NZ 619776 A NZ619776 A NZ 619776A NZ 61977612 A NZ61977612 A NZ 61977612A NZ 619776 B2 NZ619776 B2 NZ 619776B2
Authority
NZ
New Zealand
Prior art keywords
oxadiazolyl
piperidinyl
chloro
isopropyl
indazole
Prior art date
Application number
NZ619776A
Other versions
NZ619776A (en
Inventor
Gopinadh Bhyrapuneni
Venkateswarlu Jasti
Pradeep Jayarajan
Ramasastri Kambhampati
Abdul Rasheed Mohammed
Ramakrishna Nirogi
Sriramachandra Murthy Patnala
Srinivasa Rao Ravella
Jyothsna Ravula
Praveen Kumar Roayapalley
Original Assignee
Suven Life Sciences Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Suven Life Sciences Limited filed Critical Suven Life Sciences Limited
Priority claimed from PCT/IN2012/000011 external-priority patent/WO2013042135A1/en
Publication of NZ619776A publication Critical patent/NZ619776A/en
Publication of NZ619776B2 publication Critical patent/NZ619776B2/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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • 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
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Abstract

Provided are substituted oxadiazole derivative compounds, of the general formula (I), wherein the variables are as defined in the specification. Examples of the compounds include 6-Chloro-8-{5-[1-(2-methoxy-ethyl)-piperidin-4-yl]-[1,3,4]oxadiazol-2-yl}-chroman-5-ylamine and 1-Isopropyl-3-[5-(2-piperidin-1-yl-ethyl)-[1,3,4]oxadiazol-2-yl]-1H-indazole oxalate salt. The compounds are 5-HT4 receptor ligands. The compounds may be useful in the treatment of neurological disorders such as attention deficit hyperactivity disorder, Alzheimer’s disease, cognitive disorders, dementia and schizophrenia. idin-1-yl-ethyl)-[1,3,4]oxadiazol-2-yl]-1H-indazole oxalate salt. The compounds are 5-HT4 receptor ligands. The compounds may be useful in the treatment of neurological disorders such as attention deficit hyperactivity disorder, Alzheimer’s disease, cognitive disorders, dementia and schizophrenia.

Description

HETEROARYL‘ COMPOUNDS AS 5-HT4 RECEPTOR LIGANDS Field of Invention The present inVention relates to novel compounds of formula (I) and their pharrnaceutically acceptable salts and compositions containing them, for treatment Of ' various disorders that are related to 5-HT4 receptors. ound of the Invention -HT4 receptor has been officially recognized (Humphrey et al., 1993) and identified in a variety Of tissues across many species (for review see Ford & Clarke, 1993). 5-HT4 or modulators (e.g., Iagonists and antagonists) are found to be useful for the ent of a variety of diseases such as gastroesophageal reflux disease, disease, ' gastrointestinal gastric motility disorder, non-ulcer dyspepsia, functional dyspepsia, irritable bowel syndrome, _ constipation, dyspepsia, esophagitis, gastroesophageral disease, nausea, central nervous system diseases, cognitive disorders, dementia, attention deficit hyperactivity disorder, schizophrenia and cardiovascular disorders such as cardiac failure and heart arryhthmia (Corsi.M etal., cological analysis of 5-hydroxytryptamine effects on electrically stimulated human isolated urinary r, Br;J.Pharmacol. 1991, 104(3), 719-725; Waikar.M.V et al., Evidence for an inhibitory 5-HT4 receptor in urinary bladder of rhesus and lgus monkeys, Br.J.Pharrnacol. 1994, 111( 1), 8; Anthony P. D. W. Ford et al., The 5- HT4 Receptor, Med. Res. Rev. 1993, 13(6), 633-662; Gary W. Gullikson et al., Gastrointestinal motility responses to the S and R enantiomers of zacopride a S-HT4 agonist and 5-HT; antagonist, Drug Dev. Res.‘ 1992, 26(4), 405-417; n.A.J et al., A like or in human right atrium, Naunyn-Schmiedeberg's Arch.‘ Pharmacol. 1991, 344(2), 150-159)., Patent publications WO97/17345A1, US20060194842, US20080207690, US20080255113 and 0269211 disclosed some 5-HT4 receptor compounds.
While some 5-HT4 receptor ligands have been disclosed, and there still exists a need and scope to discover new drugs with novel chemical structures for treatment of disorders affected by 5-HT4 or. It is an object of the present invention to provide a novel compound and/or a process for ation of a compound and/or a compound prepared by said method and/or a pharmaceutical composition and/or use of a compound in the manufacture of medicament for the treatment of diseases related to 5- HT4 receptors and/or use of a compound for the manufacture of a medicament for treatment of a disorder of central nervous system related to or affected by the 5-HT4 receptors. It is a further alternative object to at least provide the public with a useful choice.
Summary of the Invention The t ion relates to novel 5-HT4 ligand compounds of the formula (I), N n A N O wherein, Ar Cl Y m N is NH2 or ; R1 R1 R1 A N N is , or ; is point of ment; R1 is alkyl, R3-O-R3 or p R2; R2 is cycloalkyl or heterocyclyl, and optionally substituted with en, alkyl or -CO-OR3; R3 is alkyl; “Y” is C or O; “m” is an integer ranging from 0 to 1; with proviso when m is 0 then R1 is cycloalkyl or heterocyclyl; “n” is an integer ranging from 0 to 2; “p” is an integer ranging from 0 to 1; or a pharmaceutically acceptable salt thereof.
In one embodiment the compound is ed from the group consisting of: 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chroman- -ylamine; 1-isopropyl{5-[1-(3-methoxy propyl) piperidinyl]-[1,3,4]oxadiazolyl}- 1H-indazole; 3-[5-(1-cyclobutyl-piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl- 1H-indazole; 6-chloro[5-(3-cyclobutylaza bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazol yl] chromanyl amine; 8-Aminochloro-2,3-dihydro benzo[1,4]dioxanyl)-[1,3,4]oxadiazol- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester; -Chloro{5-[1-(tetrahydro pyranyl) piperidinyl]-[1,3,4]oxadiazol yl}-2,3-dihydro benzofuranyl amine; 6-Chloro[5-(1-cyclopentyl-piperidinylmethyl)-[1,3,4]oxadiazolyl]- chromanylamine; 6-Chloro[5-(3-isopropylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazol yl]-chromanylamine; 6-Chloro[5-(3-cyclobutylmethylaza-bicyclo[3.1.0]hexyl)- [1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazol yl}-chromanylamine; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]- [1,3,4]oxadiazolyl}-chromanylamine; -Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3- dihydro-benzofuranylamine; -Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzofuranylamine ro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3- dihydro-benzo[1,4]dioxinylamine; ro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazol yl}-2,3-dihydro-benzo[1,4]dioxinylamine; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}- 2,3-dihydro-benzo[1,4]dioxinylamine; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]- [1,3,4]oxadiazolyl}-2,3-dihydro-benzo[1,4]dioxinylamine; -Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]- [1,3,4]oxadiazolyl}-2,3-dihydro-benzofuranylamine; 4-[5-(4-Aminochloro-2,3-dihydro-benzofuranyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester; 1-Isopropyl{5-[3-(3-methoxy-propyl)aza-bicyclo[3.1.0]hexyl]- [1,3,4]oxadiazolyl}-1H-indazole; 3-[5-(3-Cyclobutylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl] pyl-1H-indazole; 3-[5-(3-Cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]- 1-isopropyl-1H-indazole; 3-[5-(3-Cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazol yl]isopropyl-1H-indazole; 1-Isopropyl{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]- [1,3,4]oxadiazolyl}-1H-indazole; 1-Isopropyl{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazol yl}-1H-indazole; 1-Isopropyl[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-1H-indazole.
The present invention relates to use of a therapeutically effective amount of compound of a (I), to manufacture a medicament in the treatment of various disorders that are related to 5-HT4 receptors.
Specifically, the compounds of this invention are useful in the treatment of various disorders such as attention t hyperactivity er, alzheimers disease, cognitive disorders, dementia or schizophrenia.
In another aspect, the ion relates to pharmaceutical compositions containing a therapeutically effective amount of at least one compound of formula (I), and their pharmaceutically acceptable salts thereof, in admixture with pharmaceutical acceptable excipient.
In still another aspect, the invention relates to the methods of treatment by using compounds of formula (I).
In yet another aspect, the invention further relates to the process for preparing compounds of a (I) and their pharmaceutically acceptable salts.
Representative compounds of the present invention include those specified below and their pharmaceutically able salts. The present invention should not be ued to be limited to them. 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chroman ylamine hemi fumarate; 6-Chloro[5-(1-cyclobutyl piperdinylmethyl)-[1,3,4]oxadiazolyl]-chromanyl amine L(+)-tartarate salt; ro[5-(1-cyclobutyl piperdinyl )-[1,3,4]oxadiazolyl]-chromanyl amine; 1-Isopropyl{5-[1-(3-methoxy propyl) piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole oxalate salt; 3-[5-(1-Cyclobutyl-piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole L(+)-tartarate salt; 6-Chloro[5-(3-cyclobutylaza o[3.1.0]hexyl)-[1,3,4]oxadiazolyl] chromanylamine oxalate salt; 4-[5-(8-Aminochloro-2,3-dihydro benzo[1,4]dioxanyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester oxalate salt; -Chloro{5-[1-(tetrahydro pyranyl) piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro benzofuranyl amine oxalate salt; 6-Chloro{5-[1-(2-methoxy-ethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman ylamine; ro{5-[1-(3-methyl-butyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman ylamine; 6-Chloro[5-(1-cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-chroman ylamine; 6-Chloro[5-(1-cyclopropylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-chroman ylamine; 6-Chloro[5-(1-isopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman- -ylamine; 6-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo [1,4]dioxinylamine; 6-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo [1,4]dioxinylamine; 6-Chloro[5-(1-cyclopentyl-piperidinyl)-[1,3,4]oxadiazolyl]-chroman ylamine; 6-Chloro[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-chromanylamine; 4-[5-(5-Aminochloro-chromanyl)-[1,3,4]oxadiazolyl]-[1,4']bipiperidinyl-1'- carboxylic acid ethyl ester; 6-Chloro[5-(3-piperidinyl-propyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-cyclopentyl-piperidinylmethyl)-[1,3,4]oxadiazolyl]-chroman ylamine oxalate salt; 6-Chloro[5-(3-isopropylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]- chromanylamine e salt; 6-Chloro[5-(3-cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazol yl]-chromanylamine oxalate salt; 6-Chloro[5-(3-cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazol yl]-chromanylamine; ro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}- chromanylamine oxalate salt; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazol yl}-chromanylamine oxalate salt; -Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzofuranylamine oxalate salt; -Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzofuranylamine oxalate salt; 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo [1,4]dioxinylamine oxalate salt; ro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzo[1,4]dioxinylamine oxalate salt; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzo[1,4]dioxinylamine oxalate salt; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazol 3-dihydro-benzo[1,4]dioxinylamine oxalate salt; -Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazol yl}-2,3-dihydro-benzofuranylamine oxalate; 4-[5-(4-Aminochloro-2,3-dihydro-benzofuranyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester oxalate; 3-[5-(1-Cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole; 1-Isopropyl{5-[1-(2-methoxy-ethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole; 3-[5-(1-Cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl[5-(1-isopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-1H-indazole; 3-[5-(1-Cyclopropylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole; ropyl{5-[1-(3-methyl-butyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole; 3-[5-(1-Cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 3-[5-(1-Cyclopentyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl{5-[3-(3-methoxy-propyl)aza-bicyclo[3.1.0]hexyl]- [1,3,4]oxadiazolyl}-1H-indazole oxalate salt; 3-[5-(3-Cyclobutylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl- azole oxalate salt; 3-[5-(3-Cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl] isopropyl-1H-indazole e salt; 3-[5-(3-Cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl] pyl-1H-indazole oxalate salt; 1-Isopropyl{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazol yl}-1H-indazole oxalate salt; 1-Isopropyl{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole oxalate salt; 1-Isopropyl[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-1H-indazole oxalate; 1-cyclobutyl piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole oxalate salt.
In another aspect the invention relates to the process for preparation of a nd of formula (I) as defined above, which comprises: (a) coupling the compound of formula (1) with compound of formula (2) in presence of dehydrating agent to form a compound of formula (I), wherein all substitutions are as defined above, (b) optionally converting the compound of formula (I) to their pharmaceutically acceptable salts.
In another aspect the invention relates to a compound of a (I) when prepared by the process above.
In another aspect the invention relates to the process for preparation of a compound of formula (I) as defined above, which comprises: - 7A - (a) coupling the compound of formula (1) with compound of a (2), in presence of suitable solvent to form a compound of formula (4), (b) cyclizing the compound of formula (4) to form a compound of formula (I), wherein all tutions are as defined above, (c) optionally converting the compound of formula (I) to their pharmaceutically acceptable salts.
In another aspect the invention relates to a compound of formula (I) when prepared by the processes above.
In another aspect the invention relates to a pharmaceutical composition comprising a compound of formula (I) and pharmaceutically acceptable excipients.
In another aspect the invention relates to use of a compound of formula (I) in the manufacture of medicament for the treatment of diseases related to 5-HT4 receptors.
In another aspect the invention s to use of a nd of formula (I) in the manufacture of a ment for treatment of a disorder of l nervous system related to or affected by the 5-HT4 receptors.
Detailed Description of the Invention Unless otherwise stated, the following terms used in the specification and claims have the meanings given below: - 7B - The term “alkyl” means ht chain or branched hydrocarbon radical consisting solely of carbon and en atoms, containing no unsaturation, having from one to three carbon atoms, and which is attached to the rest of the molecule by a single bond. Exemplary “alkyl” groups include methyl, ethyl, n-propyl, iso-propyl and the like.
The term “cycloalkyl” means non-aromatic mono cyclic ring of 3 to 8 carbon atoms. Exemplary “cycloalkyl” groups include cyclopropyl, cyclobutyl, cyclopentyl and the like.
The term “heterocyclyl” means non-aromatic mono cyclic ring of 2 to 7 carbon atoms, whose ring structures include 1 to 3 heteroatoms, these additional atoms may be repeated more than once in ring. Exemplary “heterocyclyl” groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl and the like.
The phrase "pharmaceutically able salts" tes that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, the mammal being treated therewith.
The phrase "therapeutically effective amount" is d as an amount of a compound of the present invention that (i) treats the particular disease, condition or disorder (ii) eliminates one or more symptoms of the particular disease, condition or disorder (iii) delays the onset of one or more symptoms of the particular disease, condition or disorder bed herein.
Commercial reagents were ed without further purification. Room temperature refers to 25 - 40 oC. Unless otherwise stated, all mass spectra were d out using ESI conditions. 1H-NMR spectra were recorded at 400 MHz on a Bruker instrument. Deuterated chloroform, methanol or ylsulfoxide was used as solvent.
TMS was used as internal reference standard. Chemical shift values are expressed in parts per million (d) values. The ing abbreviations are used for the multiplicity for the NMR signals: s=singlet, bs=broad singlet, d=doublet, let, q=quartet, qui=quintet, h=heptet, ble doublet, dt=double triplet, tt=triplet of triplets, m=multiplet. tography refers to column chromatography performed using 100 - 200 mesh silica gel and executed under nitrogen pressure (flash chromatography) conditions. - 7C - Unless the context clearly requires ise, throughout the description and the claims, the words “comprise”, “comprising”, and the like, are to be construed in an inclusive sense as d to an exclusive or exhaustive sense, that is to say, in the sense of “including, but not limited to”.
The reference to any prior art in the specification is not, and should not be taken as, an acknowledgement or any form of suggestion that the prior art forms part of the common general knowledge in New Zealand.
Pharmaceutical compositions In order to use the compounds of formula (I) in therapy, they will ly be formulated into a pharmaceutical ition in accordance with standard pharmaceutical practice.
The pharmaceutical compositions of the present invention may be formulated in a conventional manner using one or more pharmaceutically acceptableexcipient. The ceutically acceptable excipient is carrier or diluent. Thus, the active nds of the invention may be formulated for oral, intranasal or parenteral (e.g., intravenous, intramuscular or subcutaneous). Such pharmaceutical compositions and processes for preparing same are well known in the art (The Science and Practice of Pharmacy, D.B.
Troy, 21st Edition, Williams & Wilkins, 2006).
The dose of the active compounds can vary depending on factors such as the route of administration, age and weight of patient, nature and severity of the disease to be treated and r factors. Therefore, any reference herein to a pharmacologically - 7D - effective amount of the compounds of general formula (I) refers to the aforementioned .- factors.
: Methods of Preparation The compounds of formula (I) can be prepared by Scheme 1 & Scheme II as shown below 'SchemeI .- O N\NH2 /N__. n 7N\ 0 o + HQ > ——~——» ~ 0 (I) (,2) - In above Scheme I, all symbols are as defined above.
The compound of formula (1) is coupled with nd of formula (2) using dehydrating agent to form compound of formula (I). The dehydrating agent is ed from group consisting of aluminium phosphate, calcium oxide, cyanuric chloride, N,N'- dicyclohexylcarbodiimide, Il) chloride, ormic acid, phosphorus pentoxide or phosphoryl chloride and more preferably selected dehydrating agent is oryl chloride.
The compounds of forrnula (l) and formula (2) maytbe prepared'by using preparations 1 to 9 or commercially available or can be prepared by conventional methods or by modification, using known process.
WO 42135 Schemell .
H o o ”\an a H‘ 0 N\NH o 0 (l ) ’ . (4) _1 (2 In above Scheme II, all symbols are as defined above; The compound of formula (1) is coupled with compound of formula (2) in suitable solvent to form nd of formula (4). The compound of al(4) is cyclized in presence of atingflagent to form compound of formula (I).
In the first step of the above preparation, the solvent is selected from group consisting of ethanol, tetrahydrofuran, dichloromethane, dichloroethane, toluene, dimethylformamide, yl ide, 1,4-dioxan, tetrahydrofuran, triethylamine, toluene, pyridine, ethyl acetate, dichloromethane and the like or a mixture thereof and more preferably selected solvents are dichloromethane and triethylamine.
In the secOnd step of the above preparation, the dehydrating is selected from group consisting of aluminiumphosphate, calcium oxide, cyanuric chloride, N,N'- dicyclohexylcarbodiimide, iron(III) chloride, 'orthoformic acid, phosphorus pentoxide or phosphoryl chloride and more preferably selected dehydrating agent is phosphoryl chloride.
The compounds of formula (1) and formula (2) may be prepared by using preparations 1 to 9 or commercially ble or can be prepared by conventional methods or by modification, using known s.
If necessary, pharmaceutiCally acceptable salts for compounds of formula (I) may be prepared conventionally by on with the appropriate acid or acid derivative.
Suitable phannaceutically acceptable salts will be apparent to those d in the art and include those described in J. Pharm. Sci., 1977, 66, 1-19, such as acid addition salts formed with inorganic acids 6. g. hydrochloric, hydrobromic, ic, nitric or phosphoric acid and organic acids e.g., succinic, maleic, acetic, fumaric, citric, ,malic, tartaric, benzoic, p—toluic, p- toluenesulfonic, benzenesulfonic , acid, methanesulfonic or naphthalenesulfonic acid. The most red salts of compounds of formula (I) are oxalate, tartarate,‘ fumarate, methane sulfonate, hydrochloride and sulfate. Based on the clinical development of the compound we will select the salt form of the compound and effective dose. e salt is most preferable salt for the free base compound of Example 3 and Example 4. Fumarate salt is most preferable salt for the free base nd of Example 1. From free base compounds of Examples 1-74, the person skilled in art can easily prepare all preferred salts of this invention based on the clinincal devolpment of the compound.
Examples The novel compounds of the present invention were prepared according to the following experimental procedures, using appropriate materials and conditions.
Preparation 1: Preparation of 5-Aminochloro chromancarboxylic acid hydrazide Step (i): Preparation of Methyl 4-aminohydroxy benzoate To a stirred solution of 4-aminosa1icylic acid (50 grams, 326.7 mmol) in methanol (375 mL) at 0 °C was added trated sulfuric acid (99.7 m_L, 1.87 mmol) maintaining temperature of the reactiOn below 20°C. The reaction mixture was gradually heated to reflux and upon tion of the reaction after 6 hours it was cooled to ice bath temperature and ed with aqueous sodium hydroxide solution (10.0 N, 2145 _mL). The white precipitate that formed was filtered, washed with water, ether and dried under vacuum to obtain Methyl 44aminohydroxy benzoate (50.70 grams).
Yield: 93%. 1H - NMR (DMSO-dé): 8 10.76 (bs, 1H), 7.43 (d, J = 8.6 Hz, 1H), 6.13 (bs, 2H), 6.10 (dd, J = 8.6, 2.0 Hz, 1H), 5.99 (d, J =_ 2.0 Hz, 1H), 3.79 (s, 3H); Mass (m/z): 168 .
Step (ii): Preparation of Methyl 4-acetylamino—2-hydroxy benzoate A on of Methyl 4-aminohydroxy benzoate (50.7 grams, 303.6, mmol, ed in above step) in ethyl acetate (750 mL) was added to a stirred solution of water (250 mL) and sodium bicarbonate (34.9 grams, 415.5 mmol) cooled at 0 °C followed by acetyl chloride (29.7 mL, 415.5 mmol) over a period of 15 minutes. The reaction mixture was gradually warmed to room temperature and stirred‘for 2 hours.
The two layers were separated and the organic layer was washed with brine, dried over anhydrous sodium sulphate and the solvent was removed under reduced re to obtain methyl ylamino-2—hydroxy benzoate (63.5 grams).
Yield: 99 %. 1H -' NMR (CDCI3): 5 10.86 (bs, 1H), 7.78 (d, J = 8.6 Hz, 1H), 7.23 (s, 1H), 7.16 (bs, 1H), 7.10 (d, J = 8.6 Hz, 1H), 6.13 (bs, 1H), 3.92 (s, 3H), 2.19 (s, 3H); Mass (m/z): 208 (M—H)+.
Step (iii): Preparation of methyl 4-acetylaminochlorohydroxy benzoate To a stirred solution of methyl 4—acetylaminohydroxy benzoate (61.4 grams, 294.0 mmol, obtained in above step) in dichloroethane (1.2 ,L) was added N- chlorosuccinimide (58.8 grams, 441 mmol) and the reaction mixture was refluxed for 3 hours. The volatiles were removed under reduced pressure; the solid compound thus precipitated was diluted with water (1.0 L) and filtered. The crude product was diluted with a 1:9 e (methanol and dichloromethane)‘ and washed with brine. The organic layer was dried over anhydrous sodium. sulphate and the volatiles were removed under reduced pressure to obtain methyl 4-acetylamino-5jchlorohydroxy benzoate (67.7 grams).
Yield: 94.6 %. lH - NMR (DMSO-d5)3 5 10.49 (bs, 1H), 9.47 (s, 1H), 7.75 (s, 1H), 7.72 (s, 1H), 3.85 (s, 3H), 2.16 (s, 3H); Mass (m/z): 244, 246 (M+H)*.
Step (iv): Preparation of methyl _ 4-Acetylamino-S—chloro-Z-(prop-Z-ynyloxy) henzoate I €1.1- To a stirred solution-of methyl 4-acetylamino—5-chloro—2-hydroxy benzoate (30 grams, 123.2 mmol, obtained in above step) in dimethylformamide (246 mL) was added potassium carbonate (42.5 grams, 308 mmol). The reaction mixture was cooled to 0 °C and propargyl bromide (22.3 mL, 150.3 mmol) was added Over a period of 15 minutes. The reaction e was warmed to room ature and stirred for 5 hours before being dumped in ice cold water. The solids precipitated were filtered and the crude product was dissolved in a 1:9 mixture.(methanol:dichloromethane) and washed with brine solution. The organic layer was dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure to obtain the title compound (25.2 grams).
Yield: 73 %. 1H - NMR (DMSO-de): 8 9.60 (s, 1H), 7.91 (s, 1H), 7.76 (s, 1H), 4.82 (s, .ZH)’ 3.77 (s, 3H), 3.61 (s, 1H), 2.15 (s, 3H); Mass (m/z): 282, 284 (M+H),+.
Step (v): Preparation of methyl S-acetylamino-6—chloro-2H—chromene—S- carboxylate A stirred solution of methyl 4-acetylamino-5—chloro—2—(propynyloxy) benzoate (25 grams, 88.8 mmol, obtained in above step) in dowtherm A (127 mL) was heated to 220 °C for 3 hours. The reaction mixture was cooled to 60-70 °C and dumped - in hexane. The Solids precipitated were filtered and washed with hexane to obtain methyl 5-acetylamino—6—chloro-2H-chromene-S—carboxylate (16.2 grams).
Yield: 64.8 %. - i lH - NMR dfi): 8 9.77 (s, 1H), 7.58 (s, 1H), 6.42 (d, J = 10.1 Hz, 1H), 6.04 (m, 1H), 4.83 (s, 2H), 3.78 (s, 3H). 2.06 (s, 3H); Mass (m/z): 282, 284 (M+H)+.
Step (vi): Preparation of methyl 5—acetylamino—6-ehloro chr0mancarboxylate To a solution of methyl S-acetylaminochloro-2H-chromenecarboxylate (20.5 grams, 72.9 mmol, obtained in above step) in ethanol (300 mL) was added Pd/C (10% w/w, 8.6 grams). The en gas re was applied using balloon pressure.
The reaction mixture was stirred at room temperature for 5 hours and filtered h a pad of celite. The e was concentrated to dryness to obtain methyl 5-acetylamino- 6V-chloro ncarboxylate (18.88 grams).
Yield; 91.3 %. 1H - NMR (DMSO-dé): 5 9.65 (s, 1H), 7.55 (s, 1H), 4.16 (t, J = 4.5 Hz, 2H), 3.76 (s, 3H), 2.58 (t, J = 6.3 Hz, 2H), 2.05 (s,i 3H), 1.87 (m, 2H); Mass (m/z): 284, 286 (M+H)+.
Step (vii): Preparation of S-Aminoi6-ch10ro chroman-s-carboxylic acid To methyl ylamino46fchloro chromancarboxylate (18.88 grams, 66.6 mmol, obtained in above step), aqueous sodium hydroxide solution (1.4 N, 475 mL) was added and the reaction mixture was refluxed for 6 hours. The reaction mixture was ed with 2N hydrochloride at 0 °C and the precipitated product was filtered and dried under vacuum to yield. S-Amino—6-chloro chromancarboxylic acid (14.07 granis).
Ykkt92998 lH - NMR ds): 8 11.8 (bs, 1H), 7.48 (s, 1H), 5.74 (bs, 2H), 4.09 (t, J = 4.6 Hz, 2H), 2.43 (t, J = 6.4 Hz, 2H), 1.91 (m, 2H); Mass (m/z): 228, 230 (M+H)+.
. Step (viii): Preparation of methyl S-amin-bch10ro chroman-S-carboxylate To a stirred solution of 5-amino—6-chloro chromancarboxylic' acid (13.5 grams, 59.34 mmol, obtained in above step) in methanol (68 mL) cooled at 0 °C, cenc sulphuric acid (18.10 mL) was added drop wise. The reaction mixture was gradually warmed to room ature and stirred for 4 hours; The reaction mixture was cooled to 0 °C, diluted with water (202 mL) and basified with sodium hydroxide (10 M, 57.9 mL). The product that precipitated was filtered and dried under vacuum to obtain methyl 5-aminochloro chroman-Si-carboxylate (10.5 . lI-I - NMR (CDCl3)I 5 7.75 (s, 1H), 4.37 (bs, 2H), 4.24 (t, J = 5.0 Hz, 2H), 3.83 (s, 3H), 2.49 (t, J = 6.6 Hz, 2H), 2.10 (m, 2H); Mass (m/z): 242, 244 (M+H)+.
Step (ix): Preparation of 5-Aminochloro chroman-S-carboxylic acid hydrazide To a stirred solution of methyl 5-aminochloro chromancarboxylate (10.0 grams, 41.4 mmol, obtained in above step) in ethanol (82 mL), hydrazine hydrate (31.05 mL) was added. The reaction temperature was gradually increased to reflux and the reaction e was stirred at this temperature for 5 hours. The volatiles were removed under d pressure, the crude mass was dissolved in 10 % methanol in ' Idichloromethane and washed with water, brine, dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure to obtain 5-Amino—6-chloro- chroman-8~carboxylic acid hydrazide (9.3 grams).
Yield: 93 %. . 1H - NMR (DMSO‘dé): 5 8.85 (bs, 1H), 7.56 (s, 1H), 5.59 (bs, 2H), 4.43 (bs, 2H), 4.18 (t, J = 4.8sz, 2H), 2.45 (t, J = 6.5 Hz, 2H), 1.93 (m, 2H); Mass (m/z): 242, 244 (M+H)+.
Preparation 2: ation of methyl l-isopropyl-lH-indazolyl ylate Step (i): Preparation of methyl lH—indazol-S-yl carboxylate To a stirredvsolution of indazolecarboxilic acid (80.5 grams, 0.497 mmol, obtained in above step) in methanol (2 L) cooled at 0 °C was added thionyl chloride (120 mL, 1.59 mmol) over a period of 1 hour. The reaction temperature'was gradually ' . raised and the reaCtion mixture was refluxed for 5 hours. The volatiles were removed and the crude mass was d with dichloromethane, washed with aqueous sodium bicarbonate, dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure to obtain the title compound (80.2 grams).
Yield: 92 %. 1H - NMR (CDC13): 5 13.2 (bs, 1H), 8.23 (d, J = 8.2 Hz, 1H), 7.86‘(d, J = 8.4 Hz, 1H), 7.48 (t, J = 7.4 Hz, 1H), 7.35 (t, J = 7.6 Hz, 1H), 4.09 (s, 3H); Mass (m/z): 177 (M+H)+.
Step (ii): Preparation of methyl l-isopropyl-1H—indazolyl ylate To a d solution of methyl 1H-indazolyl carboxylate (80.0 grams, 0.454 mmol, obtained in above step) in dry‘dimethylformamide (500 mL) at'O °C, sodium hydride (60 % in l oil, 23.7 grams, 0.592 mmol) was added portion wise over a period of 30 minutes. The reaction mixture was gradually warmed to room temperature and stirred for 45’ minutes before cooling it again to 0 °C. To the 'reaction, isopropyliodide (55 .mL, 0.545 mmol) was added and was stirred atiroom temperature for 4 hours. The reaction mixture was poured into crushed ice, stirred for 10 minutes and extracted with ethyl acetate (2x250 mL). The ed organic, layer was washed with water (2 x 500 mL), brine, dried over anhydrous sodium sulphate and the solvent was d under reduced pressure to get the crude mass which was purified by silica gel column to obtain methyl 1-isopropyl-1H-indazol-3—yl ylate (40.0 grams).
Yfiddz409fi 1H - NMR (CDC13)I 5 8.24 (d, J = 8.1 Hz, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.43 (t, J = 7.2 Hz, 1H), 7.,31(t J= 7.6Hz, 1H), 4.96(m, 1H), 4.,04(s 3H), l.,66(d J= 6.7Hz, 6H); Mass (m/z): 219 (M+H).
Step (iii): Preparation of l—isopropyl-lH—indazol—3—yl carboxylic acid hydrazide To a stirred solution of methyl l-isopropyl-lH-indazolyl carboxylate (40.0 grams, 183.5 mmol, obtained in above step) in ethanol at room temperature hydrazine hydrate (130 mL, 2.56 mmol) was added. The reaction mixture was refluxed for 2 hours. The volatiles were removed under reduced pressure and the crude mass was diluted with dichloromethane, washed with water, brine, dried over anhydrous sodium sulphate and the solvent was removed under reduced re to obtain the title compound (37.52 grams). ' 3 Yield: 93 %. . lH - NMR (CDC13)I 5 8.35 (d, J = 8.1 Hz, 1H), 8.16 (bs, 1H), 7.47 (d, J = 8.4 Hz, 1H), , 7.41 (t, J = 7.0 Hz, 1H), 7.28 (t, J = 7.4Hz, 1H), 4.87 (m, 1H), 4.09 (s, 3H), 1.60 (d, J = 6.6 Hz, 6H); Mass (m/z): 219 (M+H)+.
Preparation 3: Preparation of o-S-ehloro—2.,3-dihydr0 benzofuran carboxylic acid hydrazide Step (i): Preparation of methyl 4-amino—S-chloro-2,3-dihydro Ibenzofu'ran - carboxylate To a stirred solution of 4-amino-5—chloro-2,3-dihydr0 benzofuran-74carboxylic aeid (Chem. Pharm. Bull. 1998, 46(1), 42-52; 3.93 g, 18.4 mmol) in methanol (36.8 mL), cooled at 0 °C, thionyl chloride (6.0 mL) was'added. The reaction mixture was gradually warmed to room temperature and was heated to reflux for '2 hours. The volatiles were removed under reduced re; the crude mass was diluted with s sodium bicarbonate solution and was extracted with ethyl e. The combined organic layer was dried over anhydrous sodium sulphate and the t was removed under vacuum to obtain methyl ochloro-2,3-dihydro benzofuran carboxylate (3.89 grams). Yield: 92.9 % lH - NMR (DMSO-d5)2 8 7.43 (s, 1H), 6.06 (bs, 2H), 4.60 (t, J = 8.8 HZ, .68 (s, 3H), 2.97 (t, J = 8.8 Hz, 2H); ' Mass (m/z): 228.0, 230.1 (M+H)+. -15.‘ Step (ii): Preparation of o—5—chloro—2,3-dihydr0 benzofurancarboxylic acid hydrazide To a stirred solution of methyl 4-aminochloro-2,3-dihydro benzofiJran ylate (3.88 grams, 17.07 mmol, obtained in the above step) in ethanol (34.1 mL), hydrazine hydrate (11.5 mL, 236.2) was added. The reaction temperature was gradually sed to reflux and the reaction mixture was stirred at this temperature for 5 hours.
The volatiles were removed under reduced pressure, the crude mass was ated with plenty of ether and pentane to obtain4-Aminochloro—2,3-dihydr0 benzofuran carboxylic acid hydrazide (3.76 grams).
Yield: 96 %. lH - NMR (DMSO-da): 5 8.35 (bs, 1H), 7.44 (s, 1H), 5.85 (s, 2H), 4.68 (t, J = 8.7 Hz, 2H), 4.43 (bs, 2H), 3.0 (t, J = 8.7 Hz, 2H); Mass (m/z): 228.0, 230.1 (M+H)+.
Preparation .4: Preparation of 8-Aminochloro—2,3—dihydro benzo[1,4]dioxane—5— carboxylic acid ide Step (i): ation of methyl 8-amin0chlor0-2,3-dihydro benzo[l,4]dioxane-5— carboxylate To a stirred solution of 8—Aminochloro—2,3-dihydro-benzo[l,4]d.i0xine—5- carboxylic acid (Journal of Medicinal Chemistry, 1993, 36, 4121; 2.2 grams, 9.58 mmol) in methanol (38.3 mL), cooled at 0 °C thionyl chloride (2.78 mL) IWas added.
The reaction mixture was gradually warmed to room temperature and then heated to reflux for 3 hours. The volatiles were removed under reduced pressure; the crude mass was diluted with aq. sodium bicarbonate solution and extracted with ethyl acetate. The ed organic layer was dried over anhydrous sodium sulphate and the t was d under vacuum to obtain the title compound (2.12 grams).
Yiled: 90.9% ‘H - NMR(CDC13): 5 7.52 (s, 1H), 4.47 (bs, 2H), 4.45 - 4.30 (m, 4H), 3.84 (s, 3H).
Mass (m/z): 244.1, 246.1 (M+H)+.
Step (ii): Preparation of 8-Amino-7—chloro-2,3-dihydro benzo[l,4]dioxane carboxylic acid hydrazide ' To a stirred solution of methyl 8-amino-7—chloro-2,3-dihydrobenzo [1,4]di0xinecarboxylate (2.1 grams, 8.6 mmol, obtained in the above step) in ethanol (34.4 mL), hydrazine hydrate (6.2 mL, 129.3 mmol) was added. The reaction temperature was gradually increased to reflux and the reaction mixture was stirred at this temperature for 5 hours. The volatiles were removed under reduced pressure, the crude mass was triturated with plenty of ether and pentane to obtain 8—aminochloro- 2,3-dihydro benzo[1,4]dioxane;5-carboxylic acid hydrazide (2.1 grams).
Yield: 100 %. 1H - NMR (DMSO-de): 5 8.80 (bs, 1H), 7.27 (s, 1H), 5.40 (bs,‘2H), 4.46 (bs, 2H), 4.40- 4.25 (m, 4H); Mass (m/z): 244.1, 246.1 (M+H)+.
Preparation 5: ation of l-cyclopropyl piperidine-4—carbonyl chloride Step (i): Preparation of opropyl dine—4—carbonitrile ‘ To a stirred solution of 1-cyclopropylpiperidone (Alfa Aesar, 3.0 grams, 21.5 mmol) in a mixture of 1, 2-dimethoxyethane (72 mL) and ethanol (2.2 mL) cooled at 0 °C, was added p-toluenesulfonylmethylisocyanide (5.45 grams, 27.95 mmol). Solid ium tertiary butoxide (5.54 grams, 49.45 mmol) was added over a period of 1 hour. The reaction e was stirred at this temperature for additional 1 hour and . gradually warmed to room temperature. Afier ng for 2 hours at this temperature, it was cooled to 0 °C, diluted with brine and ethyl acetate. The organic layer was separated, dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure to obtain crude product, which was d by slica gel column to yield opropyl piperidinecarbonitrile (1.32 grams).
Yield: 41.2 %.
‘H — NMR (CDC13): 8 2.82 (m, 2H), 2.63 (m, 1H), 2.49 (m, 2H), 1.98 - 1.78 (m, 4H), 1.70 - 1.58 (m, 1H), 0.50 - 0.40 (m, 2H), 0.40 — 0.35 (m, 2H); Mass (m/z): 151 (M+H)+.
Step>(ii): Preparation of l-cyclopropyl piperidine-4—carb0xylic acid A mixture of 1—cyclopropyl piperidinecarbonitrile (1.32 grams, 8.8 mmol, obtained in the above step) and hydrochloric acid (6 N, 35.2 mL) was refluxed for 3 hours. The volatiles were d under reduced pressure; the traces of water were ’ removed by co distilling with toluene. The crude product thus obtained was triturated ‘ with ether several times and dried under vacuum to obtained l-cyclopropyl piperidine- 4-carboXylic acid (2.02 grams).
Yield: 100 %. 2012/000011 lH - NMR (DMSO'd6): 5 12.54 (bs, 1H), 10.79 (bs, 1H), 3.50-3.40 (m, 2H), 3.18-30 (m, 2H), 2.—.78265 (m, 1H), 3.55- 3.45 (m: 1H), 2.10- 1.85 (m, 4H), 1.20— 1.10 (m, 2H), 080- 070 (m, 2H); Mass (m/z): 170 (M+H) .
Step (iii): Preparation of l-cyclopropyl piperidine—4—carbonyl chloride To a stirred mixture of 1-cyclopropyl piperidinecarboxylic acid (10.0 grams, 48.6 mmol, obtained in above step) in dichloromethane (198 mL) cooled at 0 °C was added dry dimethyl formamide (2 mL) ed by drop wise addition of oxalyl chloride (12.5 ‘mL, 145.8- mmol). The on mixture'was lly warmed to room temperature and stirred for 1 hour. The volatiles were removed under reduced pressure and the crude l-cyclopropyl piperidine—4-carbonyl ch1oride (11.0 grams). This crude product was used in the next reaction without purification.
Yeild: 100%. 1H - NMR (DMSO-d6): 8 10.66 (bs, 1H), 3.50 — 3.42 (m, 2H), 3.40 - 3.30 (m, 1H), 3.15 , -30(m 2H), 2.80- 2.65(m, 1H), 2.10- 1.80(m,4H), 115- , 2H), 0.80- 0.70 (m, 2H); Mass (m/z): 184 .
Preparation 6: Preparation of (l-cyclobutyl piperidinyl) acetic acid Step (i): Preparation of t-butyl 4-ethoxycarbonylmethylene pipcridine-l- carboxylate ' To a stirred solution of 1-Boc—4-piperidone (2.0 grams, 10.03 mmol) in benzene— (40 mL) at room temperature was added Wittig reagent (5.23 grams, 15 mmol). The reaction mixture was d for 10 hours and the volatiles were removed under reduced pressure 'to obtain a crude mass Which was purified by silica gel column chromatography to obtain t—butyl 4-,ethoxycarbonylmethylene piperidine-l—carboxylate (2.05 grams).
Yield: 76 %. lH - NMR (CDC13): 8 5.71 (5,1H), 4.16 (q, 2H), 3.55 - 3.45 (m, 4H), 2.94 (t, J = 5.7 Hz, 2H), 2.28 (t, J = 5.6 Hz, 2H), 1.47 (s, 9H), 1.28 (t, J = 7.1 Hz, 3H); Mass (m/z): 270 (M+H)+.
Step (ii): Preparation of l 4-ethoxycarbonylmethyl piperidine-l-carboxylate To a stirred solution of t-butyl - 4-ethoxycarbonylmethylene piperidine-l— carboxylate (2.05 grams, 7.62 mmol, obtained in above step) in ethanol (30 mL) at -_18- room temperature was added Pd/C (10 wt %, 600 mg). Hydrogen n pressure was applied on the reaction for 5 hours. The reaction mixture was d through a pad of celite and the volatiles were removed under reduced pressure to obtain t-buty1 ethoxycarbonylmethyl piperidine- 1-carboxy1ate (1.98 grams).
Yield: 95.8 %.
‘H - NMR (CDC13): 6 4.20-4.0(m, 4H), 2.83 - 2.65 (m, 2H), 2.23 (d, J = 6.8 Hz, 2H), 2.0 - 1.88 (m, 1H), 1.75 -:1.68 (m, 2H), 1.45 (s, 9H), 1.26 (t, J = 7.0 Hz, 3H), 1.25 - 1.05 (m, 2H); Mass (ni/z): 272 (M+H)+.
Step (iii): Preparation of piperidin—4-yl acetic acid ethyl ester To a stirred solution of t—butyl 4-ethoxycarbonylmethyl dine-l- carboxylate (1.98 grams, 7.3 mmol, obtainedin the above step) in isopropyl alchol (5 mL) cooled at 0 °C_, was added a solution of dry isopropanolic hydrogen chloride (~3 N, 15 mL). The reaction mixture was stirred at room ature for 16 hours. The volatiles were removed under d pressure and“ the crude product was triturated with ether several times, dried under vacuum to obtain piperidinyl acetic acid ethyl ester (1.57 grams) Yield: 100 %. 1H — NMR (DMSO-ds): 8 4.03 (q, 2H), 3.23 - 3.15 (m, 2H), 2.86- 2.78 (m, 2H), 2.24 (d, J = 6.8 Hz, 2H), 2.0 - 1.85 (m, 1H), 1.81 - 1.72 (m, 2H), 1.40 — 1.25 (m, 2H), 1.14 (t, J = 6.9 Hz, 3H); Mass (m/z): 172 (M+H)+. I Step (iv): Preparation of (l-cyclobutyl piperidin-4—yl) acetic acid ethyl ester A mixture of cyclobutanone‘(0.3 mL, 3.94 mmol) in acetic acid (0.19 mL, 328 mmol) was added to a d solution of piperidinyl acetic acid ethyl ester (562 mg, 3.28 mmol, obtained in above step) in dichloromethane cooled at 0 °C. Solid sodium triacetoxyborohydride (1.39 grams, 7.2 mmol) was added portion wise over a period of minutes. The reaction mixture was gradually warmed to room temperature and stirred for 16 hours. The reaction mixture was cooled to 0 °C and basified with ted sodium onate solution (pH:7.5).~ The two layers were separated, the organic layer was washed with brine, dried over anhydrous sodium sulphate and the volatiles were removed under reduced pressure to obtain (l-Cyclobutyl piperidinyl) acetic acid ethyl ester (652mg). _ 19 _ Yield: 88.3 3%. lH - NMR (CDC13): 8 4.13 (q, 2H), 2.90 - 2.82 (m, 2H), 2.75 ' - 2.62 (m, 1H), 2.22 (d, J = 6.9 Hz. 2H), 2.10 .95 (m, 2H), 1.95 - 1.80 (m, 2H), 1.80—1.60 (m, 7H), 1.35 - 1.20 (m, 2H), 1.27 (t, J = 7.1 Hz, 3H); Mass (m/Z): 226 (M+H)+.
Step (v): Preparation of (l-cyclobutyl piperidin-4—yl) acetic acid To a stirred mixture of (l-cyclobutyl piperidinyl) acetic acid ethyl ester (652.9 mg, 2.90 mmol, obtained in above step), tetrahydrofuran (6 mL) and water (6.0 mL) cooled at 0 °C lithium hydroxide drate (133 mg, 3.19 mmol) was added in a single lot. The reaction mixture was stirred at room temperature for 16 hours. The reaction mixture was cooled again to 0 °C and acidified with 2N hydrochloric acid to pH22-3.’The volatiles were removed under d pressure and the traces of water were removed by azeotropic distillation with e to obtain ( 1-cyclobutyl piperidin- 4-yl) acetic acid (747.9 mg).
Yield: 100 %. l.H - NMR (DMSO-ds): 5 12.25 (bs, 1H), 10.98 (bs, 1H), 3.56 - 3.45 (m, 1H), 3.30 - 3.20 (m, 2H), 3.1030 (m, 0.5H), 2.90 - 2.82 (m, 0.5H), 2.75 - 2.60 (m, 2H), 2.40 - 2.30 (m, 2H), 2.22 (d, J = 6.9 Hz, 1H), 2.17 (d, J = 6.8 Hz, 1H), 2.15 - 2.08 (m, 2H), 1.95 - 1.75 (m, 2H), 1.74 - 1.65 (m, 2H), 1.65 - 1.50 (m, 2H); Mass (m/z): 198 (M+H)+.
Preparation 7: Preparation of l—(3—methoxy propyl) din-4—carboxylic acid Step (i): Preparation of ethyl 1—(3-methoxy propyl) piperidinca‘rb0xylate To a stirred on of ethyl isonipecotate (22.0 grams, 140 mmol) in acetonitriie (250 mL) at room temperature was added cesium carbonate (97 grams, 298 mmol) followed by omethoxypropane (20 mL, 154 mmol) and the reaction mixture was heated to reflux for 4 hours. The reaction mixture was cooled to room temperature and filtered through a small pad of celite. The volatiles were removed under reduced pressUre to obtain ethyl 1-(3-methoxy propyl) piperidincarboxylate (31.0 grams).
Yield: 99% ' 1H - NMR ): 5 4.12 (q, 2H), 3.41 (t, J = 6.4 H2, 2H), 2.90 — 2.85 (m, 2H), 2.38 (t, J = 7.4VHz,I2H), 2.34 - 2.20 (m, 1H), 2.05 - 1.93 (m, 2H), 1.92 - 1.85 (m, 2H), 1.80 - - 1.70 (m, 4H), 1.23 (t, J = 7.1 Hz, 3H); -20_- Mass (m/z): 230 (M+H)".
Step (ii): Preparation of 1-(3-methoxy propyl) piperidincarboXylic acid To a stirred mixture of ethyl '1-(3—methoxy propyl) piperidin-4—carboxylate (33.0 grams, 144.1 mmol, obtained in the above step), tetrahydrofuran (200 mL) and water (200 mL) was added lithium hydroxide monohydrate (6.1 grams, 144.1 mmol).
The reaction mixture was stirred at room temperature for 16 hours before being diluted with cetate. The two layers were ted and the aqueous layer was acidified to pH: 3-4 with concentrated hydrochloric acid and the volatiles were removed under reduced re to obtain 1—(3-methoxy propyl) piperidincarboxylic acid (35.0 grams).
Yield: 100 %. - lH — NMR (DMSO-dé): 5 3.30 (t, J = 6.4 Hz, 2H), 3.19 (s, 3H), 2.80 2.70 (m, 2H), 2.25 (t, J = 7.5 Hz, 2H), 2.15 - 2.05 (m, 1H), 1.92 - 1.82 (m, 2H), 1.78 — 1.70 (m, 2H), 1.68 - 1.57 (m, 2H), 1.55 - 1.43 (m, 2H1; Mass (m/z): 202 (M+H)+.
Preparation 8: ation of 3-cyclobutylaza bicyclo[3.l.0]hexane-6— ~ carboxylic acid Step (i): Preparation of ethyl ylaza bicyclo[3.1.0]hexanecarboxylate To a stirred solution of ethyl 3-benzyl-2,4-dioxoaza bicyclo[3.1.0]hexane-6¥ carboxylate (SYNLETT, 1996, 1097; 5.0 grams, 18.3 mmol) in tetrahydrofuran (74 mL) cooled at 0 °C, BH3-DMS (2N solution in tetrahydrofuran 36 mL, 73.2 mmol) was added over a period of 30 minutes. The reaction temperature was gradually raised to reflux for 6 hours. After cooling the reaction mixture to 0 °C, it was quenched by adding aqueous ammonium chloride solution and was ted with ethyl acetate. The combined organic layer was dried over anhydrous sodium te and the solvent was removed under reduced pressure. The crude product was purified by silica gel column chromatography to obtain ethyl 3-benzylaza o[3.1.0]hexanecarboxylate (2.8 grams)‘ 3 Yield: 62.5% , lH - NMR (CDC13)I 5 7.40 - 7.20 (m, 5H), 4.14 (q, 2H), 3.61 '(s, 2H), 3.05 (d, J = 9.0 Hz, 2H), 2.44 (d, J = 8.7 Hz, 2H), 2.14 (t, J = 2.6 Hz, 1H), 1.97 (s, 2H), 1.28 (t, J = 7.1 Hz, 3H);.
Mass (m/z): 246.2 .(M+H)+. -2]- Step (ii): Preparation of ethyl 3-aza bicyclo[3.].0]hexane-6Qcarboxylate To a stirred on of ethyl 3-benzyl-3—aza bicyclo[3.1.0]hexane—6-carboxylate (2.0 grams, 8.1 mmol, obtained in the above step) in methanol (20 mL), palladium hydroxide (468 mg) was added. The reaction e was applied with hydrogen pressure using hydrogen balloon. The reaction mixture was stirred at room temperature for 2 hours and filtered through a small pad of celitc. The volatiles were removed under reduced pressure to obtain ethyl 3-aza bicyclo[3.1.0]hexanecarboxy1ate (1.22 grams) Yhddz96964 lH - NMR (CDC13): 8 4.11 (q, 2H), 3.11 (d, J = 11.6 Hz, 2H), 2.98 (d, J = 11.7 Hz, 2H), 2.02 (s, 2H), 1.49 (t, J = 3.0 Hz, 1H), 1.24 (t, J = 4.2 Hz, 3H); Mass (m/z): 156.1 .
Step (iii): Preparation of ethyl 3-cyclobutyl—3-aza bicyclo[3.l.0]hexane-6— ‘ I carboxylate A mixture of cyclobutanone (157 mg, 2.19 mmol) in acetic acid (0.11 mL, 1.56 mmol) was added to a stirred solution of ethyl 3-aza bicyclo[3.1.0]hexane carboxylate (243 mg, 1.56 mmol, obtained in the above step) in romethane, cooled at 0 °C. Solid sodium triacetoxy borohydride (727 mg, 3.43 mmol) was added portion wise over a period of 15 minutes. The reaction e was gradually warmed to room temperature and stirred for 16 hours. The reaction mixture was cooled to 0°C and basified with saturated sodium bicarbonate on (pH:7.5). The two layers were separated, the organic layer was washed with brine, dried over anhydrous sodium sulfate and the volatiles were removed under reduced pressure to obtain ethyl 3- cyclobutylaza bicyclo[3.1.0]hexanecarboxylate (219 mg). i Ykkk66096 lH - NMR (CDC13): 8 4.11 (q,2H), 3.10-2.90 (m, 3H), 2.34 (d, J = 8.8 Hz, 2H), 2.04 (s, 1H), 1.93 (s, 2H), 2.0 - 1.80 (m, 3H), 1.80- 1.55 (m, 3H), 1.25 (t, J = 7.1, Hz, 3H); Mass (m/z); 210.2 (M+H)T.
Step (iv): Preparation of 3-cyclobutylaza o[3.1.0]hexanecarboxylic acid To a stirred mixture of ethyl 3-cyclobutylaza bicyclo[3.1.0]hexane carboxylate (218 mg, 1.04 mmol, obtained in the above step), tetrahydrofuran (2 mL) and water (2.0 mL) cooled at 0 °C, lithium ide monohydrate (133 mg, 3.19 mmol) was added in a single lot. The reaction e was stirred at room temperature for 24 hours. The (reaction mixture .was cooled again to 0 °C and acidified with 2N hydrochloric acid’ to pH: 2-3. The volatiles were removed.under reduced pressure and the traces of water were removed by azeotropic distillation with toluene to obtain 3- cyclobutylaza bicyclo[3.1.0]hexane—6—carboxylic acid (180 mg).
Yield: 92 %.
IH - NMR (DMSO-dg): 5 2.98 - 2.86 (m, 1H), 2.78 (d, J = 8.5 Hz, 2H), 2.20 (d, J = 8.1 Hz, 2H), 1.90 -l.80 (m, 2H), 1.82 - 1.68 (m,- 2H), 1.65 - 1.55 (m, 2H), 1.49 (s, 2H), 1.42 (s, 111); Mass (m/z): 182.3 (M+H)+.
Preparation 9: atiOn of [l,4']Bipiperidinyl-4,1'-dicarboxy1ic acid -l'-ethyl ester Step (i): Preparation of ethyl 4—oxo piperidine—l-carboxylate To the d‘solution of piperidin—4-one hydrochloride (2.0 g, 14.7 mmol) in DCM (60 mL) cooled at 0 °C, was added triethylamine (5.15 mL, 36.75 mmol) and ethylchloroformate (1.59 mL, 16.6 mml). The reaction mixture was stirred at room temperaturefor 2 hours before being "diluted with water. The two layers were separated, the organic layer was dried over anhydrous sodium sulfate and the volatiles were d under reduced pressure to obtain ethyl 4—oxo—piperidine-l-carboxylate (3.14 grams).
Yield: 98 % IH *- NMR (CDC13): 5 4.22 (q, 2H), 3.79 (t, J = 6.0 Hz, 2H), 2.48 (t, J = 6.0 Hz, 2H), 1.31 (t, J = 7.1 Hz, 3H); Mass (m/z): 172.1 .
Step (ii): Preparation of [l,4']Bipiperidinyl-4,1'-dicarboxylic- acid diethyl ester A mixture of ethyl 4-oxo piperidine-l-carboxylate (3.14 grams, 18.3 mmol, obtained in the above step) in acetic acid (1.05 mL, 18.3 mmol) was added to a stirred solution of ethyl isonipecotate (2.87 mL, 18.3 mmol) in dichloromethane (10 mL) cooled at 0 °C. Solid sodium toxy borohydride”(11.6 grams, 54.9-mm'ol) was added portion wise over a period of 15 minutes. The reaction mixture was gradually warmed to room temperature and d for 16 hours. The reaction mixture was cooled to 0 °C and basified with saturated sodium bicarbonate solution (pH 7.5). The two layers were separated, the c layer was washed with brine, dried over anhydrous sodium sulfate and the volatiles were removed under reduced pressure. The crude product’was purified by silica gel column chromatography to obtain [1,4']Bipiperidinyl- I. 1 4, l'-dicarboxylic acid diethyl ester (5.51 grams).
Yield: 96.3 %. lH - NMR (CDC13): 8 5.60 - 5.10 (m, 2H), 4.35 - 4.15 (m, 1H), 4.20 - 4.08.(m, 4H), 3.98 - 3.85 (m, 1H), 3.20 - 3.10 (m, 1H), 3.10 - 3.0 (m, 1H), 2.86 - 2.710(m, 2H), 2.60 - 2.50 (m, 1H), 2.48 - 2.35 (m, 1H), 2.10 - 2.0 (m, 2H), 1.98 - 1.85 (m, 4H), 1.60 - 1.43 (m, 2H), 1.32 - 1.22 (m, 6H); Mass (m/z): 313.2 (M+H)+.
Step (iii): Preparation of [l,4']Bip_iperidinyl-4,1'-dicarboxylic acid 1'-ethyl ester To a stirred mixture of [l,4']Bipiperidinyl-4,1'-dicarboxylic acid diethyl ester (5.51 grams, 17.67 mmol), tetrahydrofuran (34 mL) and water (34 mL) cooled at 0 °C, lithium hydroxide monohydrate (742.0 mg, 17.67 mml) was added. The reaction mixture was stirred at room temperature for 16 hours, diluted with Ethyl acetate. The two layers Were separated, the s layer was acidified with 2N hydrochloric acid to pH: 3-4 ' and the volatiles were d under reduced pressure to obtain Bipiperidinyl—4,1'-dicarboxylic acid 1'-ethyl ester (5.0 grams).
Yield: 94 % 1H - NMR (DMSO-d6): 5 12.53 (bs,. 1H), 11.16 (bs, 1H), 4.15 - 3.98 (m, 4H), 3.47 - 3.35 (m, 3H), 3.0 -2.90 (m, 2H), 2.90 — 2.65 (m, 2H), 2.60 - 2.50 (m, 1H), 2.18 — 2.08 (m, 2H), 2.05 ~ 1.94 (m, 4H), 1.60 -1.50 (m, 2H), 1.16 (t, J = 7.0 Hz, 3H); Mass (m/z): 285.1 (M+H)+.
Example 1: ation of 6-Chloro[5-(1-cyclopropyl-piperidinyl)- [1,3,4]oxadiazolyl]-chroman-S-ylamine hemi fumarate :‘ Step (1): Preparation of N-(l-cyclopropyl piperidine—4-carbonyl)-N’-(5-amino chloro chroman-S-carbonyl) hydrazine To a stirred solution of 'S-aminochloro ncarboxylic acid hydrazide (8.0 grams, 33.1 mmol, ed in preparation 1) in dichloromethane (200 mL) cooled at 0 °C, was added triethyla’mine (13.9 mL, 99.9 mmol) and 'a solution of l- cyclopropylpiperidinecarbonyl chloride (11.0 grams) in dichloromethane (200 mL).
The reaction mixture was warmed to room temperature and stirred for 16 hours before diluting it with water (160 mL). The two layers were separated, the organic layer was dried over anhydrous sodium te and the les were removed under reduced pressure to obtain the title compound (10.5 grams).
Yield: 81 %.
]H- NMR (DMSO-dé): 5 10.16 (d, J: 3.2 Hz, 1H), 9.64 (d, J= 3 .2 Hz, 1H), 7.58 (s, 1H), 5.,73(bs 2H), 421 (t, J=4.,7Hz 2H),3.0- 2.88(m, 2H), 2.46(t, J=6.,5Hz 2H), 2.30 220(m,1H),2.18- 2.05 (m, 2H), 20-1.90(m 2H), 170- 1.60(m, 2H), 1.60- 142 (m, 3H), 0.42- 0.35 (m, 2H), 0.30- 0.22 (m, 2H); Mass (m/z): 393, 395 (M+H).
Step (ii): Preparation of 6-chlorc[5-(l-cy‘clopropyl piperidinyl)- [1,3,4]oxadiazol—Z-yI]—chroman-5—ylamine To N—(l—cyclopropyl piperidine—4-carbonyl)-N’-(5-aminochloro chroman-S- carbonyl) hydrazine (10.5 grams, 26.7 mmol, obtained in the above step) was added phosphoryl chloride (53.5 mL). The reaction temperature was gradually raised to 120 °C. The on mixture was stirred at this ature for 1h, cooled to room temperature and triturated with hexanes (3x100 mL). The crude reaction was diluted with 10% aqueous sodium bicarbonate on and extracted with a 1:9 mixture of methanol in romethane. The organic layer was dried over anhydrous sodium sulphate and t was removed under reduced re and the crude product was purified by silica gel column to obtain 6-Chloro—8-[5-(l-cyclopropyl piperidinyl)- [1,3,4]oxadiazolyl] chroman-S-yi amine (8.8 grams).
Yield: 87.9 %. 1H — NMR(CDC13): 5 7.66 (s, 1H), 4.35 (bs, '2H), 4.28 (t, J = 5.0 Hz, 2H), 3.18 - 3.10 (m', 2H), 3.08 - 2.93 (m, 1H), 2.53 (t, J = 6.6 Hz, 2H), 2.40 - 2.30 (m, 2H), 2.18 - 2.05 (m, 4H), 20- 187(m 2H), 170 1.60(m, 1H), 0.50 0.,40(m 4H); Mass (m/z): 375, 377 (M+H).
Step (iii): Preparation of 6-chloro[5-(l-cyclopropyl piperidinyl)— [1,3,4[0xadiazolyl] chroman-S-yl amine hemi fumarate A suspension of 6-chloro[5-(l-cycliopropyl‘ piperidinyl)-[l,3,4]0xadiazol- 2-yl] chroman-54yl amine (14 grams, 37.3. mmol, ed in the above step) in ethanol (280 mL) was heated to reflux until clear solution obtained. The mixture was cooled to room ature and fumaric acid (4.32 grams, 37.3 mmol) was added. The reaction mixture was heated to reflux for 1 hour. The volatiles were removed under reduced pressure and the furmarate salt, thus obtained, was recrystallized from isopropanol to obtain ro[5-(1-cyclopropy1 piperidinyl)-[1,3,4]oxadiazol—2-yl] chroman-S- yl amine hemi fumarate (14.0 grams).
Yield: 92.8 %. j . lH - NMR (DMSO-dg): 5 7.48 (s, 1H), 6.60 (s, 1H), 5.75 (s, 2H), 4.13 (t, J = 4.8»Hz, 2H), 3.0 - 2.90 (m, 3H), 2.52: - 2.42 (m,‘2H), 2.40 - 2.30 (m, 2H), 2.01 - 1.90 (m, 4H), 2(nn3PD,048-040(nn2PD,035-028(nn2Hx Mass (m/z): 375, 377 . I, Example 2: Preparation of ro[5-(l-cyclobutyl piperdin—4~ylmethyl)- [1,3,4loxadiazol-Z-yll-chroman-S-yl amine L(+)-tartarate salt Step (i): Preparation of 6-chlor0[5-(1-cyclobutyl piperdin-4—yl methyl)- [l,3,4]0xadiazol72-yl]-chroman-S-yl amine To the (l-cyclobutyl piperidin¥4—.yl) acetic acid (725 mg, 3.52 mmol, obtained in preparation 4) was added phosphoryl chloride (4 mL). The e was d for 15 minutes and 5-Amino—6—chlor0-chromanearboxylic acid hydrazide (500 mg, 2.0 mmol) was added. The reaction mixture was gradually heated to reflux for 30 minutes.
The reaction mixture was cooled to room temperature, triturated with hexanes (2 x 20 mL) and the crude mass was basified with s sodium bicarbonate on. The basified mixture was extracted with 10% methanol in dichloromethane.'The organic layer was dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure and was purified by silica gel column to obtain 6I—chloro[5-(l- cyclobutyl piperdinylmethy1)—[1,3,4]oxadiazol—2—yl] chroman-S-yi amine (250 mg).
Yield: 30 %.
'H-NMRaxxmy87@(sHQ435®sflfl428mJ=50HL2HL2% (m, 2H), 2.83 (d, J =_6.9 Hz,:2H), 2.73 — 2.62 (m, 1H), 2.54'(t, J = 6.6 Hz,‘ 2H), 2.20 - 2.10 (m, 2H), 2.08 - 2.0 (m, 2H),1.95 -1.65 (m, 9H), 1.48 - 1.35 (m, 2H); Mass (m/z): 403, 405 (M+H)+.
Step (ii): Preparation of 6-chloro—8—[5-(l-cyclobutyl piperdin—4—yl methyl)- [1,3,4]oxadiazolyll-chromanyl amine L(+)-tartarate salt To a stirred on of 6-chloro-8—[5-(l-cyclobutyl piperdinyi methyl)- [1,3,4]oxadiazolyl] chroman-S-yl amine (175.7 mg, 0.436 mmoi, obtained in the above step) in methanol (2 mL), L(+)-tartaric acid (65.4 mg, 0.436 mmol) was added.
The reaction mixture was stirred for 1 hour at room temperature and the volatiles were ' removed under reduced pressure to obtain a crude mass which was triturated several times with solvent ether to obtain 6-chloro[5-(1-cyclobutyl piperdinyl )~ [.1 ,3,4]oxadiazolyl] chroman-S-yl amine L(+)-tartarate (206.2 mg) ~26- Yield: 85.5 % lH - NMR (DMSO'dG): 5 7.46 (s, 1H), 5.79 (bs, 2H), 4.12 (t, J = 4.7 Hz, 2H), 4.06 (bs, 2H), 3.20 - 3.10 (m, 1H), 3.10 = 6.7 Hz, 2H), 2.48 (t, J = 7.6 - 3.0 (m, 2H), 2.84 (d, J Hz, 2H), 2.33 - 2.15 (m, 2H), 2.10 -2.0 (m, 2H), 2.0 - 1.85 (, 5H), 1.85 - 1.72 (m, 2H), 1.70 — 1.58 (m, 2H), 1.45 - 1.30 (m, 2H); Mass (m/z); 403, 405 .
Example 3:: Preparation of l-isopropyll{S-[1-(3-methoxy propyl) piperidinyll- [‘1,3,4]oxadiazolyl}-lH-indazole oxalate salt- Step (1): Preparation of l-isopropyl{5-[1—(3-methoxy propyl) piperidinyl]- ]oxadiazol-Z-yl}-lH—indazole To the mixture of l—isopropyl-1H-indazolecarboxylic acid hydrazide (15.0 grams, 68.8 mmol) and 1-(3-Methoxy propyl)-piperidine-4—carboxylic acid hydrochloride (20.9 grams, 88.2 mmol, ed in preparation 7) cooled at 0 °C was added phosphoryl chloride (130. mL). The reaction temperature was gradually raised to 100 °C and. stirred was 2 hours. Upon completion of the reaction, it was cooled to 0 °C and triturated with hexanes (3 x 250 mL). The crude product was basified with aqueous sodium hydroxide solution and extracted with 5% ol in dichloromethane. The combined organic layer was dried over’anhydrous sodium sulphate and the solvent was removed under reduced pressure. The crude product was purified by silica gel column tography to obtain l-isopropyl{5-[l-(3—methoxy propyl) diny1]- [1,3,4]oxadiazolyl}-1H-indazole (15.78 grams) Yield: 59 %‘, 1H - NMR (CDCl3): 5 8.35 (d, J = 8.1 Hz, 1H), 7.53 (d, J = 8.5 Hz, 1H), 7.47 (t, J = 7.0 Hz, 1H), 7.33 (t, J = 7.4 Hz, 1H), 5.05-4.90 (m, 1H), 3.44 (t, J = 6.4 Hz, 2H), 3.35 (s, 3H), 3.15-2.97 (m, 3H), 2.48 (t, J = 7.3 Hz, 2H), 2.26-2.02 (m, 6H), 1.88-1.75 (m, 2H), 1.67 (11,] = 6.7 Hz, 6H); Mass 384.5 (M+H)+.
Step (ii): Preparation of l-Isopropyl-3—{5—[l-(3-methoxy-propyl)-piperidinyll- [l,3,4]oxadiazolyl}-lH-indazole oxalate salt ' To a stirred on of 1-isopropyl—3-{5-[1-(3—methoxy propyl) piperidin-4.-yl]- [l,3,4]oxadiazolyl}-1H-indazole (12.55 grams, 32.7 mmol, obtained in the above step) in 2-propanol (200 mL); oxalic acid (4.12 grams, 32.7 mmol) was added. After stirring at room temperature for .1 hour the reaction was further diluted with 2-propanol .
F27- and refluxed for 2 hours. The crystalline product which was precipitated after cooling the on mixture to room temperature was filtered, dried under vacuum to obtain 1- isopropyl{5-{l-(3-methoxy propyl). piperidinyl]-[1,3,4]oxadiazol-2—yl}-1H- indazole oxalate salt (16.4 grams) ' Yield: 88 % lH - NMR (DMSO-de): 8 8.18 (d, J =,8.l Hz, 1H), 7.90 (d, J = 8.5 Hz, 1H), 7.54 (t, J;= 7.4 Hz, 1H), 7.38 (t, J = 7.7 Hz, 1H), 5.23 - 5.10 (m, 1H), 3.50 - 3.40 (m, 3H), 3.37 (t,J = 5.9 Hz, 2H), 3.23 (s, 3H), 3.10 -2.96 (m, 4H), 2.35 - 2.25 (m, 2H), 2.18—2.02 (m, 2H), 1.94 - 1.85 (m, 2H), 1.53 (d, J = 6.6 Hz, 6H); Mass (m/z): 384.3 (M+H)+.
Example 4: Preparation of 3-[5-(l-cyclobutyl-piperidin-4—yl )- [1,3,4]oxadiazol—2-yl]—l-isopropyl-'lH-indazole L(+)-tartarate salt Step (i): Preparation of 3-[5¥(1-cyclobutyl-piperidinyl methyl)-[1,3,4]oxadiazol- ‘ I 2—yl]isopr0pyl—lH—indazole To the mixture of l-isopropyl—lH-indazolecarboxylic acid hydrazide (120 mg, 0.55 mmol) and lobutyl piperidin-4~yl) acetic acid hydrochloride (147 mg, 0.74 mmol, obtained in preparation 6) cooled at 0 °C, wasadded oryl de (1.5 mL). The reaction temperature was gradually raised to 100 °C‘and stirred was 2 hours. Upon completion of the reaction, it was cooled to 0 °C and triturated with hexanes (3 x 25.mL). The crude product was cooled to 0 °C, basified with aqueous sodium hydroxide solution and extracted with 5% methanol in dichloromethane. The combined organic layer was dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure. The crude t was purified by silica gel column chromatography to obtain 3-[5-(l—cyclobutyl piperidin—4—yl methyl)-[l,3,4]oxadiazol yl]-l—isopropyl-lH-indazole (62 mg) Yield: 30 % ‘H - NMR(CDC13): 5 8.37 (d, J = 8.1Hz, 1H), 7.54 (d, J = 8.5 Hz, 1H), 7.48 (t, J = 6.9 Hz, 1H), 7.35 (t, J = 7.5 Hz, 1H), 5.08-4.92 (m, 1H), 2.93 (d, J = 7.1 Hz, 2H), 2.92-2.87 (m, 2H), 2.74-2.62 (m, 1H), 2.10-1.93 (m, 3H), 1.92-1.82 (m, 4H), 1.80-1.65 (m, 4H), 1.68 (d, J = 6.7 Hz, 6H), 1.52—1.40 (m, 2H); Mass (m/z): 380.2 (M+H)+.
Step (ii): ation of 3-[5-(1-cyclobutyl piperidin-4—yl methyl)-[l,3,4]oxadiazol— l-isopropyl—lH-indazole L(+)-tartarate salt To a stirred solution of 3-[5-(1-cyclobutyl piperidinyl )- 4]oxadiazolyl]isopropyl-1H-indazole (62 mg, 0.16 mmol, obtained inthe above step). in 2-propanol (5.0 mL), L(+)-tartaric acid (26 mg, 0.16 mmol) was added.
After stirring at room for 1 hour the volatiles were d under reduced pressure and the crude product was triturated several times with ether to obtain 3-[5-(1- cyclobutyl piperidinyl )-[1,3,4]oxadiazol-yl]-l-isopropyl-il H-indazole L(+)- tanannesak(811ng) ‘ Yfiddz9496 1H - NMR ds): 5 8.18 (d, J = 8.1 Hz, 1H), 7.90 (d, J = 8.5 Hz, 1H), 7.54 (t, J = 14H;1HL7380“V=16HL1HL522-SJOUmlH)4Jl($2HL330-3200m 2H), 3.20 - 3.05 (m, 2H), 3.0 (d, J = 6.8 Hz, 2H), 2.45 - 2.30 (m, 1H), 2.10 - 1.90 (m, 4HL190-1800m2HL1J8-165fim2HL150-1400m2Hx Mass (m/z): 380.2 (M+H)+.
Example 5: Preparation of 6-chloro—8-[5-(3-cyclobutylaza bicyclo[3.].0]hex—6— yl)—[1,3,4]oxadiazol-2_—yl] nesiylamine oxalic acid Step (i): Preparation of 6-chloro[5-(3-eyclobutylaza bicyclo[3.].0]hexyl)— ' " [1,3,4]oxadiazolyl] chroman-S-yl amine To 3-cyclobutylaza bicyc‘lo[3.1.0]hexane-.6-carboxylic acid (74 mg, 0.40 mmol, obtained in ation 8) was added phosphoryl chloride (1 mL). The mixture was stirred for 15 minutes and 5-amino—6-chloro chroman-8—carboxylic acid hydrazide (80 mg, 0.33 mmol) was added. The reaction mixture was gradually heated to reflux for 1 hour. The reaction mixture was cooled to room temperature, triturated with hexanes (2 x 20 mL) and the crude mass was basified with aqueous sodium bicarbonate solution. The basified mixture was extracted with 10 % ol in dichloromethane The organic layer was dried over anhydrous sodium sulphate and the solvent was removed under reduced pressure and was purified by silica gel column to obtain 6- . chloro[5-(3-cyclobutylaia bicyclo[3. 1 .0]hexyl)-[ l ,3,4]oxadiazolyl] chroman-S-yl amine (18 mg).
Yield: 14.0 %.> 1H - NMR‘(CDC13): 6 7.64 (s, 1H), 4.33 (bs,l2H), 4.27 (t, J = 5.3 Hz, 2H), 3.20 - 3.0 mL3HL270-2600m1H)2530“P=64HL2HL248-2350m2H)220-2J0 . (m, 4H), 2.0 - 1.90 (m, 2H), 1.90 - 1.80 (m, 1H), 1.80 - 1.60 (m, 2H), 1.30 — 1.20 (m, 1H) ' WO 42135 Mass (m/z): 387.1, 389.2 (M+H)+ Step (ii): Preparation of 6¥chloro[5-(3-cyclobutyl—3-aza bicyclo[3.l.0]hex-6—yl)- [1,3,4loxadiazolyl] chroman-S-yl amine e salt To a stirred solution of 6-ch10ro[5—(3-cyclobutylaza o[3.l.0]hex yl)-[1,3,4]oxadiazolyl] chroman—5-yl amine (18 mg, 0.05‘mmol, obtained in the above step) in 2-propanol (3 mL), oxalic acid (6.0 mg, 0.05 mmol) was added. After stirring at room temperature for 1 hour the reaction was further diluted with 2-propanol and refluxed for 2 hours. The volatiles were removed under reduced pressure and the crude product which was obtained was triturated with ether, dried under vacuum to obtain 6-Chloro—8-[5-(3-cyclobutyl-3 ~aza o[3.1.0]hex—6-yl)—[1,3,4]oxadiazol yl] chroman-5;y1 amine oxalate salt (21.2 mg) Yield: 95.6% 7 1H - NMR (DMS'O-da): 5 7.47 (s, 1H), 5.80 (bs, 2H), 4.28 (t, J = 5.3 Hz, 2H), 3.80 - "3.55 (m, 2H), 3.30—3.20 (m, 1H), 2.70 - 2.60 (m, 1H), 2.60 4 2.40 (m, 4H), 2.30 - 2.05 (m, 4H), 2.0 - 1.90 (m, 3H), 1.90 - 1.70 (m, 2H), 1.30 - 1.20 (m, 1H); Mass (m/z): 387.1, 389.2 (M+H)+.
Example 6; Preparation of 4—[5-(8-Aminochloro—2,3-dihydro l,4]dioxan- -yl)~[l,3,4]oxadiazolyl]-[1,4']bipiperidinyl-l'-carb0xylic acid ethyl ester oxalate salt Step (i): Preparation of 4-[5-(8-Amino-7—chloro-2,3-dihydro benzo[l,4]dioxan yl)—[1,3,4]oxadiazolyl]-[1,4']bipiperidinyl—l'-carboxylic acid ethyl ester To the [1,4']bipiperidiny1—4,1'—dicarboxy1ic acid 1'-ethyl ester (372 mg,'1.02 mmol, obtained in preparation 9) was added phosphoryl chloride (3.2 mL). The mixture was stirred for 15 minutes and 8-amino—7-chlor0-2,3-dihydro benzo[1,4]dioxane carboxylic acid ide (200 mg, 0.82 mmol) was added. The reaction mixture was gradually heated to reflux for 1 hour. The reactiOn mixture was cooled to room temperature, ated with hexanes (2 x 50 mL) and; the crude mass was basified with . aqueous sodium bicarbonate solution. The basified mixture was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulphate and the t was removed under reduced pressure and was purified by silica gel column to obtain 4- [5-(8-aminochlbro-2,3—dihydro * benzo[l,4]dioxanyl)-[1,3 ,4]oxadiazoly1]— bipiperidinyl-l'—carboxylic acid ethyl ester (1 10 mg).
Vfidd221598 lH - NMR (CDC13)Z 8 7.42 (s, 1H), 4.50 — 4.36 (m, 6H), 4.33 - 4.20 (m, 2H), 4.12 (q, 2H), 3.03 - 2.92 (m, 3H), 2.83 - 2.70 (m, 2H), 2.55 — 2.42 (m, 1H), 2.42 - 2.30 (m, 2H), 2.15 > - 2.06 (m, 2H), 2.04 - 1.90 (m, 2H), 1.86 - 1.78 (m, 2H), 1.55 - 1.40 (m, 2H), 1.26 (t, J _= 7.1 112,311); , , Mass (m/z): 492.1, 494.3 (M+H)+ Step (ii): Preparation of 4-[5-(8-aminochloro-2,3-dihydro benzo[1,4]dioxin yl)-[l,3,4]oxadiazoly|]-[l,4']bipiperidinyl-l'~carboxylic acid ethyl ester oxalate salt To a stirred solution of 4—[5-(8-aminochloro-2,3-dihydro 1,4]dioxin yl)-[1,3,4]oxadiazol—2-y1]—[1,4']bipiperidinyl-l'-carboxylic acid ethyl ester (100 mg, 0.20 mmol, obtained-in the above step) in ethanol (3 mL), oxalic acid (23 mg, 0.18 mmol) was added. Afier stirring at room temperature for 1 hour the reaction was further diluted with 2-propanol and refluxed for 2 hours. The volatiles were removed under reduced pressure and the crude product obtained was triturated with ether and dried under vacuum to obtain 4-[5-(8-aminochloro-2,3-dihydro benzo[1,4]dioxiny1)4 [1,3,4]oxadiazol—2-yl]-[1,4']bipiperidinyl-1‘—carboxy1ic acid ethyl ester oxalate salt (115 mg) ..
Yield: 97.4 % IH - NMR (DMSO-ds): 8 7.29 (s, 1H), 5.66 (bs, 2H), 4.33 (s, 4H), 4.15 - 4.05 (m, 2H), 4.03 (q, 2H), 3.40 - 3.15 (m, 4H), 3.10 - 2.90 (m, 2H), 2.90 - 2.70 (m, 2H), 2.26 - 2.18 (m, 2H), 2.08 - 1.90 (m, 4H), 1.58 -l.42 (m, 2H), 1.17 (t, J = 7.0 Hz, 3H); Mass (m/z): 492.1, 494.3 (M+H)+.
Example 7: Preparation of 5-Chloro—7-{5-[1-(tetrahydro pyranyl) piperidin ,3,4]oxadia_zolyl}-2,3—dihydro uranyl amine oxalate salt Step (i): Preparation of 5-Chloro{5-[1-(tetrahydro pyran—4-yl) piperidin-4—yl]- [l,3,4]oxadiazolyl}-2,3-dihydro uranyl amine To 1-(tetrahydro pyrany1) piperidinecarboxylic acid (168.2 mg, 0.58 mmol) was added phosphoryl de (1.76 mL). The mixture was stirred for 15 minutes and 4-aminOiS-chlorol-ZJ-dihydro benzofurancarboxylic acid ide (101.2 mg, 0.0.44 mmol, obtained in preparation 3) was added. The reaction mixture was gradually heated to reflux for 2 hours. The reaction mixture was cooled to room temperature, triturated with hexanes (2 x 20 mL) and the crude mass was basified with s sodium bicarbonate solution. The basified mixture was extracted with 10 % .'31" methanol in dichloromethane. The organic layer was dried over anhydrous sodium sulphate and the t was d under reduced pressure. The residual mass was purified by silica gel column to obtain 5—chloro—7-{5-[1—(tetrahydro pyranyl) . din—4-yl]-[l,3,4]oxadiazolyl}-2,3-dihydro benzofuranyl amine (23.5 mg).
Yield: 13.1 %.
Il-l - NMR (CDCls); 8 7.64 (s, 1H), 4.84 (t, J = 8.7 Hz, 2H), 4.31 (bs, 2H), 4.10 - 4.0 (m, 2H), 3.39 (t, J é 11.4 Hz, 2H), 3.10 (t, J '= 8.7 Hz, 2H), 3.10 - 2.95 (m, 3H), 2.62 - 2.50 (m, 1H), 2.45 -, 2.25 (m, 2H), 2.20-1.95 (m, 4H), 1.88 - 1.75 (m, 2H), 1.75 - 1.60 (rh, 2H); ' Mass (m/z): 405.2, 407.4 (M+H)+.
Step (ii): ation of 5-chloro—7-{5--[1--(tetrahydro pyranyl) piperidinyl]- [1,3,4]oxadiazolyl}--2,3-dihydro benzofuran-4—yl amine oxalate salt To a stirred solution of 5-chloro {5——[1--(tetrahydro pyran-yl) piperidinyl]- [1,3,4]oxad1azolyl}-2,3-d1hydro benzofuranylamine (20.4 mg‘, 0.05 mmol) in l (2 mL), oxalic acid (6.0 mg, 0.05 mmol) was added. After stirring 'at room temperature for 1 hour the reaction was further diluted with 2-propanol and refluxed for 2 hour; The volatiles were removed under reduced pressure and the crude product obtained was triturated with ether, dried under vacuum to obtain S-chloro-7—{5-[l- (tetrahydro pyran~4-yl) piperidinyl]-[l,3,4]oxadiazol—2-yl}—2,3-dihydro benzofuran- 4-ylamine oxalate salt (22.5 mg).
Yield: 90.3 % 1H - NMR dé): 8 7.49 (s, 1H), 6.04 (s, 2H), 4.68 (t, J = 8.7 Hz, 2H), 4.0 53.90 (m, 2H), 3.40 - 3.20 (m, 5H), 3.0 (t, J = 8.7 Hz, 2H),2.35 - 2.20 (m, 3H), 2.10 - 1.90 (m, 5H), 1.70 - 1.57 (m, 3H); Mass (m/z): 405.1, 407.2 (M+H)+.
Examples 8 - 49: The compounds of Examples 8:- 49 were prepared by following the procedures as described in es 1 to 7, with some non-critical variations 6-Chloro—8-{5-[1—(2-methoxy- lH-NMR (DMSO‘dé): 5 9.92 (bs, AlH), 7.49 (s, 1H), -piperidinyl]- 4.13 (bs, 2H),._3.'70 (t ,J-5.0Hz, 2H), 3.54 (t, J=5.5 Hz, [1,3,4]0xadiazolyl}-chroman- 2H), 3.38 (s, 3H), 3.10 2.95 (m, 3H), 2.60 (t, J=5.5 Hz, S—yiamine 2H), 2.54 (t, J=6.6Hz, 2H), 2.30 - 2.13 (m, .3H), 2.15 - 1.85.(m, 5H); ' Mass (m/z): 393.2, 395.3 (M+H)+. ._ ro{5-[1-(3-methyl- (CDC13.): 6 7.67(s, 1H), 4.35 (s, 2H), 4.28 (t, » butyl)-piperidinyl]- J= 4.8 Hz, 2H), 3.05 - 2.90 (m, 3H), 2.54 (t, J=6.5 Hz, [l,3,4]oxadiazoly1}-chroman— 2H), 2.40 -2.32 (m, 2H), 2.18 - 2.06 (m, 6H), 2.06 - -ylamine 1.92 (m, 2H), 1.65 - 1.55 (m, 1H), 1.48 — 1.36(m, 2H), 0.92 (d, J= 6.5 Hz,6H); Mass (m/z): 405.4, 407.4 (M+H)+. 6-Chloro[5-( l - lH—NMR (DMSO-d5)2 5 7.48 (s, 1H), 5.79.(s, 2H), 4.14 cyclobutylmethyl-piperidin (t, J= 4.8Hz, 2H), 4.12 (5,2H), 3.12 — 2.92 (m, 5H), 2.70 yl)-[l,3,4]oxadiazolyl]- - 2.50 (m, 4H), 2.10 - 2.0 (m,7H), 2.0 - 1.93 (m, 2H), chroman-S-y13mine 1.90 - 1.75 (m, 4H), .62 (m,2H) Mass (m/z): 403.3 405.3 (M+H)+. 6-Chloro[5-(1- lH-NMR (DMSO-d6): 5 7.49 (s', 1H), 5.79 (s,- 2H), cyclopropylmethyl—piperidin 4.13 (t, J=4.8 Hz, 2H), 4.10 (s, 2H), 3.30 - 3.15 (m,2H). : y1)-[1,3,4]0xadiazoly1]- 3.15 - 3.05 (m, 3H), 2.70 ’- 2.55 (m, 4H), 2.18 - 2.07 (m, chroman—S-}‘I1am ine 2H), 2.0 - 1.80 (m, 4H), 1.0 - .090 (m,1H), 0.60 - 0.50 . (m, 2H), 0.25 - 0.16 (m, 2H); _ Mass (m/z): 389.3, 391.4, (M+H)+. 6-Ch10ro[5—(1-isopropy1- 1H—NMR (DMSO-d6): 5 7.49 (s, H), 5.80 (s, 2H), 4.22 piperidin—4-yl)—[1,3,4]oxadiazol— (t, J= 4.8 Hz, 2H), 4.05 (s, 2H), 3.25 - 3.05 (m, 6H), 2-y1]-chr0man—5—ylamine 2.88 - 2.70 (m, 2H),'2.25 - 2.10 (m, 2H), 12.0 - 1.80 (m, . 4H), 1.12 (d, J= 6.5 Hz, H); Mass (m/z): 377.3 ,379.5 (M+H)+. :33- ro{5-[1-(3-meth0xy- 5 7.48 (s, 1H), 5.79 (s, 2H), 4.13 , (DMSO-d6): ' propyl)-piperidin¥4-yl]- (bs, 4H), 3.34 (t, J=6.0 Hz, 2H), 3.30 - 3.22 (m, 1H), ]oxadiazol-_2-yl}-chroman- 3.21 (s, 3H), 3.20 - 3.0 (m, 4H), 2.70 - 2.60 (m,'2H), S—ylamine 2.60 - 2.50 (m, 2H), 2.15 - 2.03(m, 2H), 2.0’ - 1.90 (m, 2H), 1.90 - 1.70 (1n, 4H); Mass (m/z): 407.3, 409.2 . 6—Chloro—8-[5-(1-cyclobutyl- ' 1H-NMR (DMSO-d6): 5 7.48 (s, 1H), 4.14 (s, 2H), 4.13 piperidiny1)-[1,3,4]oxadiazol- (t, J: 4.8 Hz, 2H), 3.10 - 2.90 (m, 4H), 2.47 (t, J: 8.0 2-y1]-chr0man-5—ylamine Hz, 2H), 2.30 - 2.15. (m, 2H), 2.12 — 2.0 (m, 4H), 2.0 — 1.85 (m, 4H), 1.85—1.72 (m, 2H),‘1.70 - 1.58 (m, 2H) ; Mass (m/z): 389.3, 391.4 (M+H)+. 6-Chloro—8-[5—( l - lH—NMR (DMSO—d6): 5 7.29 (s, 1H), 5.65 (5,211), cyclobutylmethyl-piperidin-4— 4.33(s,4H), 4.11(s; 2H), 3.15 - 2.95(m,3H), 2.70 - y1)-[1,3,4]0xadiazoly1]-2,3-' 2.55(m,3H), dihydrb—benzo[ 1 ,4]dioxin-5 - 2.50-2.30(m,2H), 2.10—2.0(m, 2H); ylamine Mass (m/Z): 405.3, 407.4 (M-hH)+. 6-Ch10ro[5-(1-cyclobuty1- 'H-NMR (DMSO-d6): 5 7.29 (s, 1H), 5.65(s,2H), 3.10 - piperidiny1)-[1,3 ,4]oxadiaz01- 3.0(m, 1H), 3.0 - 2.85 (m, 3H), 2.30 - 2.12 (m, 2H), 2-y1]-2,3—dihydro- 2.10 -1.95 (m, 4H), 1.95 - 1.70 (m, 4H), 1.70 - 1.53 (m, benzo[1,4]dioxin—5—ylamine 2H); Mass (m/z): 391.4, 393.3 (M+H)+. 6-Chloro-8¥[5-(1-cyclopenty1- IH-NMR (DMSO-d6): 5 7.49 (s, 1H), 5.79 (s, 2H), piperidiny1)-[1,3,4]oxadiazol- 4.13 (t, J= 4.6 Hz, 2H), 4.0 (s, 2H), 3.25 - 3.08 (m, 3H), 2-y1]-chromanylamine 3.05 - 2.90 (m, 1H), 2.68 - 2.53 (m, 2H), 2.47 (1, J=7.8 tartarate salt Hz, 2H), 2.18 -2.0 (m, 2H), 2.0 - 1.80 (m, 6H), 1.70 - 1.60 (m, 2H), 1.58 -140 (m, 4H); Mass (m/z): 403.2, 405.7 (M+H)+. l6-Chloro[5-(2-piperidin-l—yl- 'H—NMR(CDC13): 6 7.69 (s, 1H), 4.35 (bs, 2H), 4.28 0 ethyl)-[ l,3,4]oxadiazolyl]- ,J= 4.5 Hz, 2H), 3.20 - 3.05 (m, 2H), 2.90 - 2.80 (m, chroman-S-ylamine 2H), 2.80 -2.70 (m, 2H), 2.65 - 2.50 (m, 4H), 2.20 - 2.10 (m, 2H), 2.10 —1.95 (m, 1H), 1.60 - 1.48 (m, 1H), 1.50 - 1.40 (in, 2H); Mass (m/z):_ 363.2, 365.2. (M+H)*. 4-[5-(5-Aminochloro- ‘H-NMR (DMSO-d6): 8 7.48 (s, 1H), 5.79 (s, 2H), chromanyl)-[l,3,4]oxadiazol- 4.21 (s, 2H), 4.13 0, J: 4.5 Hz, 2H), 4.10 - 3.95 (m, [ l ,4']bipiperidinyl- l '- 4H), 3.20 —-3.05 (m, 4H), 2.90 - 2.72 (m, 4H), 2.70 — carboxylic acid ethyl ester 2.60 (m, 2H), 2.20 - 2.08 (m, 2H), 20-190 (m, 2H), 1.90 - 1.75 (m, 4H), 1.50 - 1.35 (m, 2H), 1.16 0, J= 7.0 Hz, 3H); Mass (m/z): 490.3, 492.3 (M+H)-+. .6-Chl0r0-8—[5-(3-piperidin- l ~yl- 'H-NMR d6): 5 7.48 (s, 1H), 5.79 (s, 2H), propyl)—[ l ,3,4]oxadiazolyl]— 4.13 0, J: 4.8 Hz, 2H), 4.07 (s, 2H), 2.90 0, J=7.2 Hz, chroman-‘S-ylamine tartarate salt 2H), 2.85-2.70 (m, 6H), 2.47 0, J= 6.5 Hz, 2H), 2.08 — 1.90 (m, 4H), 1.63 - 1.53 (m,4H), 1.50 — 1.38 (m, 2H); Mass (m/z): 349.2, 351.4 (M+H)+. 6-Chloro[5-( l -cyclopentyl- ‘H-NMR d6)': 5 7.47 (s, 1H), 5.80(s, 2H), piperidinylmethyl)- 4.130, J=4.8 Hz, 2H), 3.50 - 3.35 (m, 3H),-3.0 - 2.82 [I ,3,4]0xadiazolyl]—chroman- ‘ (m, 4H), 2.47 0, J= 6.7 Hz, 2H), 2.10 -' 1.88 (m, 7H), -ylamine oxalate salt 1.70-1.40 (m, 8H); Mass (m/z): 417.3, 419.4 (M+H)*. 6-Chldr0—8—[5-(3-isopropyl lH—NMR (DMSO-d6): 5 7.47 (5,1H), 5.79 (s, 2H), aza-bicyclo[3. l .O]hexyl)- 4.120, J= 4.6 Hz, 2H), 3.75 - 3.55 (m, 2H), 3.40 - 3.20 [l,3,4]0xadiazolyl]-chroman— (m, 3H), 2.58 (s, 1H), 2.460, I: 6.6 Hz, 2H), 2.42 (s, -ylamine oxalate salt 2H), 2.0 - 1.90(m, 2H), 1.21 (d, J= 6.0 Hz, 6H); Mass (m/z): 375.2, 377.2 (M+H)*. 2012/000011 6—Chloro[5-(3- ‘H-NMRI(DMSO-d6): 8 7.46 (s, 1H); 5.79(s, 2H), cyclobutylmethyl—3—aza- 4.120, I: 4.8 Hz), 3.60-3.45 :(m, 2H), 3.20—1.90 (m, .bicyclo{3. l .O]hex—6—yl)- 4H), 2.55 (s,1H), 2.46 (1, J= 6.9 Hz, 2H), 2.32 (s, 2H), [ l,3,4]oxadiazolyl]-Chroman- 2.10 — 2.0(m, 2H), 2.0 -1.90 (m, 3H), 1.90 —,1.80(m,. -ylamine oxalate salt lH),1.80 - 1.67(m, 3H); Mass (m/z): 400.9, 403.1 v(M+H)+. r‘o[5-(3- lH-NMR(CDC13): 6 7.65 (s, 1H), 4.33 (s, 2H), 4.27 (t, cyclopropylmethyl-3Haza- J: 4.9 Hz, 2H), 3.28 (d, J: 8.0 H), 2.70 — 2.63 (m, bicyclo[3.l .0]hexyl)- 1H), 2.530, I: 6.5Hz ,2H),2.50-2.42(m, 2H), 2.38 - ]oxadiazol—2—yl]-chroman- 2.30(m, 2H), 2.17 - 2.07 (m, 4H), 0.90 - 0.80(m, 1H), S-ylamine 0.51 - 0.42(m, 2H), 0.15 — 0.06(m,2H); Mass (m/z): 387.1, 389.1 (M+H‘)*. 6-Chlor048- { 5 -[ l -(tetrahydro- - 'H-NMR (DMSO—d6): 5 7.49(s, 1H), 5.81 (s, 2H), 4.13 pyran~4~yl)-piperidinyl]— (t, J: 4.5 Hz, 2H), 4.0 - 3.90(m, 2H), 3.60 - 3.40 [-l,3,4]0xadiazolyl}-chroman-‘ (m,4H), 330(1, J: 11.2 Hz, 2H),- 3.20 — 3.10 (m, 2H), —ylamine oxalate salt 2490, J: 6.7 Hz, 2H), 2.30 — 2.20(m, 2H), 2.10 — 1.88 (m,6H), 1.70 — 1.58(m,2H); Mass (m/z): 419.2, 421.2 (MJFH)+ 6-Chloro{5-[1~(tetrahydro# ‘H—NMR (DMSO-d6): 5 7.49 (s, 1H), 5.81(s, 2H), 4.10 pyran—4-ylmethyl)—piperidin -3.9(m, 2H), 3.90 — 3.80 (m, 2H), 3.27 (1, J= 11.1 Hz, yl]-[1,3,4]oxadiazol-2—yl}- 2H), 3.15 - 1.90 (m, 5H), 2.49 (t, J: 6.6 Hz, 2H), 2.30 - chroman-S-ylamine e salt 2.20 (m,2H),'2.18—1.90(m, 4H); .60(m, 2H), 1.30 - l.10(m, 2H); _’ Mass (m/z):43'3.3, 435.2 (M+H)+.
—Chloro[5-( 1 -cyclopropyl- xH-NMR (DMSO-6): 5 7.49 (s, 1H), 6.02 (s, 2H), 4.68 piperidinyl)—[ l ,3,4]oxadiazol- (t, J=8.8‘Hz, 2H), 3.30 — 3.20 (m, 2H), 3.20 — 3.10 (m, 2-yl]-2,3~d,ihydro-benzofuran 2H), 3.05 (t, J=8.8 Hz, 2H), 2.90 -. 2.75 (m, 2H), 2.304 ylamine oxalate salt 2.20 (m, 1H), 2.18-2.07 (m, 2H), 1.90 - 1.77 (m, 2H),» 0.70 - 0.58 (m, 4H); Mass (m/z): 361.1, 363.1 (M+H)*. —36.—‘ 2012/000011 -Chloro—7-[5-(1-cyclobutyl— lH-NMR (DMSO-dé): 5 7.49 (s, 1H), 6.0 (s, 2H), 4.68 piperidihyl)-[1,3 ,4]oxadiazol- (t, J= 8.8 Hz, 2H), 3.60 - 3.50 (m, 1H), 3.40 - 3.15 (m, 2-yl]-2,3-dihydro—benzofuran 3H), 3.05 (t, J_=18.8 Hz, 2H), 2.90 - 2.70 (m, 2H), 2.30 - ylamine oxalate salt 2.10 (m,6H), 2.05-1.90 (m, 2H), 1.80 - 1.60(m, 2H); Mass (m/z): 375.3, 377.0 (M+H)+. 6-Chloro[5-( l -c'yclopropyl- 1H-NMR(DMSO-d6): 5 7.29 (s, 1H), 5.65 (s, 2H), 4.33 piperidinyl)-[1,3,410xadiazol- (s, 4H), 3.35 -' - 3.25 (m, 2H), 3.25 - 3.13 (m, 1H), 2.93 2—yl]-2,3~dihydr0— 2.82 (m, 2H), 2.37 - 2.28 (m, 1H), 2.20 - 2.10 (m, 2H), benzo[1,4]dioxinylamine 1.90 - 1.80 (m, 52H), 0.70 - 0.60 (m, 4H); oxalate salt ' Mass (m/z): 377.1, 379.0 (M+H)*. 6-Chloro{5-[1-(tetrahydro— ‘H-NMR (DMSO-d6): a 7.30 (s, 1H), 5.66 (s, 2H), 4.33 pyranyl)-piperidin—4-yl]- (s, 4H), 4.0 - 3.20(1n, 6H), 3.10 - - 3.90 (m, 2H), 3.40 [1,3,4]oxadiazoly1}—2,3- 3.0(m, 2H), 2.30 - 2.20 (m, 2H), 2.10 - 1.92 (m, 2H), dihydro-benzo[1,4]dioxin 1.92 - 1.85 (m,2H), 1.70-1.55 (m, 2H) ; e Oxalate salt Mass (m/z): 421.1, 423.2 (M+H)+. 6—Chloro{5-[1-(3-methoxy- 1H-NMR (DMSO-d6): 5 7.30 (s, 1H), 5.68 (s, 2H), )—piperidin—4-yl]- 4.33 (s, 4H), 3.50 - 3.40 (m, 2H),, 3.37 (t, J= 5.8 Hz, [1,3,4]oxadiazol-2—yl}-2,3- 2H), 3.38 - 3.28 (m, 1H), 3.23 (s, 3H), 3.10 - 2.95 (m, » Idihydro-benzo[1,4]dioxin 4H),'2.30 - 2.20 (m, 2H), 2.10 - 1.92 (m, 2H), 1.90- ylamine oxalate salt - 1.82 (m, 2H) ; Mass (m/z): 409.1 411.0 (M+H)+. ro{5-[l-(tetrahydro- 1, R (DMSO-d6): 8 7.29 (s, 1H), 5.68 (s, 2H), 4.33 pyran-4—ylmethyl)-piperidin (s, 4H), 3.90 - 3.80 (m, 2H), 3.50 - 3.30 (m, 2H), 3.291 yl]-[l,3,4]oxadiazolyl}-2,3— (t, J=l 1 1.4 Hz, 2H), 3.08 - 2.90 (m, 1H), 2.90 - 2.80 (m, dihydro-benzo[1,4]dioxin 2H), 2.28 -2.16(m, 2H),2.12 - 1.95 (m, 3H), 1.70 — 1.60 ylamlne oxalate salt (m, 2H), 1.28-1.10 (m,2H); Mass (m/z):435.2, 437.3 (M+H)+.
-Chloro{5-[1-(tetrahydro- .‘H—NMR (CDC13): 5 7.49.(s,.1H), 6.030, 2H),V4.68(t, pyranylmethyl)—piperidin :J=8.8 Hz, 2H), 3.90 - 3.80 (m, 2H), 3.40 - 3.20 (m, 5H), yl]-[1,3,4]0xadiazol—2-yl}-2,3- 3.05 0, J= 8.8 Hz, 2H), 3.0 - 2.90 (m, 2H), 2.90 - dihydro-benzofuranylamine '2.80(m, 2H), 2.25-2.15 (m, 2H), 2.10 — 1.92 (m, 3H), oxalate 1.70: 1.60 (m, 2H), 1.30-l.l3(m, 2H); ' Mass (m/z): 419.1, 421.2 (Mi-Hf. 4-[5-(43Amino-5 o-2,3- MR (DMSO-d6): a 7.49 (s, 1H), 6.03 (s,2H), dihydfo-benzofuranyl)- ' 4.680, J= 8.7 Hz, 2H), 4.15 — 4.00 (m, 4H), 3.40 - [1,3,4]0xadiazol-2—yl]— 3.15011, 4H), 3.050, J= 8.7 Hz, 2H), 3.05 - 2.92 (m, [l,4']bipiperidinyl- 1 '—carboxylic 2H), 2.90 - 2.70011, 2H), 2.30-220 (m,2H), 2.10 — acid ethyl ester e 1.90011. 4H), 1.60 — 1.42(m,2H), 1.170, J= 7.0 Hz, 3H); Mass (m/z): 476.1, 478.2 (M+H)+. 3-[5-(l-Cyclobutylmethyl- ‘H-NMR d6): 5 8.17 (d, J= 8.1 Hz, 1H), piperidin-4—yl)—[ 1 ,3,4]oxadiazol- 7.9001,J=8.56 Hz, 1H), 7.540, J= 7.5 Hz, 1H), 7.38(t, 2-yl]—l —isopropyl-1H-indazole J= 7.6 Hz, 1H), 5.2 1H), 4.12 (s, 2H), 3.3 - — 5.13(m, 3.2(m, 2H), 3.12 - 3.02(m, 2H), 2.7 — , 2H), 2.6 - 2.55(m, 1H), 2.48 - .- 2.43(m, 1H), 2.2 - 2.1 (m, 2H), 2.1 2.0 (m, 2H), 1.98—1.87(m, 2H), 1.86 - 1.72 (m, 1H), 1.71 — 1.63 (m, 2H), 1.5401, J= 6.5 Hz, 6H); _' Mass (m/z): 380 (M+H)+. ' l-Isopropyl{5-[l—(2-meth0xy- <‘H-NMR (CDC13): 8~8.35(d, J=8.1 Hz, 1H), 7.53 (d, -piperidinyl]— EJ=8.3 Hz, 1H), 7.47 0, J=6.9 Hz,1H), 7.330, J=7.3 Hz, [1,3,4]oxadiazolyl}-lH- :lH), 5.0 -4.91(m,1H), 3.550, J=5.3 Hz, 2H), 3.380, indazole 3H), 3.12 - - 3.1(m, 3H), 2.630, J=5.2Hz, 2H), 2.25‘ 2.12 (m,6H), 1.66 (d, J=6.6 Hz, 6H); Mass (m/z): 370 (M+H)+. 2012/000011 3-[5-(l-Cyclobutyl—piperidin ‘ 1H-NMR(CD.C13): 5 8.33 01, J= 8.2 Hz, 1H), 7.5201, J: yl)-[l,3,4]oxadiazolyl] 8.5 Hz, 1H), 7.470, J= 7.9 Hz, 1H), 7.330, J=7.5 Hz, pyl-lH-indazole 1H), 5.0 —4.93(m, 1H), 3.15-3.08(m, 1H), 2.98 - 2.96(m,2H), 2.8 -2.7(m-, 1H), 2.3 - 2.2011, 2H), 2.16 — 2.05 (m, 5H), 2.0 - 1.9 011,311), 1.75 - 1.69 (m, 211), 1.67(d, J= 6.7 Hz, 6H); Mass (m/z): 366.4 (M+H)+. l-Isopropyl[5—(l-isopropyl— ‘H-NMR (CDC13): 5 , J: 8.16 Hz, 1H), 7.53» (d, piperidin-4—yl)-[1,3,4]oxadiazol- I: 8.48 Hz, 1H), 7.47 9t, I: 7.97 Hz, 1H), 7.330, ’1: 2-yl]-1H-indazolc 7.48 Hz, 1H), 5.00 - 4.93011, 1H), 3.06 - 2.96 (m, 3H), 2.8 - 2.75011, 2H), 2.35 - 2.30011, 2H), 2.25 — 2.17 (m, 2H), 2.12 -2.03(m,2H), 1.65(d, 1: 6.8 Hz, 6H), 1.87(d, J: 6.5 Hz, 6H); Mass (m/z): 354 (M+H)+. 3-[5-( l -Cyclopropylmethyl- ‘H-NMR (c1303): 58.35 (d, J=8.1‘Hz, 1H), 7.5301, piperidinyl)-[1,3,4]oxadiazol- J=8.4, Hz, 1H), 7.470, J= 6.7 Hz, 1H),7.33(t, J=7.46 Hz, 2-yl]isopropyl-lH-indazole 1H), 5.03 - 4.95(m, 1H), 3.18 - 315 (m, 2H), 3.15 - 3.07(m,;1H),'2.31(d, J=6.5'Hz, 2H), 2.22 - 2.1(m, 6H), 1.67(d, J=6.68 Hz,6H), 0.98 — 0.91(m,lH), 0.57 — 0.52 (m, 2H), 0.14 -0.11(m, 2H); Mass (m/z): 366 (M+H)+. 1-[sopropyl{5.-[1—(3-methyl- ‘H-NMR (CDC13): 5 8.35(d, J= 8.1 Hz, 1H), 7.5301, J= butyl)-piperidinyl]— 8.46 Hz, 1H), 7.470, J= 6.91 Hz, 1H), 7.330, J=7.6 Hz, [1,3,4]0xadiazolz2—yl}—l H- 1H), 5.0 m,1H), 3.10 — 2.98(m, 3H),- indazole '1.67(d,J=6.68 Hz, 6H), 1.45 - ,'2H), 0.9201, J=6.57 Hz, 6H); Mass (m/z): 382 (M+H)*. 3-[5-(1-Cyclopropyl—piperid,in 1H-NMR(CDC13): 5 8.35(d, J= 8.17 Hz, 1H), 7.53(d, yl)-[l,3,4]oxadiazolyl]-l- J=8.48 Hz, 1H), 7.47(t, J=6.96 Hz, 1H), 7.33(t, J=_ 7.47 isopropyl-lH-indazole ' HZ, 1H), 5.0 - 4.93(m,1H), 3.16 ‘ 3.05(m, 3H), 2.37 (t, J: 11.3 HZ,2H), 2.19 — 2.16(m,2H), 2.07-1.98(m, 2H), 1.67(d, J= 6.6 Hz,6H), 1.63 - 1.57 (m, 1H), 051 ~ (m. 4H); Mass (m/z): 352 (M+H)+. 3-[5-(1—Cyclopentyl-piperidin—4- 1H-NMR (CDCb): 5 8.35 (d, J: 9.39 HZ, 1H), 7.53011, yl)—[l,3,4]oxadiaz01yl] ' J= 8.49 Hz, 1H), 7.470, J=8.0 Hz, 1H), 7.33 (t, J=7.43 isopropyl-lH-indazole Hz), 5.0-4.93 (m, 1H), 3.15 - 3.05(m, 3H), 2.6 — 2.53(m, 1H), 2.22 - 2.10(m, 6H), 1.98 - 1.90(m, 2H), 1.74 - , 2H), 1.67(d, Hz, 6H), 1.60—1.55 (m,2H), 1.45 - 1.38 (m, 2H); Mass (m/z): 380 (M+H)+. 1-Isopropyl{5-[3-(3-methoxy- lH-NMR (DMSO-dé): 5 8.15(d, J=8.2 Hz, 1H), 7.89(d, propyl)-3—aza—bicyclo[3 .1 .0]hex- J=8.1 Hz, 1H), 753(1, J.=7.5Hz, 1H), 7.36 (t, J = 7.4 Hz, [l,3,4]0xadiazolyl}-1H- # 1H), 5.20-5.10 (m, 1H), 3.60 - 350 (m, 2H), 3.36 (t, J indazole oxalate salt 5.8 Hz, 2H), 3.23 (s, 3H), 3.10 - 3.0 (m, 2H), 3.0-2.86 (m, 2H), 2.68 (s, 1H), 2.43 (s, 2H), 1.85-1.72 (m, 2H), 1.53 (d, J = 6.3 Hz, 6H); ' 'Mass (m/z): 382.3 (M+H)+. _ 3-[5-(3-Cyclobutyl—3-aza— 1H-NMR (ch13): 8 8.34 (d, J = 8.1 Hz, 1H), 7.52 (d,-J o[3.1 .0]hexyl)- = 8.4 Hz, 1H), 7.46 (t, J = 8.0 Hz, 1H), 7.29 (t, J = 7.3 ]oxadiazoly11—1— . "Hz, 1H), 5.0 - 4.90 (m, 1H), 3.11 (d, J = 9.0 Hz, 2H), isopropyi-lH-indazole oxalate 3.10 - 3.0 (m, 1H), 2.73 (s, 1H), 2.41 (d, J = 8.3 Hz, salt 2.06 - 1.95 1.95—1.80 (m, . 2H), "2.2 (s, 2H), (m, 2H), 2H), 1.75 - 1.65 (m, 2H), 1.67 (d, J =6.7>Hz, 6H); Mass (m/z): 364.2 (M+H)+. 3-[5-(3-Cyclobutylmethyl-,3—aza— (DMSO-d6): 5 8.14 (d, J =.8.1 Hz, 1H), 7.89 bicyclo[3.1.0]hexyl)- (d, J = 8.4 Hz, -1 H), 75311, J = 7.4 Hz, 1H), 7.36 (t, J = ‘ [1,3,4]oxadiazo1-2—y11 7.6 Hz, 1H), 5.20 — 5.10 (m, 1H), 3.55 - 3.40 (m, 2H), isopropyl— 1 H-indazole oxalate 3.10 - 3.0' (m, 2H), 2.95 - 2.85 (m, 2H), 2.67 (s, 1H), salt 2.60 - 2.50‘(m, 1H), 2.41 (s, 2H), 2.10- 1.98 (m, 2H), 1.90 - 1.80 (m, 1H), 1.85 - 1.65 (m, 3H), 1.52 (d, J = 6.0 Hz, 6H); Mass (m/z): 378.2 (M+H)+. 3_-[5-(3-Cyclopropylméthyl—3- lH-NMR(CDC13): a 8.34 (d, J = 811 Hz, 1H), 7.52 (d, J cyclo[3.1.0]hexyl)- = 8.3 Hz, 1H), 7.46 (t, J = 8.1 Hz, 1H), 7.32 (t, J = 7.5 [1,3,4]oxadiazol-29y11 Hz, 1H), 5.0 - 4.90 (tn, 1H), 3.30 (d, J = 8.9 Hz, 2H), isopropyl—lH-indazolé oxalate 2.77 (s, 1H), 2.48 (d, J = 8.6 Hz, 2H), 2.20 (s, 2H), 1.67 salt (d, J: 6.0 Hz, 6H), 0.90 - 0.80 (m, 1H), 0.50 — 0.40 (m, 2H), 0.18 - 0.08 (m, 2H); Mass (m/z): 364.1 (M+H)f. _ 1-Isopropyl-3~{5-[1-(tetrahydro- 1IH—NMR(DMSO-d6): 8 8.18 (d, J = 8.03 Hz, 1H), 7.90 pyranylmethyl)-piperidin—4- '= 8.0 Hz, (d, J = 8.4 Hz, 1H), 7.54 (t, J 1H), 7.38 (t, J = ’ y'l]-[1,3,4]oxadiazolyl}-1H- 7.3 Hz, 1H), 5.20- 5.14 (m, 1H), 3.90 — 3.80 (m, 2H), indazole oxalate salt 3.37 — 3.27 (m, 5H), 2.98 - 2.89 (m, 2H), 2.88 - 2.77 (m, 2H), 2.29 — 2.23 (m, 2H), 2.27-2.03 (m, 3H), 1.67 — 1.63 (m, 2H), 1.54 (d, J = 6.4 Hz, 6H), 1.22-1.15 (m, 2H); Mass (m/z): 410.1 (M+H)+. l-Isopropyl-3 - { 5-[ l -(tetrahydro- 1H-NMR(DMSO-d6): 88.18 (d, J = 8.1 Hz, 1H), 7.9 (d, pyranyl)-piperidinyl]- J = 8.5 Hz, 1H), 7.54 (t, J = 7.1 Hz, 1H), 7.38 (t, J = 7.3 [l,3,4]oxadiazolyl}-1H- Hz, 1H), 5.20 — 5.14 (m, 1H), 4.0 — 3.90 (m, 2H), 3.47 — indazole e salt 3.40 (m, 3H), 3.40 - 3.20 (m, 3H), 3.15 - 3.01 (m, 2H), 2.38 - 2.25 (m, 2H), 2.11 -. 2.06 (m, 2H), 1.95 — 1.86 (m, 2H), 1.66 - 1.60 (m, 2H), 1.54 (d, J = 65 Hz, 6H); Mass (m/z): 396.2 (M+H)+. j .) 1-Isopropyl[5-(2-piperidin- l- ‘H-NMR (DMSO-d6): 5 8.19(d, J= 8.1 Hz, 1H), yl-ethyl)-[ l ,3,4]oxadiazol-2—yl]— 7.91(d, J= 8.5 Hz,1H), 7.55(t, J= 7.5 Hz, 1H), 7.38(t,J= lH-indazole oxalate 7.6 Hz, 1H), 5.21 - 5.14(m, 1H), 3.49-3.41(m,4H), 3.15 - , 4H), 1.76 - l.64(m, 4H), l.54(d, J=6.59 Hz, 6H), 1.54 - , 2H); Mass (m/z): 340(M+H)+. ical Assays Example 50: ination of ECSO values for 5-HT4 receptor: A stable CHO cell line expressing recombinant human 5-HT4 receptor and pCRE-Luc reporter system was used for cell-based assay. The assay offers a non- radioactive based ch to determine binding of a compound to GPCRs. In this specific assay, the leVel of intracellular cyclic AMP which is modulated by activation or inhibition of the receptor is measured. The inant cells harbor luciferase reporter gene under the control of CAMP response element.
The above cells were grown in 96 well clear bottom white plates in‘Hams F12 medium containing 10% fetal bovine serum (FBS). Prior to the addition of compounds or standard agonist, cells were serum starved overnight. Increasing concentrations of test compounds were added in OptiMEM medium to the cells. The incubation was continued at 37 °C in C02 incubator for 4 hours. Medium was removed and cells were washed with phosphate buffered'saline. The cells Were lysed and luciferase activity was ed in a Luminometer. Luminescence units were plotted. against the compound concentrations using Graphpad software. EC50 values of the compounds were defined as the concentration required in stimulating the luciferase activity by 50 %. v—Iy—dy—Ao N A m \l ._.\ ' '204 )—| J} w H U: . g";-N h—Ih—d \lO\ . 113.3 . ”‘5"“w MO. 104 Nl—‘3' '7L2 NN . A00 NU) NO N9 MN'\IU‘ N9 ._.. as [\Jfl NOQ . ._.. C) N\O 21.5 U) 0 DJ 3L 169 b) w NP.‘ w 4:. u: U) U! 169- WW \IO\ 143 -127 w m bk»CC ,116 WO 42135 Example 51: Rodent Pharmacokinetic Study Male Wister rats (225 i 25 grams) were used as an experimental animal. Three to five animals were housed in each cage. Two days prior to dosing day, male wister rats (225’ - 250 grams) were anesthetized with isoflurane for surgical placement of jugular vein catheter. Animals were fasted over night before oral dosing (p.o) and food pellets were allowed 2 hours post , whereas intravenous dosing food and water were provided as ad libitum. Three rats were dosed with compounds of formula (I) (10 mg/kg) orally and intravenously (05 mg/kg). _At each time point blood was ted through jugular vein and immediately replenish with an equivalent volume of normal saline from freely moving rats. ted blood was transferred into a labeled eppendr off containing 10 ”L of heparin as anticoagulant. Typically blood‘samples were collected as following time points: Pre dose, 0.08 (only i.v.), 0.25, 0.5, l, 2, 4, 6, 8, and 24 hours post dose (n=3). Blood was centrifuged at 4000 rpm for 10 minutes. Plasma was prepared and stored frozen at -20 °C until analysis. The concentrations of the compounds of formula (I) were quantified in plasma by qualified LC-MS/MS method using suitable extraction technique. The compounds of a (I) were quantified in the calibration range around 2-2000 ng/mL in plasma. Study samples were analyzed using calibration samples in the batch and quality l samples spread across the batch.
Phamiacokinetic parameters Cm”, Tmax, AUCt, Ty; and Bioavailability were ated by non—compartmental model using standard non-compartmental model by using WinNonLin 5.0.1 or PhOeni-x WinNonlin 6.2'version Software package.
Ekam Strain Dose Vehic Route of Cum Bioavailabi administrat (ng/m Numb Gende ‘ g) (%) Ili-.34i13_' (gavage) 2 :l: 120, intravenous 0.0 1117 (bolus) 8 :t 285 oral (gavage) intravenous (bolus) oral (gaVage) " enous (bolus) oral 0.96 42 :l: 10 (gavage) intravenous 783 :1: 0.0 773345 . (bolus) 1.24 8 d: oral 1758 :1: 0.5 4814 :1: e) 264 0 :h 30 - 0.0 intravenous 1858 i 0.0 2373 i (bolus) 115 8 :1: 90 oral 0.2 790 i (gavage) intravenous (bolus) oral .553 i 0.4 1490 i (gavage) 53 2 :t 72 v WO 42135 intravenous (bolus) oral (gavage) intravenous (bolus) Wistar oral / Male (gavage) intravenous (bolus) oral (gavage) _47- Steril intravenous 0 0 1004 :1: 1.44 e (bolus) 8 :t 109 oral . 521' (gavage) ' l 1 l Steril intravenous e (bolus) Example 52: Rodent Brain Penetration Study Male Wister rats (225 i 25 grams) Were used as an experimental animal. Three animals were housed in each cage. Animals were given water and food ad libitum hout the experiment, and maintained on a 12 hours light/dark cycle.
Brain penetration was determined in te manner in rats. One day prior to ' dosing day, male wistar rats (225 - 250 grams) were acclimatized. Afier acclimatizatidn the rats were grouped according to the weight in each group, 3 animals were kept in individual cage and allowed free access to food and water. At each time point (0.50, 1, and 2 hours) n = 3 s were used.
The compounds of forrnula (I) was suitably preformulated and administered orally at (free base equivalent) 10 mg/kg. Blood s were removed via, cardiac puncture by using isoflurane anesthesia the animals were sacrificed to collect brain tissue. Plasma was separated and Brain samples were homogenized and stored frozen at -20 °C until analysis. The concentrations of the NCE compound in plasma and Brain were determined using MS method. ' The compounds of formula (I) were quantified in plasma and brain homogenate by qualified MS method using suitable extraction technique. The compounds of formula (I) were quantified in the ation range of 1-500 ng/mL in plasma and brain homogenates Study samples were analyzed using calibration} samples in the batch and quality control s spread across the batch. Extent of brain-plasma ratio was calculated (Cb/C1,).
Strain/ Route of: Single dose Brain Gender administration Penetration ) Wistar / Male Reagent oral (gavage) 3.88 :1: 0.26 ' grade water Wistar / Male Sterile intravenous water for (bolus) injection Wistar / Male 10 Reagent oral (gavage) 0.56 £0.08 ' grade water Wistar / Male Sterile intravenous water for (bolus) T injection Wistar/ Male ‘1 10 Reagent oral (gavage): 2.24 3: 0.09 grade water Wistar / Male 5 Sterile intravenous water for (bolus) Wistar / Male Reagent oral (gavage) 0.50 :I: 0.07 _ grade water Wistar / Male Sterile intravenOus water for (bolus) injection _ Wistar / Male - Reagent oral (gavage) 0.62 d: 0.05 grade water Wistar / Male Sterile intravenous . water for (bolus) injection- Wistar / Male oral (gavage) 5.68 d: 1.74 Wistar / Male Sterile intravenous water for (bolus) injection Wistar / Male t oral (gavage) 4.69 :t 0.69 grade lwater Wistar / Male Sterile intravenous water for (bolus) injection Wistar / Male l—Re’agent oral e) 3.15 i057 grade water Wistar / Male fSterile intravenous ' for water (bolus) injection Wistar / Male Reagent oral (gavage) 2.55 i 0.32 grade _ water ‘ Wistar / Male - Sterile intravenous water for (bolus) injection Example 53: Object Recognition Task Model 'The cognition ing 'properties of compounds of this invention were estimated by using this model.
Male Wister rats (230 — 280 grams) were used as experimental s. Four s were housed in each cage; Animals Were kept on 20 % food deprivation before v one day and given water ad libitum throughout the experiment and maintained on a 12 hours light/dark'cycle. Also the rats were habituated to individual arenas for 1 hour in the absence ofany objects.
One group of 12 rats received vehicle (1 mL/Kg) orally and’ another set of animals received nd of the a (I) either orally or i.p.-, before one hour of the familiar (Tl) and choice trial (T2).
The experiment was carried out in a 50 x 50 x 50 cm open field made up of acrylic. In the familiarization phase/(Tl), the rats were placed individually in the open field for 3 minutes, in which two identical objects (plastic bottles, 12.5 cm height x 5.5 cm diameter) covered in yellow masking tape alone (a1 and a2) were positioned in two adjacent comers, 10‘ cms from the walls. After 24 hours of the (T1) trial for long-term memory test, the same rats were placed in the same arena as they were placed in T1 trial. Choice phase (T2) rats were allowed to e the open field for 3 s in presence of one familiar object (a3) and one novel object (b) (Amber color glass bottle, 12 cm high and 5 cm in diameter)- Familiar objects ted similar textures, colors and sizes. During the Tl and T2 trial, explorations of each object (defined as sniffing, licking, chewing or having moving, vibrissae whilst directing the nose towards‘the object at a distance of less than 1 cm) were recorded separately by stopwatch. Sitting on an object was not regarded as exploratory ty, however, it was rarely observed.
T1 is the total time spent exploring the familiar objects (a1 + a2).
T2 is the total time spent exploring the familiar object and novel object (a3 +b).
‘ The object recognition test was performed as described by Ennaceur, A., A new ial test fOr neurobiological studies of memory in rats - . Delacour, 1., 1988, Behavioural data, Behavi Brain Res, 31,_47-59. m.3mg/kg,p.o. 11.21i2.18 l6.47:t1.18 ' 2012/000011 - .. e 54: Radial arm maze The cognition enhancing properties of compounds of this invention were estimated by using this model.
Radial arm maze consists of a central hub of 45 cm er. Bach arm was of dimension 42.5.x 15, x 24 cm. The maze'was elevated to a height of l m above the ground. The animals were place on a cted diet until they reached approximately 85 % of their free feeding weight. During this diet restriction period animals were habituated to the novel feed (pellets). Once the rats reached approximately85 % of their free feeding weight rats were habituated to the maze on the 1St & 2nd day._The animals that did not eat the pellets were ed from the study. Animals were randomized on day 2. On the subsequent days the treatment was given as per the allotment. Each animal was introduced into the maze individually for a period of 10 minutes. The arms were baited only: once and the animal had to learn. the rule that repeated arm entries would not be rewarded. The trial ended once the rat had visited 16 arms or 10 minutes were over or all the pellets were eaten. The arm entries were recorded using the software. Once the trial was over the rat was removed and the maze was cleaned using soap water.
Example Reversal of Scopolamine Induced 3. 1 - 3 mg/kg, p.o. 14. 1 --10 mg/kg, p.o.

Claims (18)

What we claim is:
1. A compound of the general formula (I): wherein, is or ; is , or ; is point of attachment; R1 is alkyl, R3-O-R3 or ; R2 is cycloalkyl or heterocyclyl, and optionally substituted with hydrogen, alkyl or -CO-OR3; R3 is alkyl; “Y” is C or O; “m” is an r ranging from 0 to 1; with proviso when m is 0 then R1 is cycloalkyl or heterocyclyl; “n” is an integer ranging from 0 to 2; “p” is an integer g from 0 to 1; or a pharmaceutically acceptable salt thereof.
2. The compound according to claim 1, which is selected from the group consisting of: ro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 1-isopropyl{5-[1-(3-methoxy propyl) piperidinyl]-[1,3,4]oxadiazolyl}-1H-indazole; 3-[5-(1-cyclobutyl-piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 6-chloro[5-(3-cyclobutylaza bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl] n yl amine; 4-[5-(8-Aminochloro-2,3-dihydro benzo[1,4]dioxanyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester; 5-Chloro{5-[1-(tetrahydro pyranyl) piperidinyl]-[1,3,4]oxadiazolyl}-2,3-dihydro benzofuranyl amine; ro[5-(1-cyclopentyl-piperidinylmethyl)-[1,3,4]oxadiazolyl]-chroman ylamine; 6-Chloro[5-(3-isopropylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]-chroman ylamine; 6-Chloro[5-(3-cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]- chromanylamine; ro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman ylamine; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}- chromanylamine; 5-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydro-benzofuran- 4-ylamine; 5-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydro-benzofuran ylamine; 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo [1,4]dioxinylamine; 6-Chloro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3-dihydrobenzo [1,4]dioxinylamine; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3-dihydrobenzo [1,4]dioxinylamine; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzo[1,4]dioxinylamine; 5-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzofuranylamine; 4-[5-(4-Aminochloro-2,3-dihydro-benzofuranyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester; ropyl{5-[3-(3-methoxy-propyl)aza-bicyclo[3.1.0]hexyl]-[1,3,4]oxadiazol -indazole; 3-[5-(3-Cyclobutylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole; 3-[5-(3-Cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl- azole; 3-[5-(3-Cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl- 1H-indazole; 1-Isopropyl{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}- 1H-indazole; 1-Isopropyl{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole; 1-Isopropyl[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-1H-indazole.
3. The compound according to claim 1, which is ed from the group consisting of: 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine hemi fumarate; 6-Chloro[5-(1-cyclobutyl piperdinylmethyl)-[1,3,4]oxadiazolyl]-chromanyl amine L(+) tartarate salt; 6-Chloro[5-(1-cyclobutyl piperdinyl methyl)-[1,3,4]oxadiazolyl]-chromanyl amine; 1-Isopropyl{5-[1-(3-methoxy propyl) piperidinyl]-[1,3,4]oxadiazolyl}-1H-indazole oxalate salt; 1-Cyclobutyl-piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole L(+)- tartarate salt; 6-Chloro[5-(3-cyclobutylaza bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl] chroman ylamine oxalate salt; 4-[5-(8-Aminochloro-2,3-dihydro benzo[1,4]dioxanyl)-[1,3,4]oxadiazolyl]- [1,4']bipiperidinyl-1'-carboxylic acid ethyl ester oxalate salt; 5-Chloro{5-[1-(tetrahydro pyranyl) piperidinyl]-[1,3,4]oxadiazolyl}-2,3-dihydro benzofuranyl amine oxalate salt; 6-Chloro{5-[1-(2-methoxy-ethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chromanylamine; 6-Chloro{5-[1-(3-methyl-butyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman ylamine; 6-Chloro[5-(1-cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-cyclopropylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-isopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chromanylamine; 6-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo ioxinylamine; 6-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydro-benzo[1,4]dioxin- 5-ylamine; 6-Chloro[5-(1-cyclopentyl-piperidinyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-chromanylamine; 4-[5-(5-Aminochloro-chromanyl)-[1,3,4]oxadiazolyl]-[1,4']bipiperidinyl-1'-carboxylic acid ethyl ester; 6-Chloro[5-(3-piperidinyl-propyl)-[1,3,4]oxadiazolyl]-chromanylamine; 6-Chloro[5-(1-cyclopentyl-piperidinylmethyl)-[1,3,4]oxadiazolyl]-chromanylamine oxalate salt; 6-Chloro[5-(3-isopropylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]-chroman e oxalate salt; 6-Chloro[5-(3-cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]-chroman- 5-ylamine oxalate salt; 6-Chloro[5-(3-cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]- chromanylamine; 6-Chloro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman ylamine oxalate salt; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-chroman- 5-ylamine oxalate salt; 5-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydro-benzofuran ylamine oxalate salt; 5-Chloro[5-(1-cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydro-benzofuran ylamine e salt; 6-Chloro[5-(1-cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-2,3-dihydrobenzo [1,4]dioxinylamine oxalate salt; 6-Chloro{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzo[1,4]dioxinylamine oxalate salt; 6-Chloro{5-[1-(3-methoxy-propyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3-dihydrobenzo [1,4]dioxinylamine e salt; 6-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzo[1,4]dioxinylamine oxalate salt; 5-Chloro{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-2,3- dihydro-benzofuranylamine oxalate; 4-[5-(4-Aminochloro-2,3-dihydro-benzofuranyl)-[1,3,4]oxadiazolyl]-[1,4']bipiperidinyl- 1'-carboxylic acid ethyl ester oxalate; 1-Cyclobutylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl{5-[1-(2-methoxy-ethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H-indazole; 3-[5-(1-Cyclobutyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl[5-(1-isopropyl-piperidinyl)-[1,3,4]oxadiazolyl]-1H-indazole; 3-[5-(1-Cyclopropylmethyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl{5-[1-(3-methyl-butyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H-indazole; 3-[5-(1-Cyclopropyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 3-[5-(1-Cyclopentyl-piperidinyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole; 1-Isopropyl{5-[3-(3-methoxy-propyl)aza-bicyclo[3.1.0]hexyl]-[1,3,4]oxadiazolyl}-1H- indazole oxalate salt; 3-[5-(3-Cyclobutylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole e salt; 3-[5-(3-Cyclobutylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl-1H- le oxalate salt; 3-[5-(3-Cyclopropylmethylaza-bicyclo[3.1.0]hexyl)-[1,3,4]oxadiazolyl]isopropyl-1H- indazole oxalate salt; 1-Isopropyl{5-[1-(tetrahydro-pyranylmethyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H- indazole oxalate salt; 1-Isopropyl{5-[1-(tetrahydro-pyranyl)-piperidinyl]-[1,3,4]oxadiazolyl}-1H-indazole oxalate salt; 1-Isopropyl[5-(2-piperidinyl-ethyl)-[1,3,4]oxadiazolyl]-1H-indazole e salt and 3-[5-(1-cyclobutyl piperidinyl methyl)-[1,3,4]oxadiazolyl]isopropyl-1H-indazole oxalate salt; or their pharmaceutically acceptable salts.
4. The process for preparation of a compound of formula (I) as claimed in claim 1, which comprises: (a) coupling the compound of formula (1) with compound of formula (2) in presence of dehydrating agent to form a nd of formula (I), wherein all substitutions are as defined in claim 1, (b) optionally converting the compound of formula (I) to their pharmaceutically able salts.
5. The process for preparation of a compound of formula (I) as claimed in claim 1, which comprises: (a) coupling the compound of formula (1) with compound of formula (2) in ce of suitable solvent to form a compound of formula (4), (b) cyclizing the compound of formula (4) to form a compound of formula (I), wherein all substitutions are as defined in claim 1, (c) optionally converting the compound of formula (I) to their pharmaceutically acceptable salts.
6. A pharmaceutical composition comprising a compound according to any of claims 1 to 3 and pharmaceutically acceptable excipients.
7. The pharmaceutical composition according to claim 6, for the treatment of clinical conditions mediated through 5-HT4 receptors such as attention deficit hyperactivity disorder, mers disease, cognitive disorders, dementia or schizophrenia.
8. Use of a compound according to any one of the claims 1 to 3 in the manufacture of medicament for the ent of diseases related to 5-HT4 receptors.
9. The use of compound according to the claim 8, for the treatment of clinical ions such as attention deficit hyperactivity disorder, alzheimers disease, cognitive disorders, dementia or phrenia.
10. Use of a nd of formula (I) as d in claim 1, in the manufacture of a medicament for treatment of a disorder of central nervous system related to or affected by the 5-HT4 ors.
11. A compound of formula (I) when prepared by the s of claim 4.
12. A compound of formula (I) when prepared by the s of claim 5.
13. A compound of the general formula (I) as claimed in claim 1, substantially as hereinbefore described with particular reference to any one of the Examples.
14. The process for preparation of a compound of formula (I) as claimed in claim 4, substantially as hereinbefore described with particular reference to any one of the Examples.
15. The process for preparation of a compound of formula (I) as claimed in claim 5, substantially as hereinbefore described with particular nce to any one of the Examples.
16. A pharmaceutical composition as d in claim 6, substantially as hereinbefore described with particular reference to any one of the Examples.
17. Use of a compound as d in claim 8, substantially as hereinbefore described with particular reference to any one of the Examples.
18. Use of a compound as claimed in claim 10, substantially as hereinbefore described with particular reference to any one of the Examples.
NZ619776A 2011-09-19 2012-01-05 Heteroaryl compounds as 5-ht4 receptor ligands NZ619776B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
IN3203/CHE/2011 2011-09-19
IN3203CH2011 2011-09-19
PCT/IN2012/000011 WO2013042135A1 (en) 2011-09-19 2012-01-05 Heteroaryl compounds as 5-ht4 receptor ligands

Publications (2)

Publication Number Publication Date
NZ619776A NZ619776A (en) 2015-04-24
NZ619776B2 true NZ619776B2 (en) 2015-07-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012311057B2 (en) Heteroaryl compounds as 5-HT4 receptor ligands
AU701127B2 (en) Muscarine antagonists
US7700591B2 (en) Benzoxazinyl-amidocyclopentyl-heterocyclic modulators of chemokine receptors
EP1501803B1 (en) Tetrahydropyranyl cyclopentyl tetrahydroisoquinoline modulators of chemokine receptor activity
WO2005072361A2 (en) Aminocyclopentyl pyridopyrazinone modulators of chemokine receptor activity
TW202120501A (en) 4-substituted indole and indazole sulfonamido derivatives as parg inhibitors
CA2935071A1 (en) Piperidine-dione derivatives
CZ289479B6 (en) Use of piperidine derivatives for preparing pharmaceutical preparations, piperidine derivatives and pharmaceutical preparation per se
CA2218391A1 (en) Alpha 1a adrenergic receptor antagonists
AU2003286701A1 (en) Tetrahydropyranyl cyclopentyl benzylamide modulators of chemokine receptor activity
KR20110000749A (en) Novel class of spiro piperidines for the treatment of neurodegenerative diseases
WO2011030349A1 (en) L -dihydro-2-oxoquinoline compounds a 5-ht4 receptor ligands
CN115109032B (en) Quinoline derivative and application thereof in medicine
NZ226681A (en) Diazepinone derivatives: preparatory processes and pharmaceutical compositions
NZ619776B2 (en) Heteroaryl compounds as 5-ht4 receptor ligands
Nirogi et al. Heteroaryl compounds as 5-HT4 receptor ligands
US6326372B1 (en) Lactam and cyclic urea derivatives useful as alpha 1a adrenoceptor antagonists
CA2907620A1 (en) 5-amino-quinoline-8-carboxamide derivatives as 5-ht4 receptor agonists
WO2022152852A1 (en) Antagonists of mrgx2
JP2009502899A (en) Quinoline derivatives as neurokinin receptor antagonists