WO2009081259A1 - Phenoxy-pyridyl derivatives - Google Patents

Phenoxy-pyridyl derivatives Download PDF

Info

Publication number
WO2009081259A1
WO2009081259A1 PCT/IB2008/003537 IB2008003537W WO2009081259A1 WO 2009081259 A1 WO2009081259 A1 WO 2009081259A1 IB 2008003537 W IB2008003537 W IB 2008003537W WO 2009081259 A1 WO2009081259 A1 WO 2009081259A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
mmol
acceptable salt
alkyl
Prior art date
Application number
PCT/IB2008/003537
Other languages
French (fr)
Inventor
Nancy Sue Barta
Brian Michael Campbell
Beth Dounay Amy
David Lawrence Firman Gray
Stevin Howard Zorn
Original Assignee
Pfizer Inc.
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 Pfizer Inc. filed Critical Pfizer Inc.
Publication of WO2009081259A1 publication Critical patent/WO2009081259A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the present invention comprises a novel class of compounds having the structure of formula I (including tautomers and salts of those compounds) and pharmaceutical compositions comprising a compound of formula I.
  • the present invention also comprises methods of treating a subject by administering a therapeutically effective amount of a compound of formula I to the subject. These compounds are useful for the conditions disclosed herein.
  • the present invention further comprises methods for making the compounds of formula I and corresponding intermediates.
  • Monoamine neurotransmitters include, for example, norepinephrine (noradrenaline), serotonin (5-HT), and dopamine. Lower-than-normal levels of these neurotransmitters are associated with a variety of symptoms including lack of energy, motivation, and interest in life. Thus, a normal level of monoaminergic neurotransmission is essential to maintaining drive and capacity for reward. These neurotransmitters travel from the terminal of a neuron across a small gap (i.e., the synaptic cleft) and bind to receptor molecules on the surface of a second neuron.
  • a small gap i.e., the synaptic cleft
  • This binding elicits intracellular changes that initiate or activate a response or change in the postsynaptic neuron. Inactivation occurs primarily by transport (i.e., reuptake) of the neurotransmitter back into the presynaptic neuron.
  • Abnormality in noradrenergic transmission results in various types of depression, mental, behavioral, and neurological disorders attributed to a variety of symptoms including a lack of energy, motivation, and interest in life. See generally, R. J. Baldessarini, "Drugs and the Treatment of Psychiatric Disorders: Depression and Mania” in Goodman and Gilman's The Pharmacological Basis of Therapeutics, McGraw-Hill. N.Y., N.Y., pp. 432 439 (1996).
  • Drugs that impact monoamine neurotransmission either directly as a result of efficacy at postsynaptic receptors or indirectly through inhibition of reuptake into presynaptic terminals are widely used to treat disorders including attention deficit hyperactivity disorder (ADHD), depression, and anxiety.
  • ADHD stimulant medications that increase norepinephrine and dopamine neurotransmission throughout the brain are commonly prescribed, but carry a burden of abuse liability.
  • norepinephrine reuptake inhibitors (NRI) which more selectively increase norepinephrine neurotransmission with less impact on dopamine, are also used to treat ADHD albeit with less efficacy and a slower onset of action than stimulant medications.
  • NRI agents are widely used to treat symptoms of depression and anxiety, although again taking many days before full efficacy is demonstrated.
  • compounds with serotoniniA (5-HT1A) receptor partial agonist properties are commonly used to treat symptoms of depression and anxiety, and evidence indicates that they could be used to treat symptoms of ADHD and other disorders associated with deficits in cognitive function like schizophrenia, bipolar disorders and Alzheimer's disease as well.
  • NRI and 5-HT1A partial agonist properties are more effective than NRI agents alone.
  • the present invention describes agents possessing NRI and 5-HT1A partial agonist properties that will be useful in treating symptoms of disorders wherein modulation of monoamine neurotransmission is required such as ADHD, depression, anxiety, bipolar disorders and Alzheimer's disease.
  • the invention is directed to a class of compounds, including the pharmaceutically acceptable salts of the compounds, having the structure of formula I:
  • X 1 is O or S(O) P ; is either a single bond or a double bond, wherein, when is a single bond, Y is N or CR 3 , and when is a double bond, Y is C; each Ri is independently selected from the group consisting of hydrogen, halogen, alkoxy, -CN, and alkyl, wherein each Ri alkyl is optionally independently substituted by one to three halogens; each R 2 is independently selected from the group consisting of halogen, -NO 2 , -CN, -N(R 4 ) 2 , R 4 , and -OR 4 ; or two R 2 substituents on adjacent carbons taken together with the adjacent carbons form a carbocyclic or heterocyclic ring optionally substituted with halogen, hydroxy, -CN, alkyl, or alkoxy; wherein each R 4 is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heterocycloalkyl and heteroary
  • each R 3 is independently selected from the group consisting of hydrogen, halogen, -(CH 2 ) t OH, -(CH 2 ) t CF 3 , -(CH 2 ) t C ⁇ N, -NO 2 , -(CH 2 ) t N[(CH 2 ) t R 4 ] 2l -(CH 2 ),-alkyl, -(CH 2 ) t O[(CH 2 ) t R 4 ], -(CH 2 )»(C 3 -Ci 2 )cycloalkyl > -(CH 2 )t-aryl, -(CH 2 ) t -heterocycloalkyl and -(CH 2 ) t heteroaryl; or two R 3 groups each attached to the same carbon atom of the ring containing Y, taken
  • n is 1. In another embodiment of the invention n is 2.
  • R 4 is hydrogen
  • n1 is 1.
  • n1 is 2.
  • p is 0. In another embodiment of the invention p is 1. In another embodiment of the invention p is 2.
  • Xi is O.
  • Xi is S.
  • each Ri is hydrogen. In another embodiment of the invention each R 2 is hydrogen.
  • each Ri is independently halogen or CrC 6 alkyl.
  • n is 1 and Ri is fluorine, chlorine, alkyl such as methyl, or trifluoroalkyl.
  • n is 2 and each Ri is fluorine.
  • n is 2 and each Ri is alkyl such as methyl.
  • n is 2 wherein one Ri is fluorine and the other R 1 is chlorine.
  • n is 2 wherein one Ri is alkyl such as methyl and the other Ri is chlorine or fluorine.
  • Y is N and each R 2 is independently halogen or alkyl.
  • each R 2 is independently fluorine, chlorine, or methyl.
  • Y is CH and each R 2 is independently halogen or alkyl. In one example of this embodiment, each R 2 is independently fluorine, chlorine, or methyl.
  • n 1
  • n is 2.
  • each R 2 is independently halogen, alkyl optionally substituted with one to three halogens, heteroaryl, or alkoxy optionally substituted with one to three halogens.
  • m is 1 and R 2 is fluorine, chlorine, alkyl optionally substituted with one to three halogens such as methyl or trifluoromethyl, or alkoxy optionally substituted with one to three halogens such as methoxy or trifluoromethoxy.
  • m is 2 and each R 2 is fluorine.
  • m is 2 and each R 2 is chlorine.
  • m is 2 and each R 2 is alkyl such as methyl.
  • m is 2 wherein one R 2 is fluorine and the other R 2 is chlorine.
  • m is 2 wherein one R 2 is alkyl such as methyl and the other R 2 is chlorine or fluorine. In another example of this embodiment, m is 2 wherein one R 2 is alkoxy such as methoxy and the other R 2 is chlorine or fluorine.
  • each R 3 is hydrogen.
  • Y is C.
  • Exemplary compounds according to the invention include the compounds disclosed in Table 1 herein or pharmaceutically acceptable salts thereof.
  • the compounds of formula I are useful for the treatment of a variety of neurological and psychiatric disorders, including but not limited to: acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, mild cognitive impairment, obesity,
  • the invention provides a method for treating a condition in a mammal, such as a human, selected from the conditions above, comprising administering a compound of formula I to the mammal.
  • the mammal is preferably a mammal in need of such treatment.
  • the invention provides a method for treating attention deficit/hyperactivity disorder, schizophrenia and Alzheimer's Disease.
  • the present invention provides methods of treating neurological and psychiatric disorders associated with norepinephrine reuptake inhibition, 5HT1a agonist activity, or both comprising: administering to a patient in need thereof an amount of a compound of formula I effective in treating such disorders.
  • the compound of formula I is optionally used in combination with another active agent.
  • Such an active agent may be, for example, an atypical antipsychotic or a cholinesterase inhibitor.
  • Such atypical antipsychotics include, but are not limited to, ziprasidone, clozapine, olanzapine, risperidone, quetiapine, aripiprazole, paliperidone, and such cholinesterase inhibitors include but are not limited to donepezil and galantamine.
  • the invention is also directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula I, and a pharmaceutically acceptable carrier.
  • the composition may be, for example, a composition for treating a condition selected from the group consisting of acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including but not limited to cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol,
  • alkyl refers to a linear or branched-chain saturated hydrocarbyl substituent (i.e., a substituent obtained from a hydrocarbon by removal of a hydrogen) containing from one to twenty carbon atoms; in one embodiment from one to twelve carbon atoms; in another embodiment, from one to ten carbon atoms; in another embodiment, from one to six carbon atoms; and in another embodiment, from one to four carbon atoms.
  • substituents include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl and tert-butyl), pentyl, iso-amyl, hexyl and the like.
  • benzyl refers to methyl radical substituted with phenyl, i.e.,
  • cycloalkyl refers to a saturated carbocyclic substituent having three to fourteen carbon atoms. In one embodiment, a cycloalkyl substituent has three to ten carbon atoms. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The term “cycloalkyl” also includes substituents that are fused to a C 6 -
  • Cio aromatic ring or to a 5-10-membered heteroaromatic ring wherein a group having such a fused cycloalkyl group as a substituent is bound to a carbon atom of the cycloalkyl group.
  • a fused cycloalkyl group is substituted with one or more substituents, the one or more substitutents, unless otherwise specified, are each bound to a carbon atom of the cycloalkyl group.
  • a cycloalkyl may be a single ring, which typically contains from 3 to 6 ring atoms. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Alternatively, 2 or 3 rings may be fused together, such as bicyclodecanyl and decalinyl.
  • aryl refers to an aromatic substituent containing one ring or two or three fused rings.
  • the aryl substituent may have six to eighteen carbon atoms. As an example, the aryl substituent may have six to fourteen carbon atoms.
  • aryl may refer to substituents such as phenyl, naphthyl and anthracenyl.
  • aryl also includes substituents such as phenyl, naphthyl and anthracenyl that are fused to a C 4 -Ci 0 carbocyclic ring, such as a C 5 or a C 6 carbocyclic ring, or to a 4-10-membered heterocyclic ring, wherein a group having such a fused aryl group as a substituent is bound to an aromatic carbon of the aryl group.
  • the one or more substitutents are each bound to an aromatic carbon of the fused aryl group.
  • aryl groups include accordingly phenyl, naphthalenyl, tetrahydronaphthalenyl (also known as "tetralinyl”), indenyl, isoindenyl, indanyl, anthracenyl, phenanthrenyl, benzonaphthenyl (also known as "phenalenyl”), and fluorenyl.
  • the number of carbon atoms in a hydrocarbyl substituent is indicated by the prefix “C x -Cy-,” wherein x is the minimum and y is the maximum number of carbon atoms in the substituent.
  • C x -Cy- refers to an alkyl substituent containing from 1 to 6 carbon atoms.
  • C 3 -C 6 -cycloalkyl refers to saturated cycloalkyl containing from 3 to 6 carbon ring atoms.
  • the number of atoms in a cyclic substituent containing one or more heteroatoms is indicated by the prefix "X-Y-membered", wherein wherein x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent.
  • X-Y-membered wherein wherein x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent.
  • 5-8-membered heterocycloalkyl refers to a heterocycloalkyl containing from 5 to 8 atoms, including one ore more heteroatoms, in the cyclic moiety of the heterocycloalkyl.
  • hydrogen refers to hydrogen substituent, and may be depicted as -H.
  • hydroxy refers to -OH.
  • the prefix "hydroxy” indicates that the substituent to which the prefix is attached is substituted with one or more hydroxy substituents.
  • Compounds bearing a carbon to which one or more hydroxy substituents include, for example, alcohols, enols and phenol.
  • hydroxyalkyl refers to an alkyl that is substituted with at least one hydroxy substituent.
  • examples of hydroxyalkyl include hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl.
  • cyano (also referred to as “nitrile”) means -CN, which also referred to as "nitrile).
  • carbonyl means -C(O)-, which also may be depicted as:
  • amino refers to -NH 2 .
  • alkylamino refers to an amino group, wherein at least one alkyl chain is bonded to the amino nitrogen in place of a hydrogen atom.
  • alkylamino substituents include monoalkylamino such as methylamino (exemplified by the formula -NH(CH 3 )), which may also be
  • dialkylamino such as dimethylamino
  • halogen refers to fluorine (which may be depicted as -F), chlorine (which may be depicted as -Cl), bromine (which may be depicted as -Br), or iodine (which may be depicted as -I).
  • the halogen is chlorine.
  • the halogen is a fluorine.
  • the prefix "halo" indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen substituents.
  • haloalkyl refers to an alkyl that is substituted with at least one halogen substituent. Where more than one hydrogen is replaced with halogens, the halogens may be the identical or different.
  • haloalkyls include chloromethyl, dichloromethyl, difluorochloromethyl, dichlorofluoromethyl, trichloromethyl, 1 -bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, difluoroethyl, pentafluoroethyl, difluoropropyl, dichloropropyl, and heptafluoropropyl.
  • haloalkoxy refers to an alkoxy that is substituted with at least one halogen substituent.
  • haloalkoxy substituents include chloromethoxy, 1 -bromoethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy (also known as "perfluoromethyloxy"), and 2,2,2-trifluoroethoxy. It should be recognized that if a substituent is substituted by more than one halogen substituent, those halogen substituents may be identical or different (unless otherwise stated).
  • oxy refers to an ether substituent, and may be depicted as -O-.
  • alkoxy refers to an alkyl linked to an oxygen, which may also be represented as
  • alkoxy examples include methoxy, ethoxy, propoxy and butoxy.
  • heterocycloalkyl refers to a saturated or partially saturated ring structure containing a total of 3 to 14 ring atoms. At least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur.
  • a heterocycloalkyl alternatively may comprise 2 or 3 rings fused together, wherein at least one such ring contains a heteroatom as a ring atom (i.e., nitrogen, oxygen, or sulfur).
  • the ring atom of the heterocycloalkyl substituent that is bound to the group may be the at least one heteroatom, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom.
  • the group or substituent may be bound to the at least one heteroatom, or it may be bound to a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom.
  • heterocycloalkyl also includes substituents that are fused to a C ⁇ -Cio aromatic ring or to a 5-10-membered heteroaromatic ring, wherein a group having such a fused heterocycloalkyl group as a substituent is bound to a heteroatom of the heterocyclocalkyl group or to a carbon atom of the heterocycloalkyl group.
  • a fused heterocycloalkyl group is substituted with one more substituents, the one or more substitutents, unless otherwise specified, are each bound to a heteroatom of the heterocyclocalkyl group or to a carbon atom of the heterocycloalkyl group.
  • heteroaryl refers to an aromatic ring structure containing from 5 to 14 ring atoms in which at least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur.
  • a heteroaryl may be a single ring or 2 or 3 fused rings.
  • heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl; 5-membered ring substituents such as triazolyl, imidazolyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1 ,2,3-, 1 ,2,4-, 1 ,2,5-, or 1 ,3,4-oxadiazolyl and isothiazolyl; 6/5-membered fused ring substituents such as benzothiofuranyl, isobenzothiofuranyl, benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl; and 6/6-membered fused rings such as quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and
  • the ring atom of the heteroaryl substituent that is bound to the group may be the at least one heteroatom, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom.
  • heteroaryl also includes pyridyl N-oxides and groups containing a pyridine N-oxide ring.
  • single-ring heteroaryls include furanyl, dihydrofuranyl, tetradydrofuranyl, thiophenyl (also known as "thiofuranyl"), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thia ⁇ diazolyl, ox
  • 2-fused-ring heteroaryls include, indolizinyl, pyrindinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl (including pyrido[3,4-b]-pyridinyl, pyrido[3,2-b]-pyridinyl, or pyrido[4,3-b]-pyridinyl), and pteridinyl, indolyl, isoindolyl, indoleninyl, isoindazolyl, benzazinyl, phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl, benzopyranyl, benzothiopyranyl, benzoxazolyl, indoxazinyl, anthranilyl, benzodioxolyl, benzodioxanyl, benzo
  • 3-fused-ring heteroaryls or heterocycloalkyls include 5,6-dihydro-4H-imidazo[4,5, 1 -ij]quinoline, 4,5-dihydroimidazo[4,5, 1 -h ⁇ ndole, 4,5,6,7-tetrahydroimidazo[4,5,1-jk][1]benzazepine, and dibenzofuranyl.
  • fused-ring heteroaryls include benzo-fused heteroaryls such as indolyl, isoindolyl (also known as “isobenzazolyl” or “pseudoisoindolyl”), indoleninyl (also known as “pseudoindolyl”), isoindazolyl (also known as “benzpyrazolyl”), benzazinyl (including quinolinyl (also known as “1 -benzazinyl”) or isoquinolinyl (also known as "2-benzazinyl”)), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (also known as “1 ,2-benzodiazinyl”) or quinazolinyl (also known as “1 ,3-benzodiazinyl”)), benzopyranyl (including “chromanyl” or “isochromanyl”), benzopyr
  • heteroaryl also includes substituents such as pyridyl and quinolinyl that are fused to a C 4 -Ci 0 carbocyclic ring, such as a C 5 or a C 6 carbocyclic ring, or to a 4-10-membered heterocyclic ring, wherein a group having such a fused aryl group as a substituent is bound to an aromatic carbon of the heteroaryl group or to a heteroatom of the heteroaryl group.
  • the one or more substitutents are each bound to an aromatic carbon of the heteroaryl group or to a heteroatom of the heteroaryl group.
  • a substituent is "substitutable" if it comprises at least one carbon, sulfur, oxygen or nitrogen atom that is bonded to one or more hydrogen atoms. Thus, for example, hydrogen, halogen, and cyano do not fall within this definition. If a substituent is described as being “substituted,” a non-hydrogen substituent is in the place of a hydrogen substituent on a carbon, oxygen, sulfur or nitrogen of the substituent. Thus, for example, a substituted alkyl substituent is an alkyl substituent wherein at least one non-hydrogen substituent is in the place of a hydrogen substituent on the alkyl substituent.
  • monofluoroalkyl is alkyl substituted with a fluoro substituent
  • difluoroalkyl is alkyl substituted with two fluoro substituents. It should be recognized that if there is more than one substitution on a substituent, each non-hydrogen substituent may be identical or different (unless otherwise stated). If a substituent is described as being “optionally substituted,” the substituent may be either (1 ) not substituted, or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the carbon (to the extent there are any) may separately and/or together be replaced with an independently selected optional substituent.
  • a nitrogen of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the nitrogen (to the extent there are any) may each be replaced with an independently selected optional substituent.
  • One exemplary substituent may be depicted as -N R' R," wherein R' and R" together with the nitrogen atom to which they are attached, may form a heterocyclic ring.
  • the heterocyclic ring formed from R' and R" together with the nitrogen atom to which they are attached may be partially or fully saturated.
  • the heterocyclic ring consists of 3 to 7 atoms.
  • the heterocyclic ring is selected from the group consisting of pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridyl and thiazolyl.
  • a group of substituents are collectively described as being optionally substituted by one or more of a list of substituents, the group may include: (1) unsubstitutable substituents, (2) substitutable substituents that are not substituted by the optional substituents, and/or (3) substitutable substituents that are substituted by one or more of the optional substituents.
  • a substituent is described as being optionally substituted with up to a particular number of non-hydrogen substituents, that substituent may be either (1 ) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituent, whichever is less.
  • substituent may be either (1 ) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituent, whichever is less.
  • any heteroaryl with less than 3 substitutable positions would be optionally substituted by up to only as many non-hydrogen substituents as the heteroaryl has substitutable positions.
  • tetrazolyl (which has only one substitutable position) would be optionally substituted with up to one non-hydrogen substituent.
  • an amino nitrogen is described as being optionally substituted with up to 2 non- hydrogen substituents, then the nitrogen will be optionally substituted with up to 2 non-hydrogen substituents if the amino nitrogen is a primary nitrogen, whereas the amino nitrogen will be optionally substituted with up to only 1 non-hydrogen substituent if the amino nitrogen is a secondary nitrogen.
  • alkylcycloalkyl contains two moieties: alkyl and cycloalkyl.
  • a d-C 6 - prefix on CrC 6 -alkylcycloalkyl means that the alkyl moiety of the alkylcycloalkyl contains from 1 to 6 carbon atoms; the CrC 6 - prefix does not describe the cycloalkyl moiety.
  • the prefix "halo" on haloalkoxyalkyl indicates that only the alkoxy moiety of the alkoxyalkyl substituent is substituted with one or more halogen substituents.
  • the compound may exist in the form of optical isomers (enantiomers).
  • the present invention comprises enantiomers and mixtures, including racemic mixtures of the compounds of formula I.
  • the present invention comprises diastereomeric forms (individual diastereomers and mixtures thereof) of compounds.
  • geometric isomers may arise.
  • the present invention comprises the tautomeric forms of compounds of formula I.
  • tautomeric isomerism 'tautomerism'
  • This can take the form of proton tautomerism in compounds of formula I containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
  • the various ratios of the tautomers in solid and liquid form is dependent on the various substituents on the molecule as well as the particular crystallization technique used to isolate a compound.
  • the compounds of this invention may be used in the form of salts derived from inorganic or organic acids.
  • a salt of the compound may be advantageous due to one or more of the salt's physical properties, such as enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil.
  • a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
  • the salt preferably is pharmaceutically acceptable.
  • pharmaceutically acceptable salt refers to a salt prepared by combining a compound of formula I with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption.
  • Pharmaceutically acceptable salts are particularly useful as products of the methods of the present invention because of their greater aqueous solubility relative to the parent compound.
  • salts of the compounds of this invention are non-toxic “pharmaceutically acceptable salts.”
  • Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid.
  • Suitable pharmaceutically acceptable acid addition salts of the compounds of the present invention when possible include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids.
  • Suitable organic acids generally include, for example, aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids.
  • suitable organic acids include acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, mesylate, stearate, salicylate, p-hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2-hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, ⁇ -hydroxybutyric acid, galactarate, galacturonate, adipate, alginate, butyrate, camphorate
  • suitable pharmaceutically acceptable salts thereof may include alkali metal salts, i.e., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts.
  • base salts are formed from bases which form non-toxic salts, including aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
  • Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.
  • secondary, tertiary or quaternary amine salts such as tromethamine, diethylamine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine.
  • Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (CrC 6 ) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (i.e., dimethyl, diethyl, dibuytl, and diamyl sulfates), long chain halides (i.e., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (i.e., benzyl and phenethyl bromides), and others.
  • lower alkyl (CrC 6 ) halides e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides
  • dialkyl sulfates i.e., dimethyl, die
  • hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • the compounds of the invention and their salts may exist in both unsolvated and solvated forms.
  • prodrugs of the compound of the invention.
  • certain derivatives of the compound of the invention which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into the compound of the invention having the desired activity, for example, by hydrolytic cleavage.
  • Such derivatives are referred to as “prodrugs.” Further information on the use of prodrugs may be found in "Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and "Bioreversible Carriers in Drug Design,” Pergamon Press, 1987 (ed. E B Roche, American Pharmaceutical Association).
  • Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of any of formula I with certain moieties known to those skilled in the art as "pro-moieties” as described, for example, in “Design of Prodrugs” by H Bundgaard (Elsevier, 1985).
  • the present invention also includes isotopically labelled compounds, which are identical to those recited in formula I, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F, and 36 CI, respectively.
  • Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention.
  • Certain isotopically labelled compounds of the present invention, for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • Isotopically labelled compounds of formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • a compound of the invention is administered in an amount effective to treat a condition as described herein.
  • the compounds of the invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • Therapeutically effective doses of the compounds required to treat the progress of the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
  • the compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
  • the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally.
  • the compounds of the invention can also be administered intranasally or by inhalation.
  • the compounds of the invention may be administered rectally or vaginally.
  • the compounds of the invention may also be administered directly to the eye or ear.
  • the dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions. In one embodiment, the total daily dose of a compound of the invention (administered in single or divided doses) is typically from about 0.01 to about 100 mg/kg.
  • total daily dose of the compound of the invention is from about 0.1 to about 50 mg/kg, and in another embodiment, from about 0.5 to about 30 mg/kg (i.e., mg compound of the invention per kg body weight). In one embodiment, dosing is from 0.01 to 10 mg/kg/day. In another embodiment, dosing is from 0.1 to 1.0 mg/kg/day. Dosage unit compositions may contain such amounts or submultiples thereof to make up the daily dose. In many instances, the administration of the compound will be repeated a plurality of times in a day (typically no greater than 4 times). Multiple doses per day typically may be used to increase the total daily dose, if desired.
  • compositions may be provided in the form of tablets containing 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 75.0, 100, 125, 150, 175, 200, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1mg to about 100 mg of active ingredient.
  • doses may range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
  • Suitable subjects according to the present invention include mammalian subjects. Mammals according to the present invention include, but are not limited to, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development. Use in the Preparation of a Medicament
  • the invention comprises the use of one or more compounds of the invention for the preparation of a medicament for the treatment of the conditions recited herein.
  • the compound of the invention can be administered as compound per se.
  • pharmaceutically acceptable salts are suitable for medical applications because of their greater aqueous solubility relative to the parent compound.
  • the present invention comprises pharmaceutical compositions.
  • Such pharmaceutical compositions comprise a compound of the invention presented with a pharmaceutically-acceptable carrier.
  • the carrier can be a solid, a liquid, or both, and may be formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compounds.
  • a compound of the invention may be coupled with suitable polymers as targetable drug carriers. Other pharmacologically active substances can also be present.
  • the compounds of the present invention may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • the active compounds and compositions may be administered orally, rectally, parenterally, or topically.
  • Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention.
  • the oral administration may be in a powder or granule form.
  • the oral dose form is sub-lingual, such as, for example, a lozenge.
  • the compounds of formula I are ordinarily combined with one or more adjuvants.
  • Such capsules or tablets may contain a controlled-release formulation.
  • the dosage forms also may comprise buffering agentsor may be prepared with enteric coatings.
  • oral administration may be in a liquid dose form.
  • Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (i.e., water).
  • Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
  • the present invention comprises a parenteral dose form.
  • Parenteral administration includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion.
  • injectable preparations i.e., sterile injectable aqueous or oleaginous suspensions
  • topical administration includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration.
  • compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams.
  • a topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas.
  • administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol.
  • Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958, by Finnin and Morgan (October 1999).
  • Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in suitable carrier.
  • a typical formulation suitable for ocular or aural administration may be in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant.
  • Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2- tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the present invention comprises a rectal dose form.
  • rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • Other carrier materials and modes of administration known in the pharmaceutical art may also be used.
  • Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks.
  • the compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment of various conditions or disease states.
  • the compound(s) of the present invention and other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially.
  • An exemplary therapeutic agent may be, for example, a metabotropic glutamate receptor agonist.
  • the administration of two or more compounds "in combination” means that the two compounds are administered closely enough in time that the presence of one alters the biological effects of the other.
  • the two or more compounds may be administered simultaneously, concurrently or sequentially. Additionally, simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration.
  • simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration.
  • the phrases “concurrent administration,” “co-administration,” “simultaneous administration,” and “administered simultaneously” mean that the compounds are administered in combination.
  • Kits The present invention further comprises kits that are suitable for use in performing the methods of treatment described above.
  • the kit contains a first dosage form comprising one or more of the compounds of the present invention and a container for the dosage, in quantities sufficient to carry out the methods of the present invention.
  • the kit of the present invention comprises one or more compounds of the invention.
  • the invention relates to the novel intermediates useful for preparing the compounds of the invention.
  • the compounds of the formula I may be prepared by the methods described below, together with synthetic methods known in the art of organic chemistry, or modifications and derivatisations that are familiar to those of ordinary skill in the art.
  • the starting materials used herein are commercially available or may be prepared by routine methods known in the art (such as those methods disclosed in standard reference books such as the COMPENDIUM OF ORGANIC SYNTHETIC METHODS, Vol. I-VI (published by Wiley-lnterscience)). Preferred methods include, but are not limited to, those described below.
  • Preferred methods include, but are not limited to, those described below.
  • n-Butyllithium (924 ml_, 2.3 mol, 2.5 M in hexane) was added slowly to a solution of diisopropylamine (323.3 ml_, 3.2 mol) in THF (6 L) at -78 °C and stirred for 1 h.
  • a solution of tert-butyl 4-oxopiperidine-i-carboxylate (400 g, 2.01 mol) in THF (2 L) was added and the mixture was stirred for 1 h at -78 0 C.
  • the reaction mixture was heated at 100 ° C overnight, cooled to room temperature, and the solvent was evaporated. To the residue was added ethyl acetate, and the suspension was filtered through a pad of Celite. The filtrate was washed with water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure.
  • the crude product was purified by flash column chromatography (hexanes/ ethyl acetate, 15:1 - 8:1) to give the te/t-butyl 4-(2-phenoxypyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (1.86 g, 53 %).
  • Ci 6 H 16 F 2 N 2 O-1.7HCI C 54.74 %; H 5.07 %; N 7.98 %, and found: C 54.93 %;
  • Ci 7 H 19 FN 2 O 2 /2HCI 302.99 [M+1] + .
  • Table 1 shows the structure of the compounds and relevant biological data that were measured in each case either on the compound as a free base or on the pharmaceutically acceptable salt of the compound disclosed in the Table.
  • S refers to SPA Binding Assay
  • F refers to Filtration Binding Assay. Each assay is disclosed in greater detail hereinbelow.
  • Cell pellets (7 to 15 g) were thawed on ice and resuspended in 3 ml of membrane prep buffer (20 mM HEPES, pH 7.4 @24°C, 1 complete protease inhibitor tablet ((Roche catalog #11697498001) /50 ml buffer) per ml of packed cell volume, using a vortex mixer to disperse the cell pellet. The cells were then allowed to swell on ice for 10 min. The suspension was homogenized for 4 x 10 second bursts at 25,000 rpm on a PT3100 polytron. The homogenate was spun at 3,000 rpm for 20 min in a SS34 rotor cooled to 4 0 C.
  • membrane prep buffer 20 mM HEPES, pH 7.4 @24°C, 1 complete protease inhibitor tablet ((Roche catalog #11697498001) /50 ml buffer) per ml of packed cell volume, using a vortex mixer to disperse the cell pellet.
  • the supernatant was decanted into new 50 ml centrifuge tubes and the pellets were rehomogenized in 10-15 ml of membrane prep buffer and spun as described above. The supernatants were then pooled and spun at 17,500 rpm in the SS34 rotor (35,000 x g) for 30 min at 4 0 C. The supernatants were discarded and the cell pellets were resuspended by vigorous pipetting in 1 ml membrane prep buffer per 1 ml of original packed cell volume. Protein concentrations were determined with the BCA Protein Assay Kit (Pierce, #23227) utilizing the microplate assay procedure. The membranes were stored in 1 ml aliquots at -8O 0 C until use.
  • SPA beads Wood germ agglutinin coated polyvinyltoluene
  • HEK-293 EBNA cell membrane (Catalog # RBHS1AM) stably expressing the 5HT1a receptor was purchased from Perkin Elmer. The cells were grown under standard cell culture techniques, harvested, and received frozen suspended in 5OmM TRIS-HCI, 0.5mM EDTA, 1OmM MgCI 2 , 10% sucrose, pH to 7.4. The company provides both receptor concentration (Bmax), ligand affinity (Kd), and also calculates the protein concentration. Subsequently, these membrane preparations are stored at -8O 0 C until needed. Lyophilized wheat germ agglutinin SPA beads (WGA-SPA) are reconstituted in 10 ml_ of assay buffer (5OmM TRIS MgCI 2 , pH 7.4).
  • WGA-SPA Lyophilized wheat germ agglutinin SPA beads
  • the bead suspension is added to the cell membranes to maintain a bead concentration of 500 ⁇ g/well and a protein concentration of 10 ⁇ g/ well (384-well plate format). This bead/membrane mixture is rocked gently for 30 minutes at 4 0 C. After 30 minutes the suspension is then centrifuged at 1000 rpm at 25 0 C. The supernatant is carefully discarded and the remaining pellet is diluted in assay buffer and aliquoted into assay plates to initiate incubation.
  • Binding studies were carried out in 50 ⁇ l_ assay volume using Costar 3705 (white, clear bottom) 384 well plates to which 0.5-1.0 ml_ of compound in 100% DMSO was prespotted, 20 ⁇ L of [3H]-8-OH-DPAT (final concentration of 5nM and 30 ⁇ L of bead/membrane slurry are added sequentially using Titertek Multidrops. Plates are sealed with clear adhesive and allowed to incubate overnight at 25 0 C. The nonspecific binding was defined by using 1 ⁇ M Lisuride.
  • the plates were counted in a Packard Trilux Microbeta Scintillation Counter using a normalized protocol for 3H-SPA counting
  • the data was analyzed using an Excel macro which used the statistical average for the high control (Total Binding) values and the 1 ⁇ M lisuride values as the low control (Nonspecific binding) values. Z factors were determined for each plate using these values. Active compounds were further titrated to determine Ki values. Ki values were determined by analysis using a least squares nonlinear regression curve fit assuming a one site competition ligand binding model. 5HT1A Filtration Binding Assay
  • HeLa cells transfected with the human 5HT1A receptor were incubated in a solution containing 50OuI of 50 mM Tris-HCI, 10 mM MgSO4, 0.5 mM EDTA, 0.1% Ascorbic acid at pH 7.4 (solution A) and the diluted test agent, plus 1.5 nM [3H]8-OH-DPAT in a 96- well format. In cases where non-specific binding was determined 1OuM of [3H]8-OH-DPAT was used. The solution was left to incubate at 4C for 1 hour. The assay is terminated by filtration of solution over GF/C filters that have been pre-soaked in 0.3% polyethylenimine with solution A.
  • CHO cells transfected with human 5 HT1 A receptor were maintained in Ultra CHO (Cambrex, 12-724Q) media supplemented with 10% dialyzed FBS (Gibco 26400 036), 1% Geneticin (Gibco 10131-027), and 0.5% Pen/Strep (Gibco 15140 122).
  • Cells are grown in 15OmM cell culture dishes and harvested in ice-cold PBS by scraping the plates when the cells are approx. 80% confluent. Cells are collected in PBS and centrifuged at 4 0 C at low speed (2500 rpm) until pellet forms.
  • the pellet is then homogenized in tissue buffer with a polytron, and centrifuged at 4 0 C at high speed (20,000 rpm) for 15 min. Repeat this washing at least 4 times. After final wash, the protein can be measured and the homogenate aliquoted appropriately. .
  • Working stocks were prepared as followed:

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Neurology (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compounds and pharmaceutically acceptable salts of the compounds are disclosed, wherein the compounds have the structure of Formula (I), as defined in the specification. Corresponding pharmaceutical compositions, methods of treatment, methods of synthesis, and intermediates are also disclosed.

Description

PHENOXY-PYRIDYL DERIVATIVES
FIELD OF THE INVENTION
The present invention comprises a novel class of compounds having the structure of formula I (including tautomers and salts of those compounds) and pharmaceutical compositions comprising a compound of formula I. The present invention also comprises methods of treating a subject by administering a therapeutically effective amount of a compound of formula I to the subject. These compounds are useful for the conditions disclosed herein. The present invention further comprises methods for making the compounds of formula I and corresponding intermediates.
BACKGROUND OF THE INVENTION
Many types of mental, behavioral, and neurological disorders originate from disturbances in neuronal circuits that convey signals using certain monoamine neurotransmitters. Monoamine neurotransmitters include, for example, norepinephrine (noradrenaline), serotonin (5-HT), and dopamine. Lower-than-normal levels of these neurotransmitters are associated with a variety of symptoms including lack of energy, motivation, and interest in life. Thus, a normal level of monoaminergic neurotransmission is essential to maintaining drive and capacity for reward. These neurotransmitters travel from the terminal of a neuron across a small gap (i.e., the synaptic cleft) and bind to receptor molecules on the surface of a second neuron. This binding elicits intracellular changes that initiate or activate a response or change in the postsynaptic neuron. Inactivation occurs primarily by transport (i.e., reuptake) of the neurotransmitter back into the presynaptic neuron. Abnormality in noradrenergic transmission results in various types of depression, mental, behavioral, and neurological disorders attributed to a variety of symptoms including a lack of energy, motivation, and interest in life. See generally, R. J. Baldessarini, "Drugs and the Treatment of Psychiatric Disorders: Depression and Mania" in Goodman and Gilman's The Pharmacological Basis of Therapeutics, McGraw-Hill. N.Y., N.Y., pp. 432 439 (1996).
Drugs that impact monoamine neurotransmission either directly as a result of efficacy at postsynaptic receptors or indirectly through inhibition of reuptake into presynaptic terminals are widely used to treat disorders including attention deficit hyperactivity disorder (ADHD), depression, and anxiety. For example, in ADHD stimulant medications that increase norepinephrine and dopamine neurotransmission throughout the brain are commonly prescribed, but carry a burden of abuse liability. Alternatively, norepinephrine reuptake inhibitors (NRI) which more selectively increase norepinephrine neurotransmission with less impact on dopamine, are also used to treat ADHD albeit with less efficacy and a slower onset of action than stimulant medications. In addition, NRI agents are widely used to treat symptoms of depression and anxiety, although again taking many days before full efficacy is demonstrated. Finally compounds with serotoniniA (5-HT1A) receptor partial agonist properties are commonly used to treat symptoms of depression and anxiety, and evidence indicates that they could be used to treat symptoms of ADHD and other disorders associated with deficits in cognitive function like schizophrenia, bipolar disorders and Alzheimer's disease as well.
Studies indicate that adding 5-HT1A partial agonist properties to a norepinephrine reuptake inhibitor will produce efficacy comparable to stimulants with a fast onset of action in treating the primary symptoms of ADHD, but without the abuse liability associated with stimulant medications. Adding 5-HT1 A partial agonism to an NRI agent increases extracellular levels of both norepinephrine and dopamine within the prefrontal cortex, a region of the brain important for attention and executive function, to a degree similar to stimulant medications. In contrast and unlike stimulants, this combination of pharmacologies does not impact extracellular dopamine levels within the nucleus accumbens, a region of the brain associated with reward behavior and abuse liability, to a degree greater than seen with NRI agents alone. Moreover, preclinical behavioral studies measuring antidepressant efficacy and cognitive function demonstate that compounds possessing both NRI and 5-HT1A partial agonist properties are more effective than NRI agents alone. Thus, the present invention describes agents possessing NRI and 5-HT1A partial agonist properties that will be useful in treating symptoms of disorders wherein modulation of monoamine neurotransmission is required such as ADHD, depression, anxiety, bipolar disorders and Alzheimer's disease.
Accordingly, it would be desirable to have compounds acting as both NRI and 5-HT1A agonists to treat individuals suffering from a variety of conditions where inhibiting reuptake of norepinephrine, and having 5-HT1A agonist activity, provides a benefit while reducing or eliminating the adverse side effects associated with conventional norepinephrine reuptake inhibitors. There also is a need for the use of such compounds to treat disorders of the central nervous system that are associated with deficits or imbalances in catecholamine neurotransmission. Furthermore, there is a need for pharmaceutical compositions containing such compounds. Still further, there is a need for medicaments containing such compounds, and the use of such compositions in the manufacture of such compounds.
SUMMARY OF THE INVENTION
The invention is directed to a class of compounds, including the pharmaceutically acceptable salts of the compounds, having the structure of formula I:
Figure imgf000005_0001
I or a pharmaceutically acceptable salt thereof, wherein:
X 1 is O or S(O)P; is either a single bond or a double bond, wherein, when is a single bond, Y is N or CR3, and when is a double bond, Y is C; each Ri is independently selected from the group consisting of hydrogen, halogen, alkoxy, -CN, and alkyl, wherein each Ri alkyl is optionally independently substituted by one to three halogens; each R2 is independently selected from the group consisting of halogen, -NO2, -CN, -N(R4)2, R4, and -OR4; or two R2 substituents on adjacent carbons taken together with the adjacent carbons form a carbocyclic or heterocyclic ring optionally substituted with halogen, hydroxy, -CN, alkyl, or alkoxy; wherein each R4 is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heterocycloalkyl and heteroaryl; or two R4 groups on the same nitrogen atom may be taken together with the nitrogen atom to form a 5 to 8 membered heterocyclic ring which optionally has 1 to 3 additional ring heteroatoms selected from the group consisting of N, O, and S; and each R4 alkyl, cycloalkyl, aryl, heterocycloalkyl and heteroaryl is optionally independently substituted by one to three substituents independently selected from the group consisting of halogen, hydroxy, -CN, -(CrCβ)alkyl, -(CrC6)alkoxy, -CF3, -OCF3, -N[(CH2)tR4]2, -NO2, -(CH2)tN[(CH2)tR4](C=O)[(CH2),R4], -S(Ci-C6)alkyl, -(S=O)(CrCβ)alkyl, -S(=O)2(C1-C6)alkyl, -(C=O)O(Ci -C6)alkyl, -O(C=O)(CrC6)alkyl,
-(C=O)(Ci -C6)alkyl, cycloalkyl, aryl, heterocycloalkyl and heteroaryl; each R3 is independently selected from the group consisting of hydrogen, halogen, -(CH2)tOH, -(CH2)tCF3, -(CH2)tC≡N, -NO2, -(CH2)tN[(CH2)tR4]2l -(CH2),-alkyl, -(CH2)tO[(CH2)tR4], -(CH2)»(C3-Ci2)cycloalkyl> -(CH2)t-aryl, -(CH2)t-heterocycloalkyl and -(CH2)theteroaryl; or two R3 groups each attached to the same carbon atom of the ring containing Y, taken together with the carbon atom, form a three to seven membered carbocyclic ring; n is an integer selected from 0, 1 and 2; m is an integer selected from 0, 1 , 2, 3 and 4; n1 is an integer selected from 1 , 2, and 3; p is an integer selected from 0, 1 and 2; and each t is an integer independently selected from 0, 1 , 2, 3, 4, and 5.
In one embodiment of the invention n is 1. In another embodiment of the invention n is 2.
In another embodiment of the invention R4 is hydrogen.
In another embodiment of the invention n1 is 1.
In another embodiment of the invention n1 is 2.
In another embodiment of the invention p is 0. In another embodiment of the invention p is 1. In another embodiment of the invention p is 2.
In one embodiment of the invention Xi is O.
In another embodiment of the invention Xi is S.
In another embodiment of the invention each Ri is hydrogen. In another embodiment of the invention each R2 is hydrogen.
In another embodiment of the invention, each Ri is independently halogen or CrC6 alkyl. In one example of this embodiment, n is 1 and Ri is fluorine, chlorine, alkyl such as methyl, or trifluoroalkyl. In another example of this embodiment, n is 2 and each Ri is fluorine. In another example of this embodiment, n is 2 and each Ri is alkyl such as methyl. In another example of this embodiment, n is 2 wherein one Ri is fluorine and the other R1 is chlorine. In another example of this embodiment, n is 2 wherein one Ri is alkyl such as methyl and the other Ri is chlorine or fluorine.
In another embodiment of the invention Y is N and each R2 is independently halogen or alkyl.
In one example of this embodiment, each R2 is independently fluorine, chlorine, or methyl.
In another embodiment of the invention Y is CH and each R2 is independently halogen or alkyl. In one example of this embodiment, each R2 is independently fluorine, chlorine, or methyl.
In another embodiment of the invention, m is 1.
In another embodiment of the invention, m is 2.
In another embodiment of the invention, each R2 is independently halogen, alkyl optionally substituted with one to three halogens, heteroaryl, or alkoxy optionally substituted with one to three halogens.
In one example of this embodiment, m is 1 and R2 is fluorine, chlorine, alkyl optionally substituted with one to three halogens such as methyl or trifluoromethyl, or alkoxy optionally substituted with one to three halogens such as methoxy or trifluoromethoxy. In another example of this embodiment, m is 2 and each R2 is fluorine. In another example of this embodiment, m is 2 and each R2 is chlorine. In another example of this embodiment, m is 2 and each R2 is alkyl such as methyl. In another example of this embodiment, m is 2 wherein one R2 is fluorine and the other R2 is chlorine. In another example of this embodiment, m is 2 wherein one R2 is alkyl such as methyl and the other R2 is chlorine or fluorine. In another example of this embodiment, m is 2 wherein one R2 is alkoxy such as methoxy and the other R2 is chlorine or fluorine.
In another embodiment of the invention, each R3 is hydrogen.
In another embodiment of the invention, is a single bond and Y is N.
In another embodiment of the invention, is a single bond and Y is CR3.
In another embodiment of the invention, is a double bond and
Y is C. Exemplary compounds according to the invention include the compounds disclosed in Table 1 herein or pharmaceutically acceptable salts thereof.
The compounds of formula I are useful for the treatment of a variety of neurological and psychiatric disorders, including but not limited to: acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, mild cognitive impairment, obesity, schizophrenia, anxiety (including generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive compulsive disorder), mood disorders (including depression, mania, bipolar disorders), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hype ractivity disorder, autism, Asperger's disease, and conduct disorder. Accordingly, in one embodiment, the invention provides a method for treating a condition in a mammal, such as a human, selected from the conditions above, comprising administering a compound of formula I to the mammal. The mammal is preferably a mammal in need of such treatment. As examples, the invention provides a method for treating attention deficit/hyperactivity disorder, schizophrenia and Alzheimer's Disease.
In another embodiment the present invention provides methods of treating neurological and psychiatric disorders associated with norepinephrine reuptake inhibition, 5HT1a agonist activity, or both comprising: administering to a patient in need thereof an amount of a compound of formula I effective in treating such disorders. The compound of formula I is optionally used in combination with another active agent. Such an active agent may be, for example, an atypical antipsychotic or a cholinesterase inhibitor. Such atypical antipsychotics include, but are not limited to, ziprasidone, clozapine, olanzapine, risperidone, quetiapine, aripiprazole, paliperidone, and such cholinesterase inhibitors include but are not limited to donepezil and galantamine.
The invention is also directed to a pharmaceutical composition comprising a compound of formula I, and a pharmaceutically acceptable carrier. The composition may be, for example, a composition for treating a condition selected from the group consisting of acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including but not limited to cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, mild cognitive impairment, obesity, schizophrenia, anxiety (including generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive compulsive disorder), mood disorders (including depression, mania, bipolar disorders), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hyperactivity disorder, autism, Asperger's disease, and conduct disorder, wherein the composition contains an amount of the compound of formula I that is effective in the treatment of such conditions. DETAILED DESCRIPTION OF THE INVENTION
This detailed description of embodiments is intended only to acquaint others skilled in the art with Applicants' invention, its principles, and its practical application so that others skilled in the art may adapt and apply the invention in its numerous forms, as it may be best suited to the requirements of a particular use. This invention, therefore, is not limited to the embodiments described in this specification, and may be variously modified.
Abbreviations and Definitions TABLE A - Abbreviations
Figure imgf000011_0001
Figure imgf000012_0001
The term "alkyl" refers to a linear or branched-chain saturated hydrocarbyl substituent (i.e., a substituent obtained from a hydrocarbon by removal of a hydrogen) containing from one to twenty carbon atoms; in one embodiment from one to twelve carbon atoms; in another embodiment, from one to ten carbon atoms; in another embodiment, from one to six carbon atoms; and in another embodiment, from one to four carbon atoms. Examples of such substituents include methyl, ethyl, propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, sec-butyl and tert-butyl), pentyl, iso-amyl, hexyl and the like.
The term "benzyl" refers to methyl radical substituted with phenyl, i.e.,
the following structure:
Figure imgf000013_0001
The term "cycloalkyl" refers to a saturated carbocyclic substituent having three to fourteen carbon atoms. In one embodiment, a cycloalkyl substituent has three to ten carbon atoms. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The term "cycloalkyl" also includes substituents that are fused to a C6-
Cio aromatic ring or to a 5-10-membered heteroaromatic ring, wherein a group having such a fused cycloalkyl group as a substituent is bound to a carbon atom of the cycloalkyl group. When such a fused cycloalkyl group is substituted with one or more substituents, the one or more substitutents, unless otherwise specified, are each bound to a carbon atom of the cycloalkyl group. The fused C6-Ci0 aromatic ring or to a 5-10-membered heteroaromatic ring may be optionally substituted with halogen, CrC6 alkyl, C3-Ci0 cycloalkyl, or =O.
A cycloalkyl may be a single ring, which typically contains from 3 to 6 ring atoms. Examples include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Alternatively, 2 or 3 rings may be fused together, such as bicyclodecanyl and decalinyl.
The term "aryl" refers to an aromatic substituent containing one ring or two or three fused rings. The aryl substituent may have six to eighteen carbon atoms. As an example, the aryl substituent may have six to fourteen carbon atoms. The term "aryl" may refer to substituents such as phenyl, naphthyl and anthracenyl. The term "aryl" also includes substituents such as phenyl, naphthyl and anthracenyl that are fused to a C4-Ci0 carbocyclic ring, such as a C5 or a C6 carbocyclic ring, or to a 4-10-membered heterocyclic ring, wherein a group having such a fused aryl group as a substituent is bound to an aromatic carbon of the aryl group. When such a fused aryl group is substituted with one more substituents, the one or more substitutents, unless otherwise specified, are each bound to an aromatic carbon of the fused aryl group. The fused C4-Ci0 carbocyclic or 4-10-membered heterocyclic ring may be optionally substituted with halogen, d-Cβ alkyl, C3- C10 cycloalkyl, or =0. Examples of aryl groups include accordingly phenyl, naphthalenyl, tetrahydronaphthalenyl (also known as "tetralinyl"), indenyl, isoindenyl, indanyl, anthracenyl, phenanthrenyl, benzonaphthenyl (also known as "phenalenyl"), and fluorenyl.
In some instances, the number of carbon atoms in a hydrocarbyl substituent (i.e., alkyl, alkenyl, cycloalkyl, cycloalkenyl, aryl, etc.) is indicated by the prefix "Cx-Cy-," wherein x is the minimum and y is the maximum number of carbon atoms in the substituent. Thus, for example, "CrCβ-alky!" refers to an alkyl substituent containing from 1 to 6 carbon atoms. Illustrating further, C3-C6-cycloalkyl refers to saturated cycloalkyl containing from 3 to 6 carbon ring atoms.
In some instances, the number of atoms in a cyclic substituent containing one or more heteroatoms (i.e., heteroaryl or heterocycloalkyl) is indicated by the prefix "X-Y-membered", wherein wherein x is the minimum and y is the maximum number of atoms forming the cyclic moiety of the substituent. Thus, for example, 5-8-membered heterocycloalkyl refers to a heterocycloalkyl containing from 5 to 8 atoms, including one ore more heteroatoms, in the cyclic moiety of the heterocycloalkyl. The term "hydrogen" refers to hydrogen substituent, and may be depicted as -H.
The term "hydroxy" or "hydroxyl" refers to -OH. When used in combination with another term(s), the prefix "hydroxy" indicates that the substituent to which the prefix is attached is substituted with one or more hydroxy substituents. Compounds bearing a carbon to which one or more hydroxy substituents include, for example, alcohols, enols and phenol.
The term "hydroxyalkyl" refers to an alkyl that is substituted with at least one hydroxy substituent. Examples of hydroxyalkyl include hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl.
The term "cyano" (also referred to as "nitrile") means -CN, which also
may be depicted:
Figure imgf000015_0001
The term "carbonyl" means -C(O)-, which also may be depicted as:
Figure imgf000015_0002
The term "amino" refers to -NH2.
The term "alkylamino" refers to an amino group, wherein at least one alkyl chain is bonded to the amino nitrogen in place of a hydrogen atom.
Examples of alkylamino substituents include monoalkylamino such as methylamino (exemplified by the formula -NH(CH3)), which may also be
depicted:
Figure imgf000015_0003
and dialkylamino such as dimethylamino,
(exemplified by the formula
-N(CH3J2, which may also be depicted:
Figure imgf000015_0004
.
The term "halogen" refers to fluorine (which may be depicted as -F), chlorine (which may be depicted as -Cl), bromine (which may be depicted as -Br), or iodine (which may be depicted as -I). In one embodiment, the halogen is chlorine. In another embodiment, the halogen is a fluorine. The prefix "halo" indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen substituents. For example, haloalkyl refers to an alkyl that is substituted with at least one halogen substituent. Where more than one hydrogen is replaced with halogens, the halogens may be the identical or different. Examples of haloalkyls include chloromethyl, dichloromethyl, difluorochloromethyl, dichlorofluoromethyl, trichloromethyl, 1 -bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, difluoroethyl, pentafluoroethyl, difluoropropyl, dichloropropyl, and heptafluoropropyl. Illustrating further, "haloalkoxy" refers to an alkoxy that is substituted with at least one halogen substituent. Examples of haloalkoxy substituents include chloromethoxy, 1 -bromoethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy (also known as "perfluoromethyloxy"), and 2,2,2-trifluoroethoxy. It should be recognized that if a substituent is substituted by more than one halogen substituent, those halogen substituents may be identical or different (unless otherwise stated). The term "oxo" refers to =0.
The term "oxy" refers to an ether substituent, and may be depicted as -O-. The term "alkoxy" refers to an alkyl linked to an oxygen, which may also be represented as
-O-R, wherein the R represents the alkyl group. Examples of alkoxy include methoxy, ethoxy, propoxy and butoxy.
The term "heterocycloalkyl" refers to a saturated or partially saturated ring structure containing a total of 3 to 14 ring atoms. At least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur. A heterocycloalkyl alternatively may comprise 2 or 3 rings fused together, wherein at least one such ring contains a heteroatom as a ring atom (i.e., nitrogen, oxygen, or sulfur). In a group that has a heterocycloalkyl substituent, the ring atom of the heterocycloalkyl substituent that is bound to the group may be the at least one heteroatom, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom. Similarly, if the heterocycloalkyl substituent is in turn substituted with a group or substituent, the group or substituent may be bound to the at least one heteroatom, or it may be bound to a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom.
The term "heterocycloalkyl" also includes substituents that are fused to a Cβ-Cio aromatic ring or to a 5-10-membered heteroaromatic ring, wherein a group having such a fused heterocycloalkyl group as a substituent is bound to a heteroatom of the heterocyclocalkyl group or to a carbon atom of the heterocycloalkyl group. When such a fused heterocycloalkyl group is substituted with one more substituents, the one or more substitutents, unless otherwise specified, are each bound to a heteroatom of the heterocyclocalkyl group or to a carbon atom of the heterocycloalkyl group. The fused C6-Ci0 aromatic ring or 5-10-membered heteroaromatic ring may be optionally substituted with halogen, d-C6 alkyl, C3-Ci0 cycloalkyl, CrC6 alkoxy, or =0.
The term "heteroaryl" refers to an aromatic ring structure containing from 5 to 14 ring atoms in which at least one of the ring atoms is a heteroatom (i.e., oxygen, nitrogen, or sulfur), with the remaining ring atoms being independently selected from the group consisting of carbon, oxygen, nitrogen, and sulfur. A heteroaryl may be a single ring or 2 or 3 fused rings. Examples of heteroaryl substituents include 6-membered ring substituents such as pyridyl, pyrazyl, pyrimidinyl, and pyridazinyl; 5-membered ring substituents such as triazolyl, imidazolyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1 ,2,3-, 1 ,2,4-, 1 ,2,5-, or 1 ,3,4-oxadiazolyl and isothiazolyl; 6/5-membered fused ring substituents such as benzothiofuranyl, isobenzothiofuranyl, benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl; and 6/6-membered fused rings such as quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and 1 ,4-benzoxazinyl. In a group that has a heteroaryl substituent, the ring atom of the heteroaryl substituent that is bound to the group may be the at least one heteroatom, or it may be a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom. Similarly, if the heteroaryl substituent is in turn substituted with a group or substituent, the group or substituent may be bound to the at least one heteroatom, or it may be bound to a ring carbon atom, where the ring carbon atom may be in the same ring as the at least one heteroatom or where the ring carbon atom may be in a different ring from the at least one heteroatom. The term "heteroaryl" also includes pyridyl N-oxides and groups containing a pyridine N-oxide ring. Examples of single-ring heteroaryls include furanyl, dihydrofuranyl, tetradydrofuranyl, thiophenyl (also known as "thiofuranyl"), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, isopyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, isoimidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, dithiolyl, oxathiolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiaθdiazolyl, oxathiazolyl, oxadiazolyl (including oxadiazolyl, 1 ,2,4-oxadiazolyl (also known as "azoximyl"), 1 ,2,5-oxadiazolyl (also known as "furazanyl"), or 1 ,3,4-oxadiazolyl), oxatriazolyl (including 1 ,2,3,4-oxatriazolyl or 1 ,2,3,5-oxatriazolyl), dioxazolyl (including 1 ,2,3-dioxazolyl, 1 ,2,4-dioxazolyl, 1 ,3,2-dioxazolyl, or 1 ,3,4-dioxazolyl), oxathiazolyl, oxathiolyl, oxathiolanyl, pyranyl (including 1 ,2-pyranyl or 1 ,4-pyranyl), dihydropyranyl, pyridinyl (also known as "azinyl"), piperidinyl, diazinyl (including pyridazinyl (also known as "1 ,2-diazinyl"), pyrimidinyl (also known as "1 ,3-diazinyl" or "pyrimidyl"), or pyrazinyl (also known as "1 ,4-diazinyl")), piperazinyl, triazinyl (including s-triazinyl (also known as "1 ,3,5-triazinyl"), as-triazinyl (also known 1 ,2,4-triazinyl), and v-triazinyl (also known as "1 ,2,3-triazinyl")), oxazinyl (including 1 ,2,3-oxazinyl, 1 ,3,2-oxazinyl, 1 ,3,6-oxazinyl (also known as "pentoxazolyl"), 1 ,2,6-oxazinyl, or 1 ,4-oxazinyl), isoxazinyl (including o-isoxazinyl or p-isoxazinyl), oxazolidinyl, isoxazolidinyl, oxathiazinyl (including 1 ,2,5-oxathiazinyl or 1 ,2,6-oxathiazinyl), oxadiazinyl (including 1 ,4,2-oxadiazinyl or 1 ,3,5,2-oxadiazinyl), morpholinyl, azepinyl, oxepinyl, thiepinyl, and diazepinyl.
Examples of 2-fused-ring heteroaryls include, indolizinyl, pyrindinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl (including pyrido[3,4-b]-pyridinyl, pyrido[3,2-b]-pyridinyl, or pyrido[4,3-b]-pyridinyl), and pteridinyl, indolyl, isoindolyl, indoleninyl, isoindazolyl, benzazinyl, phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl, benzopyranyl, benzothiopyranyl, benzoxazolyl, indoxazinyl, anthranilyl, benzodioxolyl, benzodioxanyl, benzoxadiazolyl, benzofuranyl, isobenzofuranyl, benzothienyl, isobenzothienyl, benzothiazolyl, benzothiadiazolyl, benzimidazolyl, benzotriazolyl, benzoxazinyl, benzisoxazinyl, and tetrahydroisoquinolinyl.
Examples of 3-fused-ring heteroaryls or heterocycloalkyls include 5,6-dihydro-4H-imidazo[4,5, 1 -ij]quinoline, 4,5-dihydroimidazo[4,5, 1 -hφndole, 4,5,6,7-tetrahydroimidazo[4,5,1-jk][1]benzazepine, and dibenzofuranyl.
Other examples of fused-ring heteroaryls include benzo-fused heteroaryls such as indolyl, isoindolyl (also known as "isobenzazolyl" or "pseudoisoindolyl"), indoleninyl (also known as "pseudoindolyl"), isoindazolyl (also known as "benzpyrazolyl"), benzazinyl (including quinolinyl (also known as "1 -benzazinyl") or isoquinolinyl (also known as "2-benzazinyl")), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (also known as "1 ,2-benzodiazinyl") or quinazolinyl (also known as "1 ,3-benzodiazinyl")), benzopyranyl (including "chromanyl" or "isochromanyl"), benzothiopyranyl (also known as "thiochromanyl"), benzoxazolyl, indoxazinyl (also known as "benzisoxazolyl"), anthranilyl, benzodioxolyl, benzodioxanyl, benzoxadiazolyl, benzofuranyl (also known as "coumaronyl"), isobenzofuranyl, benzothienyl (also known as "benzothiophenyl," "thionaphthenyl," or "benzothiofuranyl"), isobenzothienyl (also known as "isobenzothiophenyl," "isothionaphthenyl," or "isobenzothiofuranyl"), benzothiazolyl, benzothiadiazolyl, benzimidazolyl, benzotriazolyl, benzoxazinyl (including 1 ,3,2-benzoxazinyl, 1 ,4,2-benzoxazinyl, 2,3,1 -benzoxazinyl, or 3,1 ,4-benzoxazinyl), benzisoxazinyl (including 1 ,2-benzisoxazinyl or 1 ,4-benzisoxazinyl), tetrahydroisoquinolinyl , carbazolyl, xanthenyl, and acridinyl. The term "heteroaryl" also includes substituents such as pyridyl and quinolinyl that are fused to a C4-Ci0 carbocyclic ring, such as a C5 or a C6 carbocyclic ring, or to a 4-10-membered heterocyclic ring, wherein a group having such a fused aryl group as a substituent is bound to an aromatic carbon of the heteroaryl group or to a heteroatom of the heteroaryl group. When such a fused heteroaryl group is substituted with one more substituents, the one or more substitutents, unless otherwise specified, are each bound to an aromatic carbon of the heteroaryl group or to a heteroatom of the heteroaryl group. The fused C4-Ci0 carbocyclic or 4-10-membered heterocyclic ring may be optionally substituted with halogen, d-Cβ alkyl, C3- Ci0 cycloalkyl, or =0.
A substituent is "substitutable" if it comprises at least one carbon, sulfur, oxygen or nitrogen atom that is bonded to one or more hydrogen atoms. Thus, for example, hydrogen, halogen, and cyano do not fall within this definition. If a substituent is described as being "substituted," a non-hydrogen substituent is in the place of a hydrogen substituent on a carbon, oxygen, sulfur or nitrogen of the substituent. Thus, for example, a substituted alkyl substituent is an alkyl substituent wherein at least one non-hydrogen substituent is in the place of a hydrogen substituent on the alkyl substituent. To illustrate, monofluoroalkyl is alkyl substituted with a fluoro substituent, and difluoroalkyl is alkyl substituted with two fluoro substituents. It should be recognized that if there is more than one substitution on a substituent, each non-hydrogen substituent may be identical or different (unless otherwise stated). If a substituent is described as being "optionally substituted," the substituent may be either (1 ) not substituted, or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the carbon (to the extent there are any) may separately and/or together be replaced with an independently selected optional substituent. If a nitrogen of a substituent is described as being optionally substituted with one or more of a list of substituents, one or more of the hydrogens on the nitrogen (to the extent there are any) may each be replaced with an independently selected optional substituent. One exemplary substituent may be depicted as -N R' R," wherein R' and R" together with the nitrogen atom to which they are attached, may form a heterocyclic ring. The heterocyclic ring formed from R' and R" together with the nitrogen atom to which they are attached may be partially or fully saturated. In one embodiment, the heterocyclic ring consists of 3 to 7 atoms. In another embodiment, the heterocyclic ring is selected from the group consisting of pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, isoxazolyl, pyridyl and thiazolyl.
This specification uses the terms "substituent," "radical," and "group" interchangeably.
If a group of substituents are collectively described as being optionally substituted by one or more of a list of substituents, the group may include: (1) unsubstitutable substituents, (2) substitutable substituents that are not substituted by the optional substituents, and/or (3) substitutable substituents that are substituted by one or more of the optional substituents.
If a substituent is described as being optionally substituted with up to a particular number of non-hydrogen substituents, that substituent may be either (1 ) not substituted; or (2) substituted by up to that particular number of non-hydrogen substituents or by up to the maximum number of substitutable positions on the substituent, whichever is less. Thus, for example, if a substituent is described as a heteroaryl optionally substituted with up to 3 non-hydrogen substituents, then any heteroaryl with less than 3 substitutable positions would be optionally substituted by up to only as many non-hydrogen substituents as the heteroaryl has substitutable positions. To illustrate, tetrazolyl (which has only one substitutable position) would be optionally substituted with up to one non-hydrogen substituent. To illustrate further, if an amino nitrogen is described as being optionally substituted with up to 2 non- hydrogen substituents, then the nitrogen will be optionally substituted with up to 2 non-hydrogen substituents if the amino nitrogen is a primary nitrogen, whereas the amino nitrogen will be optionally substituted with up to only 1 non-hydrogen substituent if the amino nitrogen is a secondary nitrogen. A prefix attached to a multi-moiety substituent only applies to the first moiety. To illustrate, the term "alkylcycloalkyl" contains two moieties: alkyl and cycloalkyl. Thus, a d-C6- prefix on CrC6-alkylcycloalkyl means that the alkyl moiety of the alkylcycloalkyl contains from 1 to 6 carbon atoms; the CrC6- prefix does not describe the cycloalkyl moiety. To illustrate further, the prefix "halo" on haloalkoxyalkyl indicates that only the alkoxy moiety of the alkoxyalkyl substituent is substituted with one or more halogen substituents. If the halogen substitution may only occur on the alkyl moiety, the substituent would be described as "alkoxyhaloalkyl." If the halogen substitution may occur on both the alkyl moiety and the alkoxy moeity, the substituent would be described as "haloalkoxyhaloalkyl."
When a substituent is comprised of multiple moieties, unless otherwise indicated, it is the intention for the final moiety to serve as the point of attachment to the remainder of the molecule. For example, in a substituent A- B-C, moiety C is attached to the remainder of the molecule. In a substituent A-B-C-D, moiety D is attached to the remainder of the molecule. Similarly, in a substituent aminocarbonylmethyl, the methyl moiety is attached to the remainder of the molecule, where the substituent may also be be depicted as
Figure imgf000023_0001
In a substituent trifluoromethylaminocarbonyl, the carbonyl moiety is attached to the remainder of the molecule, where the
substituent may also be depicted as
Figure imgf000023_0002
If substituents are described as being "independently selected" from a group, each substituent is selected independent of the other. Each substituent therefore may be identical to or different from the other substituent(s). Isomers
When an asymmetric center is present in a compound of formula I, hereinafter referred to as the compound of the invention, the compound may exist in the form of optical isomers (enantiomers). In one embodiment, the present invention comprises enantiomers and mixtures, including racemic mixtures of the compounds of formula I. In another embodiment, for compounds of formulae I that contain more than one asymmetric center, the present invention comprises diastereomeric forms (individual diastereomers and mixtures thereof) of compounds. When a compound of formula I contains an alkenyl group or moiety, geometric isomers may arise. Tautomeric Forms
The present invention comprises the tautomeric forms of compounds of formula I. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of formula I containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism. The various ratios of the tautomers in solid and liquid form is dependent on the various substituents on the molecule as well as the particular crystallization technique used to isolate a compound.
Salts
The compounds of this invention may be used in the form of salts derived from inorganic or organic acids. Depending on the particular compound, a salt of the compound may be advantageous due to one or more of the salt's physical properties, such as enhanced pharmaceutical stability in differing temperatures and humidities, or a desirable solubility in water or oil. In some instances, a salt of a compound also may be used as an aid in the isolation, purification, and/or resolution of the compound.
Where a salt is intended to be administered to a patient (as opposed to, for example, being used in an in vitro context), the salt preferably is pharmaceutically acceptable. The term "pharmaceutically acceptable salt" refers to a salt prepared by combining a compound of formula I with an acid whose anion, or a base whose cation, is generally considered suitable for human consumption. Pharmaceutically acceptable salts are particularly useful as products of the methods of the present invention because of their greater aqueous solubility relative to the parent compound. For use in medicine, the salts of the compounds of this invention are non-toxic "pharmaceutically acceptable salts." Salts encompassed within the term "pharmaceutically acceptable salts" refer to non-toxic salts of the compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid.
Suitable pharmaceutically acceptable acid addition salts of the compounds of the present invention when possible include those derived from inorganic acids, such as hydrochloric, hydrobromic, hydrofluoric, boric, fluoroboric, phosphoric, metaphosphoric, nitric, carbonic, sulfonic, and sulfuric acids, and organic acids such as acetic, benzenesulfonic, benzoic, citric, ethanesulfonic, fumaric, gluconic, glycolic, isothionic, lactic, lactobionic, maleic, malic, methanesulfonic, trifluoromethanesulfonic, succinic, toluenesulfonic, tartaric, and trifluoroacetic acids. Suitable organic acids generally include, for example, aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids.
Specific examples of suitable organic acids include acetate, trifluoroacetate, formate, propionate, succinate, glycolate, gluconate, digluconate, lactate, malate, tartaric acid, citrate, ascorbate, glucuronate, maleate, fumarate, pyruvate, aspartate, glutamate, benzoate, anthranilic acid, mesylate, stearate, salicylate, p-hydroxybenzoate, phenylacetate, mandelate, embonate (pamoate), methanesulfonate, ethanesulfonate, benzenesulfonate, pantothenate, toluenesulfonate, 2-hydroxyethanesulfonate, sufanilate, cyclohexylaminosulfonate, algenic acid, β-hydroxybutyric acid, galactarate, galacturonate, adipate, alginate, butyrate, camphorate, camphorsulfonate, cyclopentanepropionate, dodecylsulfate, glycoheptanoate, glycerophosphate, heptanoate, hexanoate, nicotinate, 2-naphthalesulfonate, oxalate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, thiocyanate, tosylate, and undecanoate. Furthermore, where the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts, i.e., sodium or potassium salts; alkaline earth metal salts, e.g., calcium or magnesium salts; and salts formed with suitable organic ligands, e.g., quaternary ammonium salts. In another embodiment, base salts are formed from bases which form non-toxic salts, including aluminum, arginine, benzathine, choline, diethylamine, diolamine, glycine, lysine, meglumine, olamine, tromethamine and zinc salts.
Organic salts may be made from secondary, tertiary or quaternary amine salts, such as tromethamine, diethylamine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), and procaine. Basic nitrogen-containing groups may be quaternized with agents such as lower alkyl (CrC6) halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (i.e., dimethyl, diethyl, dibuytl, and diamyl sulfates), long chain halides (i.e., decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides), arylalkyl halides (i.e., benzyl and phenethyl bromides), and others.
In one embodiment, hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. The compounds of the invention and their salts may exist in both unsolvated and solvated forms.
Prodrugs
Also within the scope of the present invention are so-called "prodrugs" of the compound of the invention. Thus, certain derivatives of the compound of the invention which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into the compound of the invention having the desired activity, for example, by hydrolytic cleavage. Such derivatives are referred to as "prodrugs." Further information on the use of prodrugs may be found in "Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T Higuchi and W Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press, 1987 (ed. E B Roche, American Pharmaceutical Association). Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of any of formula I with certain moieties known to those skilled in the art as "pro-moieties" as described, for example, in "Design of Prodrugs" by H Bundgaard (Elsevier, 1985).
Isotopes
The present invention also includes isotopically labelled compounds, which are identical to those recited in formula I, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine and chlorine, such as 2H, 3H, 13C, 11C, 14C, 15N, 18O, 17O, 31P, 32P, 35S, 18F, and 36CI, respectively. Compounds of the present invention, prodrugs thereof, and pharmaceutically acceptable salts of said compounds or of said prodrugs which contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically labelled compounds of the present invention, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half- life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of formula I of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
Administration and Dosing
Typically, a compound of the invention is administered in an amount effective to treat a condition as described herein. The compounds of the invention are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. Therapeutically effective doses of the compounds required to treat the progress of the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts. The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth. In another embodiment, the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
In another embodiment, the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermally. In another embodiment, the compounds of the invention can also be administered intranasally or by inhalation. In another embodiment, the compounds of the invention may be administered rectally or vaginally. In another embodiment, the compounds of the invention may also be administered directly to the eye or ear.
The dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions. In one embodiment, the total daily dose of a compound of the invention (administered in single or divided doses) is typically from about 0.01 to about 100 mg/kg. In another embodiment, total daily dose of the compound of the invention is from about 0.1 to about 50 mg/kg, and in another embodiment, from about 0.5 to about 30 mg/kg (i.e., mg compound of the invention per kg body weight). In one embodiment, dosing is from 0.01 to 10 mg/kg/day. In another embodiment, dosing is from 0.1 to 1.0 mg/kg/day. Dosage unit compositions may contain such amounts or submultiples thereof to make up the daily dose. In many instances, the administration of the compound will be repeated a plurality of times in a day (typically no greater than 4 times). Multiple doses per day typically may be used to increase the total daily dose, if desired.
For oral administration, the compositions may be provided in the form of tablets containing 0.01 , 0.05, 0.1 , 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 75.0, 100, 125, 150, 175, 200, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, or in another embodiment, from about 1mg to about 100 mg of active ingredient. Intravenously, doses may range from about 0.1 to about 10 mg/kg/minute during a constant rate infusion.
Suitable subjects according to the present invention include mammalian subjects. Mammals according to the present invention include, but are not limited to, canine, feline, bovine, caprine, equine, ovine, porcine, rodents, lagomorphs, primates, and the like, and encompass mammals in utero. In one embodiment, humans are suitable subjects. Human subjects may be of either gender and at any stage of development. Use in the Preparation of a Medicament
In another embodiment, the invention comprises the use of one or more compounds of the invention for the preparation of a medicament for the treatment of the conditions recited herein.
Pharmaceutical Compositions
For the treatment of the conditions referred to above, the compound of the invention can be administered as compound per se. Alternatively, pharmaceutically acceptable salts are suitable for medical applications because of their greater aqueous solubility relative to the parent compound. In another embodiment, the present invention comprises pharmaceutical compositions. Such pharmaceutical compositions comprise a compound of the invention presented with a pharmaceutically-acceptable carrier. The carrier can be a solid, a liquid, or both, and may be formulated with the compound as a unit-dose composition, for example, a tablet, which can contain from 0.05% to 95% by weight of the active compounds. A compound of the invention may be coupled with suitable polymers as targetable drug carriers. Other pharmacologically active substances can also be present. The compounds of the present invention may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The active compounds and compositions, for example, may be administered orally, rectally, parenterally, or topically. Oral administration of a solid dose form may be, for example, presented in discrete units, such as hard or soft capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of at least one compound of the present invention. In another embodiment, the oral administration may be in a powder or granule form. In another embodiment, the oral dose form is sub-lingual, such as, for example, a lozenge. In such solid dosage forms, the compounds of formula I are ordinarily combined with one or more adjuvants. Such capsules or tablets may contain a controlled-release formulation. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agentsor may be prepared with enteric coatings.
In another embodiment, oral administration may be in a liquid dose form. Liquid dosage forms for oral administration include, for example, pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art (i.e., water). Such compositions also may comprise adjuvants, such as wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.
In another embodiment, the present invention comprises a parenteral dose form. "Parenteral administration" includes, for example, subcutaneous injections, intravenous injections, intraperitoneally, intramuscular injections, intrasternal injections, and infusion. Injectable preparations (i.e., sterile injectable aqueous or oleaginous suspensions) may be formulated according to the known art using suitable dispersing, wetting agents, and/or suspending agents. In another embodiment, the present invention comprises a topical dose form. "Topical administration" includes, for example, transdermal administration, such as via transdermal patches or iontophoresis devices, intraocular administration, or intranasal or inhalation administration. Compositions for topical administration also include, for example, topical gels, sprays, ointments, and creams. A topical formulation may include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. When the compounds of this invention are administered by a transdermal device, administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958, by Finnin and Morgan (October 1999).
Formulations suitable for topical administration to the eye include, for example, eye drops wherein the compound of this invention is dissolved or suspended in suitable carrier. A typical formulation suitable for ocular or aural administration may be in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (i.e., absorbable gel sponges, collagen) and non-biodegradable (i.e., silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
For intranasal administration or administration by inhalation, the active compounds of the invention are conveniently delivered in the form of a solution or suspension from a pump spray container that is squeezed or pumped by the patient or as an aerosol spray presentation from a pressurized container or a nebulizer, with the use of a suitable propellant. Formulations suitable for intranasal administration are typically administered in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1 ,1 ,1 ,2- tetrafluoroethane or 1 ,1 ,1 ,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
In another embodiment, the present invention comprises a rectal dose form. Such rectal dose form may be in the form of, for example, a suppository. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate. Other carrier materials and modes of administration known in the pharmaceutical art may also be used. Pharmaceutical compositions of the invention may be prepared by any of the well-known techniques of pharmacy, such as effective formulation and administration procedures. The above considerations in regard to effective formulations and administration procedures are well known in the art and are described in standard textbooks. Formulation of drugs is discussed in, for example, Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania, 1975; Liberman, et al., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Kibbe, et al., Eds., Handbook of Pharmaceutical Excipients (3rd Ed.), American Pharmaceutical Association, Washington, 1999.
Co-administration The compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment of various conditions or disease states. The compound(s) of the present invention and other therapeutic agent(s) may be may be administered simultaneously (either in the same dosage form or in separate dosage forms) or sequentially. An exemplary therapeutic agent may be, for example, a metabotropic glutamate receptor agonist.
The administration of two or more compounds "in combination" means that the two compounds are administered closely enough in time that the presence of one alters the biological effects of the other. The two or more compounds may be administered simultaneously, concurrently or sequentially. Additionally, simultaneous administration may be carried out by mixing the compounds prior to administration or by administering the compounds at the same point in time but at different anatomic sites or using different routes of administration. The phrases "concurrent administration," "co-administration," "simultaneous administration," and "administered simultaneously" mean that the compounds are administered in combination.
Kits The present invention further comprises kits that are suitable for use in performing the methods of treatment described above. In one embodiment, the kit contains a first dosage form comprising one or more of the compounds of the present invention and a container for the dosage, in quantities sufficient to carry out the methods of the present invention. In another embodiment, the kit of the present invention comprises one or more compounds of the invention.
Intermediates
In another embodiment, the invention relates to the novel intermediates useful for preparing the compounds of the invention. General Synthetic Schemes
The compounds of the formula I may be prepared by the methods described below, together with synthetic methods known in the art of organic chemistry, or modifications and derivatisations that are familiar to those of ordinary skill in the art. The starting materials used herein are commercially available or may be prepared by routine methods known in the art (such as those methods disclosed in standard reference books such as the COMPENDIUM OF ORGANIC SYNTHETIC METHODS, Vol. I-VI (published by Wiley-lnterscience)). Preferred methods include, but are not limited to, those described below. During any of the following synthetic sequences it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This can be achieved by means of conventional protecting groups, such as those described in T. W. Greene, Protective Groups in Organic Chemistry, John Wiley & Sons, 1981 ; T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons, 1991 , and T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Chemistry, John Wiley & Sons, 1999, which are hereby incorporated by reference.
Compounds of formula I, or their pharmaceutically acceptable salts, can be prepared according to the reaction Schemes discussed herein below.
Unless otherwise indicated, the substituents in the Schemes are defined as above. Isolation and purification of the products is accomplished by standard procedures, which are known to a chemist of ordinary skill.
It will be understood by one skilled in the art that the various symbols, superscripts and subscripts used in the schemes, methods and examples are used for convenience of representation and/or to reflect the order in which they are introduced in the schemes, and are not intended to necessarily correspond to the symbols, superscripts or subscripts in the appended claims.
The schemes are representative of methods useful in synthesizing the compounds of the present invention. They are not to constrain the scope of the invention in any way.
Working Examples
The following illustrate the synthesis of various compounds of the present invention. Additional compounds within the scope of this invention may be prepared using the methods illustrated in these Examples, either alone or in combination with techniques generally known in the art.
Scheme 1
Figure imgf000035_0001
Scheme 2
Figure imgf000036_0001
Figure imgf000036_0002
Pd-C/H2/MeOH
Figure imgf000036_0003
Scheme 3.
Figure imgf000037_0001
Figure imgf000037_0002
Scheme 4
Figure imgf000037_0003
Figure imgf000037_0004
Pd2dba3, Dave-Phos, f-BuOK/ toluene
600C
Figure imgf000037_0005
Experi mentals
General Procedure A:
Figure imgf000037_0006
A mixture of 3-bromo-2-chloro pyridine, the appropriate phenol and potassium carbonate in Λ/,Λ/-dimethylformamide was heated at 1500C overnight. The reaction was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layer was dried over anhydrous magnesium sulfate and concentrated in vacuo. The crude product was purified by silica gel chromatography to afford the desired bromopyridine. General Procedure A':
Figure imgf000038_0001
A mixture of 3-bromo-2-chloropyridine, the appropriate phenol and cesium carbonate in dimethyl sulfoxide was heated at 120 0C overnight. The reaction was cooled to room temperature and diluted with three volumes of water, causing the product to precipitate out of solution. This precipitate was filtered off and washed with water, then dissolved in ethyl acetate. This solution was dried over anhydrous magnesium sulfate and concentrated in vacuo to give the crude product with >95% purity. General Procedure B:
Figure imgf000038_0002
A mixture of appropriate bromopyridine, tert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate and cesium fluoride in Λ/,Λ/-dimethylformamide was degassed for 30 minutes, and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) was added. The reaction mixture was heated at 100 °C overnight. The reaction mixture was cooled to room temperature, and the solvent was evaporated. To the residue was added ethyl acetate, and the suspension was filtered through a pad of Celite. The filtrate was washed with water, dried over anhydrous sodium sulfate, and concentrated in vacuo. The crude product was purified by silica gel chromatography to afford the desired te/t-butyl carboxylate protected dihydropyridine. General Procedure C:
Figure imgf000039_0001
To a solution of appropriate te/t-butyl carboxylate protected dihydropyridine in methanol was added palladium on carbon and the suspension was hydrogenated at 30 psi until the reduction was complete. The mixture was filtered through a pad of Celite, and the filtrate was concentrated in vacuo. The crude product was used directly in the next step or was purified by silica gel chromatography to afford the desired te/t-butyl carboxylate protected piperidine. General Procedure D:
Figure imgf000039_0002
X = N; CH To dry methanol under N2 at 0 °C was added acetyl chloride. The solution was stirred at room temperature for 30 minutes, and then transferred to a flask containing appropriate te/t-butyl carboxylate protected piperidine or te/t-butyl carboxylate protected piperazine. The mixture was stirred overnight at room temperature. The solvent was evaporated to afford target compound. General Procedure E:
Figure imgf000040_0001
A mixture of appropriate bromopyridine, te/ϊ-butyl piperazine-1- carboxylate and sodium tert-butoxide in toluene was flushed with N2 for 10 min. To the reaction mixture, tris(dibenzylideneacetone)dipalladium and Dave-Phos was added and heated to 60 °C overnight. The reaction mixture was cooled to room temperature and diluted with ethyl acetate and water. The organic layer was extracted with ethyl acetate, dried over anhydrous sodium sulfate and concentrated in vacuo. The crude product was purified by silica gel chromatography to afford the desired tert-butyl carboxylate protected piperazine.
General Procedure F:
Figure imgf000040_0002
To dry methanol under N2 at 0 °C was added appropriate tert-butyl carboxylate protected piperidine. To the solution, 2.4M solution of HCI in MeOH was added and the mixture was stirred overnight. The solvent was removed in vacuo to result in the desired product.
Example #1
7ert-Butyl-4-(5,5-dimethyl-1,3,2-dioxaborinan-2-yl)-5,6- dihydropyridine-1 (2H)-carboxylate:
Figure imgf000041_0001
n-Butyllithium (924 ml_, 2.3 mol, 2.5 M in hexane) was added slowly to a solution of diisopropylamine (323.3 ml_, 3.2 mol) in THF (6 L) at -78 °C and stirred for 1 h. A solution of tert-butyl 4-oxopiperidine-i-carboxylate (400 g, 2.01 mol) in THF (2 L) was added and the mixture was stirred for 1 h at -78 0C. A solution of Λ/-phenyltriflimide (753 g, 2.11 mol) in THF (2 L) was added at -78 0C and the reaction mixture was allowed to warm to rt and stirred at rt overnight. The reaction mixture was diluted with ethyl acetate and washed with brine, and the separated organic phase was dried over Na2SO4, filtered, and concentrated. The crude residue was purified by silica gel column chromatography (hexanes/ethyl acetate, 5:1) to afford tert-butyl 4- (trifluoromethylsulfonyloxy)-5,6-dihydropyridine-1 (2H)-carboxylate as a brown oil. To an oven dried flask containing tert-butyl 4-(trifluoromethylsulfonyloxy)- 5,6-dihydropyridine-1(2H)-carboxylate (12 g, 36.25 mmol), 2-(5,5-dimethyl- 1 ,3,2-dioxaborinan-2-yl)-5,5-dimethyl-1 ,3,2-dioxaborinane (9.82 g, 43.5 mmol) and K3PO4 (24.62 g, 116 mmol) were added toluene (100 mL) and dioxane (40 mL). While stirring the reaction mixture at room temperature, the air in the flask was removed and refilled with N2. This process was repeated three times, followed by the addition of [1 ,1'- Bis(diphenylphosphino)ferrocene]palladium(ll)chloride (4.89 g, 5.35 mmol) and the reaction mixture was stirred overnight at 110 0C, cooled to room temperature and filtered through a pad of Celite, and the filtrate was evaporated in vacuo. The dark red oily material was chromatographed on silica column (ethyl acetate/hexanes 4:96 to 10:90) to yield compound tert- Butyl-4-(5,5-dimethyl-1 ,3(2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 [2H)- carboxylate (7.59 g, 71%) as an orange solid. 1H NMR (400 MHz, CDCI3) δ 6.41 (br. s, 1 H), 3.94 (s, 2H), 3.63 (s, 4H), 3.42 (s, 2H), 2.18 (s, 2H), 1.44 (s, 9H), 0.97 (s, 6H).
3-Bromo-2-phenoxypyridine:
Figure imgf000042_0001
A mixture of 3-bromo-2-chloropyridine (19.24 g, 0.1 mol), phenol (9.41 g, 0.1 mol) and potassium carbonate (20.73 g, 0.15 mol) in N, N- dimethylformamide (100 ml_) was heated with stirring at 150 °C overnight. The reaction mixture was cooled to room temperature, diluted with water, and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were dried over anhydrous magnesium sulfate, and the solvent was removed under reduced pressure. The crude product was purified by silica gel column chromatography (hexanes/ethyl acetate, 1 :0 - 30:1 ) to give 3-bromo-2- phenoxypyridine (12 g, 58 %). 1H NMR (400 MHz, CDCI3): δ 8.07 (dd, J = 1.6 & 5.2 Hz, 1 H), 7.93 (dd, J = 1.6 & 7.6 Hz, 1 H), 7.42 (t, 2H), 7.23 (t, 1 H), 7.16 (d, 2H), 6.89 (q, J = 5.2 & 7.6 Hz, 1 H). ES-MS (m/z) for C11H8BrO: 249.79 & 251.78 [M+1]+.
Terf-butyl 4-(2-phenoxypyridin-3-yl)-5,6-dihydropyridine-1 (2H) carboxylate:
Figure imgf000042_0002
A mixture of 3-bromo-2-phenoxypyridine (2.5 g, 0.01 mol), tert-butyl 4- (5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (2.95 g, 0.01 mol) and cesium fluoride (2.28 g, 0.015 mol) in N1N- dimethylformamide (30 mL) was degassed for 30 minutes, and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (1.1 g, 0.0015 mol) was added. The reaction mixture was heated at 100 °C overnight, cooled to room temperature, and the solvent was evaporated. To the residue was added ethyl acetate, and the suspension was filtered through a pad of Celite. The filtrate was washed with water, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude product was purified by flash column chromatography (hexanes/ ethyl acetate, 15:1 - 8:1) to give the te/t-butyl 4-(2-phenoxypyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (1.86 g, 53 %). 1H NMR (400 MHz, CDCI3): δ 8.07 (dd, 1 H), 7.58 (dd, 1 H), 7.39 (t, 2H), 7.19 (t, 1 H), 7.10 (d, 2H), 6.99 (m, 1 H), 5.98 (s, 1 H), 4.08 (br s, 2H), 3.61 (m, 2H), 2.59 (br s, 2H), 1.49 (s, 9H). ES-MS (m/z) for C2IH24N2O3: 352.99 [M+1]+.
Terf-butyl 4-(2-phenoxypyridin-3-yl)piperϊdine-1-carboxylate:
Figure imgf000043_0001
To a solution of fe/t-butyl 4-(2-phenoxypyridin-3-yl)-5,6-dihydropyridine- 1 (2H)-carboxylate (0.8 g, 2.3 mmol) in methanol (30 ml.) was added palladium (0.2 g, 10 wt % on activated carbon), and the suspension was hydrogenated at 25 psi until the reduction was complete. The mixture was filtered through a pad of Celite, and the filtrate was concentrated under reduced pressure. The residue was purified by flash column chromatography (hexanes/ethyl acetate, 20:1 - 8:1 ) to give tert-butyl 4-(2-phenoxypyridin-3- yl)piperidine-1-carboxylate (0.63 g, 78 %). 1H NMR (400 MHz, CDCI3): δ 8.02 (dd, 1 H), 7.55 (dd, 1 H), 7.40 (t, 2H), 7.19 (t, 1 H), 7.10 (d, 2H), 6.98 (q, 1 H), 4.38 (s, 2H), 3.14 (m, 1 H), 2.85 (m, 2H), 1.95 (d, 2H), 1.64 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for C21H26N2O3: 355.01 [M+1]+.
2-Phenoxy-3-(piperidin-4-yl)pyridine hydrochloride:
Figure imgf000044_0001
To a cold solution of dry methanol (25 mL) under N2 was added acetyl chloride (1 mL) at 0 °C. The solution was stirred for 30 minutes, and then transferred to a flask containing tert-butyl 4-(2-phenoxypyridin-3-yl)piperidine- 1 -carboxylate (0.22 g, 0.62 mmol). The mixture was stirred overnight at room temperature. The solvent was evaporated to afford target 2-phenoxy-3-
(piperidin-4-yl)pyridine (0.13 g, 72%). 1H NMR (400 MHz, CDCI3): δ 7.96 (dd,
1 H), 7.77 (m, 1 H), 7.41 (t, 2H), 7.21 (t, 1 H), 7.13 (m, 1 H), 7.07 (d, 2H), 3.52
(d, 2H), 3.31 (m, 1 H), 3.18 (t, 2H), 2.20 (d, 2H), 2.06 (m, 2H). ES-MS (m/z) for Ci6H18N2O/1.35HCI: 254.95 [M+1]+. CHN calculated for Ci6Hi8N2O/1.35HCI:
C 63.31 %; H 6.43 %; N 9.23 %; found: C 63.27 %; H 6.49 %; N 8.85 %.
Example #2 3-(Piperidin-4-yl)-2-(p-tolyloxy)pyridine trifluoroacetic acid salt:
Figure imgf000044_0002
Using General Procedure D, terf-butyl 4-(2-(p-tolyloxy)pyridin-3- yl)piperidine-1 -carboxylate (90 mg, 0.22 mmol) was reacted with acetyl chloride (17.5 mg, 0.223 mmol) in methanol (6 ml) then purified with HPLC with 0.025% trifluoroacetic acid to afford the desired product as an oil (46 mg, 49%). 1H NMR (400 MHz, CD3OD) δ 2.03 (qd, J=12.86, 4.15 Hz, 2 H), 2.14 - 2.24 (m, 2 H), 2.34 (s, 3 H), 3.18 (td, J=12.96, 2.70 Hz, 2 H), 3.26 - 3.37 (m, 1 H), 3.47 - 3.57 (m, 2 H), 6.91 - 6.99 (m, 2 H), 7.10 (dd, J=7.46, 4.98 Hz, 1 H), 7.21 (d, J=7.88 Hz, 2 H), 7.74 (dd, J=7A6, 1.66 Hz, 1 H), 7.94 (dd, J=AU, 1.87 Hz, 1 H). MS-APCI (m/z): 269 [M+1]+. Example # 3 3-Bromo-2-(4-(trifluoromethoxy)phenoxy)pyridine:
Figure imgf000045_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (3.03 g, 15.7 mmol) was reacted with 4-(trifluoromethoxy)phenol (3.93 g, 22 mmol) and potassium carbonate (4.35 g, 31.5 mmol) in Λ/,Λ/-dimethylformamide (20 ml) to afford the desired product as oil (3.65 g, 69%). 1H NMR (400 MHz, CDCI3) δ
6.88 (dd, J=7.60, 4.87 Hz, 1 H), 7.14 - 7.19 (m, 2 H), 7.21 - 7.27 (m, 2 H),
7.90 (dd, J=7.80, 1.75 Hz, 1 H), 8.04 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 335 [M+1]+.
Terf-butyl 4-(2-(4-(trifluoromethoxy)phenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000045_0002
Using General Procedure B, 3-bromo-2-(4- (trifluoromethoxy)phenoxy)pyridine (551.7 mg, 1.651 mmol) was reacted with fert-butyl 4-(5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)- carboxylate (585 mg, 1.98 mmol), cesium fluoride (502 mg, 3.30 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (67.4 mg, 0.0826 mmol) in Λ/,Λ/-dimethylformamide (5 ml) to afford the desired product as an oil (440mg, 61 %). 1H NMR (400 MHz, CDCI3) δ 1.46 (s, 9 H), 2.50 - 2.59 (m, 2 H), 3.59 (t, J=5.65 Hz, 2 H), 4.06 (d, J=2.73 Hz, 2 H), 5.89 - 5.99 (m, 1 H), 7.00 (dd, J=7.41 , 4.87 Hz, 1 H), 7.05 - 7.14 (m, 2 H), 7.18 - 7.24 (m, 2 H), 7.58 (dd, J=7.41 , 1.95 Hz, 1 H), 8.03 (dd, J=4.97, 1.85 Hz, 1 H). MS-APCI (m/z): 302 [M+1]+. Te/t-butyl 4-(2-(4-(trifluoromethoxy)phenoxy)pyridin-3- yl)piperidine-1 -carboxylate:
Figure imgf000046_0001
Using General Procedure C, te/t-butyl 4-(2-(4- (trifluoromethoxy)phenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (430 mg, 0.985 mmol) was reduced to the desired product as oil (410 mg, 95%). 1H NMR (400 MHz, CD3OD) δ 1.44 (s, 9 H), 1.66 (qd, J=12.82, 12.65, 4.39 Hz, 2 H), 1.84 - 1.95 (m, 2 H), 2.77 - 2.97 (m, 2 H), 3.14 (tt, J=11.89, 3.31 Hz, 1 H), 4.17 - 4.27 (m, 2 H), 7.07 - 7.16 (m, 3 H), 7.26 - 7.33 (m, 2 H), 7.73 (dd, J=7.21 , 1.56 Hz, 1 H), 7.92 (dd, J=4.87, 1.75 Hz, 2 H).
3-(Piperidin-4-yl)-2-(4-(trifluoromethoxy)phenoxy)pyridine hydrochloride salt:
Figure imgf000046_0002
Using General Procedure D, tert-butyl 4-(2-(4- (trifluoromethoxy)phenoxy)pyridin-3-yl)piperidine-1 -carboxylate (400.6 mg, 0.914 mmol) was reacted with acetyl chloride (71.7 mg, 0.914 mmol) in methanol (6 ml) to afford the desired product as a solid (250 mg, 73%). 1H NMR (400 MHz, CD3OD) δ 2.06 (qd, J=12.86, 4.15 Hz, 2 H), 3.19 (td, J=12.96, 2.70 Hz, 2 H), 3.31 - 3.41 (m, 1 H), 3.49 - 3.58 (m, 2 H), 7.19 - 7.27 (m, 3 H), 7.36 (d,
Figure imgf000046_0003
1.66 Hz, 1 H), 8.02 (dd, J=4.98, 1.66 Hz, 1 H). MS-APCI (m/z): 339 [M+1]+.
Example #4
3-Bromo-2-(2-chloro-4-methylphenoxy)pyridine:
Figure imgf000047_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (3.10 g, 16.1 mmol) was reacted with 2-chloro-4-cresol (3.22 g, 22.6 mmol) and potassium carbonate (5.57 g, 40.3 mmol) in Λ/,Λ/-dimethylformamide (20 ml) to afford the desired product was a solid (4.33 g, 90%). MS-APCI (m/z): 297 [M+1]+.
Terf-butyl 4-(2-(2-chloro-4-methylphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000047_0002
Using General Procedure B, 3-bromo-2-(2-chloro-4- methylphenoxy)pyridine (538.3 mg, 1.803 mmol) was reacted with te/t-butyl 4- (5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (639 mg, 2.16 mmol), cesium fluoride (548 mg, 3.61 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (74 mg, 0.09 mmol) in N, N- dimethylformamide (5 ml) to afford the desired product as oil (240 mg, 33%). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 2.33 (s, 3 H), 2.59 - 2.68 (m, 2 H), 3.62 (t, .£=5.46 Hz, 2 H), 4.05 - 4.13 (m, 2 H), 5.97 - 6.04 (m, 1 H), 6.94 (dd, J=7.41 , 4.87 Hz, 1 H), 7.04 (d, J=8.19 Hz, 1 H), 7.07 - 7.1 1 (m, 1 H), 7.26 (d, J=1.36 Hz, 1 H), 7.55 (dd, J=7.41 , 1.95 Hz, 1 H), 7.98 (dd, J=Λ.87, 1.75 Hz, 1 H). MS-APCI (m/z): 401 [M+1]+. 2-(2-Chloro-4-methylphenoxy)-3-(piperidin-4-yl)pyrϊdine:
Figure imgf000047_0003
First, te/t-butyl 4-(2-(2-chloro-4-methylphenoxy)pyridin-3-yl)piperidine- 1 -carboxylate was synthesized as oil (355.3 mg, 88 %) using General Procedure C with tert-butyl 4-(2-(2-chloro-4-methylphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate (400 mg, 0.998 mmol). Then using General Procedure D te/t-butyl 4-(2-(2-chloro-4-methylphenoxy)pyridin-3- yl)piperidine-1-carboxylate (106 mg, 0.263 mmol) was reacted with acetyl chloride (15 mg, 0.191 mmol) in methanol (6 ml) to afford the desired product as a solid (78 mg, 98 %). 1H NMR (400 MHz, CD3OD-Qf4) δ ppm 2.09 (qd, J=13.27, 3.73 Hz,2 H), 2.20 - 2.30 (m, 2 H), 2.36 (s, 3 H), 3.20 (td, J=13.06, 2.90 Hz, 2 H), 3.35 (tt, J=12.44, 3.73 Hz, 1 H), 3.49 - 3.58 (m, 2 H), 7.04 - 7.13 (m, 2 H), 7.18 (dd, J=8.29, 2.07 Hz, 1 H), 7.28 - 7.35 (m, 1 H), 7.74 (dd, J=7.46, 1.66 Hz, 1 H), 7.89 (dd, J=4.98, 2.07 Hz, 1 H). MS-APCI (m/z+): 304 (M+H).
Example #5
3-Bromo-2-(2-(trifluoromethoxy)phenoxy)pyridine:
Figure imgf000048_0001
Using General Procedure A, 3-bromo-2-chloropyridine (3.9 g, 20.33 mmol) was reacted with 2-(trifluoromethoxy)phenol (3.8 g, 21.35 mmol)and potassium carbonate (5.6 g, 40.66 mmol) in Λ/,Λ/-dimethylformamide (15 ml) to afford the desired product (4.1 g, 60%).1H NMR (400 MHz, CDCI3): δ 8.07 (dd, 1 H), 7.96 (dd, 1 H), 7.41-7.24 (m, 4H), 6.92-6.85 (m, 1 H). 19F NMR (400 MHz, CDCI3) δ -58 (s). ES-MS (m/z): 333.8 [M+1]+.
Tert-butyl 4-(2-(2-(tπfluoromethoxy)phenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000048_0002
Using General Procedure B, 3-bromo-2-(2- (trifluoromethoxy)phenoxy)pyridine (2.8 g, 8.4 mmol) was reacted with tert- butyl 4-(5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)- carboxylate (2.97 g, 10.1 mmol), cesium fluoride (1.9 g, 12.6 mmol) and [1 , 1'- Bis (diphenylphosphino)ferrocene] palladium (II) (0.92 g, 1.3 mmol) in N1N- dimethylformamide (15 ml) to afford the desired product (2.3 g, 64%).1H NMR (400 MHz, CDCI3) δ 8.02 (dd, 1 H), 7.59 (dd, 1 H), 7.38-7.19 (m, 4H), 7.02-6.96 (m, 1 H), 5.99 (s, 1 H), 4.18-4.07 (br s, 2H), 3.73-3.58 (br s, 2H), 2.66-2.54 (br s, 2H), 1.44 (S, 9H). 19F NMR (400 MHz, CDCI3) δ -58 (s). ES-MS (m/z): 437.09 [M+1]+.
Tert-butyl 4-(2-(2-(trifluoromethoxy)phenoxy)pyridin-3- yl)piperidine-1-carboxylate:
Figure imgf000049_0001
Using General Procedure C, tert-butyl 4-(2-(2- (trifluoromethoxy)phenoxy)pyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (2.14 g, 4.9 mmol) was reduced to the desired product (1.42 g, 68%). 1H NMR (400 MHz, CDCI3) δ 7.99 (dd, 1 H), 7.58 (dd, 1 H), 7.39-7.18 (m, 4H), 7.02-6.96 (m, 1 H), 4.43-4.17 (br s, 2H), 3.19-3.07 (m, 1 H), 2.99-2.73 (br s, 2H), 1.98 (d, 2H), 1.77-1.54 (m, 2H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -58 (s).
3-(Piperidin-4-yl)-2-(2-(trifluoromethoxy)phenoxy)pyridine hydrochloride:
Figure imgf000049_0002
Using General Procedure D, te/t-butyl 4-(2-(2- (trifluoromethoxy)phenoxy)pyridin-3-yl)piperidine-1-carboxylate (1.4 g, 3.1 mmol) was reacted with acetyl chloride (4.4 ml_, 62.1 mmol) in methanol (40 ml) to afford the desired product (1.1 g, 91%). 1H NMR (400 MHz, CD3OD) δ 7.98 (dd, 1 H), 7.78 (dd, 1 H), 7.42-7.11 (m, 4H), 7.16-7.09 (m, 1 H), 3.56 (d, 2H), 3.42-3.28 (m, 1 H), 3.19 (t, 2H), 2.28-1.99 (m, 4H). 19F NMR (400 MHz, CD3OD) δ -59 (s) ppm. ES-MS (m/z): 339.03 [M+1]+. CHN calculated for Ci7Hi7F3N2O2-I HCI.0.33H2O: C 53.62 %; H 4.94 %; N 7.36 %, and found: C 53.49 %; H 4.98 %; N 7.51 %.
Example # 6
3-Bromo-2-(2-(trifluoromethyl)phenoxy)pyridine:
Figure imgf000050_0001
Using General Procedure A1 3-bromo-2-chloropyridine (2.37 g, 12.3 mmol) was reacted with 2-trifluoromethylphenol (2 g, 12 mmol) and potassium carbonate (4.02 g, 12.3 mol) in Λ/,Λ/-dimethylformamide (10 ml) to afford the desired product (0.3 g, 8 %). 1H NMR (400 MHz, CDCI3): δ 8.04 (dd, 1 H), 7.95 (dd, 1 H), 7.72 (d, 1 H), 7.60 (t, 1 H), 7.34 (d, 2H), 7.26 (d, 1 H), 6.92 (q, 1 H). ES-MS (m/z) for Ci2H7BrF3NO: 317.86 & 319.81 [M+1]+.
Te/t-butyl 4-(2-(2-(trifluoromethyl)phenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000050_0002
Using General Procedure B, 3-bromo-2-(2- trifluoromethylphenoxy)pyridine (0.3 g, 0.9 mmol) was reacted with te/t-butyl
4-(5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)- carboxylate (0.362 g, 1.23 mmol), cesium fluoride (215 mg, 1.42 mmol) and
[1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (0.104 g, 0.1415 mol) in
Λ/,Λ/-dimethylformamide (5 ml) to afford the desired product (0.35 g, 85 %). 1H NMR (400 MHz, CDCI3): δ 8.03 (dd, 1 H), 7.70 (d, 1 H), 7.60 (dd, 1 H), 7.56 (t, 1 H), 7.38 (t, 1 H), 7.17 (d, 1 H), 7.02 (q, 1 H), 5.98 (s, 1 H), 4.08 (br s, 2H), 3.61 (t, 2H), 2.58 (m, 2H), 1.49 (s, 9H). rerf-butyl 4-(2-(2-(trifluorornethyl)phenoxy)pyridin-3-yl)piperidine- 1-carboxylate:
Figure imgf000051_0001
Using General Procedure C, tert-butyl 4-(2-(2- (trifluoromethyl)phenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (0.3 g, 3.1 mmol) was reduced to the desired product (0.3 g, 98 %). 1H NMR (400 MHz, CDCI3): δ 8.00 (dd, 1 H), 7.71 (dd, 1 H), 7.60-7.54 (m, 2H), 7.29 (d, 1 H), 7.25 (d, 2H), 7.01 (q, 2H), 4.28 (br s, 2H), 3.14 (m, 1 H), 2.84 (t, 2H), 1.95 (d, 2H), 1.62 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for C22H25F3N2O3: 423.16 [M+1]+.
3-(Piperidin-4-yl)-2-(2-(trifluoromethyl)phenoxy)pyridine hydrochloride:
Figure imgf000051_0002
Using General Procedure D, tert-butyl 4-(2-(2- (trifluoromethyl)phenoxy)pyridin-3-yl)piperidine-1-carboxylate (0.3 g, 0.7 mmol) was reacted with acetyl chloride (1 mL) in methanol (25 ml) to afford the desired product (0.2 g, 80%). 1H NMR (400 MHz, CD3OD): δ 7.96 (d, 1 H), 7.80 (d, 1 H), 7.75 (d, 1 H), 7.66 (t, 1 H), 7.39 (t, 1 H), 7.25 (d, 1 H), 7.15 (q, 1 H), 3.56 (d, 2H), 3.34 (m, 1 H), 3.18 (t, 2H), 2.22 (d, 2H), 2.06 (m, 2H). 19F NMR (376 MHz, CD3OD): δ -63.2. ES-MS (m/z) for C17H17F3N2OzM .25HCI: 323.09 [M+1]+. CHN calculated for C17H17F3N2O/"! .25HCI: C 55.50 %; H 5.00 %; N 7.61 %, and found: C 55.56 %; H 5.43 %; N 7.42 %.
Example #7 3-Bromo-2-(2-methoxyphenoxy)pyridine:
Figure imgf000052_0001
Using General Procedure A, 3-bromo-2-chloropyridine (5 g, 26 mmol) was reacted with 2-methoxyphenol (3.2 g, 26 mmol) and potassium carbonate (5.39 g, 39 mmol) in Λ/,Λ/-dimethylformamide (25 ml) to afford the desired product (3.7 g, 51%). 1H NMR (400 MHz, CDCI3): δ 8.02 (dd, 1 H), 7.91 (dd, 1 H), 7.28-7.14 (m, 2H), 7.04-6.96 (m, 2H), 6.85 (q, 1 H), 3.75 (s, 3H). ES-MS (m/z) for C12H10BrNO2: 279.86 & 281.81 [M+1]+.
Terf-butyl 4-(2-(2-methoxyphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000052_0002
Using General Procedure B, 3-bromo-2-(2-methoxyphenoxy)pyridine
(1.07 g, 3.82 mmol) was reacted with fert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.35 g, 4.58 mmol), cesium fluoride (0.87 g, 5.7 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (0.42 g, 0.57 mmol) in N1N- dimethylformamide (10 ml) to afford the desired product (1.20 g, 82 %). 1H NMR (400 MHz, CDCI3): δ 8.00 (dd, 1 H), 7.55 (dd, 1 H), 7.24-7.08 (m, 2H), 7.08-6.88 (m, 3H), 6.02 (s, 1 H), 4.09 (br s, 2H), 3.74 (s, 3H), 3.63 (br s, 2H), 2.65 (br s, 2H), 1.49 (s, 9H). ES-MS (m/z) for Chemical Formula: C22H26N2O4: 383.08 [M+1]+. Terf-butyl 4-(2-(2-methoxyphenoxy)pyridin-3-yl)piperidine-1- carboxylate:
Figure imgf000053_0001
Using General Procedure C, tert-butyl 4-(2-(2-methoxyphenoxy)pyridin- 3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.2 g, 3.1 mmol) was reduced to the desired product (0.9 g, 74 %). 1H NMR (400 MHz, CDCI3): δ 7.95 (dd, 1 H), 7.50 (dd, 1 H), 7.18 (t, 1 H), 7.11 (d, 1 H), 6.99 (m, 2H), 6.92 (q, 1 H), 4.26 (s, 2H)1 3.17 (m, 1 H), 2.86 (br s, 2H), 2.02 (d, 2H), 1.63 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for C22H28N2O4: 385.10 [M+1]+.
2-(2-Methoxyphenoxy)-3-(piperidin-4-yl)pyridine dihydrochloride:
Figure imgf000053_0002
Using General Procedure D, te/t-butyl 4-(2-(2-methoxyphenoxy)pyridin- 3-yl)piperidine-1 -carboxylate (0.84 g, 2.2 mmol) was reacted with acetyl chloride (2 ml_) in methanol (50 ml) to afford the desired product (0.72 g, quant.). 1H NMR (400 MHz, CD3OD): δ 8.17 (d, 1 H), 8.00 (d, 1 H), 7.42-7.28 (m, 3H), 7.21 (d, 1 H), 7.09 (t, 1 H), 3.73 (s, 3H), 3.56 (d, 2H), 3.46 (m, 1 H), 3.22 (t, 2H), 2.27 (d, 2H), 2.12 (m, 2H). ES-MS (m/z) for Ci7H20N2O2/2HCI: 285.09 [M+1]+. CHN calculated for C17H2oN2θ2/2HCI/2H2O: C 51.91 %; H 6.66 %; N 7.12 %, and found: C 52.87 %; H 6.44 %; N 7.07 %.
Example #8
3-Bromo-2-(2-ethoxyphenoxy)pyridine:
Figure imgf000054_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (1.0 g, 5.3 mmol) was reacted with 2-ethoxyphenol (732 mg, 5.3 mmol) and potassium carbonate (1.1 g, 8.0 mmol) in Λ/,Λ/-dimethylformamide (10 ml) to afford the desired product (0.58 g, 37 %). 1H NMR (400 MHz, CDCI3): δ 8.04 (dd, 1 H), 7.92 (dd, 1 H), 7.24-7.16 (m, 2H), 7.05-6.96 (m, 2H), 6.86 (m, 1 H), 3.97 (q, 2H), 1.14 (t, 3H). ES-MS (m/z) for C13H12BrNO2: 293.85 & 295.84 [M+1]+.
Tert-butyl 4-(2-(2-ethoxyphenoxy)pyridin-3-yl)-5,6-dihydropyridine- 1(2H)-carboxylate:
Figure imgf000054_0002
Using General Procedure B, 3-bromo-2-(2-ethoxyphenoxy)pyridine (0.58 g, 1.97 mmol) was reacted with te/t-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (873 mg, 2.96 mmol), cesium fluoride (449 mg, 2.96 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (216 mg, 2.96 mmol) in N, N- dimethylformamide (10 ml) to afford the desired product (0.57 g, 73 %). 1H NMR (400 MHz, CDCI3): δ 8.00 (dd, 1 H), 7.55 (dd, 1 H), 7.17 (t, 2H), 7.04-6.90 (m, 3H), 6.00 (s, 1 H), 4.11 (br s, 2H), 3.95 (q, 2H), 3.64 (br s, 2H), 2.67 (br s, 2H), 1.50 (s, 9H), 1.10 (t, 3H). ES-MS (m/z) for Chemical Formula: C23H28N2O4: 397.20 [M+I]+.
Terf-butyl 4-(2-(2-ethoxyphenoxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000055_0001
Using General Procedure C, tø/t-butyl 4-(2-(2-ethoxyphenoxy)pyridin-3- yl)-5,6-dihydropyridine-1(2H)-carboxylate (0.57 g, 2.3 mmol) was reduced to the desired product (0.63 g, 78 %). 1H NMR (400 MHz, CDCI3): δ 8.02 (dd, 1 H), 7.55 (dd, 1 H), 7.40 (t, 2H), 7.19 (t, 1 H), 7.10 (d, 2H), 6.98 (q, 1 H), 4.38 (s, 2H), 3.14 (m, 1 H), 2.85 (m, 2H), 1.95 (d, 2H), 1.64 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for C23H30N2O4: 399.03 [M+1]+.
2-(2-Ethoxyphenoxy)-3-(piperidin-4-yl)pyridine hydrochloride:
Figure imgf000055_0002
Using General Procedure D, tert-butyl 4-(2-(2-ethoxyphenoxy)pyridin-3- yl)piperidine-1-carboxylate (0.22 g, 0.62 mmol) was reacted with acetyl chloride (1 ml_) in methanol (25 ml) to afford the desired product (0.30 g, quant.). 1H NMR (400 MHz, CDCI3): δ 8.14 (br s, 1 H), 7.82 (br s, 1 H), 7.36- 6.96 (m, 5H), 3.95 (q, 2H), 3.67 (br s, 2H), 3.41 (br s, 1 H), 3.08 (br s, 2H), 2.28 (br s, 4H), 1.02 (t, 3H). ES-MS (m/z) for Ci8H22N2O2/2HCI: 299.02 [M+1]+. CHN calculated for
Figure imgf000055_0003
C 58.23 %; H 6.52 %; N 7.54 %, and found: C 58.61 %; H 6.73 %; N 7.55 %.
Example #9 Te/f-butyl 4-(2-hydroxypyridin-3-yl)piperidine-1 -carboxylate:
Figure imgf000056_0001
To a solution of te/t-butyl-4-(2-phenoxypyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate (1.0 g, 2.3 mmol) in methanol (30 mL) was added palladium (0.2 g, 10 wt % on activated carbon), and the suspension was hydrogenated at 35 psi for 4 hours. The mixture was filtered through a pad of Celite, and the filtrate was concentrated under reduced pressure. The residue was purified by flash column chromatography using 10 % methanol in ethyl acetate to give tert-butyl 4-(2-hydroxypyridin-3-yl)piperidine-1- carboxylate (0.68 g, 86 %). 1H NMR (400 MHz, CDCI3): δ 11.77 (s, 1 H), 7.24 (d, J = 7.2 Hz, 2H), 6.25 (t, J = 7.2 & 6.8 Hz, 1 H), 4.23 (s, 2H), 3.02 (m, 1 H), 2.86 (m, 2H), 1.88 (d, 2H), 1.70-1.40 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for Ci5H22N2O3: 278.97 [M+1]+.
Terf-butyl 4-(2-(2-fluorophenoxy)pyridin-3-yl)piperidine-1- carboxylate:
Figure imgf000056_0002
A mixture of 4-(2-hydroxypyridin-3-yl)piperidine-1 -carboxylate (340 mg, 1.2 mmol), 2-fluoro-1-iodobenzene (272 mg, 1.22 mmol), cesium carbonate (797 mg, 2.45 mmol) and 2,2,6,6-tetramethylheptane-3,5-dione (56 mg, 0.31 mmol) in 1 -methyl-2-pyrrolidinone (10 mL) was degassed and filled with nitrogen. To this mixture was added copper (I) chloride (61 mg, 0.61 mmol), the mixture was degassed and filled with nitrogen three times. This mixture was then heated at 120 °C overnight, cooled down to room temperature, diluted with ethyl acetate, filtered through a pad of Celite. The filtrate was washed thoroughly with water. The separated organic phase was concentrated under reduced pressure and purified over silica gel (hexanes/ethyl acetate,10:1 - 4:1 ) to afford te/t-butyl 4-(2-(2- fluorophenoxy)pyridin-3-yl)piperidine-1-carboxylate (0.177 g, 39 %). 1H NMR (400 MHz, CDCI3): δ 7.96 (dd, J = 1.6 & 4.8 Hz, 1 H), 7.54 (d, J = 7.2 Hz, 1 H), 7.25-7.15 (m, 4H), 6.97 (dd, 1 H), 4.28 (br s, 2H), 3.17 (m, 1 H), 2.87 (t, 2H), 1.99 (d, 2H), 1.65 (m, 2H), 1.49 (s, 9H). 19F NMR (376 MHz, CDCI3): δ - 128.888. 2-(2-Fluorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride:
Figure imgf000057_0001
To a 0 C solution of dry methanol (25 ml_) under nitrogen was added acetyl chloride (1 ml_) at 0 °C. The solution was stirred for 30 minutes, and then transferred to a flask containing te/t-butyl 4-(2-(2-fluorophenoxy)pyridin- 3-yl)piperidine-1-carboxylate (177 mg, 0.17 mmol). The mixture was stirred overnight at room temperature. The solvent was evaporated to afford target 2- (2-fluorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride (120 mg). 1H NMR (400 MHz, CD3OD): δ 8.14 (d, J = 7.6 Hz, 1 H), 8.06 (d, J = 5.2 Hz, 1 H), 7.44- 7.26 (m, 5H), 3.57 (d, 2H), 3.45 (m, 1 H), 3.23 (m, 2H), 2.26-1.90 (m, 4H). ES- MS (m/z) for C16H17FN2OZHCI: 273.03 (M++1). 19F NMR (376 MHz, CDCI3): δ -131.586. CHN calculated for C16H17FN2O/1.2HCI: C 60.80 %; H 5.80 %; N 8.86 %, and found: C 61.11 %; H 6.07 %; N 8.51 %.
Example #10
Terf-butyl 4-(2-(2-chlorophenoxy)pyridin-3-yl)piperidine-1- carboxylate:
Figure imgf000058_0001
A mixture of 4-(2-hydroxypyridin-3-yl)piperidine-1-carboxylate (0.34 g, 1.2 mmol), 2-chloro-1-iodobenzene (292 mg, 1.22 mmol), cesium carbonate (797 mg, 2.45 mmol) and 2,2)6,6-tetramethylheptane-3,5-dione (56 mg, 0.31 mmol) in 1 -methyl-2-pyrrolidinone (10 ml_) was degassed and filled with nitrogen. To this mixture was added copper (I) chloride (61 mg, 0.61 mmol), the mixture was degassed and filled with nitrogen three times. This mixture was then heated at 120 C overnight. The mixture was cooled to room temperature, diluted with ethyl acetate, and filtered through a pad of Celite. The filtrate was washed thoroughly with water. The separated organic phase was concentrated under reduced pressure and purified over silica gel (hexanes/ethyl acetate, 10:1 - 4:1) to afford terf-butyl 4-(2-(2- chlorophenoxy)pyridin-3-yl)piperidine-1-carboxylate (66 mg, 14 %). 1H NMR (400 MHz, CDCI3): δ 7.96 (d, J= 3.6 Hz, 1 H), 7.55 (d, J= 7.2 Hz, 1 H), 7.48 (d, J = 7.2 Hz, 1 H), 7.32 (t, 1 H), 7.23-7.15 (m, 2H), 6.97 (t, 1 H), 4.28 (br s, 2H), 3.19 (m, 1 H), 2.87 (br s, 2H), 2.02 (d, 2H), 1.66 (m, 2H), 1.49 (s, 9H).
2-(2-Chlorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride salt:
Figure imgf000058_0002
To a cold solution of dry methanol (25 ml_) under nitrogen was added acetyl chloride (1 ml_) at 0 0C. The solution was stirred for 30 minutes, and then transferred to a flask containing tert-butyl 4-(2-phenoxypyridin-3- yl)piperidine-1 -carboxylate (66 mg, 0.17 mmol). The mixture was stirred overnight at room temperature. The solvent was evaporated to afford target 2- (2-chlorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride (0.044 g, 80%). 1H NMR (400 MHz, CD3OD): δ 7.90 (del, 1 H), 7.78 (dd, 1 H)1 7.50 (dd, 1 H), 7.38 (m, 1 H), 7.28-7.21 (m, 2H), 7.11 (dd, 1 H), 3.55 (d, 2H), 3.38 (m, 1 H), 3.21 (t, 2H), 2.27 (d, 2H), 2.11 (m, 2H). ES-MS (m/z) for C16H17CIN2OZHCI: 289.03 [M+1]+. 13C NMR (400 MHz, CD3OD): δ 160.4, 149.8, 144.9, 137.2, 130.4, 128.2, 127.1 , 126.8, 126.4, 124.2, 119.4, 44.4, 34.3, 27.9..
Example #11 3-Bromo-2-(2-chloro-3-(trifluoromethyl)phenoxy)pyridine:
Figure imgf000059_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (3.52 g, 18.3 mmol) was reacted with 2-chloro-3-(trifluoromethyl)phenol (4.32 g, 22 mmol) and potassium carbonate (4.29 g, 27.5 mmol) in Λ/,Λ/-dimethylformamide (15 ml) to afford the desired product as an oil (3.59 g, 56%). 1H NMR (400 MHz, CDCI3) δ 6.93 (dd, J=7.60, 4.87 Hz, 1 H), 7.41 - 7.46 (m, 2 H), 7.59 - 7.64 (m, 1 H), 7.96 (dd, ^=7.70, 1.66 Hz, 1 H), 8.00 (dd, J=4.78, 1.66 Hz, 1 H). MS- APCI (m/z): 351 (M+H).
Teit-butyl 4-(2-(2-chloro-3-(trifluoromethyl)phenoxy)pyridin-3-yl)- 5,6-dihydropyridine-1(2H)-carboxylate:
Figure imgf000059_0002
Using General Procedure B, 3-bromo-2-(2-chloro-3- (trifluoromethyl)phenoxy)pyridine (3.5 g, 9.93 mmol) was reacted with tert- butyl 4-(5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)- carboxylate (3.68 g, 11.9 mmol), cesium fluoride (2.26 g, 14.9 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (162 mg, 0.199 mmol) in Λ/,Λ/-dimethylformamide (20 ml) to afford the desired product as a solid (320 mg, 7%). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 2.60 - 2.67 (m, 2 H), 3.62 (t, J=5.65 Hz, 2 H), 4.06 - 4.08 (m, 2 H), 6.00 - 6.05 (m, 1 H), 7.00 (dd, J=7.41 , 4.87 Hz, 1 H), 7.34 - 7.42 (m, 2 H), 7.56 (dd, J=7.31 , 2.05 Hz, 1 H), 7.59 (dd, Λ=7.41 , 1 .95 Hz, 1 H), 7.96 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 455 (M+H), 354 (M-100).
Te/f-butyl 4-(2-(2-chloro-3-(trifluoromethyl)phenoxy)pyridin-3- yl)piperidine-1 -carboxylate:
Figure imgf000060_0001
Using General Procedure C, tert-butyl 4-(2-(2-chloro-3- o (trif luoromethyl)phenoxy)-pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (300 mg, 0.660 mmol) was reduced to the desired product as oil (220 mg, 73%). 1H NMR (400 MHz, CDCI3) δ 1 .17 - 1.33 (m, 2 H), 1.46 (s, 9 H), 1 .59 - 1.76 (m, 2 H), 1.97 - 2.03 (m, 2 H), 2.74 - 2.96 (m, 2 H), 3.16 (tt, dd, J=12.48, 3.31 Hz, 1 H), 7.00 (dd, dd, J=7.41 , 4.87 Hz, 1 H)1 7.39 - 7.43 (m, 2 H), 7.54 -5 7.59 (m, 2 H)1 7.92 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 457 (M+H), 357 (M+H-100).
2-(2-Chloro-3-(trifluoromethyl)phenoxy)-3-(piperidin-4-yl)pyridine hydrochloride salt:
Figure imgf000060_0002
0 Using General Procedure D, tert-butyl 4-(2-(2-chloro-3-
(trifluoromethyl)phenoxy)-pyridin-3-yl)piperidine-1 -carboxylate (220 mg, 0.521 mmol) was reacted with acetyl chloride (0.741 ml, 10.4 mmol) in methanol (5 ml) to afford the desired product as a solid (130 mg, 63%). 1H NMR (400 MHz, CD3OD) δ 2.15 (m, 2 H), 2.24 (d, J=1.75 Hz, 2 H), 3.20 (t, J=2.53 Hz, 1 H),5 3.29 (dt, .£=3.27, 1.58 Hz, 4 H), 3.54 (d, J=12.67 Hz, 1 H), 7.15 (m, 1 H), 7.55 (m, 2 H), 7.68 (d, J=1.68 Hz, 1 H), 7.77 (d, J=λ A Hz, 1.4 Hz, 1 H), 7.95 (m, 1 H). MS-APCI (m/z+): 357 (M+H).
Example #12 3-Bromo-2-(2-chloro-6-methoxyphenoxy)pyridine:
Figure imgf000061_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (485.4 mg, 2.522 mmol) was reacted with 2-chloro-6-methoxylphenol (400 mg, 252 mmol) and potassium carbonate (591 mg, 3.78 mmol) in N, N- dimethylformamide (5 ml) to afford the desired product as a solid (280 mg, 35%). 1H NMR (400 MHz, CDCI3) δ 3.76 (s, 3 H), 6.90 (dd, J=8.19, 1.56 Hz, 1 H), 6.93 (dd, J=6.63, 2.92 Hz, 1 H), 7.06 (dd, J=6.63,1.56 Hz, 1 H), 7.14 (t, J=6.63 Hz, 1 H), 7.91 (dd, J=7.60, 1.56 Hz, 1 H), 7.98 (dd, J=4.78, 1.66 Hz, 1 H). MS-APCI (m/z): 315 (M+H).
Te/t-butyl 4-(2-(2-chloro-6-methoxyphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate:
Figure imgf000061_0002
Using General Procedure B, 3-bromo-2-(2-chloro-6- methoxyphenoxy)pyridine (280 mg, 0.89 mmol) was reacted with tert-butyl 4- (5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (330 mg, 1.07 mmol), cesium fluoride (203 mg, 1.34 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (14.5 mg, 0.018 mmol) in N, N- dimethylformamide (4 ml) to afford the desired product as a solid (180 mg, 49%). 1H NMR (400 MHz, CDCI3) δ 2.61 - 2.73 (m, 2 H), 3.57 - 3.67 (m, 2 H), 3.73 (s, 3 H), 4.04 - 4.13 (m, 2 H), 6.02 - 6.07 (m, 1 H), 6.89 (dd, J=8.19, 1.56 Hz, 1 H), 6.93 (dd, J=7.41 , 4.87 Hz, 1 H), 7.05 (dd, J=8.19, 1.56 Hz, 1 H), 7.12 (t, J=8.19 Hz, 1 H), 7.54 (dd, J=7.41 , 1.95 Hz, 1 H), 7.93 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z): 417 (M+H), 317 (M+H-100).
7erf-butyl 4-(2-(2-chloro-6-methoxyphenoxy)pyridin-3- yl)piperidine-1 -carboxylate:
Figure imgf000062_0001
Using General Procedure C, tert-butyl 4-(2-(2-chloro-6- methoxyphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (180 mg, 0.432 mmol) was reduced to the desired product as glass (180 mg, quant). 1H NMR (400 MHz, CDCI3) δ 1.49 (s, 9 H) 1.57 - 1.72 (m, 4 H), 2.01 - 2.12 (m, 2 H), 2.80 - 2.95 (m, 2 H), 3.21 (tt, J=12.09, 3.31 Hz, 1 H), 6.87 - 6.97 (m, 2 H), 7.07 (dd, J=8.19, 1.56 Hz,1 H), 7.13 (t, J=8.19 Hz,1 H), 7.52 (dd, J=7.41 , 1.36 Hz, 1 H), 7.91 (dd, J=4.97, 1.85 Hz, 1 H). MS-APCI (m/z): 419 (M+H), 319 (M+H-100).
2-(2-Chloro-6-methoxyphenoxy)-3-(piperidin-4-yl)pyridine hydrochloride salt:
Figure imgf000062_0002
Using General Procedure D, terf-butyl 4-(2-(2-chloro-6- methoxyphenoxy)pyridin-3-yl)piperidine-1 -carboxylate (200 mg, 0.477 mmol) was reacted with acetyl chloride (0.679 ml, 9.55 mmol) in methanol (5 ml) to afford the desired product as a solid (100 mg, 59%). 1H NMR (400 MHz, CD3OD) δ 2.00-2.20 (m, 4 H), 3.29 (dt, J=3.27, 1.58 Hz, 2 H), 3.35 (m, 1 H), 3.51 (d, J=3.2 Hz, 2 H), 3.71 (s, 3 H), 7.00-7.10 (m, 3H), 7.21 (t, J=3.2 Hz, 1 H), 7.70 (t, 3.2 Hz, 1 H) 7.82 (m, 1 H). MS-APCI (m/z): 319 (M+H).
Example #13 3-Bromo-2-(2,3-dichlorophenoxy)pyridine:
Figure imgf000063_0001
Using General Procedure A, 3-bromo-2-chloro pyridine (1.5 g, 7.79 mmol) was reacted with 2,3-dichlorophenol (1.78 g, 10.9 mmol) and potassium carbonate (2.37 g, 17.1 mmol) in Λ/,Λ/-dimethylformamide (7.79 ml) to afford the desired product as a solid (1.2 g, 48%). MS-APCI (m/z): 319 (M+H).
7eιf-butyl 4-(2-(2,3-dichlorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000063_0002
Using General Procedure B, 3-bromo-2-(2,3-dichlorophenoxy)pyridine (324 mg, 1.02 mmol) was reacted with tert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (377 mg, 1.22 mmol), cesium fluoride (463 mg, 3.05 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (119 mg, 0.163 mmol) in N, N- dimethylformamide (2.5 ml) to afford the desired product as a solid (428 mg, 58%). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 2.57 - 2.66 (m, 2 H), 3.56 - 3.66 (m, 2 H), 4.04 - 4.12 (m, 2 H), 5.98 - 6.04 (m, 1 H), 6.99 (dd,
Figure imgf000063_0003
, 4.97 Hz, 1 H), 7.09 (dd, Λ=8.09, 1.46 Hz, 1 H), 7.23 (t, J=8.19 Hz, 1 H), 7.33 (dd, J=8.19, 1.56 Hz, 1 H), 7.58 (dd, J=7.41 , 1.95 Hz, 1 H), 7.97 (dd, ^4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 421 (M+H)
Terf-butyl 4-(2-(2,3-dichlorophenoxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000063_0004
Using General Procedure C, tert-butyl 4-(2-(2,3- dichlorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (246 mg, 0.584 mmol) was reduced to the desired product (244 mg, 99%). MS-APCI (m/z): 423 (M+H).
2-(2,3-Dichlorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride salt:
Figure imgf000064_0001
Using General Procedure D, tert-butyl 4-(2-(2,3- dichlorophenoxy)pyridin-3-yl)piperidine-1 -carboxylate (244 mg, 0.576 mmol) was reacted with acetyl chloride (0.41 ml, 5.76 mmol) in methanol (5 ml) then purified with ion exchange column followed by silica gel chromatography to afford the desired product as gum (111 mg, 60%). 1H NMR (400 MHz, CDCI3) δ 1.61 - 1.78 (m, 2 H), 1.95 - 2.02 (m, 2 H), 2.80 (dt, J=12.28, 2.53 Hz, 2H),
3.13 (tt, J=12.16, 3.46 Hz, 1 H), 3.22 (dt, J=11.82, 3.12 Hz, 2 H), 6.98 (dd, J=7.41 , 4.87 Hz, 1 H), 7.21 - 7.25 (m, 2 H), 7.32 (dd, J=7.99, 1.56 Hz, 1 H),
7.60 (dd, J=7.41 , 1.75 Hz, 1 H), 7.92 (dd, J=4.97, 1.85 Hz, 1 H). MS-APCI
(m/z): 323 (M+H).
Example #14 3-Bromo-2-(2,4-dichlorophenoxy)pyridine:
Figure imgf000064_0002
Using General Procedure A, 3-bromo-2-chloro pyridine (1.50 g, 7.79 mmol) was reacted with 2,4-dichlorophenol (1.78 g, 10.9 mmol) and potassium carbonate (2.37 g, 17.1 mmol) in Λ/,Λ/-dimethylformamide (7.79 ml) to afford the desired product as a solid (1.49 g, 60%). 1H NMR (400 MHz, CDCI3) δ 6.90 (dd, J=7.60, 4.87 Hz, 1 H), 7.17 (d, J=8.77 Hz, 1 H), 7.29 (dd, J=8.58, 2.53 Hz, 1 H), 7.47 (d, J=2.34 Hz, 1 H), 7.93 (dd, J=7.70, 1.66 Hz, 1 H), 8.00 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 319 (M+H).
Te/t-butyl 4-(2-(2,4-dichlorophenoxy)pyιϊdin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000065_0001
Using General Procedure B, 3-bromo-2-(2,4-dichlorophenoxy)pyridine (300 mg, 0.940 mmol) was reacted with tert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (349 mg, 1.13 mmol), cesium fluoride (429 mg, 2.82 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (110 mg, 0.150 mmol) in N1N- dimethylformamide (2.35 ml) to afford the desired product as a solid (221 mg, 56%). 1H NMR (400 MHz, CDCI3) δ 1.48 (s, 9 H), 2.57 - 2.66 (m, 2 H), 3.62 (t, J=5.65 Hz, 2 H), 4.08 (q, J=2.53 Hz, 2 H), 5.97 - 6.03 (m, 1 H), 6.99 (dd, ^7.41 , 4.87 Hz, 1 H), 7.11 (d, J=8.77 Hz, 1 H), 7.26 (dd, J=8.77, 2.53 Hz, 1 H), 7.45 (d, J=2.34 Hz, 1 H), 7.57 (dd, J=7.31 , 1.85 Hz, 1 H), 7.97 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z): 421 (M+H).
Terf-butyl 4-(2-(2,4-dichlorophenoxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000065_0002
Using General Procedure C, tert-butyl 4-(2-(2,4- dichlorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (171 mg, 0.406 mmol) was reduced to the desired product as glass (160 mg, 93%). MS-APCI (m/z): 423 (M+H). 2-(2,4-Dichlorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride salt:
Figure imgf000066_0001
Using General Procedure F, te/f-butyl 4-(2-(2,4- dichlorophenoxy)pyridin-3-yl)piperidine-1 -carboxylate (171 mg, 0.404 mmol) was reacted with HCI solution (8 ml) in methanol (3 ml) to afford the desired product as a solid (21 mg, 14%). 1H NMR (400 MHz, DMSO-dfe) δ 1.91 - 2.03
(m, 2 H), 2.02 - 2.13 (m, 2 H), 3.01 - 3.15 (m, 2 H), 3.19 - 3.30 (m, 2 H), 3.33 -
3.38 (m, 1 H), 7.16 (dd, J=7.41 , 4.87 Hz, 1 H), 7.36 (d, J=8.77 Hz, 1 H), 7.50 (dd, J=8.77, 2.53 Hz, 1 H), 7.73 (dd, J=7.51 , 1.66 Hz, 1 H), 7.78 (d, J=2.53
Hz, 1 H), 7.95 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 323 (M+H).
Example #15 3-Bromo-2-(2-fluoro-4-methylphenoxy)pyridine:
Figure imgf000066_0002
Using General Procedure A, 3-bromo-2-chloropyridine (3.0 g, 15.6 mmol) was reacted with 2-fluoro-4-methylphenol (1.97 g, 15.6 mmol) and potassium carbonate (3.65 g, 23.4 mmol) in Λ/,/V-dimethylformamide (15 ml) to afford the desired product as glass (3.3 g, 75%). 1H NMR (400 MHz, CDCI3) δ 2.36 (s, 3 H), 6.87 (dd, J=7.70, 4.78 Hz, 1 H), 6.93 - 7.03 (m, 2 H), 7.11 (t, J=8.58 Hz, 1 H), 7.90 (dd, J=7.60, 1.75 Hz, 1 H), 8.02 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 283 (M+H).
Tert-butyl 4-(2-(2-fluoro-4-methylphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000067_0001
Using General Procedure B, 3-bromo-2-(2-fluoro-4- methylphenoxy)pyridine (3.3 g, 1.7 mmol) was reacted with tert-butyl 4-(5,5- dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (3.68 g, 11.9 mmol), cesium fluoride (2.26 g, 14.9 mmol) and [1 , 1'-Bis
(diphenylphosphino)ferrocene] palladium (II) (162 mg, 0.199 mmol) in N, N- dimethylformamide (20 ml) to afford the desired product as gum (2.6 g, 58%).
1H NMR (400 MHz, CDCI3) δ 1.46 (s, 9 H), 2.33 (s, 3 H), 2.55 - 2.64 (m, 2 H),
3.61 (t, J=5.65 Hz, 2 H), 4.03 - 4.12 (m, 2 H), 5.94 - 6.02 (m, 1 H), 6.92 - 7.01 (m, 3 H), 7.06 (t, J=8.58 Hz, 1 H), 7.54 (dd, J=7.31 , 1.85 Hz, 1 H), 7.99 (dd,
J=4.97, 1.85 Hz, 1 H). MS-APCI (m/z): 385 (M+H), 285 (M+1-100).
Terf-butyl 4-(2-(2-fluoro-4-methylphenoxy)pyridin-3-yl)piperidine- 1-carboxylate:
Figure imgf000067_0002
Using General Procedure C, te/t-butyl 4-(2-(2-fluoro-4- methylphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.5 g, 3.9 mmol) was reduced to the desired product as gum (1.18 g, 78%). 1H NMR (400 MHz, CDCI3) δ 1.45 (s, 9 H), 1.53 - 1.72 (m, 2 H), 1.91 - 2.01 (m, 2 H), 2.34 (s, 3 H), 2.75 - 2.95 (m, 2 H), 3.14 (tt, J=12.28, 3.51 Hz, 1 H), 4.11 - 4.40 (m, 2 H), 6.90 - 7.01 (m, 3 H), 7.07 (t, J=7.99 Hz, 1 H), 7.51 (dd, J=7.21 , 1.56 Hz, 1 H), 7.94 (dd, J=4.87, 1.75 Hz 1 H). MS-APCI (m/z+): 387 (M+H), 287 (M+1-100).
2-(2-Fluoro-4-methylphenoxy)-3-(piperidin-4-yl)pyrϊdine hydrochloride salt:
Figure imgf000068_0001
Using General Procedure D, tert-butyl 4-(2-(2-fluoro-4- methylphenoxy)pyridin-3-yl)piperidine-1 -carboxylate (1.2 g, 2.9 mmol) was reacted with acetyl chloride (4.1 ml, 58 mmol) in methanol (50 ml) to afford the desired product as a solid (830 mg, 89%). 1H NMR (400 MHz, CD3OD) δ 2.07 (td, J=13.01 , 3.61 Hz, 2 H), 2.22 (d, J=1.75 Hz, 2 H), 2.35 (s, 3 H), 3.18 (td, J=13.06, 3.12 Hz, 2 H), 3.29 (dt, J=3.27, 1.58 Hz, 1 H), 3.52 (dd, J=10.62, 1.85 Hz, 2 H), 7.00 - 7.11 (m, 4 H), 7.73 (dd, J=7.51 , 1.85 Hz, 1 H), 7.89 (dd, J=4.97, 1.85 Hz, 1 H). MS-APCI (m/z+): 287 (M+H). Example #16
Terf-butyl 4-(2-(2-ethoxy-6-fluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000068_0002
Using General Procedure A, reaction of 3-bromo-2-chloropyridine (600 mg, 3.12 mmol), 2-ethoxy-6-fluorophenol (445 mg, 3.12 mmol) and potassium carbonate (862 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (2.60 ml) afforded
3-bromo-2-(2-ethoxy-6-fluorophenoxy)pyridine as a solid (800 g, 86%). Then using General Procedure B, reaction of 3-bromo-2-(2-ethoxy-6- fluorophenoxy)pyridine (348 mg, 1.12 mmol) with te/f-butyl 4-(5,5-dimethyl- 1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (414 mg, 1.34 mmol), cesium fluoride (508 mg, 3.34 mmol) and [1 , 1'-Bis
(diphenylphosphino)ferrocene] palladium (II) (130 mg, 0.178 mmol) in N1N- dimethylformamide (2.79 ml) afforded the desired product as an oil (208 mg, 45%). 1H NMR (400 MHz, CDCI3) δ 1.15 (t, J=7.02 Hz, 3 H), 1.47 (s, 9 H), 2.59 - 2.70 (m, 2 H), 3.61 (t, J=5.36 Hz, 2 H), 3.95 (q, J=7.02 Hz, 2 H), 4.02 - 4.10 (m, 2 H), 5.94 - 6.04 (m, 1 H), 6.68 - 6.82 (m, 2 H), 6.91 (dd, Λ=7.31 , 4.97 Hz, 1 H), 7.06 (dddf J=B.A3, 6.14 Hz, 1 H), 7.51 (dd, J=7.31 , 1.85 Hz, 1 H), 7.94 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z): 415 (M+H).
2-(2-Ethoxy-6-fluorophenoxy)-3-(piperidin-4-yl)pyridine trifluoroacetic acid salt:
Figure imgf000069_0001
Using General Procedure C, reaction of te/t-butyl 4-(2-(2-ethoxy-6- fluorophenoxy)-pyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (244 mg,
0.589 mmol) afforded te/t-butyl 4-(2-(2-ethoxy-6-fluorophenoxy)pyridin-3- yl)piperidine-1-carboxylate as an oil (220 mg, 90%). Then using General
Procedure D, te/t-butyl 4-(2-(2-ethoxy-6-fluorophenoxy)pyridin-3-yl)piperidine-
1 -carboxylate (139 mg, 0.334 mmol) was reacted with acetyl chloride (26.2 mg, 0.334 mmol) in methanol (2 ml) then purified by HPLC with 0.025% trifluoroacetic acid to afford the desired product as a solid (73 mg, 46%). 1H
NMR (400 MHz, CD3OD) δ 1.10 (t, J=6.92 Hz, 3 H), 2.01 - 2.18 (m, 2 H), 2.23
- 2.27 (m, 2 H), 3.11 - 3.24 (m, 2 H), 3.25 - 3.43 (m, 2 H), 3.46 - 3.58 (m, 2
H), 3.95 (q, J=6.89 Hz, 2 H), 6.74 - 6.91 (m, 2 H), 7.05 (dd, J=7.31 , 4.97 Hz, 1 H), 7.04 - 7.18 (m, 1 H), 7.70 (d, J=7.41 Hz, 1 H), 7.84 (d, J=3.31 Hz, 1 H).
MS-APCI (m/z): 318 (M+H).
Example #17 te/t-butyl 4-(2-(4-fluoro-2-methoxyphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000070_0001
Using General Procedure A, reaction of 3-bromo-2-chloropyridine (600 mg, 3.12 mmol), 2-methoxy-4-fluorophenol (443 mg, 3.12 mmol) and potassium carbonate (862 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (3 ml) afforded 3-bromo-2-(4-fluoro-2-methoxyphenoxy)pyridine as a solid (750 mg, 81 %). Then using General Procedure B, 3-bromo-2-(4-fluoro-2- methoxyphenoxy)pyridine (460 mg, 1.54 mmol) was reacted with te/t-butyl 4- (5,5-dimethyl-1 )3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1(2H)-carboxylate (573 mg, 1.85 mmol), cesium fluoride (703 mg, 4.63 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (181 mg, 0.247 mmol) in N, N- dimethylformamide (3.86 ml) to afford the desired product (313 mg, 51 %). 1H NMR (400 MHz, CDCI3) δ 1.46 (s, 9 H), 2.54 - 2.64 (m, 2 H), 3.60 (t, J=5.36 Hz, 2 H), 3.66 (s, 3 H), 4.06 (d, J=2.53 Hz, 2 H), 5.94 - 6.01 (m, 1 H), 6.83 - 6.89 (m, 3 H), 6.93 (dd, J=7.31 , 4.97 Hz, 1 H), 7.53 (dd, J=7.31 , 1.85 Hz, 1 H), 7.96 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z): 401 (M+H), 301 (M+H- 100).
Tert-butyl 4-(2-(4-fluoro-2-methoxyphenoxy)pyridin-3- yl)piperidine-1-carboxylate:
Figure imgf000070_0002
Using General Procedure C, te/t-butyl 4-(2-(4-fluoro-2- methoxyphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (189 mg, 0.472 mmol) was reduced to the desired product as oil (144 mg, 76%). 1H NMR (400 MHz, CD3OD) δ 2.03 - 2.32 (m, 4 H), 3.20 (d, J=5.07 Hz, 2 H), 3.41 - 3.52 (m, 1 H), 3.55 (d, J=11.89 Hz, 2 H), 3.72 (s, 3 H), 7.09 - 7.19 (m, 1 H), 7.17 - 7.30 (m, 2 H), 7.45 - 7.56 (m, 1 H), 8.08 (d, J=4.87 Hz, 1 H), 8.26 - 8.36 (m, 1 H).
2-(4-Fluoro-2-methoxyphenoxy)-3-(piperidin-4-yl)pyridine:
Figure imgf000071_0001
Using General Procedure D, te/f-butyl 4-(2-(4-fluoro-2- methoxyphenoxy)pyridin-3-yl)piperidine-1-carboxylate (76.9 mg, 0.191 mmol) was reacted with acetyl chloride (15 mg, 0.191 mmol) in methanol (2 ml) to afford the desired product as a solid (65 mg, quant). 1H NMR (400 MHz, CD3OD) δ 2.05 (dq, J=13.69, 4.15 Hz, 2 H), 2.19 - 2.29 (m, 2 H), 3.19 (td, J=12.96, 2.70 Hz, 2 H), 3.28 - 3.39 (m, 1 H), 3.48 - 3.57 (m, 2 H), 3.65 (s, 3 H), 6.93 - 7.01 (m, 2 H), 7.03 - 7.11 (m, 2 H), 7.72 (d, J=6.22 Hz, 1 H), 7.88 (dd, J=4.98, 2.07 Hz, 1 H). MS-APCI (m/z): 303 (M+H).
Tert-butyl 4-(2-(2-ethyl-4-fluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate:
Figure imgf000071_0002
Using General Procedure A, reaction of 3-bromo-2-chloro pyridine (600 mg, 3.12 mmol), 2-ethyl-4-fluorophenol (437 mg, 3.12 mmol) and potassium carbonate (826 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (3 ml) afforded 3- bromo-2-(2-ethyl-4-fluorophenoxy)pyridine as an oil (778 mg, 84%). Then using General Procedure B, 3-bromo-2-(2-ethyl-4-fluorophenoxy)pyridine (250 mg, 0.844 mmol) was reacted with te/t-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (313 mg, 1.01 mmol), cesium fluoride (385 mg, 2.53 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (98.8 mg, 0.135 mmol) in N,N- dimethylformamide (2.11 ml) to afford the desired product as a solid (221 mg, 66%). 1H NMR (400 MHz, CDCI3) δ 1.13 (t, J=7.60 Hz, 3 H), 1.48 (s, 9 H), 2.50 (q, J=7.60 Hz, 2 H), 2.55 - 2.63 (m, 2 H), 3.61 (t, J=5.56 Hz, 2 H), 4.08 (d, J=2.14 Hz, 2 H), 5.93 - 5.99 (m, 1 H), 6.87 - 7.01 (m, 4 H), 7.54 (dd, J=7.41 , 1.95 Hz, 1 H), 7.98 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z+): 399 (M+H).
2-(2-Ethyl-4-fluorophenoxy)-3-(piperidin-4-yl)pyridine trifluoroacetic acid salt:
Figure imgf000072_0001
Using General Procedure C, reaction of te/t-butyl 4-(2-(2-ethyl-4- fluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (224 mg, 0.562 mmol) afforded fe/t-butyl 4-(2-(2-ethyl-4-fluorophenoxy)pyridin-3- yl)piperidine-1-carboxylate as oil (186 mg, 83%). Then using General Procedure D, tert-butyl 4-(2-(2-ethyl-4-fluorophenoxy)pyridin-3-yl)piperidine-1- carboxylate (146 mg, 0.365 mmol) was reacted with acetyl chloride (28.6 mg, 0.365 mmol) in methanol (2 ml) then purified with HPLC with 0.025% trifluoroacetic acid to afford the desired product as oil (103 mg, 63%). 1H NMR (400 MHz, CD3OD) δ 1.15 (t, J=7.67 Hz, 3 H), 2.07 (qd, J=13.27, 3.32 Hz, 2 H), 2.17 - 2.28 (m, 2 H), 2.52 (q, J=7.46 Hz, 2 H), 3.20 (td, J=13.06, 2.90 Hz, 2 H), 3.36 (tt, J=12.18, 3.42, 3.32 Hz, 1 H), 3.51 - 3.58 (m, 2 H), 6.93 - 6.99 (m, 2 H), 7.04 - 7.12 (m, 2 H), 7.75 (dd, J=7.46, 1.66 Hz, 1 H), 7.90 (dd, J=4.98, 1.66 Hz, 1 H). MS-APCI (m/z): 301 (M+H). Example #19
2-Fluoro-6-methylphenol:
Figure imgf000073_0001
To a solution of 3-fluoro-2-hydroxybenzaldehyde (5.84 g, 41.7 mmol) in methanol (60 ml_) was added 10% palladium in charcoal (4.4 g, 4.2 mmol) and the mixture was hydrogenated at 60 psi for 3 h. The mixture was filtered through a pad of Celite and the filtrate was evaporated. The crude was purified by flash column chromatography using 5% ethyl acetate in hexanes to obtain the desired product (2.89 g, 55%). 1H NMR (400 MHz, CDCI3) δ 6.99- 6.73 (m, 3H), 5.12 (d, 1 H), 2.26 (s, 3H). 19F NMR (400 MHz, CDCI3) δ -143
(S). 3-Bromo-2-(2-fluoro-6-methylphenoxy) pyridine:
Figure imgf000073_0002
Using General Procedure A', 3-bromo-2-chloropyridine (1.39 g, 7.21 mmol) was reacted with 2-fluoro-6-methylphenol (1.00 g, 7.93 mmol) and cesium carbonate (3.56 g, 10.8 mmol) in dimethyl sulfoxide (8 ml) to afford the desired product (1.93 g, 90%).1H NMR (400 MHz, CDCI3): δ 8.02 (dd, 1 H), 7.96 (dd, 1 H), 7.18-6.84 (m, 4H), 2.21 (s, 3H). 19F NMR (400 MHz, CDCI3) δ - 129 (S). ES-MS m/z 281.94 [M+1]+. re/t-butyl 4-(2-(2-fluoro-6-methylphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000073_0003
Using General Procedure B, 3-bromo-2-(2-fluoro-6- methylphenoxy)pyridine (2.5 g, 8.9 mmol) was reacted with te/t-butyl 4-(5,5- dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (3.15 g, 10.67 mmol), cesium fluoride (2.03 g, 13.34 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (0.98 g, 1.33 mmol) in N,N- dimethylformamide (15 ml) to afford the desired product (2.09 g, 62%).1H NMR (400 MHz, CDCI3) δ 7.99 (dd, 1 H), 7.58 (dd, 1 H), 7.15-6.92 (m, 4H), 5.99 (s, 1 H), 4.18-4.07 (br s, 2H), 3.73-3.58 (br s, 2H), 2.66-2.54 (br s, 2H), 2.21 (s, 3H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -130 (s).
Terf-butyl 4-(2-(2-fluoro-6-methylphenoxy)pyridin-3-yl)piperidine- 1-carboxylate:
Figure imgf000074_0001
Using General Procedure C, tert-butyl 4-(2-(2-fluoro-6- methylphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (2 g, 5.21 mmol) was reduced to the desired product (1.83 g, 92%). 1H NMR (400 MHz, CDCI3) δ 7.99 (dd, 1 H), 7.58 (dd, 1 H), 7.19-6.91 (m, 4H), 4.43-4.17 (br s, 2H), 3.22-3.11 (m, 1 H), 2.99-2.73 (br s, 2H), 2.21 (s, 3H), 1.98 (d, 2H), 1.81-1.58 (m, 2H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -130 (s).
2-(2-Fluoro-6-methylphenoxy)-3-(piperidin-4-yl)pyridine dihydrochloride:
Figure imgf000074_0002
Using General Procedure D, terf-butyl 4-(2-(2-fluoro-6- methylphenoxy)pyridin-3-yl)piperidine-1-carboxylate (1.8 g, 4.7 mmol) was reacted with acetyl chloride (6.7 ml_, 94 mmol) in methanol (50 ml) to afford the desired product (1.51 g, 95%). 1H NMR (400 MHz, CD3OD) δ 7.92 (dd,
1 H), 7.78 (dd, 1 H), 7.19-6.98 (m, 4H), 3.58 (d, 2H), 3.42-3.34 (m, 1 H), 3.19 (t, 2H)1 2.31-2.04 (m, 7H). 19F NMR (400 MHz, CD3OD) δ -133 (s). ES-MS m/z 287.11 [M+1]+. CHN calculated for C17H19FN2CI .5HCI.0.17H2O: C 59.35 %; H 6.10 %; N 8.14 %, and found: C 59.41 %; H 6.22 %; N 8.15 %.
Example #20 3-Bromo-2-(2-fluoro-4-methoxyphenoxy)pyridine:
Figure imgf000075_0001
Using General Procedure A, 3-bromo-2-chloropyridine (500 mg, 2.60 mmol) was reacted with 2-fluoro-4-methoxylphenol (443 mg, 3.12 mmol) and potassium carbonate (718 mg, 5.20 mmol) in Λ/,Λ/-dimethylformamide (5 ml) to afford the desired product as a solid (685 mg, 88%). 1 H NMR (400 MHz, CDCI3) δ 3.80 (s, 3 H,) 6.66 - 6.77 (m, 2 H), 6.86 (dd, J=7.70, 4.78 Hz, 1 H), 7.14 (t, J=8.77 Hz, 1 H), 7.90 (dd, J=7.70, 1.66 Hz, 1 H)1 8.02 (dd, J=4.78, 1.66 Hz, 1 H). MS-APCI (m/z): 299 (M+H).
Teit-butyl 4-(2-(2-fluoro-4-methoxyphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate:
Figure imgf000075_0002
Using General Procedure B, 3-bromo-2-(2-fluoro-4- methoxyphenoxy)pyridine (580 mg, 1.95 mmol) was reacted with tert-butyl 4- (5,5-dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (686 mg, 2.33 mmol), cesium fluoride (887 mg, 5.84 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (285 mg, 0.389 mmol) in N1N- dimethylformamide (10 ml) to afford the desired product as a solid (628 mg, 81 %). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 2.56 - 2.64 (m, 2 H), 3.62 (t, J=5.46 Hz, 2 H), 3.79 (s, 3 H), 4.05 - 4.11 (m, 2 H)1 5.95 - 6.01 (m, 1 H), 6.66 - 6.76 (m, 2 H), 6.94 (dd, J=7.31 , 4.97 Hz, 1 H), 7.10 (t, J=8.77 Hz, 1 H), 7.54 (dd, J=7.31 , 1.85 Hz, 1 H), 7.99 (dd, J=4.97, 1.85 Hz, 1 H). rerf-butyl 4-(2-(2-fluoro-4-methoxyphenoxy)pyridin-3- yl)piperidine-1 -carboxylate:
Figure imgf000076_0001
Using General Procedure C, te/t-butyl 4-(2-(2-fluoro-4- methoxyphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (610 mg,
1.52 mmol) was reduced to the desired product as a solid (190 mg, 31 %). 1H
NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 1.56 - 1.70 (m, 2 H), 1.91 - 2.00 (m, 2
H), 2.78 - 2.92 (m, 2 H), 3.14 (tt, dd, J=12.09, 3.31 Hz, 1 H), 3.79 (s, 3 H),
4.16 - 4.40 (m, 2 H), 6.67 - 6.76 (m, 2 H), 6.93 (dd, J=7.41 , 4.87 Hz1 1 H), 7.10 (t, dd, J=8.77, 1 H), 7.50 (dd, J=7.41 , 1.17 Hz, 1 H), 7.94 (dd, J=4.97,
1.85 Hz, 1 H). MS-APCI (m/z): 403 (M+H).
2-(2-Fluoro-4-methoxyphenoxy)-3-(piperidin-4-yl)pyridine:
Figure imgf000076_0002
Using General Procedure D, te/f-butyl 4-(2-(2-fluoro-4- methoxyphenoxy)pyridin-3-yl)piperidine-1 -carboxylate (175 mg, 0.435 mmol) was reacted with acetyl chloride (341 mg, 4.35 mmol) in methanol (10 ml) then purified with ion exchange column to afford the desired product as a solid
(128 mg, 97%). 1H NMR (400 MHz, CDCI3) δ 1.62 - 1.74 (m, 2 H), 1.99 (dt,
J=13.26, 2.93 Hz, 2 H), 2.83 (dt, J=12.18, 2.53 Hz, 2 H), 3.15 (tt, J=12.09, 3.51 Hz, 1 H), 3.23 (dt, J= 11.70, 3.22 Hz, 2 H), 3.82 (s, 3 H), 6.69 - 6.80 (m, 2
H), 6.96 (dd, J=7.41 , 5.07 Hz, 1 H), 7.13 (t, J=8.58 Hz, 1 H), 7.57 (dd, J=7.41 ,
1.95 Hz, 1 H), 7.96 (dd, J=5.07, 1.95 Hz, 1 H). MS-APCI (m/z): 303 (M+H).
Example #21 3-Bromo-2-(2-chloro-6-fluorophenoxy)pyridine:
Figure imgf000077_0001
Using General Procedure A, 3-bromo-2-chloropyridine (3.0 g, 16 mmol) was reacted with 2-chloro-6-fluorophenol (2.28 g, 15.6 mmol) and potassium carbonate (3.23 g, 23.4 mol) in Λ/,Λ/-dimethylformamide (10 ml) to afford the desired product (1.5 g, 31%). 1H NMR (400 MHz, CDCI3): δ 8.01 (dd, 1 H), 7.96 (dd, 1 H), 7.30-7.10 (m, 3H), 6.93 (q, 1 H). 19F NMR (376 MHz, CDCI3): δ - 125.25. ES-MS (m/z) for CnH9BrCIFNO: 301.79, 303.81 [M+1]+.
Terf-butyl 4-(2-(2-chloro-6-fluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000077_0002
Using General Procedure B, 3-bromo-2-(2-chloro-6- fluorophenoxy)pyridine (1.45 g, 4.8 mmol) was reacted with te/f-butyl 4-(5,5- dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.7 g, 0.00575 mol), cesium fluoride (1.09 g, 7.2 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (0.527 g, 0.72 mmol) in N1N- dimethylformamide (10 ml) to afford the desired product (1.49 g, 82%). 1H NMR (400 MHz, CDCI3): δ 7.96 (dd, 1 H), 7.59 (dd, 1 H), 7.30-7.06 (m, 3H), 7.00 (q, 1 H), 6.05 (m, 1 H), 4.11 (br s, 2H), 3.65 (t, 2H), 2.66 (br s, 2H), 1.50 (s, 9H). 19F NMR (376 MHz, CDCI3): δ -125.144. ES-MS (m/z) for C21 H22CIFN2O3: 404.95 [M+1 ]+.
Terf-butyl 4-(2-(2-chloro-6-fluorophenoxy)pyridin-3-yl)piperidine-1- carboxylate:
Figure imgf000078_0001
Using General Procedure C, te/f-butyl 4-(2-(2-chloro-6- fluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (0.3 g, 0.7 mmol) was reduced to the desired product (0.29 g, 96 %). 1H NMR (400 MHz, CDCI3): δ 7.93 (dd, 1 H), 7.56 (dd, 1 H), 7.30-7.08 (m, 3H), 6.99 (q, 1 H), 4.30 (s, 2H), 3.79 (s, 3H), 3.20 (m, 1 H), 2.88 (br s, 2H), 2.04 (d, 2H), 1.67 (m, 2H), 1.49 (S, 9H). 19F NMR (376 MHz, CDCI3): δ -125.452. ES-MS (m/z) for C2IH24CIFN2O3: 406.88 [M+1]+.
2-(2-Chloro-6-fluorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride:
Figure imgf000078_0002
Using General Procedure D, te/t-butyl 4-(2-(2-chloro-6- fluorophenoxy)pyridin-3-yl)piperidine-1-carboxylate (0.29 g, 0.713 mmol) was reacted with acetyl chloride (1 ml) in methanol (25 ml) to afford the desired product (0.20 g, 82%). 1H NMR (400 MHz, CD3OD): δ 7.90 (d, 1 H), 7.78 (d, 1 H), 7.37-7.18 (m, 3H), 7.13 (q, 1 H), 3.55 (d, 2H), 3.39 (m, 1 H), 3.25 (t, 2H), 2.27 (d, 2H), 2.13 (m, 2H). 19F NMR (376 MHz, CDCI3): δ -127.996. ES-MS (m/z) for C16H16CIFN2O: 306.96 [M+1]+. CHN calculated for C16H16CIFN2O/1.4HCI: C 53.71 %; H 4.90 %; N 7.83 %, and found: C 53.87 %; H 5.14 %; N 7.69 %. Example #22
3-Bromo-2-(2,6-difluorophenoxy)pyridine:
Figure imgf000079_0001
Using General Procedure A, 3-bromo-2-chloropyridine (500 mg, 2.60 mmol) was reacted with 2,6-difluorophenol (406 mg, 3.12 mmol) and potassium carbonate (718 mg, 5.20 mmol) in Λ/,Λ/-dimethylformamide (5 ml) to afford the desired product as a solid (560 mg, 75%). 1H NMR (400 MHz, CDCI3) δ 6.91 (dd, J=7.60, 4.87 Hz, 1 H), 6.97 - 7.03 (m, 2 H), 7.15 - 7.20 (m, 1 H), 7.93 (dd, J=7.70, 1.66 Hz, 1 H), 8.00 (dd, J=4.78, 1.66 Hz, 1 H). MS- APCI (m/z+): 285 (M+H).
Tert-butyl 4-(2-(2,6-difluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1 (2H)-carboxylate:
Figure imgf000079_0002
Using General Procedure B, 3-bromo-2-(2,6-difluorophenoxy)pyridine (480 mg, 1.47 mmol) was reacted with terf-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (520 mg, 1.76 mmol), cesium fluoride (669 mg, 4.40 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (215 mg, 0.295 mmol) in N,N- dimethylformamide (10 ml) to afford the desired product as a solid (315 mg, 62%). 1H NMR (400 MHz, CDCI3) δ 1.48 (s, 9 H), 2.56 - 2.67 (m, 2 H), 3.63 (t, J=5.65 Hz, 2 H), 4.05 - 4.14 (m, 2 H), 5.98 - 6.05 (m, 1 H), 6.93 - 7.04 (m, 3 H), 7.09 - 7.19 (m, 1 H), 7.57 (dd, J=7.41 , 1.95 Hz, 1 H), 7.96 (dd, J=4.87, 1.75 Hz, 1 H).
Te/t-butyl 4-(2-(2,6-difluorophenoxy)pyridin-3-yl)piperidine-1- carboxylate:
Figure imgf000080_0001
Using General Procedure C, tert-butyl 4-(2-(2,6- difluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (350 mg, 0.901 mmol) was reduced to the desired product as gum (99 mg, 28%). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 1.57 - 1.71 (m, 2 H), 2.79 - 2.93 (m, 2 H), 3.17 (tl, J =12.28, 3.31 Hz, 1 H), 4.08 - 4.36 (m, 1 H), 6.95 - 7.04 (m, 3 H), 7.09 - 7.20 (m, 1 H), 7.53 (dd, J=7.12, 1.46 Hz, 1 H), 7.92 (dd, J=4.97, 1.85 Hz, 1 H). MS-APCI (m/z): 391 (M+H).
2-(2,6-Difluorophenoxy)-3-(piperidin-4-yl)pyridine:
Figure imgf000080_0002
Using General Procedure D, tert-butyl 4-(2-(2,6- difluorophenoxy)pyridin-3-yl)piperidine-1 -carboxylate (99 mg, 0.25 mmol) was reacted with acetyl chloride (0.180 ml, 2.54 mmol) in methanol (5 ml) then purified with ion exchange column to afford the desired product as a solid (67 mg, 91%). 1H NMR (400 MHz, CDCI3) δ 1.59 - 1.74 (m, 2 H), 1.98 - 2.01 (m, 2 H), 2.81 (dt, J=12.09, 2.34 Hz, 2 H), 3.14 (tt, ./=12.09, 3.51 Hz, 1 H), 3.20 - 3.23 (m, 2 H), 6.93 - 7.04 (m, 2 H), 7.08 - 7.18 (m, 1 H), 7.58 (dd, J=7.41 , 1.95 Hz, 1 H), 7.91 (dd, J=5.07, 1.95 Hz, 1 H). MS-APCI (m/z+): 291 (M+H).
Example #23
3-Bromo-2-(2,3-difluorophenoxy)pyridine:
Figure imgf000080_0003
Using General Procedure A, 3-bromo-2-chloro pyridine (5.91 g, 30.7 mmol) was reacted with 2,3-difluorophenol (4.2 g, 32 mmol) and potassium carbonate (8.5 g, 61 mmol) in Λ/,Λ/-dimethylformamide (20 ml) to afford the desired product (4.85 g, 56%).1H NMR (400 MHz, CDCI3): δ 8.05 (dd, 1 H), 7.96 (dd, 1 H), 7.18-6.09 (m, 4H). 19F NMR (400 MHz, CDCI3) δ -136 (s), -152
(S).
Tert-butyl 4-(2-(2,3-difluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000081_0001
Using General Procedure B, 3-bromo-2-(2,3-difluorophenoxy)pyridine
(2.78 g, 9.72 mmol) was reacted with tert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (3.4 g, 11.7 mmol), cesium fluoride (2.2 g, 14.6 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (1.07 g, 1.45 mmol) in N,N- dimethylformamide (15 ml) to afford the desired product (2.72 g, 73%).1H NMR (400 MHz, CDCI3) δ 8.02 (dd, 1 H), 7.61 (dd, 1 H), 7.26-6.84 (m, 4H), 6.02 (s, 1 H), 4.21 -4.06 (br s, 2H), 3.73-3.57 (br s, 2H), 2.71 -2.54 (br s, 2H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -137 (s), -152 (s).
Te/f-butyl 4-(2-(2,3-dif luorophenoxy)pyridin-3-yl)piperϊdine-1 - carboxylate:
Figure imgf000081_0002
Using General Procedure C, tert-butyl 4-(2-(2,3- difluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (2.69 g, 6.93 mmol) was reduced to the desired product (2.15 g, 80%). 1H NMR (400 MHz, CDCI3) δ 7.99 (dd, 1 H), 7.58 (dd, 1 H), 7.18-6.92 (m, 4H), 4.43-4.17 (br s, 2H), 3.19-3.07 (m, 1 H), 2.99-2.73 (br s, 2H), 1.98 (d, 2H)1 1.77-1.54 (m, 2H), 1.44 (s, 9H).
2-(2,3-Difluorophenoxy)-3-(piperidin-4-yl)pyridine dihydrochloride:
Figure imgf000082_0001
Using General Procedure D, te/t-butyl 4-(2-(2,3- difluorophenoxy)pyridin-3-yl)piperidine-1-carboxylate (2.0 g, 5.1 mmol) was reacted with acetyl chloride (7.3 ml_, 103 mmol) in methanol (50 ml) to afford the desired product (1.6 g, 89%). 1H NMR (400 MHz, CD3OD) δ 7.98 (dd, 1 H),
7.78 (dd, 1 H), 7.25-7.02 (m, 4H), 3.56 (d, 2H), 3.42-3.25 (m, 1 H), 3.19 (t, 2H),
2.28-1.99 (m, 4H). 19F NMR (400 MHz, CD3OD) δ -140 (s), -156 (s). ES-MS (m/z) 291.01 [M+1]+. HPLC purity: 95.62%. CHN calculated for
Ci6H16F2N2O-1.7HCI: C 54.74 %; H 5.07 %; N 7.98 %, and found: C 54.93 %;
H 5.37 %; N 7.98 %.
Example #24 & 25
3-Bromo-2-(2-fluoro-6-methoxyphenoxy)pyridine:
Figure imgf000082_0002
Using General Procedure A, 3-bromo-2-chloropyridine (3 g, 0.0156 mol) was reacted with 2-fluoro-6-methoxyphenol (2.216 g, 0.0156 mol) and potassium carbonate (3.23 g, 0.0234 mol) in Λ/,Λ/-dimethylformamide (10 ml) to afford the desired product (1.0 g, 23 %). 1H NMR (400 MHz, CDCI3): δ 8.01 (del, 1 H), 7.92 (dd, 1 H), 7.17 (m, 1 H), 6.88 (q, 1 H)1 6.86-6.78 (m, 2H), 3.80 (s, 3H). ES-MS (m/z) for Ci2H9BrFNO2: 297.82 & 299.77 [M+1]+.
7erf-butyl 4-(2-(2-fluoro-6-methoxyphenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000083_0001
Using General Procedure B, 3-bromo-2-(2-fluoro-6- methoxyphenoxy)pyridine (1.0 g, 3.4 mmol) was reacted with tert-butyl 4-(5,5- dimethyl-1 ,3,2-dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.2 g, 4.0 mmol), cesium fluoride (0.763 g, 5.03 mmol) and [1 , 1 '-Bis (diphenylphosphino)ferrocene] palladium (II) (0.368 g, 0.503 mmol) in N1N- dimethylformamide (10 ml) to afford the desired product (1.1 g, 82 %). 1H NMR (400 MHz, CDCI3): δ 7.98 (dd, 1 H), 7.56 (dd, 1 H), 7.15 (m, 1H), 6.95 (q, 1 H), 6.90-6.67 (m, 2H), 6.05 (s, 1 H), 4.10 (br s, 2H), 3.78 (s, 3H), 3.60 (br s, 2H), 2.65 (br s, 2H), 1.49 (s, 9H). ES-MS (m/z) for C22H25FN2O4: 401.11 [M+1]+.
Te/t-butyl 4-(2-(2-fluoro-6-methoxyphenoxy)pyridin-3- yl)piperidine-1 -carboxylate:
Figure imgf000083_0002
Using General Procedure C, fert-butyl 4-(2-(2-fluoro-6- methoxyphenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.1 g,
3.1 mol) was reduced to the desired product (1.1 g, 100 %). 1H NMR (400
MHz, CDCI3): δ 7.94 (dd, 1 H), 7.52 (dd, 1 H), 7.15 (m, 1 H), 6.95 (q, 1 H), 6.86- 6.78 (m, 2H), 4.27 (s, 2H), 3.79 (s, 3H), 3.20 (m, 1 H), 2.88 (br s, 2H), 2.04 (d, 2H), 1.64 (m, 2H), 1.48 (s, 9H). ES-MS (m/z) for C22H27FN2O4: 403.00 [M+1]+.
2-(2-Fluoro-6-methoxyphenoxy)-3-(piperidin-4-yl)pyridine dihydrochloride:
Figure imgf000084_0001
Using General Procedure D, tert-butyl 4-(2-(2-fluoro-6- methoxyphenoxy)pyridin-3-yl)piperidine-1 -carboxylate (1.1 g, 2.7 mmol) was reacted with acetyl chloride (4 ml_) in methanol (100 ml) to afford the desired product (0.70 g, 85%). 1H NMR (400 MHz, CD3OD): δ 7.88 (d, 1 H), 7.77 (d, 1 H), 7.23 (m, 1 H), 7.11 (q, 1 H), 6.94 (d, 1 H), 6.85 (t, 1 H), 3.75 (s, 3H), 3.55
(d, 2H), 3.37 (m, 1 H), 3.20 (t, 2H), 2.27 (d, 2H), 2.09 (m, 2H). ES-MS (m/z) for
Ci7H19FN2O2/2HCI: 302.99 [M+1]+. CHN calculated for Ci7Hi9FN2O2/2HCI: C
54.41 %; H 5.64 %; N 7.46 %, and found: C 54.80 %; H 5.60 %; N 7.66 %.
Alternatively, purification by HPLC with 0.025% trifluoroacetic acid afforded the desired product as the trifluoroacetate salt.
Example #26
3-Bromo-2-(2,5-difluorophenoxy)pyridine:
Figure imgf000084_0002
Using General Procedure A, 3-bromo-2-chloropyridine (4.7 g, 24.2 mmol) was reacted with 2,5-difluorophenol (3.47 g, 26.7 mmol) and potassium carbonate (6.7 g, 48.5 mmol) in /V,Λ/-dimethylformamide (20 ml) to afford the desired product (4.2 g, 62%).1H NMR (400 MHz, CDCI3): δ 8.05 (dd, 1 H), 7.96
(dd, 1 H), 7.18-6.09 (m, 4H). ES-MS m/z 287.74 [M+1]+. Te/t-butyl 4-(2-(2,5-difluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000085_0001
Using General Procedure B, 3-bromo-2-(2,5-difluorophenoxy)pyridine (2.0 g, 7.0 mmol) was reacted with te/t-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (2.5 g, 8.4 mmol), cesium fluoride (1.6 g, 10 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (0.77 g, 1.1 mmol) in Λ/,Λ/-dimethylformamide (15 ml) to afford the desired product (2.1 g, 78%).1H NMR (400 MHz, CDCI3) δ 8.02 (dd, 1 H), 7.61 (dd, 1 H), 7.16-6.84 (m, 4H), 6.02 (s, 1 H), 4.18-4.03 (br s, 2H), 3.69-3.54
(br s, 2H), 2.64-2.56 (br s, 2H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -
118 (s), -134 (s).
Terf-butyl 4-(2-(2,5-dif luorophenoxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000085_0002
Using General Procedure C, te/t-butyl 4-(2-(2,5- difluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (1.38 g, 3.53 mmol) was reduced to the desired product (1.34 g, 98%). 1H NMR (400 MHz, CDCI3) δ 7.99 (dd, 1 H), 7.58 (dd, 1 H), 7.18-6.83 (m, 3H), 4.24-4.11 (br s, 2H), 3.19-3.07 (m, 1 H), 2.99-2.76 (br s, 2H), 1.98 (d, 2H), 1.77-1.57 (m, 2H), 1.44 (s, 9H). 19F NMR (400 MHz, CDCI3) δ -117 (s), -134 (s). 2-(2,5-Difluorophenoxy)-3-(piperidin-4-yl)pyridine dihydrochloride:
Figure imgf000086_0001
Using General Procedure D, tert-butyl 4-(2-(2,5- difluorophenoxy)pyridin-3-yl)piperidine-1-carboxylate (1.57 g, 4.02 mmol) was reacted with acetyl chloride (5.7 ml_, 80.5 mmol) in methanol (50 ml) to afford the desired product (1.21 g, 85%). 1H NMR (400 MHz, CD3OD) δ 7.98 (dd,
1 H)1 7.78 (dd, 1 H), 7.29-6.98 (m, 3H), 3.56 (d, 2H), 3.41 -3.25 (m, 1 H), 3.19 (t,
2H), 2.26-1.99 (m, 4H). 19F NMR (400 MHz, CD3OD) δ -119 (s), -136 (s). ES-
MS (m/z) 290.95 [M+1]+. CHN calculated for C16H16F2N2O.2HCI: C 52.91 %; H 4.99 %; N 7.71 %, and found: C 52.60 %; H 5.07 %; N 7.50 %.
Example #27 3-Bromo-2-(2,4-difluorophenoxy)pyridine:
Figure imgf000086_0002
Using General Procedure A, 3-bromo-2-chloro pyridine (3.0 g, 16 mmol) was reacted with 2,4-difluorophenol (2.0 g, 16 mmol) and potassium carbonate (3.23 g, 23.4 mol) in Λ/,Λ/-dimethylformamide (10 ml) to afford the desired product (2.2 g, 48 %). 1H NMR (400 MHz, CDCI3): δ 8.05 (dd, 1 H), 7.96 (dd, 1 H), 7.26 (m, 1 H), 7.02-6.88 (m, 3H). ES-MS (m/z) for C11H6BrF2NO: 285.78 & 287.80 [M+1]+. Te/t-butyl 4-(2-(2,4-difluorophenoxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000087_0001
Using General Procedure B, 3-bromo-2-(2,4-difluorophenoxy)pyridine (1.91 g, 6.68 mmol) was reacted with tert-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1(2H)-carboxylate (2.36 g, 8.01 mmol), cesium fluoride (1.52 g, 10.0 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (0.73 g, 1.0 mmol) in N, N- dimethylformamide (15 ml) to afford the desired product (2.08 g, 80 %). 1H NMR (400 MHz, CDCI3): δ 8.01 (dd, 1 H), 7.59 (dd, 1 H), 7.18 (m, 1 H), 7.04- 6.86 (m, 3H), 6.00 (s, 1 H), 4.10 (br s, 2H), 3.64 (s, 3H), 2.61 (br s, 2H), 1.49 (s, 9H). ES-MS (m/z) for Chemical Formula: C2IH24F2N2O3: 388.94 [M+1]+.
Te/f-butyl 4-(2-(2,4-dif luorophenoxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000087_0002
Using General Procedure C, te/t-butyl 4-(2-(2,4- difluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (2.0 g, 5.2 mmol) was reduced to the desired product (1.8 g, 90 %). 1H NMR (400 MHz, CDCI3): δ 7.95 (dd, 1 H), 7.54 (dd, 1 H), 7.19 (m, 1 H), 7.02-6.88 (m, 3H), 4.29 (s, 2H), 3.15 (m, 1 H), 2.87 (br s, 2H), 1.97 (d, 2H), 1.65 (m, 2H), 1.49 (s, 9H). ES-MS (m/z) for C2i H24F2N2O3: 391.02 [M+1]+. 2-(2,4-Difluorophenoxy)-3-(piperidin-4-yl)pyridine hydrochloride:
Figure imgf000088_0001
Using General Procedure D, tert-butyl 4-(2-(2,4- difluorophenoxy)pyridin-3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (1.8 g, 4.6 mmol) was reacted with acetyl chloride (4 ml_) in methanol (100 ml) to afford the desired product (1.5 g, quant). 1H NMR (400 MHz, CD3OD): δ 8.02-7.96
(m, 2H)1 7.38 (m, 1 H), 7.26 (q, 1 H)1 7.17 (m, 1 H), 7.06 (m, 1 H), 3.56 (d, 2H),
3.40 (m, 1 H), 3.22 (t, 2H), 2.22 (d, 2H), 2.12 (m, 2H). 19F NMR (376 MHz,
CD3OD): δ-115.22 and -126.32. ES-MS (m/z) for Ci6H17 F2N2O/2HCI: 290.95
[M+1]+. CHN calculated for Ci6H17 F2N2O/2HCI/2.5H2O: C 47.07 %; H 5.68 %; N 6.86 %, and found: C 46.98 %; H 5.25 %; N 6.76 %.
Example #28 3-Bromo-2-(naphthalen-1-yloxy)pyridine:
Figure imgf000088_0002
Using General Procedure A, 3-bromo-2-chloro pyridine (2.00 g, 10.4 mmol) was reacted with 1-naphthol (2.1 g, 15 mmol) and potassium carbonate
(3.16 g, 22.9 mmol) in Λ/,Λ/-dimethylformamide (10.4 ml) to afford the desired product as a solid (1.41 g, 45%). 1H NMR (400 MHz, CDCI3) δ 6.88 (dd,
J=7.41 , 5.07 Hz, 1 H), 7.29 (dd, J=7.60, 0.97 Hz, 1 H), 7.42 - 7.53 (m, 3 H),
7.73 - 7.77 (m, 1 H), 7.86 - 7.91 (m, 1 H), 7.92 - 7.95 (m, 1 H), 7.96 - 8.00 (m, 2 H). MS-APCI (m/z): 300 [M+1]+.
Terf-butyl 4-(2-(naphthalen-1 -yloxy)pyridin-3-yl)-5,6- dihydropyridine-1(2H)-carboxylate:
Figure imgf000089_0001
Using General Procedure B, 3-bromo-2-(naphthalen-1-yloxy)pyridine (320 mg, 1.06 mmol) was reacted with te/t-butyl 4-(5,5-dimethyl-1 ,3,2- dioxaborinan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (395 mg, 1.28 mmol), cesium fluoride (485 mg, 3.20 mmol) and [1 , 1'-Bis (diphenylphosphino)ferrocene] palladium (II) (125 mg, 0.17 mmol) in N1N- dimethylformamide (2.66 ml) to afford the desired product as a gum (221 mg, 65%). 1H NMR (400 MHz, CDCI3) δ 1.45 (s, 9 H), 2.56 - 2.66 (m, 2 H), 3.52 (t, J=5.56 Hz, 2 H), 3.96 - 4.03 (m, 2 H), 6.02 - 6.08 (m, 1 H), 7.07 - 7.17 (m, 2 H), 7.41 - 7.54 (m, 3 H), 7.686 - 7.77 (m, 2 H), 7.89 (dd, J=7.8, 5.8 Hz, 2 H), 8.09 (d, J=4.24 Hz 1 H). MS-APCI (m/z): 403 [M+1]+.
Te/t-butyl 4-(2-(naphthalen-1 -yloxy)pyridin-3-yl)piperidine-1 - carboxylate:
Figure imgf000089_0002
Using General Procedure C, terf-butyl 4-(2-(naphthalen-1-yloxy)pyridin-
3-yl)-5,6-dihydropyridine-1(2H)-carboxylate (277 mg, 0.688 mmol) was reduced to the desired product (300 mg, quant). 1H NMR (400 MHz, CDCI3) δ 1.42 - 1.50 (m, 2 H), 1.47 (s, 9 H), 1.68 - 1.78 (m, 2 H)1 2.01 - 2.09 (m, 2 H), 2.75 - 2.91 (m, 2 H), 3.27 (tt, J=12.32, 3.97 Hz, 1 H), 6.97 (dd, J=7.41 , 4.87 Hz, 1 H), 7.14 (dd, J=7.54, 1.12 Hz, 1 H), 7.40 - 7.51 (m, 3 H), 7.59 (dd, Λ=7.51 , 1.85 Hz, 1 H), 7.70 (d, J=8.19 Hz, 1 H), 7.86 - 7.91 (m, 2 H), 7.93 (dd, J=4.87, 1.95 Hz, 1 H). MS-APCI (m/z): 405 [M+1]+.
2-(Naphthalen-1-yloxy)-3-(piperϊdin-4-yl)pyridine hydro chloride:
Figure imgf000090_0001
Using General Procedure D, ferf-butyl 4-(2-(naphthalen-1-yloxy)pyridin- 3-yl)piperidine-1-carboxylate (300 mg, 0.742 mmol) was reacted with acetyl chloride (1.06 ml, 14.8 mmol) in methanol (5 ml) to afford the desired product as a solid (230 mg, 91%). 1H NMR (400 MHz, CD3OD) δ 1.15-2.40 (m, 4 H), 3.20 (m, 2 H), 3.40-3.60 (m, 3 H), 7.15 (m, 2 H), 7.4-7.6 (m, 3H), 7.85-8.00 (m 5H). MS-APCI (m/z): 305 [M+1]+.
Example #29
Terf-butyl 4-(2-(2,3-dichlorophenoxy)pyridin-3-yl)piperazine-1 - carboxylate:
Figure imgf000090_0002
Using General Procedure E, 3-bromo-2-(2,3-dichlorophenoxy)pyridine (200mg, 0.627 mmol), tert-butyl piperazine-1 -carboxylate (140 mg, 0.752 mmol) and sodium tert-butoxide (90.4 mg, 0.940 mmol) in toluene (2 ml) were combined, followed by the addition of tris(dibenzylideneacetone)dipalladium (28.7 mg, 0.0313 mmol) and Dave-Phos (37.0 mg, 0.0940 mmol) to afford the desired product as a solid (174 mg, 64%). 1H NMR (400 MHz, CDCI3) δ 3.13 - 3.20 (m, 4 H), 3.56 - 3.63 (m, 4 H), 6.97 (dd, J=7.80, 4.87 Hz, 1 H), 7.12 (dd, J=8.19, 1.56 Hz, 1 H), 7.20 - 7.26 (m, 2 H), 7.33 (dd, J=7.99, 1.56 Hz, 1 H), 7.71 (dd, J=4.87, 1.56 Hz, 1 H). MS-APCI (m/z+): 424 [M+1]+. 1-(2-(2,3-Dichlorophenoxy)pyridin-3-yl)piperazine:
Figure imgf000090_0003
Using General Procedure D, tert-butyl 4-(2-(2,3- dichlorophenoxy)pyridin-3-yl)piperazine-1-carboxylate (170 mg, 0.401 mmol) was reacted with acetyl chloride (0.285 ml, 4.01 mmol) in methanol (5 ml) then purified with ion exchange column to afford the desired product as a solid (89 mg, 68%). 1H NMR (400 MHz, CDCI3) δ 2.99 - 3.08 (m, 4 H), 3.14 - 3.21 (m, 4 H), 6.95 (dd, J=7.80, 4.87 Hz, 1 H), 7.10 (dd, J=8.19, 1.56 Hz, 2 H), 7.19 - 7.26 (m, 2 H), 7.30 (dd, J=7.99, 1.56 Hz, 1 H), 7.68 (dd, J=4.78, 1.66 Hz, 1 H). MS-APCI (m/z): 324 [M+1]+.
Example #30 Tert-butyl 4-(2-(3-fluoro-2-methoxyphenoxy)pyridin-3- yl)piperazine-1 -carboxylate:
Figure imgf000091_0001
Using General Procedure A, reaction of 3-bromo-2-chloro pyridine (600 mg, 3.12 mmol), 3-fluoro-2-methoxyphenol (593 mg, 3.74 mmol) and potassium carbonate (862 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (3 ml) afforded 3-bromo-2-(3-fluoro-2-methoxyphenoxy)pyridine as a solid (655 mg, 71 %). Then using General Procedure E, 3-bromo-2-(3-fluoro-2- methoxyphenoxy)pyridine (350 mg, 1.17 mmol), te/f-butyl piperazine-1- carboxylate (262 mg, 1.41 mmol) and sodium tert-butoxide (169 mg, 1.76 mmol) in toluene (2 ml) were combined, followed by the addition of tris(dibenzylideneacetone)dipalladium (54 mg, 0.059 mmol) and Dave-Phos (69.3 mg, 0.176 mmol) to afford the desired product as a solid (284 mg, 67%). MS-APCI (m/z): 404 [M+1]+, 304 [M+1-100]+.
1-(2-(3-Fluoro-2-methoxyphenoxy)pyridin-3-yl)piperazine:
Figure imgf000092_0001
Using General Procedure D, tert-butyl 4-(2-(3-fluoro-2- methoxyphenoxy)pyridin-3-yl)piperazine-1-carboxylate (295 mg, 0.731 mmol) was reacted with acetyl chloride (114 mg, 1.46 mmol) in methanol (8 ml) then treated with 2M NH3 in MeOH after the purification to afford the desired product as oil (99 mg, 50%). 1H NMR (400 MHz, CD3OD) δ 2.97 (d, J=5.1 Hz, 2 H), 3.14 (d, J=4.9 Hz, 2 H), 3.16 (br. 2, 1 H), 3.33 (s, 3 H), 3.71 (d, JM .4 Hz, 2 H), 6.88 (dt, J=7.8, 1.7 Hz, 1 H), 7.03 (m, 3 H), 7.38 (dd, J=7.8, 1.6 Hz, 1 H), 7.61 (dd, J=4.9, 1.8 Hz, 1 H) Example #31
1-(2-(2-Ethoxy-6-fluorophenoxy)pyridin-3-yl)piperazine:
Figure imgf000092_0002
Using General Procedure A, reaction of 3-bromo-2-chloro pyridine (600 mg, 3.12 mmol), 2-ethoxy-6-fluorophenol (445 mg, 3.12 mmol) and potassium carbonate (862 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (2.60 ml) afforded 3-bromo-2-(2-ethoxy-6-fluorophenoxy)pyridine as a solid (800 mg, 86%). Then using General Procedure E, 3-bromo-2-(2-ethoxy-6- fluorophenoxy)pyridine (352 mg, 1.09 mmol) was reacted with tert-butyl piperazine-1-carboxylate (243 mg, 1.31 mmol), sodium terf-butoxide (157 mg, 1.63 mmol), tris(dibenzylideneacetone)dipalladium (49.4 mg, 0.054 mmol) and Dave-Phos (64.1 mg, 0.163 mmol) in toluene (2 ml) to result in tert-butyl 4-(2- (2-ethoxy-6-fluorophenoxy)pyridin-3-yl)piperazine-1-carboxylate as a solid (298 mg, 67%). Then using General Procedure D, terf-butyl 4-(2-(2-ethoxy-6- fluorophenoxy)pyridin-3-yl)piperazine-1-carboxylate (253 mg, 0.605 mmol) was reacted with acetyl chloride (2.1 ml, 28.8 mmol) in methanol (8 ml) then treated with 2M NH3 in MeOH after the purification to afford the desired product as gum (33 mg, 17%). 1H NMR (400 MHz, CD3OD) δ 1.14 (t, J=6.92 Hz, 3 H), 2.98 - 3.03 (m, 4 H), 3.16 - 3.23 (m, 4 H), 3.97 (q, J=7.02 Hz, 2 H), 6.81 (dd, J=8.38, 1.36 Hz, 1 H), 6.86 (dt, J=8.48, 1.32 Hz, 1 H), 7.00 (dd, J=7.80, 4.87 Hz, 1 H), 7.15 (td, J=8.48, 6.04 Hz, 1 H), 7.37 (dd, J=7.80, 1.56 Hz, 1 H).
Example #32 rerf-butyl 4-(2-(4-fluoro-2-methoxyphenoxy)pyridin-3- yl)piperazine-1-carboxylate:
Figure imgf000093_0001
Using General Procedure A, reaction of 3-bromo-2-chloropyridine (500 mg, 2.60 mmol), 2-fluoro-6-methoxyphenol (443 mg, 3.12 mmol) and potassium carbonate (718 mg, 5.20 mmol) in Λ/,Λ/-dimethylformamide (5 ml) afforded 3-bromo-2-(4-fluoro-2-methoxyphenoxy)pyridine as a solid (1.49 g, 60%). Then using General Procedure E, 3-bromo-2-(4-fluoro-2- methoxyphenoxy)pyridine (313 mg, 1.05 mmol), te/t-butyl piperazine-1- carboxylate (235 mg, 1.26 mmol) and sodium tert-butoxide (151 mg, 1.58 mmol) in toluene (2 ml) was reacted together followed by the addition of tris(dibenzylideneacetone)dipalladium (48.5 mg, 0.053 mmol) and Dave-Phos (62.2 mg, 0.158 mmol) to afford the desired product as a solid (290 mg, 69%). MS-APCI (m/z): 404 [M+1]\ 304 [M+H-100]+.
1-(2-(4-Fluoro-2-methoxyphenoxy)pyridin-3-yl)piperazine:
Figure imgf000093_0002
Using General Procedure D, tert-butyl 4-(2-(4-fluoro-2- methoxyphenoxy)pyridin-3-yl)piperazine-1 -carboxylate (262 mg, 0.649 mmol) was reacted with acetyl chloride (101 mg, 1.29 mmol) in methanol (8 ml) then treated with 2M NH3 in MeOH after the purification to afford the desired product a solid (116 mg, 59%). 1H NMR (400 MHz, CD3OD) δ 2.93 - 2.99 (m, 4 H), 3.11 - 3.18 (m, 4 H), 3.67 (s, 3 H), 6.87 (dd, J=8.97, 3.12 Hz, 1 H), 6.90 - 6.96 (m, 1 H), 7.01 (dd, J=7.80, 4.87 Hz, 1 H), 7.05 (dd, J=8.97, 5.07 Hz, 1 H), 7.37 (dd, J=7.70, 1.66 Hz, 1 H), 7.58 (dd, J=4.87, 1.56 Hz, 1 H). MS-APCI (m/z): 304 [M+1]+. Example #33
Tert-butyl 4-(2-(2-ethyl-4-fluorophenoxy)pyridin-3-yl)piperazine-1- carboxylate:
Figure imgf000094_0001
Using General Procedure A, reaction of 3-bromo-2-chloropyridine (600 mg, 3.12 mmol), 2-ethyl-4-fluorophenol (437 mg, 3.12 mmol) and potassium carbonate (862 mg, 6.24 mmol) in Λ/,Λ/-dimethylformamide (3 ml) afforded 1-
(2-bromophenoxy)-2-ethyl-4-fluorobenzene as an oil (778 g, 84%). Then using General Procedure E, 3-bromo-2-(2-ethyl-4-fluorophenoxy)pyridine (311 mg, 1.05 mmol), tert-butyl piperazine-1-carboxylate (235 mg, 1.26 mmol) and sodium te/t-butoxide (151 mg, 1.58 mmol) in toluene (2 ml) were combined, followed by the addition of tris(dibenzylideneacetone)dipalladium (48.5 mg,
0.053 mmol) and Dave-Phos (62.2 mg, 0.158 mmol), and the reaction afforded the desired product as a solid (306 mg, 73%). 1H NMR (400 MHz,
CD3OD) δ 1.13 (t, J=4.97 Hz, 3 H), 1.46 (s, 9 H), 2.51 (q, J=7.47 Hz, 2 H), 3.06 - 3.15 (m, 4 H), 3.50 - 3.62 (m, 4 H), 6.87 - 6.93 (m, 2 H), 6.97 - 7.05 (m,
2 H), 7.37 (dd, J=7.80, 1.56 Hz, 1 H), 7.60 (dd, J=4.97, 1.66 Hz, 1 H). MS-
APCI (m/z): 402 [M+I]+, 302 [M+1-100]+. 1-(2-(2-Ethyl-4-fluorophenoxy)pyridin-3-yl)piperazine:
Figure imgf000095_0001
Using General Procedure D, te/t-butyl 4-(2-(2-ethyl-4- fluorophenoxy)pyridin-3-yl)piperazine-1 -carboxylate (260 mg, 0.648 mmol) was reacted with acetyl chloride (2.2 ml, 31 mmol) in methanol (13 ml) then treated with 2M NH3 in MeOH after the purification to afford the desired product a gum (151 mg, 77%). 1H NMR (400 MHz, CD3OD) δ 1.14 (t, J=4.97 Hz, 3 H), 2.52 (q, J=7.54 Hz, 2 H), 2.94 - 3.00 (m, 4 H), 3.11 - 3.18 (m, 4 H), 6.89 - 6.94 (m, 2 H), 6.99 - 7.06 (m, 2 H), 7.39 (dd, J=7.80, 1.56 Hz, 1 H), 7.60 (dd, J=4.97, 1.66 Hz, 1 H). MS-APCI (m/z): 302 [M+1]+.
Example #34
Te/t-butyl 4-(2-(2-chloro-6-fluorophenoxy)pyridin-3-yl)piperazine- 1-carboxylate:
Figure imgf000095_0002
Using General Procedure E, 3-bromo-2-(2-chloro-6- fluorophenoxy)pyridine (310mg, 1.04 mmol), fe/t-butyl piperazine-1- carboxylate (232 mg, 1.25 mmol) and sodium fert-butoxide (175 mg, 1.56 mmol) in toluene (1 ml) were combined, followed by the addition of tris(dibenzylideneacetone)dipalladium (47.6 mg, 0.0520 mmol) and Dave- Phos (61.4 mg, 0.156 mmol), and the reaction afforded the desired product as a solid (130 mg, 31 %). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 3.13 - 3.21 (m, 4 H), 3.58 - 3.65 (m, 4 H), 6.94 (dd, J=7.60, 4.87 Hz, 1 H), 7.07 - 7.18 (m, 2 H), 7.21 (dd, J=7.70, 1.66 Hz, 1 H), 7.26 (dt, J=7.80, 1.75 Hz, 1 H), 7.67 (dd, J=4.78, 1.66 Hz, 1 H). MS-APCI (m/z): 408 [M+1]+, 308 [M+H-100]+. 1-(2-(2-Chloro-6-fluorophenoxy)pyridin-3-yl)piperazine:
Figure imgf000096_0001
Using General Procedure D, te/t-butyl 4-(2-(2-chloro-6- fluorophenoxy)pyridin-3-yl)piperazine-1-carboxylate (130 mg, 0.319 mmol) was reacted with acetyl chloride (0.227 ml, 3.19 mmol) in methanol (5 ml) then purified with ion exchange column to afford the desired product as a solid
(59 mg, 60%). 1H NMR (400 MHz, CDCI3) δ 3.06 - 3.08 (m, 4 H), 3.17 - 3.24
(m, 4 H), 6.94 (dd, J=7.80, 4.68 Hz, 1 H), 7.06 - 7.17 (m, 1 H), 7.22 (dd,
J=7.80, 1.56 Hz, 1 H), 7.26 (dt, J=8.19, 1.95 Hz, 1 H), 7.64 (dd, J=4.68, 1.56 Hz, 1 H). MS-APCI (m/z): 308 [M+1]+.
Example #35
7eιt-butyl 4-(2-(2-fluoro-6-methoxyphenoxy)pyridin-3- yl)piperazine-1 -carboxylate:
Figure imgf000096_0002
Using General Procedure E, 3-bromo-2-(2-fluoro-6- methoxyphenoxy)pyridine (287mg, 0.963 mmol), te/t-butyl piperazine-1 - carboxylate (269 mg, 1.44 mmol) and sodium te/t-butoxide (162 mg, 144 mmol) in toluene (1 ml) were combined, followed by the addition of tris(dibenzylideneacetone)dipalladium (44.1 mg, 0.048 mmol) and Dave-Phos (56.8 mg, 0.144 mmol), and the reaction afforded the desired product as a solid (410 mg, quant). 1H NMR (400 MHz, CDCI3) δ 1.47 (s, 9 H), 3.12 - 3.20 (m, 4 H), 3.58 - 3.64 (m, 4 H), 3.76 (s, 3 H), 6.76 - 6.84 (m, 2 H), 6.90 (dd, J=7.80, 4.87 Hz, 1 H), 7.13 (td, J=8.43, 6.14 Hz, 1 H), 7.17 (dd, J=7.70, 1.66 Hz, 1 H), 7.68 (dd, J=4.87, 1.75 Hz, 1 H). MS-APCI (m/z): 404 [M+1]+, 304 [M+1 -100]+.
1-(2-(2-Fluoro-6-methoxyphenoxy)pyridin-3-yl)piperazine:
Figure imgf000097_0001
Using General Procedure D, te/t-butyl 4-(2-(2-fluoro-6- methoxyphenoxy)pyridin-3-yl)piperazine-1-carboxylate (410 mg, 1.02 mmol) was reacted with acetyl chloride (0.723 ml, 10.2 mmol) in methanol (10 ml) then purified with ion exchange column to afford the desired product as a solid (252 mg, 82%). 1H NMR (400 MHz, CDCI3) δ 3.07 - 3.13 (m, 4 H), 3.19 - 3.27 (m, 4 H), 3.76 (s, 3 H), 6.75 - 6.84 (m, 2 H), 6.90 (dd, J=7.60, 4.87 Hz, 1 H), 7.12 (td, J=8.43, 6.14 Hz, 1 H), 7.19 (dd, J=7.70, 1.66 Hz, 1 H), 7.66 (dd, J=4.87, 1.56 Hz, 1 H). MS-APCI (m/z): 304 [M+1]+.
Table 1 shows the structure of the compounds and relevant biological data that were measured in each case either on the compound as a free base or on the pharmaceutically acceptable salt of the compound disclosed in the Table. In the table, (S) refers to SPA Binding Assay; (F) refers to Filtration Binding Assay. Each assay is disclosed in greater detail hereinbelow.
TABLE 1
Figure imgf000098_0001
Example #5 94.2 (S) 23.2 17600 25.0
3-(Piperidin-4-yl)-2-(2- (F) 61.8 (trif luorom ethoxy)phenox) pyridine hydrochloride
Figure imgf000099_0001
Example # 6 98.4 (S) 80.4
3-(Piperidin-4-yl)-2-(2- (F) 89.6 (trifluoromethyl)phenoxy) pyridine hydrochloride
Figure imgf000099_0002
Example #7 93.5 (S) 54.0
2-(2-Methoxyphenoxy)-3- (F) 61.2
(piperidin-4-yl)pyridine dihydrochloride
Example #8 97.5 (S) 5.1
2-(2-Ethoxyphenoxy)-3-
(piperidin-4-yl)pyridine hydrochloride
Example #9 89.0 (S) 81.2 163 91
2-(2-Fluorophenoxy)-3- (F) 96.0
(piperidin-4-yl)pyridine hydrochloride
Example #10 98.4 (S) 81.2 372 39.9
2-(2-Chlorophenoxy)-3- (F) 96.0 (piperidin-4-yl)pyridine hydrochloride salt
Figure imgf000099_0003
Example #11 (F) 67.5
2-(2-Chloro-3- (trifluoromethyl)phenoxy)- 3-(piperidin-4-yl)pyridine hydrochloride salt
Figure imgf000100_0001
Example #12 95.1 (F) 79.1 2120 19.9
2-(2-Chloro-6- methoxyphenoxy)-3- (piperidin-4-yl)pyridine hydrochloride salt
Figure imgf000100_0002
Example #13 102 (F) 91.0 415 45.7
2-(2,3-Dichlorophenoxy)- 3-(piperidin-4-yl)pyridine hydrochloride salt
Figure imgf000100_0003
Example #14 92.7 (F) 90.4
2-(2,4-Dichlorophenoxy)- 3-(piperidin-4-yl)pyridine hydrochloride salt
Figure imgf000100_0004
Example #15 87.3 (F) 92.1
2-(2-Fluoro-4- methylphenoxy)-3- (piperidin-4-yl)pyridine hydrochloride salt
Example #16 93.1 (F) 48.2
2-(2-Ethoxy-6- fluorophenoxy)-3- (piperidin-4-yl)pyridine trifluoroacetic acid salt
Figure imgf000100_0005
Example #17 93.6
2-(4-Fluoro-2- methoxyphenoxy)-3-
(piperidin-4-yl)pyridine
Example #18 97.3 (F) 70.9
2-(2-Ethyl-4- fluorophenoxy)-3- (piperidin-4-yl)pyridine trifluoroacetic acid salt
Figure imgf000101_0001
Example #19 99.0 (S) 84.6 129 59.6
2-(2-Fluoro-6- (F) 97.4 methylphenoxy)-3- (piperidin-4-yl)pyridine dihydrochloride
Figure imgf000101_0002
Example #20 52.9 (S) 54.4 829 61.2
2-(2-Fluoro-4- (F) 94.1 methoxyphenoxy)-3-
(piperidin-4-yl)pyridine
Figure imgf000101_0003
Example #21 99.9 (S) 89.6 88.7 25.8
2-(2-Chloro-6- (F) 98.2 fluorophenoxy)-3- (piperidin-4-yl)pyridine hydrochloride
Example #22 96.2 (S) 80.9
2-(2,6-Difluorophenoxy)- 3-(piperidin-4-yl)pyridine
Figure imgf000101_0004
Example #23 76.9 (S) 68.2
2-(2,3-Difluorophenoxy)- 3-(piperidin-4-yl)pyridine dihydrochloride
Figure imgf000102_0001
Example #24 88.1
2-(2-Fluoro-6- methoxyphenoxy)-3- (piperidin-4-yl)pyridine trifluoroacetic acid salt
Example #25 97.1 (S) 90.4 176 68.8
2-(2-Fluoro-6- (F) 95.3 methoxyphenoxy)-3- (piperidin-4-yl)pyridine dihydrochloride
Example #26 97.5 (S) 64.9
2-(2,5-Difluorophenoxy)- 3-(piperidin-4-yl)pyridine dihydrochloride
Example #27 96.9 (S) 74.7 1010 62.3
2-(2,4-Difluorophenoxy)- (F) 94.4 3-(piperidin-4-yl)pyridine hydrochloride
Figure imgf000102_0002
Example #28 97.9 (F) 90.6
2-(Naphthalen-1 -yloxy)-3- (piperidin-4-yl)pyridine hydro chloride
Figure imgf000102_0003
Figure imgf000103_0001
Example #35 60.8 (S) 87.8 93.9 25.0
1-(2-(2-Fluoro-6- (F) 98.4 methoxyphenoxy)pyridin-
3-yl)piperazine
Figure imgf000104_0001
When introducing elements of the present invention or the exemplary embodiment(s) thereof, the articles "a," "an," "the" and "said" are intended to mean that there are one or more of the elements. The terms "comprising," "including" and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements. Although this invention has been described with respect to specific embodiments, the details of these embodiments are not to be construed as limitations to the invention, the scope of which is defined by the appended claims. Biological Protocols
NET Binding Assay
Cell pellets (7 to 15 g) were thawed on ice and resuspended in 3 ml of membrane prep buffer (20 mM HEPES, pH 7.4 @24°C, 1 complete protease inhibitor tablet ((Roche catalog #11697498001) /50 ml buffer) per ml of packed cell volume, using a vortex mixer to disperse the cell pellet. The cells were then allowed to swell on ice for 10 min. The suspension was homogenized for 4 x 10 second bursts at 25,000 rpm on a PT3100 polytron. The homogenate was spun at 3,000 rpm for 20 min in a SS34 rotor cooled to 40C. The supernatant was decanted into new 50 ml centrifuge tubes and the pellets were rehomogenized in 10-15 ml of membrane prep buffer and spun as described above. The supernatants were then pooled and spun at 17,500 rpm in the SS34 rotor (35,000 x g) for 30 min at 40C. The supernatants were discarded and the cell pellets were resuspended by vigorous pipetting in 1 ml membrane prep buffer per 1 ml of original packed cell volume. Protein concentrations were determined with the BCA Protein Assay Kit (Pierce, #23227) utilizing the microplate assay procedure. The membranes were stored in 1 ml aliquots at -8O0C until use.
SPA beads (Wheat germ agglutinin coated polyvinyltoluene;
Amersham #RPNQ0001 ) (500 mg) were added to 10 ml of 40C assay buffer (30 mM HEPES, 180 mM NaCI, pH 7.4) and placed on ice. Membrane/bead slurry was prepared by addition of 6 ml of 50 mg/ml bead slurry with 2 ml of 3 mg/ml stock membrane preparation in the presence of 2 ml of cold assay buffer (0.6 mg membrane/ml in slurry), and was incubated for 2 hours at 40C on a top-to-tail shaker. The slurry was spun at 1200 rpm for 5 min at 40C, and the pellet was washed 2X in cold assay buffer and resuspended to the original slurry volume.
Compound concentration-response 11 -point curves were tested in Vz log scale, starting at 10 μM final concentration in the reaction mixture. Columns 1 and 24 in a Costar 3706 384-well white, clear bottom plate were left for vehicle controls and the non-specific binding component. Samples concentrations were added in single wells in duplicate plating. Wells contained:
0.5 μl 100% DMSO spots (total binding, compound, or standard) 30 μl [3H]Nisoxetine in assay buffer 20 μl bead/membrane slurry in assay buffer
After all additions were made, the plate was sealed and shaken for 1 hour at room temperature.
The plate was then dark adapted for 6 to 10 hours prior to counting in the Wallac Trilux Microbeta scintillation counter. 5HT1 A SPA Binding Assay
HEK-293 EBNA cell membrane (Catalog # RBHS1AM) stably expressing the 5HT1a receptor was purchased from Perkin Elmer. The cells were grown under standard cell culture techniques, harvested, and received frozen suspended in 5OmM TRIS-HCI, 0.5mM EDTA, 1OmM MgCI2, 10% sucrose, pH to 7.4. The company provides both receptor concentration (Bmax), ligand affinity (Kd), and also calculates the protein concentration. Subsequently, these membrane preparations are stored at -8O0C until needed. Lyophilized wheat germ agglutinin SPA beads (WGA-SPA) are reconstituted in 10 ml_ of assay buffer (5OmM TRIS MgCI2, pH 7.4). To initiate coupling of the beads to the membrane, the bead suspension is added to the cell membranes to maintain a bead concentration of 500 μg/well and a protein concentration of 10μg/ well (384-well plate format). This bead/membrane mixture is rocked gently for 30 minutes at 40C. After 30 minutes the suspension is then centrifuged at 1000 rpm at 250C. The supernatant is carefully discarded and the remaining pellet is diluted in assay buffer and aliquoted into assay plates to initiate incubation. Binding studies were carried out in 50μl_ assay volume using Costar 3705 (white, clear bottom) 384 well plates to which 0.5-1.0 ml_ of compound in 100% DMSO was prespotted, 20μL of [3H]-8-OH-DPAT (final concentration of 5nM and 30μL of bead/membrane slurry are added sequentially using Titertek Multidrops. Plates are sealed with clear adhesive and allowed to incubate overnight at 250C. The nonspecific binding was defined by using 1μM Lisuride. The plates were counted in a Packard Trilux Microbeta Scintillation Counter using a normalized protocol for 3H-SPA counting The data was analyzed using an Excel macro which used the statistical average for the high control (Total Binding) values and the 1 μM lisuride values as the low control (Nonspecific binding) values. Z factors were determined for each plate using these values. Active compounds were further titrated to determine Ki values. Ki values were determined by analysis using a least squares nonlinear regression curve fit assuming a one site competition ligand binding model. 5HT1A Filtration Binding Assay
As an alternate binding affinity assay, HeLa cells transfected with the human 5HT1A receptor were incubated in a solution containing 50OuI of 50 mM Tris-HCI, 10 mM MgSO4, 0.5 mM EDTA, 0.1% Ascorbic acid at pH 7.4 (solution A) and the diluted test agent, plus 1.5 nM [3H]8-OH-DPAT in a 96- well format. In cases where non-specific binding was determined 1OuM of [3H]8-OH-DPAT was used. The solution was left to incubate at 4C for 1 hour. The assay is terminated by filtration of solution over GF/C filters that have been pre-soaked in 0.3% polyethylenimine with solution A. Next the filters are washed three times with 500 ul of an ice cold solution containing 50 mM Tris- HCI pH 7.4. Filters are then dried, placed into a vial containing scintillant fluid and radiation levels are determined using a scintillation counter. Affinity of the compounds for the human 5HT1A receptor is calculated using GraphPad and Ligand data analysis programs. GTPΎS-5HT1 A Binding Assay
CHO cells transfected with human 5 HT1 A receptor were maintained in Ultra CHO (Cambrex, 12-724Q) media supplemented with 10% dialyzed FBS (Gibco 26400 036), 1% Geneticin (Gibco 10131-027), and 0.5% Pen/Strep (Gibco 15140 122). Cells are grown in 15OmM cell culture dishes and harvested in ice-cold PBS by scraping the plates when the cells are approx. 80% confluent. Cells are collected in PBS and centrifuged at 40C at low speed (2500 rpm) until pellet forms. The pellet is then homogenized in tissue buffer with a polytron, and centrifuged at 40C at high speed (20,000 rpm) for 15 min. Repeat this washing at least 4 times. After final wash, the protein can be measured and the homogenate aliquoted appropriately. . Working stocks were prepared as followed:
• Compounds were diluted in assay buffer to a final concentration of 10 uM
• 100μM Guanosine 5'-diphsophate, sodium salt (GDP, Sigma, Catalog No. G7127) in assay buffer • 1 nM 35S-GTPγS (NEN#NEG-030H, 1250 Ci/mmol) in assay buffer
• On the day of the assay, the cell homogenate was thawed and washed two additional times in assay buffer (20 mM Hepes, pH 7.4, 120 mM NaCI, 10 mM MgCI2J mM EDTA). Membranes were suspended at a final concentration of 0.2mg/ml_ in assay buffer. To each well of a 96-well plate, the following was added: 33μl compound 20μl GDP 77μl assay buffer 20μl 35S-GTPγS
50μl membranes (10μg)
All of the above components are added to the plate 96-well for a final volume of 200μl. The plates are incubated at 3O0C for 60 minutes. To terminate the assay, the plates are aspirated and filtered into (non PEI coated) plates with ice cold 5OmM Tris (pH-7) and washed 3X. The dried plates are counted on TopCount.

Claims

CLAIMS We claim:
1. A compound or Formula I:
Figure imgf000109_0001
or a pharmaceutically acceptable salt thereof, wherein:
X 1 is O or S(O)P; is either a single bond or a double bond, wherein, when irmzz jS a single bond, Y is N or CR3, and when is a double bond, Y is C; each Ri is independently selected from the group consisting of hydrogen, halogen, alkoxy, -CN, and alkyl, wherein each Ri alkyl is optionally independently substituted by one to three halogens; each R2 is independently selected from the group consisting of halogen, -NO2, -CN, -N(R4)2, R4, and -OR4; or two R2 substituents on adjacent carbons taken together with the adjacent carbons form a carbocyclic or heterocyclic ring optionally substituted with halogen, hydroxy, -CN, alkyl, or alkoxy; wherein each R4 is independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, heterocycloalkyl and heteroaryl; or two R4 groups on the same nitrogen atom may be taken together with the nitrogen atom to form a 5 to 8 membered heterocyclic ring which optionally has 1 to 3 additional ring heteroatoms selected from the group consisting of N, O, and S; and each R4 alkyl, cycloalkyl, aryl, heterocycloalkyl and heteroaryl is optionally independently substituted by one to three substituents independently selected from the group consisting of halogen, hydroxy, -CN, -(Ci-Cβ)alkyl, -(Ci-C6)alkoxy, -CF3, -OCF3, -N[(CH2),R4]2, -NO2, -(CH2),N[(CH2),R4](C=O)[(CH2)tR4], -S(CrCβ)alkyl, -(S=O)(Ci -C6)alkyl, -S(=O)2(Ci-Cβ)alkyl, -(C=O)O(Ci -C6)alkyl, -O(C=O)(d-C6)alkyl,
-(C=O)(Ci -C6)alkyl, cycloalkyl, aryl, heterocycloalkyl and heteroaryl; each R3 is independently selected from the group consisting of hydrogen, halogen, -(CH2)tOH, -(CH2),CF3, -(CH2)tC≡N, -NO2, -(CH2),N[(CH2)tR4]2) -(CH2)t-alkyl, -(CH2),O[(CH2),R4], -(CH2)t(C3-Ci2)cycloalkyl, -(CH2)t-aryl,
Figure imgf000110_0001
and -(CH2)theteroaryl; or two R3 groups each attached to the same carbon atom of the ring containing Y, taken together with the carbon atom, form a three to seven membered carbocyclic ring n is an integer selected from 0, 1 and 2; m is an integer selected from 0, 1 , 2, 3 and 4; n1 is an integer selected from 1 , 2, and 3; p is an integer selected from 0, 1 and 2; and each t is an integer independently selected from 0, 1 , 2, 3, 4, and 5.
2. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein n is 1.
3. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein n is 2.
4. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein R4 is hydrogen.
5. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein n1 is 1.
6. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein n1 is 2.
7. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein X1 is O.
8. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein Xi is S.
9. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein p is 0.
10. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein p is 1.
11. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein p is 2.
12. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein each R1 is hydrogen.
13. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein each R2 is hydrogen.
14. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein each R1 is independently halogen or C1 -Ce alkyl.
15. A compound of Claim 14 or pharmaceutically acceptable salt thereof wherein n is 1 and R1 is fluorine, chlorine, alkyl or trifluoroalkyl.
16. A compound of Claim 14 or pharmaceutically acceptable salt thereof wherein n is 2 and each R1 is fluorine.
17. A compound of Claim 14 or pharmaceutically acceptable salt thereof wherein n is 2 and each R1 is alkyl.
18. A compound of Claim 14 or pharmaceutically acceptable salt thereof wherein n is 2 and one R1 is fluorine and the other R1 is chlorine.
19. A compound of Claim 14 or pharmaceutically acceptable salt thereof wherein n is 2 and one R1 is alkyl and the other R1 is chlorine or fluorine.
20. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein Y is N and each R2 is independently halogen or alkyl.
21 . A compound of claim 20 or pharmaceutically acceptable salt thereof wherein each R2 is independently fluorine, chlorine, or methyl.
22. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein Y is CH and each R2 is independently halogen or alkyl.
23.A compound of Claim 22 or pharmaceutically acceptable salt thereof wherein each R2 is independently fluorine, chlorine, or methyl.
24. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein m is 1.
25. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein m is 2.
26. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein each R2 is independently halogen, alkyl optionally substituted with one to three halogens, heteroaryl, or alkoxy optionally substituted with one to three halogens.
27. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 1 and R2 is fluorine, chlorine, alkyl optionally substituted with one to three halogens, or alkoxy optionally substituted with one to three halogens.
28. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 and each R2 is fluorine.
29. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 and each R2 is chlorine.
30. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 and each R2 is alkyl.
31. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 wherein one R2 is fluorine and the other R2 is chlorine.
32. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 wherein one R2 is alkyl and the other R2 is chlorine or fluorine.
33. A compound of Claim 26 or pharmaceutically acceptable salt thereof wherein m is 2 and one R2 is alkoxy and the other R2 is chlorine or fluorine.
34.A compound of claim 1 or pharmaceutically acceptable salt thereof wherein each R3 is hydrogen.
35. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein z=z=z js a single bond and Y is N.
36. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein is a single bond and Y is CR3.
37. A compound of claim 1 or pharmaceutically acceptable salt thereof wherein is a double bond and Y is C.
38. A compound selected from the group consisting of the compounds disclosed in Table 1 herein, or a pharmaceutically acceptable salt thereof.
39. A method for the treatment of a condition selected from the group consisting of acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, mild cognitive impairment, obesity, schizophrenia, anxiety (including generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive compulsive disorder), mood disorders (including depression, mania, bipolar disorders), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hyperactivity disorder, autism, Asperger's disease, and conduct disorder, comprising administering a compound of claim 1 or a pharmaceutically acceptable salt thereof to the mammal.
40. The method of claim 39, wherein the condition is cognitive disorders, including cognitive disorders associated with schizophrenia, bipolar disorders, Alzheimer's Disease, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, and post-traumatic pain), attention deficit/hyperactivity disorder, autism, and Asperger's disease.
41. A method for treating neurological and psychiatric disorders associated with norepinephrine reuptake inhibition, 5HT1a agonist activity, or both, comprising administering to a patient in need thereof an amount of a compound of claim 1 or a pharmaceutically acceptable salt thereof effective in treating such disorders.
42. The method of claim 41 , further comprising administering an atypical antipsychotic
43. A pharmaceutical composition comprising a compound of claim 1 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
44.A composition for treating a condition selected from the group consisting of of acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, including cognitive disorders associated with schizophrenia and bipolar disorders, idiopathic and drug- induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, mild cognitive impairment, obesity, schizophrenia, anxiety (including generalized anxiety disorder, social anxiety disorder, panic disorder, post-traumatic stress disorder and obsessive compulsive disorder), mood disorders (including depression, mania, bipolar disorders), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hyperactivity disorder, autism, Asperger's disease, and conduct disorder in a mammal, wherein the composition contains an amount of the compound of claim 1 or a pharmaceutically acceptable salt thereof that is effective in the treatment of such conditions.
45. The composition of claim 44, further comprising an atypical antipsychotic.
46. The composition of claim 44, further comprising a cholinesterase inhibitor.
PCT/IB2008/003537 2007-12-21 2008-12-15 Phenoxy-pyridyl derivatives WO2009081259A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US1640107P 2007-12-21 2007-12-21
US61/016,401 2007-12-21

Publications (1)

Publication Number Publication Date
WO2009081259A1 true WO2009081259A1 (en) 2009-07-02

Family

ID=40428329

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2008/003537 WO2009081259A1 (en) 2007-12-21 2008-12-15 Phenoxy-pyridyl derivatives

Country Status (1)

Country Link
WO (1) WO2009081259A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8053438B2 (en) 2008-11-14 2011-11-08 Amgen Inc. Pyrazine compounds as phosphodiesterase 10 inhibitors
US8247433B2 (en) 2008-11-14 2012-08-21 Theravance, Inc. Process for preparing 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8318718B2 (en) 2008-11-14 2012-11-27 Amgen Inc. Pyridine and pyrimidine derivatives as phosphodiesterase 10 inhibitors
JP2013522363A (en) * 2010-03-22 2013-06-13 セラヴァンス, インコーポレーテッド 1- (2-phenoxymethylheteroaryl) piperidine compound and 1- (2-phenoxymethylheteroaryl) piperazine compound
US8637500B2 (en) 2008-12-17 2014-01-28 Amgen Inc. Aminopyridine and carboxypyridine compounds as phosphodiesterase 10 inhibitors
US8778949B2 (en) 2010-01-11 2014-07-15 Theravance Biopharma R&D Ip, Llc 1-(2-phenoxymethylphenyl)piperazine compounds
GB2594931A (en) * 2020-05-06 2021-11-17 Syngenta Crop Protection Ag Improvements in or relating to organic compounds
WO2023165874A1 (en) * 2022-03-01 2023-09-07 Syngenta Crop Protection Ag Pyrimidinyl-oxy-quinoline based herbicidal compounds
WO2023217156A1 (en) * 2022-05-09 2023-11-16 浙江同源康医药股份有限公司 Polycyclic compound and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0982030A2 (en) * 1998-08-17 2000-03-01 Pfizer Products Inc. 2,7-substituted octahydro-pyrrolo 1,2-a]pyrazine derivatives as 5ht 1a ligands
WO2003029232A1 (en) * 2001-10-04 2003-04-10 H. Lundbeck A/S Phenyl-piperazine derivatives as serotonin reuptake inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0982030A2 (en) * 1998-08-17 2000-03-01 Pfizer Products Inc. 2,7-substituted octahydro-pyrrolo 1,2-a]pyrazine derivatives as 5ht 1a ligands
WO2003029232A1 (en) * 2001-10-04 2003-04-10 H. Lundbeck A/S Phenyl-piperazine derivatives as serotonin reuptake inhibitors

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10576073B2 (en) 2008-11-14 2020-03-03 Theravance Biopharma R&D Ip, Llc Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US10722504B2 (en) 2008-11-14 2020-07-28 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8247418B2 (en) 2008-11-14 2012-08-21 Amgen Inc. Pyrazine compounds as phosphodiesterase 10 inhibitors
US9073859B2 (en) 2008-11-14 2015-07-07 Theravance Biopharma R&D Ip, Llc Crystalline form of a 4-[2-(2-fluorophenoxymethyl) phenyl]piperidine compound
US8304433B2 (en) 2008-11-14 2012-11-06 Theravance, Inc. Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US8318718B2 (en) 2008-11-14 2012-11-27 Amgen Inc. Pyridine and pyrimidine derivatives as phosphodiesterase 10 inhibitors
US8329700B2 (en) 2008-11-14 2012-12-11 Amgen Inc. Pyrazine compounds as phosphodiesterase 10 inhibitors
US9162982B2 (en) 2008-11-14 2015-10-20 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxy methyl)phenyl]piperidine compounds
US11723900B2 (en) 2008-11-14 2023-08-15 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8592596B2 (en) 2008-11-14 2013-11-26 Theravance, Inc. Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US8604058B2 (en) 2008-11-14 2013-12-10 Theravance, Inc. 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US11596624B2 (en) 2008-11-14 2023-03-07 Theravance Biopharma R&D Ip, Llc Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US8759532B2 (en) 2008-11-14 2014-06-24 Amgen Inc. Pyridine and pyrimidine derivatives as phosphodiesterase 10 inhibitors
US9187423B2 (en) 2008-11-14 2015-11-17 Theravance Biopharma R&D Ip, Llc Process for preparing 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8802857B2 (en) 2008-11-14 2014-08-12 Theravance Biopharma R&D Ip, Llc Process for preparing 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US10946007B2 (en) 2008-11-14 2021-03-16 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8304432B2 (en) 2008-11-14 2012-11-06 Theravance, Inc. 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US10946006B2 (en) 2008-11-14 2021-03-16 Theravance Biopharma R&D Ip, Llc Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US8247433B2 (en) 2008-11-14 2012-08-21 Theravance, Inc. Process for preparing 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US9675599B2 (en) 2008-11-14 2017-06-13 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US10034870B2 (en) 2008-11-14 2018-07-31 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US10206913B2 (en) 2008-11-14 2019-02-19 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US10226454B2 (en) 2008-11-14 2019-03-12 Theravance Biopharma R&D Ip, Llc Crystalline form of a 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compound
US10441579B2 (en) 2008-11-14 2019-10-15 Theravance Biopharma R&D Ip, Llc 4-[2-(2-fluorophenoxymethyl)phenyl]piperidine compounds
US8053438B2 (en) 2008-11-14 2011-11-08 Amgen Inc. Pyrazine compounds as phosphodiesterase 10 inhibitors
US8637500B2 (en) 2008-12-17 2014-01-28 Amgen Inc. Aminopyridine and carboxypyridine compounds as phosphodiesterase 10 inhibitors
US8778949B2 (en) 2010-01-11 2014-07-15 Theravance Biopharma R&D Ip, Llc 1-(2-phenoxymethylphenyl)piperazine compounds
US9012460B2 (en) 2010-01-11 2015-04-21 Theravance Biopharma R&D Ip, Llc 1-(2-phenoxymethylphenyl)piperazine compounds
JP2013522363A (en) * 2010-03-22 2013-06-13 セラヴァンス, インコーポレーテッド 1- (2-phenoxymethylheteroaryl) piperidine compound and 1- (2-phenoxymethylheteroaryl) piperazine compound
US8530663B2 (en) 2010-03-22 2013-09-10 Theravance, Inc. 1-(2-phenoxymethylheteroaryl)piperidine and piperazine compounds
GB2594931A (en) * 2020-05-06 2021-11-17 Syngenta Crop Protection Ag Improvements in or relating to organic compounds
WO2023165874A1 (en) * 2022-03-01 2023-09-07 Syngenta Crop Protection Ag Pyrimidinyl-oxy-quinoline based herbicidal compounds
WO2023217156A1 (en) * 2022-05-09 2023-11-16 浙江同源康医药股份有限公司 Polycyclic compound and use thereof

Similar Documents

Publication Publication Date Title
WO2009081259A1 (en) Phenoxy-pyridyl derivatives
US20120053165A1 (en) Novel Phenyl Imidazoles and Phenyl Triazoles As Gamma-Secretase Modulators
US7456164B2 (en) 3- or 4-monosubtituted phenol and thiophenol derivatives useful as H3 ligands
JP4173191B2 (en) 3- or 4-monosubstituted phenol and thiophenol derivatives useful as H3 ligands
WO2009004430A1 (en) N-benzyl oxazolidinones and related heterocycleic compounds as potentiators of glutamate receptors
JP5369173B2 (en) Quinoline or isoquinoline substituted P2X7 antagonists
US20080312271A1 (en) Azabenzimidazolyl compounds
DK2443092T3 (en) Bicyclic and tricyclic compounds as CAT-II inhibitors
EP3813819A1 (en) Inhibitors of cyclin-dependent kinases
US8691804B2 (en) Azetidines and cyclobutanes as histamine H3 receptor antagonists
WO2008012623A1 (en) Benzimidazolyl compounds as potentiators of mglur2 subtype of glutamate receptor
US20120202787A1 (en) Novel Heteroaryl Imidazoles And Heteroaryl Triazoles As Gamma-Secretase Modulators
WO2007135527A2 (en) Benzimidazolyl compounds
US20110224231A1 (en) Novel Lactams as Beta Secretase Inhibitors
AU2008282032B2 (en) Novel heterocyclic compounds as mGlu5 antagonists
EP2300484B1 (en) Novel class of spiro piperidines for the treatment of neurodegenerative diseases
JP2010523540A (en) Sulfonamide and pharmaceutical composition thereof
US20130150376A1 (en) Novel Sultam Compounds

Legal Events

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

Ref document number: 08864672

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08864672

Country of ref document: EP

Kind code of ref document: A1