WO2004043958A1 - Fused heterocyclic compounds - Google Patents

Fused heterocyclic compounds Download PDF

Info

Publication number
WO2004043958A1
WO2004043958A1 PCT/US2003/035543 US0335543W WO2004043958A1 WO 2004043958 A1 WO2004043958 A1 WO 2004043958A1 US 0335543 W US0335543 W US 0335543W WO 2004043958 A1 WO2004043958 A1 WO 2004043958A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
group
mmol
aryl
Prior art date
Application number
PCT/US2003/035543
Other languages
French (fr)
Inventor
Xi Chen
Xiaoqi Chen
Pingchen Fan
Juan Jaen
Leping Li
Jeffrey T. Mihalic
Original Assignee
Amgen 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 Amgen Inc. filed Critical Amgen Inc.
Priority to CA002505372A priority Critical patent/CA2505372A1/en
Priority to EP03779483A priority patent/EP1562943A1/en
Priority to NZ540328A priority patent/NZ540328A/en
Priority to BR0316070-0A priority patent/BR0316070A/en
Priority to AU2003285160A priority patent/AU2003285160B2/en
Priority to MXPA05004739A priority patent/MXPA05004739A/en
Priority to EA200500770A priority patent/EA009421B1/en
Priority to JP2004551872A priority patent/JP4547266B2/en
Publication of WO2004043958A1 publication Critical patent/WO2004043958A1/en
Priority to IS7833A priority patent/IS7833A/en
Priority to NO20052655A priority patent/NO20052655L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to compounds, compositions and methods useful in the treatment or prevention of conditions and disorders associated with eating behavior, energy homeostasis and anxiety.
  • G-protein coupled receptors play important roles in diverse signaling processes, including those involved with sensory and hormonal signal transduction. Eating disorders, which represent a major health concern throughout the world, have been linked to GPCR regulation. On the one hand, disorders such as obesity, the excess deposition of fat in the subcutaneous tissues, manifest themselves by an increase in body weight. Individuals who are obese often have, or are susceptible to, medical abnormalities including respiratory difficulties, cardiovascular disease, diabetes and hypertension. On the other hand, disorders like cachexia, the general lack of nutrition and wasting associated with chronic disease and/or emotional disturbance, are associated with a decrease in body weight.
  • MCH neuropeptide melanin-concentrating hormone
  • GPCR G-protein-coupled-receptor
  • SLC-1 which is sequentially homologous to the somatostatin receptors, is frequently referred to as melanin- concentrating hormone receptor (MCH receptor, MCHR or MCHR1) (see Chambers et al, Nature 400:261-65 (1999); Saito et al, Nature 400:265-69 (1999); and Saito et al, TEM 11(8):299-303 (2000)).
  • MCH receptor melanin- concentrating hormone receptor
  • MCH is involved in regulation of eating behavior.
  • intracerebral administration of MCH in rats resulted in stimulation of feeding.
  • mRNA corresponding to the MCH precursor is up-regulated in the hypothalamus of genetically obese mice and of fasted animals.
  • mice deficient in MCH are leaner and have a decreased food intake relative to normal mice.
  • MCH is believed to exert its activity by binding to MCHR, resulting in the mobilization of mtracelfular calcium and a concomitant reduction in cAMP levels (see Chambers et al, Nature 400:261-65 (1999); Shimada et al. Nature 396:670-74 (1998)).
  • MCH also activates inwardly rectifying potassium channels, and MCHR has been found to interact with both Gcri protein and G ⁇ q protein (Saito et al, TEM ll(8):299-303 (2000)). Moreover, analysis of the tissue localization of MCHR indicates that it is expressed in those regions of the brain involved in olfactory learning and reinforcement. The cumulative data suggest that modulators of MCHR should have an effect on neuronal regulation of food intake (see Saito et al, Nature 400:265-69 (1999)).
  • MCH has been shown to modulate behaviors other than feeding, such as anxiety (Gonzales et al. (1996) Peptides 17:171-177; Monzon et al. (1999) Physiol Behav. 67:813-817).
  • the identification of MCHR modulators is useful for the study of physiological processes mediated by MCHR and the development of therapeutic agents for the treatment or prevention of conditions and disorders associated with weight regulation, learning, anxiety and other neuronal-related functions.
  • the present invention provides fused heterocyclic compounds and compositions, and methods of use thereof to treat or prevent conditions and disorders mediated by MCHR.
  • the present invention provides compounds, compositions and methods for treating or preventing conditions and disorders associated with eating behavior, energy homeostasis and anxiety.
  • the compounds provided herein have the formula (I):
  • Q represents a single or fused aryl or heteroaryl ring;
  • Q is -N(R)- or -N(R)-(C 1 -C 3 )alkylene-;
  • L 1 is a bond, ( -C ⁇ alkylene, ( -C ⁇ alkylenoxy and (Ci- C 4 )alkylenamino;
  • L 2 is a bond, ( -C- alkylene, (C 2 -C 4 )alkenylene, (C -C 4 )alkynylene,
  • R" is hydrogen or (C ⁇ -C 8 )alkyl; each R 1 is independently selected from the group consisting of halogen, (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, fluoro(C r C 4 )alkyl, -OR 5 , -SR 5 , fluoro(C r C 4 )alkoxy, aryl, aryl(C r C 4 )alkyl, -NO 2 , -NR 5 R 6 , -C(O)R 5 , -CO 2 R 5 , - C(O)NR 5 R 6 , -N(R 6 )C(O)R 5 , -N(R 6 )CO 2 R 5 , -N(R 7 )C(O)NR 5 R 6 , -S(O) m NR 5 R 6 , -S(O) m NR 5
  • R 4 is selected from the group consisting of hydrogen, -OR 11 , -C(O)R ⁇ , - CO 2 R ⁇ , -C(O)NR ⁇ R 12 , -CN, (C r C 4 )alkyl and aryl; X and Y are independently selected from the group consisting of (C ⁇ -
  • C 8 )alkyl (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, -CO 2 R 13 and -C(O)NR 13 R 14 ; optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms independently selected from the group consisting of N, O and S;
  • R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 and R 17 are independently selected from the group consisting of hydrogen, (C 1 -C 8 )alkyl, (C 2 -
  • C 8 alkenyl, (C 2 -C 8 )alkynyl, cyclo(C 3 -C 6 )alkyl, fluoro(C ⁇ -C )alkyl, hetero(C ⁇ -C 4 )alkyl, cyclohetero(C 3 -C 6 )alkyl, aryl and aryl(C ⁇ -C 4 )alkyl;
  • R 18 is a 5- or 6-membered ring containing from 0 to 4 heteroatoms selected from the group consisting of N, O and S (e.g. tetrazole); optionally, when two R groups selected from the group consisting of R 5 ,
  • R 6 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 and R 17 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from the group consisting of N, O and S; the subscript m is 1 or 2; and the subscript n is 0, 1 or 2.
  • In certain embodiments represents a benzene, naphthalene, pyrrole, pyrazole, imidazole, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, pyridine, pyrimidine, benzotbiazole, purine, benzimidazole, indole, isoquinoline, quinoxaline or quinoline ring.
  • In certain embodiments represents a benzene ring.
  • Q is -N(R)-.
  • R" is hydrogen and R 3 is hydrogen.
  • L 2 is a bond, ( -G alkylene, (C 2 -C )alkenylene, (C 2 -C 4 )alkynylene, (C ⁇ -C 4 )al ylenoxy (e.g. -OCH 2 CH 2 -) or (CrC ⁇ alkylenamino (e.g.
  • R" is hydrogen or (CrC ⁇ alkyl; each R 1 is independently selected from the group consisting of halogen, (C ⁇ -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, fluoro(C r C 4 )alkyl, -OR 5 , -SR 5 , fluoro(C r C 4 )alkoxy, aryl, aryl(C 1 -C 4 )alkyl, -NO 2 , -NR 5 R 6 , -C(O)R 5 , -CO 2 R 5 , - C(O)NR 5 R 6 , -N(R 6 )C(O)R 5 , -N(R 6 )CO 2 R 5 , -N(R 7 )C(O)NR 5 R 6 , -S(O) m NR 5 R 6 , - S(O) m NR 5 R 6
  • R 4 is selected from the group consisting of hydrogen, -OR 11 , -C(O)R ⁇ , - CO 2 R n , -C(O)NR ⁇ R 12 , -CN, (C r C 4 )alkyl and aryl;
  • X and Y are independently selected from the group consisting of ( - C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, -CO 2 R 13 and -C(O)NR 13 R 14 ; optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms independently selected from the group consisting of N, O and S;
  • R l ⁇ is a 5- or 6-membered ring containing from 0 to 4 heteroatoms selected from the group consisting of N, O and S (e.g. tetrazole); optionally, when two R groups selected from the group consisting of R 5 , R 6 , R 8 , R 9 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 and R 17 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from the group consisting of N, O and S; the subscript m is 1 or 2; and the subscript n is 0, 1 or 2.
  • compositions provided herein comprise a pharmaceutically acceptable carrier or excipient in combination with a compound of formula I or II.
  • Methods for treating or preventing a condition or disorder selected from the group consisting of obesity, an eating disorder, an anxiety disorder and a mood disorder are provided herein.
  • the methods comprise administering to a subject in need thereof a therapeutically effective amount of one of the foregoing compounds or pharmaceutical compositions.
  • FIG. 1 provides the structures of exemplary compounds of the invention.
  • MCHR refers to the melanin-concentrating hormone receptor protein 1 (MCHR1), unless otherwise stated.
  • treat refers to a method of alleviating or abrogating a disease and/or its attendant symptoms.
  • prevent refers to a method of decreasing the probability or eliminating the possibility that a disease will be contracted.
  • MCHR-mediated condition or disorder refers to a condition or disorder characterized by inappropriate, e.g., less than or greater than normal, MCHR activity.
  • An MCHR-mediated condition or disorder may be completely or partially mediated by inappropriate MCHR activity.
  • an MCHR-mediated condition or disorder is one in which modulation of MCHR results in some effect on the underlying condition or disease (e.g., an MCHR antagonist results in some improvement in patient well-being in at least some patients).
  • MCHR-mediated conditions and disorders include obesity, eating disorders and other behavioral disorders, such as anxiety disorders and mood disorders.
  • terapéuticaally effective amount refers to that amount of the compound being administered sufficient to prevent development of or alleviate to some extent one or more of the symptoms of the condition or disorder being treated.
  • the term "obesity” refers to the excessive accumulation of body fat. Obesity may have genetic, environmental (e.g., expending less energy than is consumed) and regulatory determinants. Cardiovascular disorders, lipid disorders and metabolic disorders, such as hypertension, hyperlidemia, coronary artery disease and diabetes, are commonly associated with obesity.
  • the terms "eating disorder”, “feeding disorder”, and the like refer to an emotional and/or behavioral disturbance associated with an excessive decrease in body weight and/or inappropriate efforts to avoid weight gain, e.g., fasting, self-induced vomiting, laxative or diuretic abuse. Depression is commonly associated with eating disorders. Exemplary eating disorders include anorexia nervosa and bulimia. As used herein, the term “anxiety disorder” refers to an emotional and/or behavioral disturbance characterized by persistent and pervasive worry or restlessness, tension or irritability about, e.g., health, work, money or family, for no clear reason. An anxiety disorder may be accompanied by tachycardia or dyspnea. Exemplary anxiety disorders include anxiety, generalized anxiety disorder, panic attacks, panic disorder and obsessive-compulsive disorder (OCD).
  • OCD obsessive-compulsive disorder
  • the term "mood disorder” refers to an emotional and/or behavioral disturbance characterized by persistent and pervasive bouts of euphoria and/or depression.
  • exemplary mood disorders include depression and bipolar disorders. Anxiety is frequently associated with mood disorders, such as depression.
  • modulate refers to the ability of a compound to increase or decrease the function, or activity, of MCHR. Modulation, as described herein, includes the antagonism or agonism of MCHR, either directly or indirectly.
  • Antagonist are compounds that, e.g., partially or totally block stimulation, decrease, prevent, delay activation, inactivate, inhibit, desensitize, or down-regulate signal transduction.
  • Agonists are compounds that, e.g., stimulate, increase, activate, open, facilitate, enhance activation, sensitize or up-regulate signal transduction.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which is fully saturated, having the number of carbon atoms designated (i.e. C ⁇ -C 8 means one to eight carbons).
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n- pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • (C 1 -C 8 )alkyl refers to an alkyl group having from one to eight carbon atoms and includes, e.g., (C 1 -C 4 )alkyl.
  • alkenyl by itself or as part of another substituent, means a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be mono- or polvunsaturated, having the number of carbon atoms designated (i.e. C 2 -C 8 means two to eight carbons) and one or more double bonds.
  • alkenyl groups include vinyl, allyl, 2-propenyl, crotyl, 2-isopentenyl, 2- (butadienyl), 2,4-pentadienyl, 3-(l,4-pentadienyl) and higher homologs and isomers thereof.
  • alkynyl by itself or as part of another substituent, means a straight or branched chain hydrocarbon radical, or combination thereof, which may be mono- or polvunsaturated, having the number of carbon atoms designated (i.e. C 2 -C 8 means two to eight carbons) and one or more triple bonds.
  • alkynyl groups include ethynyl, 1- and 3-propynyl, 3-butynyl and higher homologs and isomers thereof.
  • alkylene by itself or as part of another substituent means a divalent radical derived from alkyl, as exemplified by -CH 2 CH 2 CH CH 2 -.
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having seven or fewer carbon atoms.
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, Si and S, wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group.
  • the heteroatom Si may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule.
  • heteroalkylene by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified by -CH 2 - CH 2 -S-CH 2 CH 2 - and -CH 2 -S-CH 2 -CH 2 -NH-CH 2 -.
  • heteroalkylene groups heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied.
  • cycloalkyl and heterocycloalkyl represent, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl”, respectively. Accordingly, a cycloalkyl group has the number of carbon atoms designated (i.e., C 3 -C 8 means three to eight carbons) and a heterocycloalkyl group consists of the number of atoms designated (i.e., C 2 -C 8 means two to eight carbons) and from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • heterocycloalkyl a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.
  • cycloalkyl include cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include 1 -(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3- piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.
  • halo and halogen by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
  • haloalkyl are meant to include alkyl substituted with halogen atoms, which can be the same or different, in a number ranging from one to (2m' + 1), where m' is the total number of carbon atoms in the alkyl group.
  • halo(C 1 -C 4 )alkyl is mean to include trifluoromethyl, 2,2,2- trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • haloalkyl includes monohaloalkyl (alkyl substituted with one halogen atom) and polyhaloalkyl (alkyl substituted with halogen atoms in a number ranging from two to (2m' + 1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group).
  • fluoro(C 1 -C 4 ) alkyl includes fluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 1,1-difluoroethyl, and the like.
  • perhaloalkyl means, unless otherwise stated, alkyl substituted with (2m' + 1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group.
  • perhalo(C 1 -C 4 )alkyl is meant to include trifluoromethyl, pentachloroethyl, l,l,l-trifluoro-2-bromo-2-chloroethyl, and the like.
  • aryl means, unless otherwise stated, a polvunsaturated, typically aromatic, hydrocarbon substituent which can be a single ring or multiple rings (up to three rings) which are fused together or linked covalently.
  • aryl groups include phenyl, 1-naphthyl, 2-naphthyl and 4-biphenyl.
  • heteroaryl refers to aryl groups (or rings) that contain from zero to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • heteroaryl groups include 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4- pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5- indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl and 6- quinoly
  • aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
  • arylalkyl is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l-naphthyloxy)propyl, and the like).
  • alkyl group e.g., benzyl, phenethyl, pyridylmethyl, and the like
  • an oxygen atom e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l-na
  • R', R" and R'" each independently refer to hydrogen, unsubstituted (C 1 -C 8 )alkyl and heteroalkyl, fluoro(Ci-C 4 )alkyl, unsubstituted aryl, aryl substituted with one to three halogens, unsubstituted alkyl, alkoxy or thioalkoxy groups, or aryl(C ⁇ -C 4 )alkyl groups.
  • R' and R" When R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6- or 7- membered ring.
  • -NR'R is meant to include 1-pyrrolidinyl and 4- morpholinyl.
  • an alkyl or heteroalkyl group will have from zero to three substituents, with those groups having two or fewer substituents being preferred in the present invention. More preferably, an alkyl or heteroalkyl radical will be unsubstituted or monosubstituted. Most preferably, an alkyl or heteroalkyl radical will be unsubstituted. From the above discussion of substituents, one of skill in the art will understand that the term "alkyl" is meant to include groups such as trihaloalkyl (e.g., - CF 3 and -CH 2 CF 3 ).
  • an aryl or heteroaryl group will have from zero to three substituents, with those groups having two or fewer substituents being preferred in the present invention. More preferably, an aryl or heteroaryl group will be unsubstituted or monosubstituted. Most preferably, an aryl or heteroaryl group will be unsubstituted.
  • Preferred substituents for aryl and heteroaryl groups are selected from: halogen, -OR', -OC(O)R', -NR'R", -SR', -R', -CN, -NO 2 , -CO 2 R', -CONR'R", -C(O)R', -OC(O)NR'R", -NR"C(O)R', -S(O)R', -SO 2 R', -SO 2 NR'R", -NR"SO 2 R', - N 3 , -CH(Ph) 2 , perfluoro(C ⁇ .
  • substituent -CO 2 H includes bioisosteric replacements therefor, such as:
  • Two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -T-C(O)-(CH 2 ) q -U-, wherein T and U are independently -NH-, -O-, -CH 2 - or a single bond, and q is an integer of from 0 to 2.
  • two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) r -B-, wherein A and B are independently -CH 2 -, -O-, -NH-, -S-, -S(O)-, -SO 2 -, -SO 2 NR'- or a single bond, and r is an integer of from 1 to 3.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -(CH 2 ) s -X-(CH ) t -, where s and t are independently integers of from 0 to 3, and X is -O-, -NR'-, -S-, -S(O)-, -SO2-, or -SO 2 NR'-.
  • the substituent R' in -NR'- and -SO NR'- is selected from the group consisting of hydrogen or unsubstituted (Q- C 6 )alkyl.
  • heteroatom is meant to include oxygen (O), nitrogen (N), sulfur (S) and silicon (Si).
  • salts are meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pha ⁇ naceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbomc, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbomc, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydri
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge, S.M., et al. (1977) J. Pharm. Sci. 66:1-19).
  • Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • the present invention provides compounds which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention.
  • prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have improved solubility in pharmacological compositions over the parent drug.
  • prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • An example, without limitation, of a prodrug would be a compound of the present invention which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity.
  • Additional examples include peptidyl derivatives of a compound of the invention.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, enantiomers, diastereomers, geometric isomers and individual isomers are all intended to be encompassed within the scope of the present invention.
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
  • MCHR (GenBank Accession No. U71092) is expressed in brain, at moderate levels in the eye and skeletal muscle, and in low levels in tongue and the pituitary gland. Evidence suggests that MCHR is involved in, ter alia, olfactory learning, regulation of feeding behavior and energy metabolism, regulation of the hypothalmic-pituitary-adrenocortical axis following stress, arousal and the sensation of anxiety (Saito et al, TEM 11(8):299-303 (2000)).
  • the compounds of the present invention inhibit MCHR activity, and thus, are useful in, for example, the treatment or prevention of disorders associated with these processes.
  • the present invention provides compounds represented by the formula (I):
  • aryl or heteroaryl ring represents a single or fused aryl or heteroaryl ring.
  • aryl or heteroaryl ring can represent benzene, naphthalene, pyrrole, pyrazole, imidazole, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, pyridine, pyrimidine, benzothiazole, purine, benzimidazole, indole, isoquinoline, quinoxaline or quinoline ring.
  • the symbol Q represents -N(R)- or -N(R)-(C 1 -C 3 )alkylene-. In certain embodiments the symbol Q represents -N(R)-.
  • the symbol R represents .
  • L 1 represents a divalent linkage selected from a bond, (C ⁇ - C 4 )alkylene, (C ⁇ -C )alkylenoxy and ( -C ⁇ alkylenamino.
  • Exemplary L 1 groups are a single bond, methylene, ethylene, n-propylene and n-butylene.
  • the symbol L 2 represents a divelent linkage selected from a bond, (Ci-C )alkylene, (C 2 -C 4 )alkenylene, (C 2 -C )alkynylene, (C ⁇ -C 4 )alkylenoxy and (C ⁇ -C 4 )alkylenamino.
  • Exemplary L 2 groups are a a single bond, methylene, ethylene, n-propylene and n-butylene.
  • X and Y represent independently (C ⁇ -C 8 )alkyl, (C 2 - C 8 )alkenyl, (C 2 -C 8 )alkynyl, -CO 2 R 13 or -C(O)NR 13 R 14 .
  • X and Y may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from N, O and S.
  • Exemplary -C(X)(Y)(L 2 Z) groups are:
  • R" is hydrogen or (C 1 -C 8 )alkyl.
  • Each R 1 is independently halogen, (C ⁇ -C 8 )alkyl, (C -C 8 )alkenyl, (C - C 8 )alkynyl, fluoro(C 1 -C 4 )alkyl, -OR 5 , -SR 5 , fluoro(Ct-C 4 )alkoxy, aryl, aryl(d- C 4 )alkyl, -NO 2 , -NR 5 R 6 , -C(O)R 5 , -CO 2 R 5 , -C(O)NR 5 R 6 , -N(R 6 )C(O)R 5 , -N(R 6 )CO 2 R 5 , -N(R 7 )C(O)NR 5 R 6 , -S(O) m NR 5 R 6 , -S(O) m R 5 , -CN or -N(R 6 )S(O) m R 5 .
  • R 4 is hydrogen, -OR 11 , -C(O)R ⁇ , -CO 2 R ⁇ , -C(O)NR ⁇ R 12 , -CN, (C r C 4 )alkyl or aryl.
  • R 12 , R 13 , R 14 , R 16 and R 17 are independently selected from hydrogen, (C ⁇ -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, fluoro(C ⁇ - C 4 )alkyl, hetero(C 1 -C )alkyl, aryl and aryl(C ⁇ -C )alkyl and R 18 is a 5- or 6-membered ring containing from 1 to 3 heteroatoms selected from N, O and S.
  • the subscript m is 1 or 2 and the subscript n is 0, 1 or 2.
  • R groups selected from the group consisting of R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 and R 18 are attached to the same nitrogen atom
  • the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from N, O and S.
  • R" is hydrogen and R 3 is hydrogen.
  • the present invention provides compounds of formula (II):
  • L 1 represents a divalent linkage selected from a bond, (C ⁇ - C 4 )alkylene, (C ⁇ -C 4 )alkylenoxy and ( -C ⁇ alkylenamino.
  • Exemplary L 1 groups are a single bond, methylene, ethylene, n-propylene and n-butylene.
  • L 2 represents a divalent linkage selected from a bond, ( -C ⁇ alkylene, (C 2 -C 4 )alkenylene, (C 2 -C 4 )alkynylene, (C ⁇ -C 4 )alkylenoxy and (C 1 -C 4 )alkylenamino.
  • Exemplary L 2 groups are a single bond, methylene, ethylene, n-propylene and n-butylene.
  • X and Y represent independently (C 1 -C 8 )alkyl, (C 2 - C 8 )alkenyl, (C 2 -C 8 )alkynyl, -CO 2 R 13 or -C(O)NR 13 R 14 .
  • X and Y may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from N, O and S.
  • Exemplary -C(X)(Y)(L 2 Z) groups are:
  • R" is hydrogen or (Q-Q alkyl.
  • Each R 1 is independently halogen, ( -C ⁇ alkyl, (C 2 -C 8 )alkenyl, (C 2 - C 8 )alkynyl, fluoro(C 1 -C 4 )alkyl, -OR 5 , -SR 5 , fluoro(C 1 -C 4 )alkoxy, aryl, aryl(Cr C 4 )alkyl, -NO 2 , -NR 5 R 6 , -C(O)R 5 , -CO 2 R 5 , -C(O)NR 5 R 6 , -N(R 6 )C(O)R 5 , -N(R 6 )CO 2 R 5 , -N(R 7 )C(O)NR 5 R 6 , -S(O) m NR 5 R 6 , -S(O) m R 5 , -CN or -N(R 6 )S(O) m R 5 .
  • R 4 is hydrogen, -OR 11 , -C(O)R ⁇ , -CO 2 R n , -C(O)NR u R 12 , -CN, (C r
  • R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 16 and R 17 are independently selected from hydrogen, (C ⁇ -Cs)alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, fluoro(C ⁇ - C 4 )alkyl, hetero(C 1 -C 4 )alkyl, aryl and aryl(C ⁇ -C 4 )alkyl and R 18 is a 5- or 6-membered ring containing from 1 to 3 heteroatoms selected from N, O and S.
  • the subscript m is 1 or 2 and the subscript n is 0, 1 or 2.
  • R groups selected from the group consisting of R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 and R 18 are attached to the same nitrogen atom
  • the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from N, O and S.
  • Compounds of the invention feature a pyrido[4,3-b]carbazole-derived ring, minimally substituted at the 2- and 11 -positions.
  • the ring numbering system used herein is illustrated below.
  • L 1 is (C ⁇ -C 4 )alkylene.
  • L 1 is unsubstituted ( -G alkylene or -(CH 2 ) p - wherein the subscript p is an integer of from 1 to 4.
  • p is 1, 2 or 3.
  • p is 2 or 3.
  • p is 2.
  • X and Y are combined to form a 3-, 4-, 5-,
  • 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S.
  • X and Y are combined to form a 5- or 6-membered ring containing from 0 to 2 heteroatoms selected from O, N and S.
  • X and Y are combined to form a 5- or 6-membered ring containing 0 heteroatoms, 1 nitrogen atom or 1 oxygen atom.
  • L is a bond and Z is -CO 2 R or -CO 2 NR 15 R 16 .
  • R" is hydrogen.
  • R" is substituted ( - C 8 )alkyl.
  • R" is ( -C ⁇ alkyl substituted with hydroxy, alkylamino (e.g., — NHMe) or carboxy (-CO 2 H).
  • R" is (C -C 8 )alkyl substituted with hydroxy, alkylamino or carboxy.
  • R 1 is independently halogen, (C r C 4 )alkyl, fluoro(C r C 4 )alkyl, -OR 5 , fluoro(C r C 4 )alkoxy, -CO 2 R 5 , - S(O) m NR 5 R 6 , -S(O) m R 5 or -CN.
  • R 1 is independently halogen or fluoro(C 1 -C )alkyl.
  • R 1 is halogen or fluoro(C ⁇ -C 4 )alkyl and the subscript n is 0 or 1.
  • R 1 is fluoro(C 1 -C 4 )alkyl and the subscript n is 0 or 1.
  • R 2 is (C r C 4 )alkyl or aryl.
  • R 4 is hydrogen.
  • R" and R 4 are hydrogen.
  • R" and R 4 are hydrogen and R 2 is (C ⁇ -C )alkyl or aryl.
  • R" and R 4 are hydrogen, R 1 is independently halogen, (C r C 4 )alkyl, fluoro(C 1 -C 4 )alkyl, -OR 5 , fluoro(C r C 4 )alkoxy, -CO 2 R 5 , -S(O) m NR 5 R 6 , -S(O) m R 5 or-CN and R 2 is (d-C 4 )alkyl or aryl.
  • R" and R 4 are hydrogen, R 1 is halogen or fluoro(C 1 -C )alkyl, n is 1 and R 2 is (Ci-C ⁇ alkyl or aryl. In a more particularly prefe ⁇ ed embodiment, R" and R are hydrogen, R is fluoro(C ⁇ -C 4 )alkyl, n is 1 and R is (d-C )alkyl or aryl.
  • X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S, L 2 is a bond and Z is -CO 2 R 15 or -CO 2 NR 15 R 16 .
  • X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S and R 1 is independently halogen, (Q-C ⁇ alkyl, fluoro(C r C 4 )alkyl, - OR 5 , fluoro(C r C 4 )alkoxy, -CO 2 R 5 , -S(O) m NR 5 R 6 , -S(O) m R 5 or-CN.
  • X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S and R 2 is (C C 4 )alkyl or aryl.
  • X and Y are combined to form a 3-, A-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S
  • R 1 is independently halogen, (C r C 4 )alkyl, fluoro(C r C 4 )alkyl, -OR 5 , fluoro(C r C 4 )alkoxy, -CO 2 R 5 , -S(O) m NR 5 R 6 , -S(O) m R 5 or- CN and R 2 is (C r C 4 )alkyl or aryl.
  • R 1 is independently halogen, (C r C 4 )alkyl, fluoro(C r C )alkyl, -OR 5 , fluoro(C 1 -C 4 )alkoxy, - CO 2 R 5 , -S(O) m NR 5 R 6 , -S(O) m R 5 or -CN and R 2 is (C r C 4 )alkyl or aryl.
  • R 1 is independently halogen, (C r C 4 )alkyl, fluoro(C 1 -C 4 )alkyl, -OR 5 , fluoro(C r C 4 )alkoxy, - CO 2 R 5 , -S(O) m NR 5 R 6 , -S(O) m R 5 or -CN, L 2 is a bond and Z is -CO 2 R 15 or -CO 2 NR 15 R 16 .
  • R 2 is (C r C 4 )alkyl or aryl
  • L 2 is a bond
  • Z is -CO 2 R 15 or -CO 2 NR 15 R 16 .
  • R 1 is independently halogen, fluoro(C 1 -C 4 )alkyl, -OR 5 , fluoro(C r C 4 )alkoxy, - CO 2 R 5 , -S(O) m NR 5 R D , -S(O) m R 5 or -CN, R 2 is (C r C 4 )alkyl or aryl, L 2 is a bond and Z is -CO 2 R 15 or -CO 2 NR 15 R 16 .
  • the present invention provides the following compounds:
  • the present invention provides pharmaceutically acceptable salts of the above compounds.
  • the present invention provides benzenesulfonic acid salts of the above compounds.
  • the present invention provides pharmaceutical compositions comprising one or more compounds of the invention in combination with a diagnostically or pharmaceutically acceptable carrier or excipient.
  • the subject compositions are useful for treating or preventing conditions and disorders mediated by MCHR, such as obesity and eating disorders, e.g., anorexia nervosa.
  • the compounds of the present invention can be prepared and administered in a wide variety of oral and parenteral dosage forms.
  • the compounds of the present invention can be administered by injection, for example, intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally or inxraperitoneally.
  • the compounds described herein can be administered by inhalation, for example, intranasally.
  • the compounds of the present invention can be administered transdermally.
  • Other routes of administration are also contemplated for use with the compounds of the present invention, including depot administration and rectal administration.
  • pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • the powders and tablets preferably contain from about 5% or 10% to 70% of the active compound.
  • Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • the term "preparation" is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is su ⁇ ounded by a carrier, which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.
  • a low melting wax such as a mixture of fatty acid glycerides or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
  • Liquid form preparations include solutions, suspensions and emulsions, for example, water or water/propylene glycol solutions.
  • liquid preparations can be formulated in solution in aqueous polyethylene glycol solution.
  • Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.
  • viscous material such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • Such liquid forms include solutions, suspensions and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
  • the pharmaceutical preparation is preferably in unit dosage form.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • the quantity of active component in a unit dose preparation may be varied or adjusted from 0.1 mg to 1000 mg, preferably 1.0 mg to 100 mg according to the particular application and the potency of the active component.
  • the composition can, if desired, also contain other compatible therapeutic agents.
  • the compounds utilized in the pharmaceutical method of the invention are administered at the initial dosage of about 0.001 mg/kg to about 100 mg/kg daily.
  • a daily dose range of about 0.1 mg/kg to about 10 mg/kg is prefe ⁇ ed.
  • the dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated and the compound being employed. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired.
  • compositions may be advantageously combined and/or used in combination with agents useful in the treatment and/or prevention of obesity and eating disorders and pathologies associated therewith (e.g., cardiovascular disease and hypertension).
  • agents useful in the treatment and/or prevention of obesity and eating disorders and pathologies associated therewith e.g., cardiovascular disease and hypertension.
  • administration of the subject compounds or compositions in conjunction with these alternative agents enhances the efficacy of such agents.
  • the present compounds when combined or administered in combination with, e.g., anti-obesity agents, can be used in dosages which are less than the expected amounts when used alone, or less than the calculated amounts for combination therapy.
  • Suitable agents for combination therapy include those that are cu ⁇ ently commercially available and those that are in development or will be developed.
  • Exemplary agents useful in the treatment of obesity include ⁇ 3 adrenergic receptor agonists, leptin or derivatives thereof and neuropeptide Y antagonists.
  • agents useful in the treatment of anxiety and/or mood disorders include benzodiazepines, e.g., alprazolam, chlordiazepoxide, clonazepam, clorazepate, diazepam, lorazepam, oxazepam, and the like; heterocyclic antidepressants, e.g, amitriptyline, nortriptyline, imipramine, desipramine, doxepin, trimipramine, clomipramine, protryptyline, amoxapine and maprotiline; monoamine oxidase inhibitors (MAOIs), e.g., phenelzine and tranylcypromine; serotonin reuptake inhibitors (SRIs); selective serotonin reuptake inhibitors (SSRIs), e.g., fluoxetine, fluvoxamine, paroxetine and sertraline; serotonergic-noradrenergic
  • the present invention provides methods of using one or more compounds of the invention to treat or prevent a condition or disorder associated with eating behavior, energy homeostasis or anxiety.
  • exemplary conditions and disorders associated with eating behavior, energy homeostasis and anxiety include eating disorders, such as anorexia nervosa and bulimia, obesity, anxiety disorders, e.g., generalized anxiety disorder, panic attacks, panic disorder and obsessive-compulsive disorder (OCD), and mood disorders, e.g., depression and bipolar disorders.
  • eating disorders such as anorexia nervosa and bulimia
  • obesity anxiety disorders, e.g., generalized anxiety disorder, panic attacks, panic disorder and obsessive-compulsive disorder (OCD), and mood disorders, e.g., depression and bipolar disorders.
  • OCD obsessive-compulsive disorder
  • mood disorders e.g., depression and bipolar disorders.
  • the present invention provides methods of using a compound of the invention to treat or prevent a condition or disorder mediated by MCHR.
  • the methods comprise administering to a subject in need thereof a therapeutically effective amount of a compound of the invention.
  • the present invention provides methods of using a compound of the invention to modulate MCHR.
  • the methods comprise contacting a cell with a compound of the invention.
  • the compounds of the invention may also modulate G-protein coupled receptors related to MCHR, e.g., MCHR2 (see International Publication Nos. WO 00/49046 and WO 01/07606).
  • the present invention provides a process for the preparation of a compound of formula I.
  • a general synthetic route is depicted in Scheme 1, which outlines the condensation of substituted aryl moiety A, with a bicyclic structure B to produce a compound of formula C, wherein the variables are as defined as above.
  • D 1 is hydrogen, halogen, -C(O)R 7 , -CO 2 R 8 or -C(O)NR 5 R 6 , wherein R 5 , R 6 , R 7 and R 8 are defined as above, and D 2 is a bond, -N(R")-, -N(protecting group)-, -S- or -O-, wherein R" is defined as above and protecting group is an amino protecting group.
  • Conventional amino protecting groups consist of known groups which are used to protectively block an amino group during the synthesis procedures described herein. These conventional blocking groups are readily removable, i.e., they can be removed, if desired, by procedures which will not cause cleavage or other disruption of the remaining portions of the molecule. Suitable protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al,. Protective Groups in Organic Synthesis, Wiley, New York (1991).
  • E 1 is hydrogen, -C(O)R 7 , -CO 2 R 8 or -C(O)NR 5 R 6 , wherein
  • the activity of MCHR polypeptides can be assessed using a variety of in vitro and in vivo assays to determine functional, chemical and physical effects, e.g., measuring ligand binding (e.g., radioactive ligand binding), second messenger (e.g., cAMP, cGMP, IP 3 , DAG, or Ca 2+ ) levels, ion flux, phosphorylation levels, transcription levels, neurotransmitter levels, and the like. Furthermore, such assays can be used to test for antagonists and agonists of MCHR. Screening assays may be used to identify modulators that can be used as therapeutic agents, e.g., antagonists of MCHR activity.
  • ligand binding e.g., radioactive ligand binding
  • second messenger e.g., cAMP, cGMP, IP 3 , DAG, or Ca 2+
  • ion flux e.g., phosphorylation levels
  • transcription levels e.g., neurotransmitter
  • Modulators of MCHR activity can be tested using MCHR polypeptides as described above, either recombmant or naturally occurring (e.g., endogenous).
  • the protein can be isolated, expressed in a cell, expressed in a membrane derived from a cell, expressed in tissue or in an animal, either recombinant or naturally occurring. For example, kidney cells, liver cells, colon cells, transformed cells, or membranes can be used. Modulation is tested using one of the in vitro or in vivo assays described herein.
  • Signal transduction can also be examined in vitro with soluble or solid state reactions, using a chimeric molecule such as an extracellular domain of a receptor covalently linked to a heterologous signal transduction domain, or a heterologous extracellular domain covalently linked to the transmembrane and or cytoplasmic domain of a receptor. Gene amplification can also be examined. Furthermore, ligand-binding domains of the protein of interest can be used in vitro in soluble or solid state reactions to assay for ligand binding.
  • Ligand binding to MCHR, a domain, or chimeric protein can be tested in solution, in a bilayer membrane, attached to a solid phase, in a lipid monolayer, or in vesicles. Binding of a modulator can be tested using, e.g., changes in spectroscopic characteristics (e.g., fluorescence, absorbance, refractive index) hydrodynamic (e.g., shape), chromatographic, or solubility properties.
  • spectroscopic characteristics e.g., fluorescence, absorbance, refractive index
  • hydrodynamic e.g., shape
  • chromatographic chromatographic, or solubility properties
  • MCHR-G-protein interactions can also be examined, by, for example, analysis of binding of the G-protein to MCHR or its release from MCHR can be examined. For example, in the absence of GTP, an activator will lead to the formation of a tight complex of a G protein (all three subunits) with MCHR. This complex can be detected in a variety of ways, as noted above. Such an assay can be modified to search for antagonists.
  • an activator is added to MCHR and G protein in the absence of GTP, allowed to form a tight complex, and then screened for antagonists by looking at dissociation of the MCHR-G protein complex, hi the presence of GTP, release of the alpha subunit of the G protein from the other two G protein subunits serves as a criterion of activation.
  • An activated or inhibited G-protein will in turn alter the properties of downstream effectors such as proteins, enzymes and channels.
  • the classic examples are the activation of cGMP phosphodiesterase by transducin in the visual system, adenylate cyclase by the stimulatory G-protein, phospholipase C by Gq and other cognate G proteins, and modulation of diverse channels by Gi and other G proteins. Downstream consequences can also be examined such as generation of diacyl glycerol and IP3 by phospholipase C, and in turn, for calcium mobilization by IP3.
  • Activated MCHR becomes a substrate for kinases that phosphorylate the C-terminal tail of the receptor (and possibly other sites as well).
  • activators will promote the transfer of P from gamma-labeled ATP to the receptor, which can be assayed with a scintillation counter.
  • the phosphorylation of the C-terminal tail will promote the binding of arrestin-like proteins and will interfere with the binding of G- proteins.
  • the kinase/arrestin pathway plays a key role in the desensitization of many GPCR receptors.
  • Samples or assays that are treated with a potential MCHR antagonist or agonist are compared to control samples without the test compound, to examine the extent of modulation.
  • Control samples (untreated with agonists or antagonist) are assigned a relative MCHR activity value of 100.
  • Inhibition of MCHR is achieved when the MCHR activity value relative to the control is about 90%, optionally 50%, optionally 25-0%.
  • Activation of MCHR is achieved when the MCHR activity value relative to the control is 110%, optionally 150%, 200-500%, or 1000-2000%.
  • Changes in ion flux may be assessed by determining changes in polarization (i.e., electrical potential) of the cell or membrane expressing MCHR.
  • polarization i.e., electrical potential
  • One means to determine changes in cellular polarization is by measuring changes in cu ⁇ ent (thereby measuring changes in polarization) with voltage-clamp and patch-clamp techniques, e.g., the "cell-attached" mode, the "inside-out” mode, and the "whole cell” mode (see, e.g., Ackerman et al, New Engl J. Med. 336:1575-1595 (1997)).
  • Whole cell cu ⁇ ents are conveniently determined using the standard methodology (see, e.g., Hamil et al, PFlugers. Archiv. 391:85 (1981).
  • test compounds upon the function of the polypeptides can be measured by examining any of the parameters described above. Any suitable physiological change that affects MCHR activity can be used to assess the influence of a test compound on the polypeptides of this invention.
  • functional consequences are determined using intact cells or animals, one can also measure a variety of effects such as transmitter release, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as Ca , 1P3 or cAMP.
  • Preferred assays for MCHR include cells that are loaded with ion- or voltage-sensitive dyes to report receptor activity. Assays for determining activity of such receptors can also use known agonists and antagonists for other G-protein coupled receptors as negative or positive controls to assess activity of tested compounds, h assays for identifying modulatory compounds (e.g., agonists, antagonists), changes in the level of ions in the cytoplasm or membrane voltage will be monitored using an ion- sensitive or membrane voltage fluorescent indicator, respectively.
  • modulatory compounds e.g., agonists, antagonists
  • changes in the level of ions in the cytoplasm or membrane voltage will be monitored using an ion- sensitive or membrane voltage fluorescent indicator, respectively.
  • ion- sensitive indicators and voltage probes that may be employed are those disclosed in the Molecular Probes 1997 Catalog.
  • promiscuous G-proteins such as G ⁇ l5 and G ⁇ l6 can be used in the assay of choice (Wilkie et al, Proc. NatlAcad. Set USA 88:10049-10053 (1991)).
  • Such promiscuous G-proteins allow coupling of a wide range of receptors to signal transduction pathways in heterologous cells.
  • Receptor activation typically initiates subsequent intracellular events, e.g., increases in second messengers such as IP3, which releases intracellular stores of calcium ions.
  • IP3 inositol triphosphate
  • phospholipase C-mediated hydrolysis of phosphatidylinositol (Berridge & Irvine, Nature 312:315-21 (1984)).
  • IP3 in turn stimulates the release of intracellular calcium ion stores.
  • a change in cytoplasmic calcium ion levels, or a change in second messenger levels such as IP3 can be used to assess G-protein coupled receptor function.
  • Cells expressing such G-protein coupled receptors may exhibit increased cytoplasmic calcium levels as a result of contribution from both intracellular stores and via activation of ion channels, in which case it may be desirable although not necessary to conduct such assays in calcium-free buffer, optionally supplemented with a chelating agent such as EGTA, to distinguish fluorescence response resulting from calcium release from internal stores.
  • a chelating agent such as EGTA
  • cyclic nucleotide-gated ion channels e.g., rod photoreceptor cell channels and olfactory neuron channels that are permeable to cations upon activation by binding of cAMP or cGMP (see, e.g., Altenhofen et al, Proc. Natl Acad. Sci. U.S.A.
  • Cells for this type of assay can be made by co-transfection of a host cell with DNA encoding a cyclic nucleotide-gated ion channel, GPCR phosphatase and DNA encoding a receptor (e.g., certain glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors, and the like), which, when activated, causes a change in cyclic nucleotide levels in the cytoplasm.
  • a receptor e.g., certain glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors, and the like
  • changes in intracellular cAMP or cGMP can be measured using immunoassays. The method described in Offermanns & Simon, J. Biol. Chem.
  • 270:15175-15180 (1995) maybe used to determine the level of cAMP.
  • the method described in Felley-Bosco et al, Am. J. Resp. Cell andMol Biol. 11:159-164 (1994) may be used to determine the level of cGMP.
  • an assay kit for measuring cAMP and/or cGMP is described in U.S. Patent No. 4,115,538, herein incorporated by reference.
  • phosphatidyl inositol (PI) hydrolysis can be analyzed according to U.S. Patent No. 5,436,128, herein incorporated by reference.
  • transcription levels can be measured to assess the effects of a test compound on signal transduction.
  • a host cell containing the protein of interest is contacted with a test compound for a sufficient time to effect any interactions, and then the level of gene expression is measured.
  • the amount of time to effect such interactions may be empirically determined, such as by running a time course and measuring the level of transcription as a function of time.
  • the amount of transcription may be measured by using any method known to those of skill in the art to be suitable. For example, mRNA expression of the protein of interest may be detected using northern blots or their polypeptide products may be identified using immunoassays. Alternatively, transcription based assays using a reporter gene may be used as described in U.S. Patent No. 5,436,128, herein incorporated by reference.
  • the reporter genes can be, e.g., chloramphenicol acetyltransferase, firefly luciferase, bacterial luciferase, ⁇ -galactosidase and alkaline phosphatase.
  • the protein of interest can be used as an indirect reporter via attachment to a second reporter such as green fluorescent protein (see, e.g., Mistili & Spector, Nature Biotechnology 15:961- 964 (1997)).
  • the amount of transcription is then compared to the amount of transcription in either the same cell in the absence of the test compound, or it may be compared with the amount of transcription in a substantially identical cell that lacks the protein of interest.
  • a substantially identical cell may be derived from the same cells from which the recombinant (or non-recombinant) cell line was prepared but which had not been modified by introduction of heterologous DNA. Any difference in the amount of transcription indicates that the test compound has in some manner altered the activity of the protein of interest.
  • Reagents and solvents used below can be obtained from commercial sources such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA).
  • 1 H-NMR spectra were recorded on a Narian Gemini 400 MHz ⁇ MR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constant(s) in Hertz (Hz) and number of protons.
  • Election lonization (El) mass spectra were recorded on a Hewlett Packard 5989A mass spectrometer.
  • Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses). A single m/e value is reported for the M+H (or, as noted, M-H) ion containing the most common atomic isotopes. Isotope patterns correspond to the expected formula in all cases.
  • Electrospray ionization (ESI) mass spectrometry analysis was conducted on a Hewlett- Packard 1100 MSD electrospray mass spectrometer using the HP1 100 HPLC for sample delivery.
  • analyte was dissolved in methanol at 0.1 mg/mL and 1 microliter was infused with the delivery solvent into the mass spectrometer, which scanned from 100 to 1500 daltons. All compounds could be analyzed in the positive ESI mode, using 1:1 acetonitrile/water with 1% acetic acid as the delivery solvent. The compounds provided below could also be analyzed in the negative ESI mode, using 2mM NH 4 OAc in acetonitrile/water as delivery solvent. Analytical HPLC analysis was conducted on a Hewlett-Packard Series 1050 system equipped with a C18 reverse phase column (4.6 mm xl50mm) manufactured by Shiseido Co., Japan.
  • Compound 4 was prepared in 4 steps, as follows.
  • Step 1 Robinson Annulation.
  • a mixture of N-benzyl-4-piperidone (500 g, 2.65 mol) and pyrrolidine (330 mL, d 0.852, 3.96 mol) in toluene (2L) was heated at refluxing while water from elimination was removed with a Dean-Stark trap. After 8 h, 70 mL of water were collected, and the volume of collected water ceased to increase further.
  • GC analysis revealed the presence of the starting N-benzyl-4-piperidone and product enamine.
  • the solvent and excess pyrrolidine were evaporated under reduced pressure (vacuum, 60 ton; heating bath, 50°C). The residue was dissolved in 500 mL of toluene, and evaporated again to give a dark oil (630g).
  • reaction vessel was capped and heated at 90°C for 36 h until the completion of reaction, as monitored by TLC (10:1:0.1, CH 2 Cl 2 /MeOH/NH 4 OH), and ES-MS in positive mode.
  • TLC 10:1:0.1, CH 2 Cl 2 /MeOH/NH 4 OH
  • ES-MS in positive mode.
  • the reaction mixture was poured to a sti ⁇ ed solution of 1 N NaOH (some precipitates were formed).
  • the mixture was extracted with dichloromethane three times.
  • the combined organic extracts were washed with water, dried over NaSO 4 , filtered, and concentrated to give a solid.
  • the residue was triturated with CH 2 C1 2 .
  • the solid was collected by filtration.
  • the product could be purified by chromatography on silica gel column with a gradient elution of increasing polarity from 20: 1 :0.1 to 6: 1 :0.1 CH 2 Cl 2 /MeOH/NH 4 OH to obtain compound 4 as the major product.
  • Step 1 To a 500 mL flask containing iPr 2 NH (16.82 mL, 120 mmol) in THF
  • Compound 13 was synthesized in 5 steps according to the following scheme.
  • Step 1 A mixture of 4-allyl-tetrahydropyran carboxylic acid methyl ester (15.7 g, 92.4 mmol) and LiOH-H 2 O (29 g, 688 mmol) in THF (110 mL), MeOH (110 mL) and water (5 mL) was heated to 85° C in a sealed vessel overnight. Upon cooling to r.t., it was extracted with EtOAc, washed with water , dried with anhydrous Na 2 SO and concentrated by rotary evaporation to yield the corresponding carboxylic acid as a white solid (12.82 g). Step 2. To a mixture of the above acid (12.82 g, 75.4 mmol), DMF (4 drops) in
  • the obtained azide was dissolved in 100 mL of benzene and added slowly to 100 mL of refluxing benzene. The reflux continued for another 40 min at which time no more gas was released. Benzene was distilled off and 120 mL of MeOH was added to the mixture. The mixture was heated at refluxing for 36 h, cooled to r.t. and was directly chromatographed using 40% EtOAc/hexanes as eluent. The carbamate product was obtained as a white solid (14.15 g).
  • Step 4 The above aldehyde (0.38 g, 1.9 mmol) was sti ⁇ ed with amine 4 (0.587 g, 1.9 mmpl) and NaBH(OAc) 3 (1.61 g, 7.6 mmol) in C1CH 2 CH 2 C1 (15 mL) overnight. The reaction mixture was poured into a dilute aqueous ammonia solution and extracted with EtOAc.
  • Step 1 A mixture of amine 13 (0.654 g, 1.5 mmol), OHCCH 2 CH 2 CH 2 CO 2 Et (0.260 g, 2 mmol), NaBH(OAc) 3 (1.27 g, 6 mmol) and
  • the product obtained above (0.35 g, 0.63 mmol) was hydrolyzed by treating with LiOH.H O (0.50 g, 12 mmol) in dioxane (4 mL) and H 2 O (2 mL) at r.t. for 3 h.
  • the mixture was acidified with HO Ac to slightly acidic, and was concentrated to dryness.
  • the above acid ( ⁇ 0.1 mmol, contained inorganic salt) was heated with
  • the above aldehyde was converted to nitrile by the following reaction.
  • a vial containing NH 2 OH.HCl (0.083 g, 1.2 mmol) and CH 3 CN (3 mL) at 0 °C was added TEA (0.168 mL, 1.2 mmol) and the aldehyde (0.470 g, 0.91 mmol).
  • the mixture was sti ⁇ ed at 0 °C for 30 min. and at r.t. for 4 h.
  • phthalic anhydride (0.178 g, 1.2 mmol) was added and the mixture was heated to 90 °C for 1 h, poured into saturated NaHCO and extracted with EtOAc.
  • acylamidine was treated with NH 2 NH 2 ⁇ O (0.030 mL, 0.6 mmol) in AcOH (lmL) at 90 °C for 10 min. After cooling to r.t., the whole was directly loaded onto a column eluted with 10 - 30% MeOH/DCM mixed with 1 - 4% NH 4 OH to yield triazole 38 as a white solid (0.050 g).
  • Step 1 LAH (0.64 g, 16.7 mL) was added to a dry THF solution (70 mL) containing the ester compound from Example 1 (4.0 g, 8.4 mmol) at room temperature. The solution was heated at reflux for 2 h. After heating, water (0.6 mL) was added followed by a 1 N solution of NaOH (0.6 mL), and a final addition of water (1.2 mL). The resulting solid was filtered washed with copious amount of dichloromethane. The filtrate was concentrated and used in the next step without purification: ESI (MH + ) m/z 451.
  • Step 2 The resulting alcohol compound from above (0.6 g, 1.33 mmol) was dissolved in DMSO/Et N (2.5:1, 0.2 M) and was treated with SO 3 -pyridine complex (0.85 g, 5.33 mmol) and at room temperature. After stirring for 2 h, the mixture was poured into water (60 mL) and extracted with dichloromethane (3 x 100 mL). The organic layers were washed with brine, dried over Na SO 3 , and concentrated to give the aldehyde intermediate. ESI (MH*) m/z 449.
  • Step 3 Sodium hydride (0.96, 40 mmol) was added to a dry DMF (0.2 M) solution containing tiiethyl phosphonoacetate (4 mL, 20.0 mmol) at room temperature. After stirring for 10 min., the aldehyde intermediate from above (4.51 g, 10.1 mmol) was added and the mixture was sti ⁇ ed at room temperature overnight. Excess DMF was removed under vacuum and the remaimng residue was taken up in a 10% MeOH/DCM solution, washed with water, dried with Na 2 SO 3 and concentrated. A portion of this material was purified by using preparative HPLC (C 18 column, 10%- 90% acetonitrile/water gradient).
  • Lithium hydroxide (30 mg, 1.3 mmol) was added to a THF/water solution (1:1, 0.2 M) containing ester intermediate from Example 44 (0.54 g, 1.0 mmol) and heated at reflux for 3 h. After cooling to room temperature, the solution was titrated with a 3 N HC1 solution to neutral pH and concentrated to dryness using reduced pressure. This material was used in the next step without purification.
  • ESI MH +
  • HBTU (3 equiv.) was added to a dichloromethane solution (0.2 M) containing, triethylamine (3 equiv.), acid intermediate from above(l equiv.), and the respective amine (2 equiv.) at room temperature. After stirring overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient).
  • Tosylmethyl isocyanide (22 mg, 0.1 mmol) was added to a methanol solution (5 mL) containing potassium carbonate (19 mg, 1.2 mmol) and aldehyde intermediate from Step 2 of Example 44 (50 mg, 0.1 mmol). The solution was heated at reflux for 24 h.
  • the co ⁇ esponding amine compound was prepared following the same procedures as depicted for Example 15, with the exception of substituting 4-allyl- tetrahydropyran carboxylic acid methyl ester with 1 -allyl- 1-cyclohexyl carboxylic acid methyl ester.
  • Compound 76 was prepared by reducing compound 69 and oxidizing the co ⁇ esponding alcohol with SO 3 -pyridine in DMSO/TEA as described in Example 44. To the solution of compound 76 (25 mg, 0.061 mmol) in 2 mL DCM was added NaBH(OAc) 3 (26.0 mg, 0.122 mmol) followed by the addition of amine (11 mg, 0.122 mmol). The reaction mixture was kept stirring for overnight. The solvent was removed and 1 mL DMF was added to dissolve the mixture. After filtration, the solution was injected directly to reverse HPLC to render pure yellow film 77 (18 mg, 0.038 mmol) as the product.
  • Comound 83 (0.02g, 0.04 mmol) was treated with lithium aluminumhydride (0.02 g, 0.042mmol) in THF for 2 h. at room temperature. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 20:1:0.1 DCM:MeOH:NH 4 OH to yield 85 as yellow solid (0.055 g).
  • the acid (0.052g, 0.11 mmol) was reacted with 2-methoxyethylamine (0.0098 g, 0.13 mmol), under the condition of EDC (0.063 g, 0.33 mmol), HOBt (0.0147g, 0.11 mmol), and NMM (0.033 g, 0.33 mmol) in DMF (1 mL) at room temperature overnight.
  • the reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentiated and purified by flash chromatography on silica gel eluted with 20: 1 :0.1 DCM:MeOH:NH 4 OH to yield 86 as brown oil (0.003 g).
  • the MCHR modulatory activity of the compounds of the invention can be assessed using the in vitro and in vivo assay methods described above.
  • Exemplary in vitro methods include fluorometric imaging plate reader
  • FLIPR functional assays
  • G Protein-Coupled Receptors (1999) pp. 105-108 T. Haga, G. Bernstein, eds.) CRC Press; Lembo et al. (1999) Nature Cell Biol. 1:267- 271; Saito et al. (1999) Nature 400:265-269; Wood et al (2000) Eur. J. Pharmacol. 396:1-8 and Miller et al. (1999) J. Biomol Screen. 4:249-258) and radioligand binding assays (see, e.g., Receptor Binding Techniques (1999) pp. 37-47 (M.

Abstract

Compounds, compositions and methods are provided that are useful in the treatment and/or prevention of a condition or disorder mediated by a G-protein coupled receptor. In particular, the compounds of the invention are useful in the treatment and/or prevention of eating disorders, obesity, anxiety disorders and mood disorders.

Description

FUSED HETEROCYCLIC COMPOUNDS
This application claims the benefit of U.S. provisional application no. 60/424,456, the contents of which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION
The present invention relates to compounds, compositions and methods useful in the treatment or prevention of conditions and disorders associated with eating behavior, energy homeostasis and anxiety.
BACKGROUND OF THE INVENTION
G-protein coupled receptors play important roles in diverse signaling processes, including those involved with sensory and hormonal signal transduction. Eating disorders, which represent a major health concern throughout the world, have been linked to GPCR regulation. On the one hand, disorders such as obesity, the excess deposition of fat in the subcutaneous tissues, manifest themselves by an increase in body weight. Individuals who are obese often have, or are susceptible to, medical abnormalities including respiratory difficulties, cardiovascular disease, diabetes and hypertension. On the other hand, disorders like cachexia, the general lack of nutrition and wasting associated with chronic disease and/or emotional disturbance, are associated with a decrease in body weight. The neuropeptide melanin-concentrating hormone (MCH), a cyclic hypothalamic peptide involved in the regulation of several functions in the brain, has previously been found to be a major regulator of eating behavior and energy homeostasis. It has previously been determined that MCH is the natural ligand for the 353-amino acid orphan G-protein-coupled-receptor (GPCR) termed SLC-1 (also known as GPR24). Subsequent to this determination, SLC-1, which is sequentially homologous to the somatostatin receptors, is frequently referred to as melanin- concentrating hormone receptor (MCH receptor, MCHR or MCHR1) (see Chambers et al, Nature 400:261-65 (1999); Saito et al, Nature 400:265-69 (1999); and Saito et al, TEM 11(8):299-303 (2000)).
Compelling evidence exists that MCH is involved in regulation of eating behavior. First, intracerebral administration of MCH in rats resulted in stimulation of feeding. Next, mRNA corresponding to the MCH precursor is up-regulated in the hypothalamus of genetically obese mice and of fasted animals. Finally, mice deficient in MCH are leaner and have a decreased food intake relative to normal mice. MCH is believed to exert its activity by binding to MCHR, resulting in the mobilization of mtracelfular calcium and a concomitant reduction in cAMP levels (see Chambers et al, Nature 400:261-65 (1999); Shimada et al. Nature 396:670-74 (1998)). MCH also activates inwardly rectifying potassium channels, and MCHR has been found to interact with both Gcri protein and Gαq protein (Saito et al, TEM ll(8):299-303 (2000)). Moreover, analysis of the tissue localization of MCHR indicates that it is expressed in those regions of the brain involved in olfactory learning and reinforcement. The cumulative data suggest that modulators of MCHR should have an effect on neuronal regulation of food intake (see Saito et al, Nature 400:265-69 (1999)).
MCH has been shown to modulate behaviors other than feeding, such as anxiety (Gonzales et al. (1996) Peptides 17:171-177; Monzon et al. (1999) Physiol Behav. 67:813-817). The identification of MCHR modulators is useful for the study of physiological processes mediated by MCHR and the development of therapeutic agents for the treatment or prevention of conditions and disorders associated with weight regulation, learning, anxiety and other neuronal-related functions.
SUMMARY OF THE INVENTION
The present invention provides fused heterocyclic compounds and compositions, and methods of use thereof to treat or prevent conditions and disorders mediated by MCHR. In particular, the present invention provides compounds, compositions and methods for treating or preventing conditions and disorders associated with eating behavior, energy homeostasis and anxiety. The compounds provided herein have the formula (I):
Figure imgf000005_0001
wherein
Figure imgf000005_0002
represents a single or fused aryl or heteroaryl ring; Q is -N(R)- or -N(R)-(C1-C3)alkylene-;
Figure imgf000005_0003
L1 is a bond, ( -C^alkylene, ( -C^alkylenoxy and (Ci- C4)alkylenamino; L2 is a bond, ( -C- alkylene, (C2-C4)alkenylene, (C -C4)alkynylene,
(Ci-C4)alkylenoxy (e.g. -OCH2CH2-) or (d-C^alkylenamino (e.g. -NH-CH2CH2-);
R" is hydrogen or (Cι-C8)alkyl; each R1 is independently selected from the group consisting of halogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(CrC4)alkyl, -OR5, -SR5, fluoro(CrC4)alkoxy, aryl, aryl(CrC4)alkyl, -NO2, -NR5R6, -C(O)R5, -CO2R5, - C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, - S(O)mR5, -CN and -N(R6)S(O)mR5;
R2 and R3 are independently selected from the group consisting of hydrogen, halogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C -C8)alkynyl, fluoro(C1-C4)alkyl, - OR8, -SR8, fluoro(C1-C4)alkoxy, aryl, aryl(C C4)alkyl, -NO2, -NR8R9, =O, -C(O)R8, - CO2R8, -C(O)NR8R9, -N(R9)C(O)R8, -N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9, -S(O)mR8, -CN and -N(R9)S(O)mR8;
R4 is selected from the group consisting of hydrogen, -OR11, -C(O)Rπ, - CO2Rπ, -C(O)NRπR12, -CN, (CrC4)alkyl and aryl; X and Y are independently selected from the group consisting of (C\-
C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, -CO2R13 and -C(O)NR13R14; optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms independently selected from the group consisting of N, O and S;
Z is selected from the group consisting of -OR15, -NR15R16, -NR15R18, -C(O)R15, -CO2R15, -R18, -C(O)NR15R16, -C(O)NR15R18, -SO2NR15R16, -SO2NR15R18, -NR16SO2R15, -N(R15)N(R16)SO2R17, -C(O)N(R16)OR15, hydroxy - C8)alkyl, fluoro(Cι-C4)alkyl, heteroaryl, -C(=NOR15)NR16R17, -C(R16)=NOR15, -NR16(OR15), -C(O)NR17C(O)NR15R16, -NR17C(O)NR16C(O)R15 and -NR17C(O)NR15R16;
R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16 and R17 are independently selected from the group consisting of hydrogen, (C1-C8)alkyl, (C2-
C8)alkenyl, (C2-C8)alkynyl, cyclo(C3-C6)alkyl, fluoro(Cι-C )alkyl, hetero(Cι-C4)alkyl, cyclohetero(C3-C6)alkyl, aryl and aryl(Cι-C4)alkyl;
R18 is a 5- or 6-membered ring containing from 0 to 4 heteroatoms selected from the group consisting of N, O and S (e.g. tetrazole); optionally, when two R groups selected from the group consisting of R5,
R6, R8, R9, R11, R12, R13, R14, R15, R16 and R17 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from the group consisting of N, O and S; the subscript m is 1 or 2; and the subscript n is 0, 1 or 2.
In certain embodiments
Figure imgf000006_0001
represents a benzene, naphthalene, pyrrole, pyrazole, imidazole, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, pyridine, pyrimidine, benzotbiazole, purine, benzimidazole, indole, isoquinoline, quinoxaline or quinoline ring.
In certain embodiments
Figure imgf000006_0002
represents a benzene ring.
In certain embodiments Q is -N(R)-.
In further embodiments, R3 is hydrogen or =O.
In particular embodiments,
Figure imgf000006_0003
represents a benzene ring, R" is hydrogen and R3 is hydrogen.
Further compounds provided herein have the formula (II):
Figure imgf000007_0001
II wherein
L 1 i s a bond, ( -C- alkylene, (CrC^alkylenoxy and (C\- C )alkylenamino;
L2 is a bond, ( -G alkylene, (C2-C )alkenylene, (C2-C4)alkynylene, (Cι-C4)al ylenoxy (e.g. -OCH2CH2-) or (CrC^alkylenamino (e.g. -NH-CH2CH2-); R" is hydrogen or (CrC^alkyl; each R1 is independently selected from the group consisting of halogen, (Cι-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(CrC4)alkyl, -OR5, -SR5, fluoro(CrC4)alkoxy, aryl, aryl(C1-C4)alkyl, -NO2, -NR5R6, -C(O)R5, -CO2R5, - C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, - S(O)mR5, -CN and -N(R6)S(O)mR5;
R2 is selected from the group consisting of hydrogen, halogen, ( - C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, -OR8, -SR8, fluorotd- C4)alkoxy, aryl, ary -C^alkyl, -NO2, -NR8R9, =O, -C(O)R8, -CO2R8, -C(O)NR8R9, - N(R9)C(O)R8, -N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9, -S(O)mR8, -CN and - N(R9)S(O)mR8;
R4 is selected from the group consisting of hydrogen, -OR11, -C(O)Rπ, - CO2Rn, -C(O)NRπR12, -CN, (CrC4)alkyl and aryl;
X and Y are independently selected from the group consisting of ( - C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, -CO2R13 and -C(O)NR13R14; optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms independently selected from the group consisting of N, O and S;
Z is selected from the group consisting of -OR15, -NR15R16, ~NR15R18, -C(O)R15, -CO2R15, -R18, -C(O)NR15R16, -C(O)NR15R18, -SO2NR15R16, -SO2NR15R18, -NR16SO2R15, -N(R15)N(R16)SO2R17, -C(O)N(R16)OR15, hydroxy(Cι- C8)alkyl, fluoro(C1-C4)alkyl, heteroaryl, -C(=NOR15)NR16R17, -C(R16)=NOR15, -NR16(OR15), -C(O)NR17C(O)NR15R16, -NR17C(O)NR16C(O)R15 and -NR17C(O)NR15R16; R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16 and R17 are independently selected from the group consisting of hydrogen, (C1-C8)alkyl, (C2- C8)alkenyl, (C2-C8)alkynyl, cyclo(C3-C6)alkyl, fluoro(C1-C4)alkyl, hetero(C C4)alkyl, cyclohetero(C3-C6)alkyl, aryl and aryl(Cι-C4)alkyl;
R is a 5- or 6-membered ring containing from 0 to 4 heteroatoms selected from the group consisting of N, O and S (e.g. tetrazole); optionally, when two R groups selected from the group consisting of R5, R6, R8, R9, R11, R12, R13, R14, R15, R16 and R17 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from the group consisting of N, O and S; the subscript m is 1 or 2; and the subscript n is 0, 1 or 2.
The compounds provided in the above formulas are meant to include all pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof. The pharmaceutical compositions provided herein comprise a pharmaceutically acceptable carrier or excipient in combination with a compound of formula I or II.
Methods for treating or preventing a condition or disorder selected from the group consisting of obesity, an eating disorder, an anxiety disorder and a mood disorder are provided herein. The methods comprise administering to a subject in need thereof a therapeutically effective amount of one of the foregoing compounds or pharmaceutical compositions.
Other objects, features and advantages of the present invention will become apparent to those skilled in the art from the following description and claims. BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 provides the structures of exemplary compounds of the invention.
DETAILED DESCRIPTION OF THE INVENTION
Abbreviations and Definitions
The abbreviations used herein are conventional, unless otherwise defined.
The term "MCHR" refers to the melanin-concentrating hormone receptor protein 1 (MCHR1), unless otherwise stated.
The terms "treat", "treating" and "treatment" refer to a method of alleviating or abrogating a disease and/or its attendant symptoms.
The terms "prevent", "preventing" and "prevention" refer to a method of decreasing the probability or eliminating the possibility that a disease will be contracted.
As used herein, the term "MCHR-mediated condition or disorder" and the like refers to a condition or disorder characterized by inappropriate, e.g., less than or greater than normal, MCHR activity. An MCHR-mediated condition or disorder may be completely or partially mediated by inappropriate MCHR activity. However, an MCHR-mediated condition or disorder is one in which modulation of MCHR results in some effect on the underlying condition or disease (e.g., an MCHR antagonist results in some improvement in patient well-being in at least some patients). Exemplary
MCHR-mediated conditions and disorders include obesity, eating disorders and other behavioral disorders, such as anxiety disorders and mood disorders.
The term "therapeutically effective amount" refers to that amount of the compound being administered sufficient to prevent development of or alleviate to some extent one or more of the symptoms of the condition or disorder being treated.
As used herein, the term "obesity" refers to the excessive accumulation of body fat. Obesity may have genetic, environmental (e.g., expending less energy than is consumed) and regulatory determinants. Cardiovascular disorders, lipid disorders and metabolic disorders, such as hypertension, hyperlidemia, coronary artery disease and diabetes, are commonly associated with obesity.
As used herein, the terms "eating disorder", "feeding disorder", and the like refer to an emotional and/or behavioral disturbance associated with an excessive decrease in body weight and/or inappropriate efforts to avoid weight gain, e.g., fasting, self-induced vomiting, laxative or diuretic abuse. Depression is commonly associated with eating disorders. Exemplary eating disorders include anorexia nervosa and bulimia. As used herein, the term "anxiety disorder" refers to an emotional and/or behavioral disturbance characterized by persistent and pervasive worry or restlessness, tension or irritability about, e.g., health, work, money or family, for no clear reason. An anxiety disorder may be accompanied by tachycardia or dyspnea. Exemplary anxiety disorders include anxiety, generalized anxiety disorder, panic attacks, panic disorder and obsessive-compulsive disorder (OCD).
As used herein, the term "mood disorder" refers to an emotional and/or behavioral disturbance characterized by persistent and pervasive bouts of euphoria and/or depression. Exemplary mood disorders include depression and bipolar disorders. Anxiety is frequently associated with mood disorders, such as depression. The term "modulate" refers to the ability of a compound to increase or decrease the function, or activity, of MCHR. Modulation, as described herein, includes the antagonism or agonism of MCHR, either directly or indirectly. Antagonist are compounds that, e.g., partially or totally block stimulation, decrease, prevent, delay activation, inactivate, inhibit, desensitize, or down-regulate signal transduction. Agonists are compounds that, e.g., stimulate, increase, activate, open, facilitate, enhance activation, sensitize or up-regulate signal transduction.
The term "alkyl," by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which is fully saturated, having the number of carbon atoms designated (i.e. Cι-C8 means one to eight carbons). Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n- pentyl, n-hexyl, n-heptyl, n-octyl, and the like. As used herein, (C1-C8)alkyl refers to an alkyl group having from one to eight carbon atoms and includes, e.g., (C1-C4)alkyl. The term "alkenyl", by itself or as part of another substituent, means a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be mono- or polvunsaturated, having the number of carbon atoms designated (i.e. C2-C8 means two to eight carbons) and one or more double bonds. Examples of alkenyl groups include vinyl, allyl, 2-propenyl, crotyl, 2-isopentenyl, 2- (butadienyl), 2,4-pentadienyl, 3-(l,4-pentadienyl) and higher homologs and isomers thereof. The term "alkynyl", by itself or as part of another substituent, means a straight or branched chain hydrocarbon radical, or combination thereof, which may be mono- or polvunsaturated, having the number of carbon atoms designated (i.e. C2-C8 means two to eight carbons) and one or more triple bonds. Examples of alkynyl groups include ethynyl, 1- and 3-propynyl, 3-butynyl and higher homologs and isomers thereof.
The term "alkylene" by itself or as part of another substituent means a divalent radical derived from alkyl, as exemplified by -CH2CH2CH CH2-. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. A "lower alkyl" or "lower alkylene" is a shorter chain alkyl or alkylene group, generally having seven or fewer carbon atoms.
The terms "alkoxy," "alkylamino" and "alkylthio" (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.
The term "heteroalkyl," by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, Si and S, wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. The heteroatom Si may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Examples include -CH2-CH2- O-CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, -CH2-S-CH2-CH3, -CH2-CH2,- S(O)-CH3, -CH2-CH2-S(O)2-CH3, -CH=€H-O-CH3, -Si(CH3)3, -CH2-CH=N-OCH3, and -CH=CH-N(CH3)-CH3. Up to two heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3 and -CH2-O-Si(CH3)3
Similarly, the term "heteroalkylene" by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified by -CH2- CH2-S-CH2CH2- and -CH2-S-CH2-CH2-NH-CH2-. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied.
The terms "cycloalkyl" and "heterocycloalkyl", by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of "alkyl" and "heteroalkyl", respectively. Accordingly, a cycloalkyl group has the number of carbon atoms designated (i.e., C3-C8 means three to eight carbons) and a heterocycloalkyl group consists of the number of atoms designated (i.e., C2-C8 means two to eight carbons) and from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include 1 -(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3- piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like. The terms "halo" and "halogen," by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as "haloalkyl," are meant to include alkyl substituted with halogen atoms, which can be the same or different, in a number ranging from one to (2m' + 1), where m' is the total number of carbon atoms in the alkyl group. For example, the term "halo(C1-C4)alkyl" is mean to include trifluoromethyl, 2,2,2- trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Thus, the term "haloalkyl" includes monohaloalkyl (alkyl substituted with one halogen atom) and polyhaloalkyl (alkyl substituted with halogen atoms in a number ranging from two to (2m' + 1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group). Accordingly, the term "fluoro(C1-C4) alkyl" includes fluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 1,1-difluoroethyl, and the like. The term "perhaloalkyl" means, unless otherwise stated, alkyl substituted with (2m' + 1) halogen atoms, where m' is the total number of carbon atoms in the alkyl group. For example the term "perhalo(C1-C4)alkyl" is meant to include trifluoromethyl, pentachloroethyl, l,l,l-trifluoro-2-bromo-2-chloroethyl, and the like.
The term "aryl" means, unless otherwise stated, a polvunsaturated, typically aromatic, hydrocarbon substituent which can be a single ring or multiple rings (up to three rings) which are fused together or linked covalently. Non-limiting examples of aryl groups include phenyl, 1-naphthyl, 2-naphthyl and 4-biphenyl.
The term "heteroaryl" refers to aryl groups (or rings) that contain from zero to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule through a heteroatom. Non-limiting examples of heteroaryl groups include 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4- pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5- indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl and 6- quinolyl.
For brevity, the term "aryl" when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, the term "arylalkyl" is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl, and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(l-naphthyloxy)propyl, and the like).
Each of the above terms (e.g., "alkyl," "heteroalkyl," "aryl" and "heteroaryl") is meant to include both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below. Substituents for the alkyl and heteroalkyl radicals (as well as those groups referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl and heterocycloalkenyl) can be a variety of groups selected from: -OR', =O, =NR', =N-OR', -NR'R", -SR', halogen, -SiR'R"R'", -OC(O)R', -C(O)R', -CO2R', -CONR'R", -OC(O)NR'R", -NR"C(O)R', -NR'-
C(O)NR"R'", -NR'-SO2NR"R'", -NR"CO2R', -NH-C(NH2)=NH, -NR'C(NH2)=NH, - NH-C(NH2)=NR', -S(O)R', -SO2R', -SO2NR'R", -NR"SO2R', -CN and -NO2, in a number ranging from zero to three, with those groups having zero, one or two substituents being particularly preferred. R', R" and R'" each independently refer to hydrogen, unsubstituted (C1-C8)alkyl and heteroalkyl, fluoro(Ci-C4)alkyl, unsubstituted aryl, aryl substituted with one to three halogens, unsubstituted alkyl, alkoxy or thioalkoxy groups, or aryl(Cι-C4)alkyl groups. When R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6- or 7- membered ring. For example, -NR'R" is meant to include 1-pyrrolidinyl and 4- morpholinyl. Typically, an alkyl or heteroalkyl group will have from zero to three substituents, with those groups having two or fewer substituents being preferred in the present invention. More preferably, an alkyl or heteroalkyl radical will be unsubstituted or monosubstituted. Most preferably, an alkyl or heteroalkyl radical will be unsubstituted. From the above discussion of substituents, one of skill in the art will understand that the term "alkyl" is meant to include groups such as trihaloalkyl (e.g., - CF3 and -CH2CF3).
Preferred substituents for the alkyl and heteroalkyl radicals are selected from: -OR', =O, -NR'R", -SR', halogen, -SiR'R"R'", -OC(O)R', -C(O)R', -CO2R', - CONR'R", -OC(O)NR'R", -NR"C(O)R', -NR"CO2R', -NR'-SO2NR"R'", -S(O)R', - SO2R\ -SO2NR'R", -NR"SO2R', -CN and -NO2, where R' and R" are as defined above. Further preferred substituents are selected from:'-OR', =O, -NR'R", halogen, - OC(O)R', -CO2R', -CONR'R", -OC(O)NR'R", -NR"C(O)R', -NR"CO2R', -NR'- SO2NR"R'", -SO2R', -SO2NR'R", -NR"SO2R', -CN and -NO2
Similarly, substituents for the aryl and heteroaryl groups are varied and selected from: halogen, -OR', -OC(O)R\ -NR'R", -SR', -R', -CN, -NO2, -CO2R', -CONR'R", -C(O)R', -OC(O)NR'R", -NR"C(O)R', -NR"CO2R', -NR'-C(O)NR"R'", - NR'-SO2NR"R'", -NH-C(NH2)=NH, -NR'C(NH2)=NH, -NH-C(NH2)=NR', -S(O)R', - SO2R', -SO2NR'R", -NR"SO2R\ -N3, -CH(Ph)2, perfluoro(C1-C4)alkoxy and perfluoro(C1-C )alkyl, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R', R" and R'" are independently selected from hydrogen, (Cι-C8)alkyl and heteroalkyl, unsubstituted aryl and heteroaryl, (unsubstituted aryl)-(C1-C4)alkyl and (unsubstituted aryl)oxy-(C1-C4)alkyl. Typically, an aryl or heteroaryl group will have from zero to three substituents, with those groups having two or fewer substituents being preferred in the present invention. More preferably, an aryl or heteroaryl group will be unsubstituted or monosubstituted. Most preferably, an aryl or heteroaryl group will be unsubstituted.
Preferred substituents for aryl and heteroaryl groups are selected from: halogen, -OR', -OC(O)R', -NR'R", -SR', -R', -CN, -NO2, -CO2R', -CONR'R", -C(O)R', -OC(O)NR'R", -NR"C(O)R', -S(O)R', -SO2R', -SO2NR'R", -NR"SO2R', - N3, -CH(Ph)2, perfluoro(Cι.-C )alkoxy and perfluoro(C1-C4)alkyl, where R' and R" are as defined above. Further preferred substituents are selected from: halogen, -OR', - OC(O)R', -NR'R", -R', -CN, -NO2, -CO2R', -CONR'R", -NR"C(O)R', -SO2R', -SO2NR'R", -NR"SO2R', perfluoro(C C4)alkoxy and perfluoro(C C4)alkyl As used herein, the substituent -CO2H, includes bioisosteric replacements therefor, such as:
Figure imgf000016_0001
and the like. See, e.g., The Practice of Medicinal Chemistry; Wermuth, C.G., Ed.; Academic Press: New York, 1996; p. 203.
Two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -T-C(O)-(CH2)q-U-, wherein T and U are independently -NH-, -O-, -CH2- or a single bond, and q is an integer of from 0 to 2. Alternatively, two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -A-(CH2)r-B-, wherein A and B are independently -CH2-, -O-, -NH-, -S-, -S(O)-, -SO2-, -SO2NR'- or a single bond, and r is an integer of from 1 to 3. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula -(CH2)s-X-(CH )t-, where s and t are independently integers of from 0 to 3, and X is -O-, -NR'-, -S-, -S(O)-, -SO2-, or -SO2NR'-. The substituent R' in -NR'- and -SO NR'- is selected from the group consisting of hydrogen or unsubstituted (Q- C6)alkyl.
As used herein, the term "heteroatom" is meant to include oxygen (O), nitrogen (N), sulfur (S) and silicon (Si).
The term "pharmaceutically acceptable salts" is meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of phaπnaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbomc, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge, S.M., et al. (1977) J. Pharm. Sci. 66:1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts. The neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
In addition to salt forms, the present invention provides compounds which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention. Additionally, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmacological compositions over the parent drug. A wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. An example, without limitation, of a prodrug would be a compound of the present invention which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound of the invention.
Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, enantiomers, diastereomers, geometric isomers and individual isomers are all intended to be encompassed within the scope of the present invention.
The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention. Embodiments of the Invention
MCHR (GenBank Accession No. U71092) is expressed in brain, at moderate levels in the eye and skeletal muscle, and in low levels in tongue and the pituitary gland. Evidence suggests that MCHR is involved in, ter alia, olfactory learning, regulation of feeding behavior and energy metabolism, regulation of the hypothalmic-pituitary-adrenocortical axis following stress, arousal and the sensation of anxiety (Saito et al, TEM 11(8):299-303 (2000)). The compounds of the present invention inhibit MCHR activity, and thus, are useful in, for example, the treatment or prevention of disorders associated with these processes.
Compounds
In one aspect, the present invention provides compounds represented by the formula (I):
Figure imgf000019_0001
I wherein or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof. In formula I,
Figure imgf000019_0002
represents a single or fused aryl or heteroaryl ring. For instance, can represent benzene, naphthalene, pyrrole, pyrazole, imidazole, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, pyridine, pyrimidine, benzothiazole, purine, benzimidazole, indole, isoquinoline, quinoxaline or quinoline ring. In preferred
embodiments,
Figure imgf000019_0003
represents benzene. The symbol Q represents -N(R)- or -N(R)-(C1-C3)alkylene-. In certain embodiments the symbol Q represents -N(R)-. The symbol R represents
Figure imgf000020_0001
.
The symbol L1 represents a divalent linkage selected from a bond, (C\- C4)alkylene, (Cι-C )alkylenoxy and ( -C^alkylenamino. Exemplary L1 groups are a single bond, methylene, ethylene, n-propylene and n-butylene. The symbol L2 represents a divelent linkage selected from a bond, (Ci-C )alkylene, (C2-C4)alkenylene, (C2-C )alkynylene, (Cι-C4)alkylenoxy and (Cι-C4)alkylenamino. Exemplary L2 groups are a a single bond, methylene, ethylene, n-propylene and n-butylene.
The letters X and Y represent independently (Cι-C8)alkyl, (C2- C8)alkenyl, (C2-C8)alkynyl, -CO2R13 or -C(O)NR13R14. Optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from N, O and S.
The letter Z represents -OR12, -NR12R13, -CO2R12, -R15, -C(O)NR1 R13, -C(O)NR12R15, -SO2NR12R13, -NR13SO2R15, -N(R12)N(R13)SO2R14, -C(O)N(R13)OR12, fluoro(C1-C4)alkyl, heteroaryl, -C(=NOR12)NR13R14, -C(R13)=NOR12, -NR13(OR12), -C(O)NR14C(O)NR12R13, -NR14C(O)NR13C(O)R12 and -NR14C(O)NR12R13. Exemplary -C(X)(Y)(L2Z) groups are:
Figure imgf000021_0001
R" is hydrogen or (C1-C8)alkyl.
Each R1 is independently halogen, (Cι-C8)alkyl, (C -C8)alkenyl, (C - C8)alkynyl, fluoro(C1-C4)alkyl, -OR5, -SR5, fluoro(Ct-C4)alkoxy, aryl, aryl(d- C4)alkyl, -NO2, -NR5R6, -C(O)R5, -CO2R5, -C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, -S(O)mR5, -CN or -N(R6)S(O)mR5. Exemplary R1 groups are Cl and CF3.
R and R are independently selected from hydrogen, halogen, (Ci- C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoiO(Cι-C )alkyl, -OR8, -SR8, fluoro(C1- C4)alkoxy, aryl, aryl(Cι -C4)alkyl, -NO2, -NR8R9, =O, -C(O)R8, -CO2R8, -C(O)NR8R9, - N(R9)C(O)R8, -N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9, -S(O)mR8, -CN and - N(R9)S(O)mR8. Exemplary R2 groups are methyl, isopropyl, trifluoromethyl, hydroxy, methoxy, hydroxymethyl, trifluoromethoxy, phenyl and =O. certain embodiments, R3 is hydrogen or =O.
R4 is hydrogen, -OR11, -C(O)Rπ, -CO2Rπ, -C(O)NRπR12, -CN, (Cr C4)alkyl or aryl. R5, R6, R7, R8, R9, R10, R1 ! , R12, R13, R14, R16 and R17 are independently selected from hydrogen, (Cι-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(Cι- C4)alkyl, hetero(C1-C )alkyl, aryl and aryl(Cι-C )alkyl and R18 is a 5- or 6-membered ring containing from 1 to 3 heteroatoms selected from N, O and S. The subscript m is 1 or 2 and the subscript n is 0, 1 or 2. Optionally, when two R groups selected from the group consisting of R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16, R17 and R18 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from N, O and S.
In particular embodiments,
Figure imgf000022_0001
represents a benzene ring, R" is hydrogen and R3 is hydrogen.
In another aspect, the present invention provides compounds of formula (II):
Figure imgf000022_0002
II or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof. In formula II, the symbol L1 represents a divalent linkage selected from a bond, (C\- C4)alkylene, (Cι-C4)alkylenoxy and ( -C^alkylenamino. Exemplary L1 groups are a single bond, methylene, ethylene, n-propylene and n-butylene. The symbol L2 represents a divalent linkage selected from a bond, ( -C^alkylene, (C2-C4)alkenylene, (C2-C4)alkynylene, (Cι-C4)alkylenoxy and (C1-C4)alkylenamino. Exemplary L2 groups are a single bond, methylene, ethylene, n-propylene and n-butylene.
The letters X and Y represent independently (C1-C8)alkyl, (C2- C8)alkenyl, (C2-C8)alkynyl, -CO2R13 or -C(O)NR13R14. Optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from N, O and S.
The letter Z represents -OR12, -NR12R13, -CO2R12, -R15, -C(O)NR12R13, -C(O)NR12R15, -SO2NR12R13, -NR13SO2R15, -N(R12)N(R13)SO2R14, -C(O)N(R13)OR12, fluoro(C1-C4)alkyl, heteroaryl, -C(=NOR12)NR13R14, -C(R13)=NOR12, -NR13(OR12), -C(O)NR14C(O)NR12R13, -NR14C(O)NR13C(O)R12 and -NR14C(O)NR12R13. Exemplary -C(X)(Y)(L2Z) groups are:
Figure imgf000023_0001
R" is hydrogen or (Q-Q alkyl.
Each R1 is independently halogen, ( -C^alkyl, (C2-C8)alkenyl, (C2- C8)alkynyl, fluoro(C1-C4)alkyl, -OR5, -SR5, fluoro(C1-C4)alkoxy, aryl, aryl(Cr C4)alkyl, -NO2, -NR5R6, -C(O)R5, -CO2R5, -C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, -S(O)mR5, -CN or -N(R6)S(O)mR5. Exemplary R1 groups are Cl and CF3.
R2 is selected from hydrogen, halogen, ( -Cs^lkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, -OR8, -SR8, fluoro(C1-C4)alkoxy, aryl, aryl(C C4)alkyl, -NO2, -NR8R9, =O, -C(O)R8, -CO2R8, -C(O)NR8R9, -N(R9)C(O)R8, - N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9, -S(O)mR8, -CN and -N(R9)S(O)mR8. Exemplary R2 groups are methyl, isopropyl, trifluoromethyl, hydroxy, methoxy, hydroxymethyl, trifluoromethoxy, phenyl and =O. R4 is hydrogen, -OR11, -C(O)Rπ, -CO2Rn, -C(O)NRuR12, -CN, (Cr
C4)alkyl or aryl.
R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R16 and R17 are independently selected from hydrogen, (Cι-Cs)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(Cι- C4)alkyl, hetero(C1-C4)alkyl, aryl and aryl(Cι-C4)alkyl and R18 is a 5- or 6-membered ring containing from 1 to 3 heteroatoms selected from N, O and S. The subscript m is 1 or 2 and the subscript n is 0, 1 or 2. Optionally, when two R groups selected from the group consisting of R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16, R17 and R18 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from N, O and S.
Compounds of the invention feature a pyrido[4,3-b]carbazole-derived ring, minimally substituted at the 2- and 11 -positions. The ring numbering system used herein is illustrated below.
Figure imgf000024_0001
II
One of skill in the art will understand that formula II encompasses two enantiomers. The enantiomers have the structural orientations represented by the following formulae:
Figure imgf000025_0001
Within formula I or II above, a number of groups of embodiments are preferred, described below.
In one group of preferred embodiments, L1 is (Cι-C4)alkylene. In a prefeπed embodiment, L1 is unsubstituted ( -G alkylene or -(CH2)p- wherein the subscript p is an integer of from 1 to 4. In a further preferred embodiment, p is 1, 2 or 3. In a still further prefeπed embodiment, p is 2 or 3. In a particularly prefeπed embodiment, p is 2.
One group of prefeπed embodiments is represented by the formula (III):
Figure imgf000025_0002
III.
In a prefeπed embodiment, X and Y are combined to form a 3-, 4-, 5-,
6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S.
In a further prefeπed embodiment, X and Y are combined to form a 5- or 6-membered ring containing from 0 to 2 heteroatoms selected from O, N and S. In a particularly prefeπed embodiment, X and Y are combined to form a 5- or 6-membered ring containing 0 heteroatoms, 1 nitrogen atom or 1 oxygen atom.
In another prefeπed embodiment, L is a bond and Z is -CO2R or -CO2NR15R16. In another group of prefeπed embodiments, R" is hydrogen.
In another group of prefeπed embodiments, R" is substituted ( - C8)alkyl. In a prefeπed embodiment, R" is ( -C^alkyl substituted with hydroxy, alkylamino (e.g., — NHMe) or carboxy (-CO2H). In a particularly preferred embodiment, R" is (C -C8)alkyl substituted with hydroxy, alkylamino or carboxy. In another group of prefeπed embodiments, R1 is independently halogen, (CrC4)alkyl, fluoro(CrC4)alkyl, -OR5, fluoro(CrC4)alkoxy, -CO2R5, - S(O)mNR5R6, -S(O)mR5 or -CN. In a further prefeπed embodiment, R1 is independently halogen or fluoro(C1-C )alkyl. In a still further prefeπed embodiment, R1 is halogen or fluoro(Cι-C4)alkyl and the subscript n is 0 or 1. h a particularly prefeπed embodiment, R1 is fluoro(C1-C4)alkyl and the subscript n is 0 or 1.
In another group of prefeπed embodiments, R2 is (CrC4)alkyl or aryl. In another group of prefeπed embodiments, R4 is hydrogen.
Also particularly prefeπed are those embodiments that combine two or more of these prefeπed groups. Accordingly, in one group of particularly prefeπed embodiments, R" and R4 are hydrogen.
In another group of particularly prefeπed embodiments, R" and R4 are hydrogen and R2 is (Cι-C )alkyl or aryl.
In another group of particularly prefeπed embodiments, R" and R4 are hydrogen, R1 is independently halogen, (CrC4)alkyl, fluoro(C1-C4)alkyl, -OR5, fluoro(CrC4)alkoxy, -CO2R5, -S(O)mNR5R6, -S(O)mR5 or-CN and R2 is (d-C4)alkyl or aryl. In a particularly prefeπed embodiment, R" and R4 are hydrogen, R1 is halogen or fluoro(C1-C )alkyl, n is 1 and R2 is (Ci-C^alkyl or aryl. In a more particularly prefeπed embodiment, R" and R are hydrogen, R is fluoro(Cι-C4)alkyl, n is 1 and R is (d-C )alkyl or aryl.
One group of particularly prefeπed embodiments is represented by the formula (IV):
Figure imgf000026_0001
IV wherein p, R1, R2, L2, X, Y and Z have the meanings and prefeπed groupings provided above.
In a particularly preferred embodiment, X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S, L2 is a bond and Z is -CO2R15 or -CO2NR15R16.
In another particularly preferred embodiment, X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S and R1 is independently halogen, (Q-C^alkyl, fluoro(CrC4)alkyl, - OR5, fluoro(CrC4)alkoxy, -CO2R5, -S(O)mNR5R6, -S(O)mR5 or-CN.
In another particularly prefeπed embodiment embodiment, X and Y are combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S and R2 is (C C4)alkyl or aryl. In another particularly prefeπed embodiment embodiment, X and Y are combined to form a 3-, A-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from O, N and S, R1 is independently halogen, (CrC4)alkyl, fluoro(CrC4)alkyl, -OR5, fluoro(CrC4)alkoxy, -CO2R5, -S(O)mNR5R6, -S(O)mR5 or- CN and R2 is (CrC4)alkyl or aryl. In another particularly prefeπed embodiment embodiment, R1 is independently halogen, (CrC4)alkyl, fluoro(CrC )alkyl, -OR5, fluoro(C1-C4)alkoxy, - CO2R5, -S(O)mNR5R6, -S(O)mR5 or -CN and R2 is (CrC4)alkyl or aryl.
In another particularly prefeπed embodiment embodiment, R1 is independently halogen, (CrC4)alkyl, fluoro(C1-C4)alkyl, -OR5, fluoro(CrC4)alkoxy, - CO2R5, -S(O)mNR5R6, -S(O)mR5 or -CN, L2 is a bond and Z is -CO2R15 or -CO2NR15R16.
In another particularly prefeπed embodiment embodiment, R2 is (Cr C4)alkyl or aryl, L2 is a bond and Z is -CO2R15 or -CO2NR15R16.
In another particularly prefeπed embodiment embodiment, R1 is independently halogen,
Figure imgf000027_0001
fluoro(C1-C4)alkyl, -OR5, fluoro(CrC4)alkoxy, - CO2R5, -S(O)mNR5RD, -S(O)mR5 or -CN, R2 is (CrC4)alkyl or aryl, L2 is a bond and Z is -CO2R15 or -CO2NR15R16.
In a particular embodiment, the present invention provides the following compounds:
Figure imgf000028_0001
In further particular embodiments, the present invention provides pharmaceutically acceptable salts of the above compounds. For example, in a certain embodiment the present invention provides benzenesulfonic acid salts of the above compounds.
Compositions
In another aspect, the present invention provides pharmaceutical compositions comprising one or more compounds of the invention in combination with a diagnostically or pharmaceutically acceptable carrier or excipient. The subject compositions are useful for treating or preventing conditions and disorders mediated by MCHR, such as obesity and eating disorders, e.g., anorexia nervosa. The compounds of the present invention can be prepared and administered in a wide variety of oral and parenteral dosage forms. Thus, the compounds of the present invention can be administered by injection, for example, intravenously, intramuscularly, intracutaneously, subcutaneously, intraduodenally or inxraperitoneally. Also, the compounds described herein can be administered by inhalation, for example, intranasally. Additionally, the compounds of the present invention can be administered transdermally. Other routes of administration are also contemplated for use with the compounds of the present invention, including depot administration and rectal administration.
For preparing pharmaceutical compositions from the compounds of the present invention, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances which may also act as diluents, flavoring agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material. In powders, the carrier is a finely divided solid which is in a mixture with the finely divided active component. In tablets, the active component is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
The powders and tablets preferably contain from about 5% or 10% to 70% of the active compound. Suitable carriers are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like. The term "preparation" is intended to include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component with or without other carriers, is suπounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid dosage forms suitable for oral administration.
For preparing suppositories, a low melting wax, such as a mixture of fatty acid glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
Liquid form preparations include solutions, suspensions and emulsions, for example, water or water/propylene glycol solutions. For parenteral injection, liquid preparations can be formulated in solution in aqueous polyethylene glycol solution. Aqueous solutions suitable for oral use can be prepared by dissolving the active component in water and adding suitable colorants, flavors, stabilizers, and thickening agents as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, and other well-known suspending agents. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
The pharmaceutical preparation is preferably in unit dosage form. In such form the preparation is subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
The quantity of active component in a unit dose preparation may be varied or adjusted from 0.1 mg to 1000 mg, preferably 1.0 mg to 100 mg according to the particular application and the potency of the active component. The composition can, if desired, also contain other compatible therapeutic agents.
In therapeutic use for the treatment or prevention of conditions and disorders associated with MCHR, the compounds utilized in the pharmaceutical method of the invention are administered at the initial dosage of about 0.001 mg/kg to about 100 mg/kg daily. A daily dose range of about 0.1 mg/kg to about 10 mg/kg is prefeπed. The dosages, however, may be varied depending upon the requirements of the patient, the severity of the condition being treated and the compound being employed. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day, if desired.
The compositions may be advantageously combined and/or used in combination with agents useful in the treatment and/or prevention of obesity and eating disorders and pathologies associated therewith (e.g., cardiovascular disease and hypertension). In many instances, administration of the subject compounds or compositions in conjunction with these alternative agents enhances the efficacy of such agents. Accordingly, in some instances, the present compounds, when combined or administered in combination with, e.g., anti-obesity agents, can be used in dosages which are less than the expected amounts when used alone, or less than the calculated amounts for combination therapy.
Suitable agents for combination therapy include those that are cuπently commercially available and those that are in development or will be developed. Exemplary agents useful in the treatment of obesity include β3 adrenergic receptor agonists, leptin or derivatives thereof and neuropeptide Y antagonists. Exemplary agents useful in the treatment of anxiety and/or mood disorders include benzodiazepines, e.g., alprazolam, chlordiazepoxide, clonazepam, clorazepate, diazepam, lorazepam, oxazepam, and the like; heterocyclic antidepressants, e.g, amitriptyline, nortriptyline, imipramine, desipramine, doxepin, trimipramine, clomipramine, protryptyline, amoxapine and maprotiline; monoamine oxidase inhibitors (MAOIs), e.g., phenelzine and tranylcypromine; serotonin reuptake inhibitors (SRIs); selective serotonin reuptake inhibitors (SSRIs), e.g., fluoxetine, fluvoxamine, paroxetine and sertraline; serotonergic-noradrenergic antidepressants, e.g., venlafaxine; 5-HT2 antagonists, e.g., trazadone, nefazodone and mirtazapine; and catecholaminergic antidepressants, e.g., buproprion.
Methods of Use
In yet another aspect, the present invention provides methods of using one or more compounds of the invention to treat or prevent a condition or disorder associated with eating behavior, energy homeostasis or anxiety. Exemplary conditions and disorders associated with eating behavior, energy homeostasis and anxiety include eating disorders, such as anorexia nervosa and bulimia, obesity, anxiety disorders, e.g., generalized anxiety disorder, panic attacks, panic disorder and obsessive-compulsive disorder (OCD), and mood disorders, e.g., depression and bipolar disorders. Methods of using a compound of the invention to treat or prevent a condition or disorder associated with eating behavior include methods of modifying eating behavior or food intake, for example, stimulating or suppressing eating behavior or increasing or decreasing food intake. The methods comprise administering to a subject in need thereof a therapeutically effective amount of a compound of the invention.
In another aspect, the present invention provides methods of using a compound of the invention to treat or prevent a condition or disorder mediated by MCHR. The methods comprise administering to a subject in need thereof a therapeutically effective amount of a compound of the invention.
In still another aspect, the present invention provides methods of using a compound of the invention to modulate MCHR. The methods comprise contacting a cell with a compound of the invention.
The compounds of the invention may also modulate G-protein coupled receptors related to MCHR, e.g., MCHR2 (see International Publication Nos. WO 00/49046 and WO 01/07606).
Preparation of the Compounds
The present invention provides a process for the preparation of a compound of formula I.
Scheme 1
Figure imgf000032_0001
A general synthetic route is depicted in Scheme 1, which outlines the condensation of substituted aryl moiety A, with a bicyclic structure B to produce a compound of formula C, wherein the variables are as defined as above. In formula A, D1 is hydrogen, halogen, -C(O)R7, -CO2R8 or -C(O)NR5R6, wherein R5, R6, R7 and R8 are defined as above, and D2 is a bond, -N(R")-, -N(protecting group)-, -S- or -O-, wherein R" is defined as above and protecting group is an amino protecting group. Conventional amino protecting groups consist of known groups which are used to protectively block an amino group during the synthesis procedures described herein. These conventional blocking groups are readily removable, i.e., they can be removed, if desired, by procedures which will not cause cleavage or other disruption of the remaining portions of the molecule. Suitable protecting groups for the compounds of the present invention will be recognized from the present application taking into account the level of skill in the art, and with reference to standard textbooks, such as Greene, T. W. et al,. Protective Groups in Organic Synthesis, Wiley, New York (1991). In foπnula B, E1 is hydrogen, -C(O)R7, -CO2R8 or -C(O)NR5R6, wherein
R5, R6, R7 and R8 are defined as above, and E2 is =O or -NR5R6, wherein R5 and R6 are
1 9 defined as above. When a compound of formula A, wherein D is hydrogen and D is - N(R")- or -N(protecting group)-, -S-, or -O-, reacts with a compound of formula B, wherein E1 is hydrogen and E2 is =O or a protected version thereof (e.g., an acetal), under the typical Fisher indolization conditions, a compound of formula C is produced. One of skill in the art will understand that the synthesis provided above can be modified to use different starting materials and alternate reagents to accomplish the desired transfonnations. For example, a compound of formula A, wherein D1 is a leaving group such as Cl, Br, I or toluenesulfonate, can react with a compound of formula B, wherein E2 is =O or a protected version thereof, via a palladium-catalyzed coupling reaction to produce a compound of formula C. Also, a compound of formula
• 1 • ■ 9
A, wherein D is a leaving group and D is a nitro group, can react with a compound of formula B, wherein E1 is CO2R and wherein E2 is =O or a protected version thereof, to produce a compound of formula C. Accordingly, the synthesis and reagents described herein are all expressed as non-limiting embodiments.
Materials represented by formula A are available commercially (Aldrich Chemical), or can be obtained synthetically following literature procedures.
One way to prepare compounds represented by formula B is by the Robinson annulation process between a cyclic ketone and a substituted enone followed by saturation of the double bond. One of the skill in the art will readily appreciate that other methods are available. The relative stereochemistry and absolute stereochemistry can be controlled in the process. The individual forms of compounds of formula B, e.g., diastereomers and enantiomers, can be formed by stereoconxrolled reactions, or may be separated, e.g., by chromatographic techniques (diastereomers) and by resolution (enantiomers).
Analysis of the Compounds
The activity of MCHR polypeptides can be assessed using a variety of in vitro and in vivo assays to determine functional, chemical and physical effects, e.g., measuring ligand binding (e.g., radioactive ligand binding), second messenger (e.g., cAMP, cGMP, IP3, DAG, or Ca2+) levels, ion flux, phosphorylation levels, transcription levels, neurotransmitter levels, and the like. Furthermore, such assays can be used to test for antagonists and agonists of MCHR. Screening assays may be used to identify modulators that can be used as therapeutic agents, e.g., antagonists of MCHR activity. Modulators of MCHR activity can be tested using MCHR polypeptides as described above, either recombmant or naturally occurring (e.g., endogenous). The protein can be isolated, expressed in a cell, expressed in a membrane derived from a cell, expressed in tissue or in an animal, either recombinant or naturally occurring. For example, kidney cells, liver cells, colon cells, transformed cells, or membranes can be used. Modulation is tested using one of the in vitro or in vivo assays described herein. Signal transduction can also be examined in vitro with soluble or solid state reactions, using a chimeric molecule such as an extracellular domain of a receptor covalently linked to a heterologous signal transduction domain, or a heterologous extracellular domain covalently linked to the transmembrane and or cytoplasmic domain of a receptor. Gene amplification can also be examined. Furthermore, ligand-binding domains of the protein of interest can be used in vitro in soluble or solid state reactions to assay for ligand binding.
Ligand binding to MCHR, a domain, or chimeric protein can be tested in solution, in a bilayer membrane, attached to a solid phase, in a lipid monolayer, or in vesicles. Binding of a modulator can be tested using, e.g., changes in spectroscopic characteristics (e.g., fluorescence, absorbance, refractive index) hydrodynamic (e.g., shape), chromatographic, or solubility properties.
MCHR-G-protein interactions can also be examined, by, for example, analysis of binding of the G-protein to MCHR or its release from MCHR can be examined. For example, in the absence of GTP, an activator will lead to the formation of a tight complex of a G protein (all three subunits) with MCHR. This complex can be detected in a variety of ways, as noted above. Such an assay can be modified to search for antagonists. In one embodiment, an activator is added to MCHR and G protein in the absence of GTP, allowed to form a tight complex, and then screened for antagonists by looking at dissociation of the MCHR-G protein complex, hi the presence of GTP, release of the alpha subunit of the G protein from the other two G protein subunits serves as a criterion of activation. An activated or inhibited G-protein will in turn alter the properties of downstream effectors such as proteins, enzymes and channels. The classic examples are the activation of cGMP phosphodiesterase by transducin in the visual system, adenylate cyclase by the stimulatory G-protein, phospholipase C by Gq and other cognate G proteins, and modulation of diverse channels by Gi and other G proteins. Downstream consequences can also be examined such as generation of diacyl glycerol and IP3 by phospholipase C, and in turn, for calcium mobilization by IP3.
Activated MCHR becomes a substrate for kinases that phosphorylate the C-terminal tail of the receptor (and possibly other sites as well). Thus, activators will promote the transfer of P from gamma-labeled ATP to the receptor, which can be assayed with a scintillation counter. The phosphorylation of the C-terminal tail will promote the binding of arrestin-like proteins and will interfere with the binding of G- proteins. The kinase/arrestin pathway plays a key role in the desensitization of many GPCR receptors. For a general review of GPCR signal transduction and methods of assaying signal transduction, see, e.g., Methods in Enzymology, vols. 237 and 238 (1994) and volume 96 (1983); Bourne et al, Nature 10:349:117-27 (1991); Bourne et al, Nature 348:125-32 (1990); Pitcher et al, Annu. Rev. Biochem. 67:653-92 (1998).
Samples or assays that are treated with a potential MCHR antagonist or agonist are compared to control samples without the test compound, to examine the extent of modulation. Control samples (untreated with agonists or antagonist) are assigned a relative MCHR activity value of 100. Inhibition of MCHR is achieved when the MCHR activity value relative to the control is about 90%, optionally 50%, optionally 25-0%. Activation of MCHR is achieved when the MCHR activity value relative to the control is 110%, optionally 150%, 200-500%, or 1000-2000%.
Changes in ion flux may be assessed by determining changes in polarization (i.e., electrical potential) of the cell or membrane expressing MCHR. One means to determine changes in cellular polarization is by measuring changes in cuπent (thereby measuring changes in polarization) with voltage-clamp and patch-clamp techniques, e.g., the "cell-attached" mode, the "inside-out" mode, and the "whole cell" mode (see, e.g., Ackerman et al, New Engl J. Med. 336:1575-1595 (1997)). Whole cell cuπents are conveniently determined using the standard methodology (see, e.g., Hamil et al, PFlugers. Archiv. 391:85 (1981). Other known assays include radiolabeled ion flux assays and fluorescence assays using voltage-sensitive dyes (see, e.g., Nestergaπd-Bogind et al, J. Membrane Biol 88:67-75 (1988); Gonzales & Tsien, Chem. Biol. 4:269-277 (1997); Daniel et al, J. Pharmacol. Meth. 25:185-193 (1991); Holevinsky et al, J. Membrane Biology 137:59-70 (1994)). Generally, the compounds to be tested are present in the range from 1 pM to 100 mM. The effects of the test compounds upon the function of the polypeptides can be measured by examining any of the parameters described above. Any suitable physiological change that affects MCHR activity can be used to assess the influence of a test compound on the polypeptides of this invention. When the functional consequences are determined using intact cells or animals, one can also measure a variety of effects such as transmitter release, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as Ca , 1P3 or cAMP.
Preferred assays for MCHR include cells that are loaded with ion- or voltage-sensitive dyes to report receptor activity. Assays for determining activity of such receptors can also use known agonists and antagonists for other G-protein coupled receptors as negative or positive controls to assess activity of tested compounds, h assays for identifying modulatory compounds (e.g., agonists, antagonists), changes in the level of ions in the cytoplasm or membrane voltage will be monitored using an ion- sensitive or membrane voltage fluorescent indicator, respectively. Among the ion- sensitive indicators and voltage probes that may be employed are those disclosed in the Molecular Probes 1997 Catalog. For G-protein coupled receptors, promiscuous G- proteins such as Gαl5 and Gαl6 can be used in the assay of choice (Wilkie et al, Proc. NatlAcad. Set USA 88:10049-10053 (1991)). Such promiscuous G-proteins allow coupling of a wide range of receptors to signal transduction pathways in heterologous cells. Receptor activation typically initiates subsequent intracellular events, e.g., increases in second messengers such as IP3, which releases intracellular stores of calcium ions. Activation of some G-protein coupled receptors stimulates the formation of inositol triphosphate (IP3) through phospholipase C-mediated hydrolysis of phosphatidylinositol (Berridge & Irvine, Nature 312:315-21 (1984)). IP3 in turn stimulates the release of intracellular calcium ion stores. Thus, a change in cytoplasmic calcium ion levels, or a change in second messenger levels such as IP3 can be used to assess G-protein coupled receptor function. Cells expressing such G-protein coupled receptors may exhibit increased cytoplasmic calcium levels as a result of contribution from both intracellular stores and via activation of ion channels, in which case it may be desirable although not necessary to conduct such assays in calcium-free buffer, optionally supplemented with a chelating agent such as EGTA, to distinguish fluorescence response resulting from calcium release from internal stores.
Other assays can involve determining the activity of receptors which, when activated, result in a change in the level of intracellular cyclic nucleo tides, e.g., cAMP or cGMP, by activating or inhibiting downstream effectors such as adenylate cyclase. There are cyclic nucleotide-gated ion channels, e.g., rod photoreceptor cell channels and olfactory neuron channels that are permeable to cations upon activation by binding of cAMP or cGMP (see, e.g., Altenhofen et al, Proc. Natl Acad. Sci. U.S.A. 88:9868-9872 (1991) and Dhallan et α/., Nature 347:184-187 (1990)). In cases where activation of the receptor results in a decrease in cyclic nucleotide levels, it may be preferable to expose the cells to agents that increase intracellular cyclic nucleotide levels, e.g., forskolin, prior to adding a receptor-activating compound to the cells in the assay. Cells for this type of assay can be made by co-transfection of a host cell with DNA encoding a cyclic nucleotide-gated ion channel, GPCR phosphatase and DNA encoding a receptor (e.g., certain glutamate receptors, muscarinic acetylcholine receptors, dopamine receptors, serotonin receptors, and the like), which, when activated, causes a change in cyclic nucleotide levels in the cytoplasm. In one embodiment, changes in intracellular cAMP or cGMP can be measured using immunoassays. The method described in Offermanns & Simon, J. Biol. Chem. 270:15175-15180 (1995) maybe used to determine the level of cAMP. Also, the method described in Felley-Bosco et al, Am. J. Resp. Cell andMol Biol. 11:159-164 (1994) may be used to determine the level of cGMP. Further, an assay kit for measuring cAMP and/or cGMP is described in U.S. Patent No. 4,115,538, herein incorporated by reference. In another embodiment, phosphatidyl inositol (PI) hydrolysis can be analyzed according to U.S. Patent No. 5,436,128, herein incorporated by reference. In another embodiment, transcription levels can be measured to assess the effects of a test compound on signal transduction. A host cell containing the protein of interest is contacted with a test compound for a sufficient time to effect any interactions, and then the level of gene expression is measured. The amount of time to effect such interactions may be empirically determined, such as by running a time course and measuring the level of transcription as a function of time. The amount of transcription may be measured by using any method known to those of skill in the art to be suitable. For example, mRNA expression of the protein of interest may be detected using northern blots or their polypeptide products may be identified using immunoassays. Alternatively, transcription based assays using a reporter gene may be used as described in U.S. Patent No. 5,436,128, herein incorporated by reference. The reporter genes can be, e.g., chloramphenicol acetyltransferase, firefly luciferase, bacterial luciferase, β-galactosidase and alkaline phosphatase. Furthermore, the protein of interest can be used as an indirect reporter via attachment to a second reporter such as green fluorescent protein (see, e.g., Mistili & Spector, Nature Biotechnology 15:961- 964 (1997)).
The amount of transcription is then compared to the amount of transcription in either the same cell in the absence of the test compound, or it may be compared with the amount of transcription in a substantially identical cell that lacks the protein of interest. A substantially identical cell may be derived from the same cells from which the recombinant (or non-recombinant) cell line was prepared but which had not been modified by introduction of heterologous DNA. Any difference in the amount of transcription indicates that the test compound has in some manner altered the activity of the protein of interest.
The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of noncritical parameters that could be changed or modified to yield essentially similar results.
EXAMPLES
Reagents and solvents used below can be obtained from commercial sources such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA). 1H-NMR spectra were recorded on a Narian Gemini 400 MHz ΝMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), coupling constant(s) in Hertz (Hz) and number of protons. Election lonization (El) mass spectra were recorded on a Hewlett Packard 5989A mass spectrometer. Mass spectrometry results are reported as the ratio of mass over charge, followed by the relative abundance of each ion (in parentheses). A single m/e value is reported for the M+H (or, as noted, M-H) ion containing the most common atomic isotopes. Isotope patterns correspond to the expected formula in all cases. Electrospray ionization (ESI) mass spectrometry analysis was conducted on a Hewlett- Packard 1100 MSD electrospray mass spectrometer using the HP1 100 HPLC for sample delivery. Normally the analyte was dissolved in methanol at 0.1 mg/mL and 1 microliter was infused with the delivery solvent into the mass spectrometer, which scanned from 100 to 1500 daltons. All compounds could be analyzed in the positive ESI mode, using 1:1 acetonitrile/water with 1% acetic acid as the delivery solvent. The compounds provided below could also be analyzed in the negative ESI mode, using 2mM NH4OAc in acetonitrile/water as delivery solvent. Analytical HPLC analysis was conducted on a Hewlett-Packard Series 1050 system equipped with a C18 reverse phase column (4.6 mm xl50mm) manufactured by Shiseido Co., Japan. Gradient elution was performed using variable percentage of acetonitrile and water (each with 0.1% trifluoroacetic acid added) as a mobile phase. Optical purity analysis was also conducted on a Hewlett-Packard Series 1050 system equipped with a chiral HLPC column (ChiralPak AD, 4.6 mm x 150mm) purchased from Chiral Technology. Isopropanol (3%) and hexane (97%) containing 0.1% diethylamine was used as a mobile phase.
Example 1
Figure imgf000040_0001
Compound 4 was prepared in 4 steps, as follows.
Step 1. Robinson Annulation. A mixture of N-benzyl-4-piperidone (500 g, 2.65 mol) and pyrrolidine (330 mL, d 0.852, 3.96 mol) in toluene (2L) was heated at refluxing while water from elimination was removed with a Dean-Stark trap. After 8 h, 70 mL of water were collected, and the volume of collected water ceased to increase further. GC analysis revealed the presence of the starting N-benzyl-4-piperidone and product enamine. The solvent and excess pyrrolidine were evaporated under reduced pressure (vacuum, 60 ton; heating bath, 50°C). The residue was dissolved in 500 mL of toluene, and evaporated again to give a dark oil (630g).
The resulting enamine was dissolved in anhydrous dioxane (2 L) and filtered into a 5-L three-necked flask equipped with a mechanic stiπer, a condenser and an addition funnel. 3-Penten-2-one (333 g, 2.78 mol) was added to the reaction vessel in 20 min. The reaction mixture was heated to reflux for 25 h. After cooled to near r.t, NaOMe (6.7 g, 0.125 mol) was added and the mixture was heated to reflux for 6 h. A premixed solution of AcONa (200 g) in 400 mL water and glacial AcOH (400 mL) was added to the reaction mixture after cooling to near room temperature. The reaction mixture was heated to reflux for an additional 5 h. Approximately 1 L of solvent (and possibly pyπolidine) was distilled out, the rest of the reaction mixture was cooled to r.t„ brought to slightly basic (pH 8-9) with 2 N NaOH (2.5 L). After layer separation, aqueous phase was extracted with AcOEt (3 L). The organic extracts were combined, washed with brine, and filtered through a short silica gel plug to give a dark clear solution. The filtrate was concentrated under reduced pressure to a thick oil (670 g). This material was used in the resolution step directly. Step 2. Resolution
Scheme 2
Figure imgf000041_0001
To a stiπed hot solution of the racemic isoquinolinone free base (626 g, 2.45 mol) in 95% ethanol (800 mL) was added solution of di-O-p-toluoyl-L-tartaric acid (945 g, 2.45 mol) in hot ethanol (1500 mL). Precipitation of the less soluble diasteromeric salt occurred generally as soon of the mixing was completed. The mixture was heated in a hot water bath (80°C) with gentle stirring for 1 h and allowed to cool to r.t. slowly (typically overnight). The precipitate was collected by filtration and rinsed wih cold 95% ethanol (800 mL). The solid (off-white) was triturated in hot 95% ethanol (1500 mL) and collected after cooling (typically after standing at r.t. overnight) by filtration and washed with cold ethanol. An off-white solid (340g, ca. 98% ee) was obtained after two triturations.
Compound 2a was liberated from the salt by neutralization with NaOH and extraction with AcOEt. Step 3. Hydrogenation
Figure imgf000041_0002
A flask for the Parr shaker hydrogenation apparatus was charged with the N-benzylisoqinolinone compound from previous step (120 g, 0.47 mol), 10% Pd/C (12 g, contains 50% water), di-t-butyl dicarbonate (133 g, 0.61 mol), and ethanol (1200 mL, 200 proof). The reactions took place under a hydrogen pressure of 60 psi. The hydrogen presure dropped quickly in the first 2 h, frequent recharges were needed. The reaction was typically left to go undisturbed for 8 h or longer. No further H2 consumption was observed. The reaction mixture was filtered through a Celite pad, rinsed with ethanol. Filtrate was concentrated to the product as a thick oil, which ' solidified on standing to give a white solid. This material was used in the next step without further purification.
Step 4. Fisher indole synthesis
Figure imgf000042_0001
S 4
The solution of the N-Boc-isoquinolinone compound from previous step in propanol (200 mL) was placed in a pressure resistant vessel. Concentrated H2SO4 (13 mL) was added slowly. Gas release took place immediately and subsided after 45 min. 4-Trifluorophenylhydrazine (16.56 g, 94.0 mmol) was added. The mixture was stiπed for 1 h at r.t followed by 3 h at refluxing or until gas evolution stopped. At this time, MS analysis indicted hydrozone as the major component of the reaction mixture. The reaction vessel was capped and heated at 90°C for 36 h until the completion of reaction, as monitored by TLC (10:1:0.1, CH2Cl2/MeOH/NH4OH), and ES-MS in positive mode. At the completion of the reaction, the reaction mixture was poured to a stiπed solution of 1 N NaOH (some precipitates were formed). The mixture was extracted with dichloromethane three times. The combined organic extracts were washed with water, dried over NaSO4, filtered, and concentrated to give a solid. The residue was triturated with CH2C12. The solid was collected by filtration. The product could be purified by chromatography on silica gel column with a gradient elution of increasing polarity from 20: 1 :0.1 to 6: 1 :0.1 CH2Cl2/MeOH/NH4OH to obtain compound 4 as the major product. 1H NMR δ 11.2 (s, 1H), 7.80 (s, 1H), 7.43 (d, J = 5.4 Hz, 1H), 7.28 (d, J = 5.4 Hz, 1H), 3.40 (d, J = 6.0 Hz, 1H), 3.01 (d, J - 8.0Hz, 1H), 2.75 (d, J = 10 Hz, 1H), 2.60 (m, 2H), 2.40 (m, 2H), 1.82 (d, J = 8 Hz, 1H), 1.56 (m, 1H), 1.38 (d, J = 5.4 Hz, 3H), 1.25 (m, 2H). MS (ES): 309 [M+H]+. Example 2
Figure imgf000043_0001
5
Compound 5 was synthesized in 3 steps, as follows. Step 1. To a 500 mL flask containing iPr2NH (16.82 mL, 120 mmol) in THF
(200 mL) at -78° C was added n-BuLi (48 mL, 2.5 M/hexanes, 120 mmol). After stirring for 30 min at -78° C, 4-Tetrahydro-pyran-4-carboxylic acid methyl ester (11.86 mL, 100 mmol) was added. After stirring for an additional 45 min, HMPA (10 mL) and allyl iodide (11.9 mL, 130 mmol) were added. The reaction was maintained for 20 min at the low temperature and allowed to warm up to r.t. The reaction mixture was poured into water and extracted with ether. The organic layer was washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20-30% EtOAc/hexanes to yield a clear oil (16.6 g).
Step 2. The above alkylation product (15.26 g, 83 mmol) was stiπed with NaIO
(39.0 g, 182 mmol) and OsO4 (70 mg) in iPrOH (400 mL) and H2O (400 mL) overnight. The reaction mixture was poured into water and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 40-70% EtOAc/hexanes to yield the product, 4-(2-oxo-ethyl)- tetrahydropyran-4-carboxylic acid methyl ester, as an oil (8.8 g). Step 3. The above aldehyde (1.86 g, 10 mmol) was stiπed with amine 4 (3.08 g, 10 mmol) and NaBH(OAc)3 (8.48 g, 40 mmol) in C1CH2CH2C1 (50 mL) overnight. It was poured into a dilute aq. ammonia solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of EtOAc, 10% MeOH/EtOAc, 10% MeOH/CH2Cl2, 20% MeOH/CH2Cl2 and 30% MeOH/CH2Cl2 to yield 5 as a solid (3.0 g). 1H NMR δ (DMSO, 400 MHz), 11.20 (s, 1H), 7.88 (s, 1H), 7.44 (d, J = 8.5 Hz, 1H), 7.30 (d, J = 8.5 Hz, 1H), 3.73 (m, 3H), 3.68 (s, 3H), 3.33 (m, 2H), 3.22 (m, 1H), 2.88 (m, 1H), 2.75 (m, 1H), 2.60 (m, 1H), 2.42 (m, 1H), 2.26 (m, 2H), 1.98 (m, 2H), 1.7 - 1.95 (m, 5H), 1.51 (m, 2H), 1.38 (d, J 6.5 Hz, 3H), 1.39 (m, 1H), 1.20 (m, 1H). MS (ES): 479 [M+H].
Example 3
Figure imgf000044_0001
A sample of compound 5 (0.080 g, 0.17 mmol) in THF (2 mL) was reduced with LiAlH4 (0.400 mL, 1M/THF, 0.40 mmol) in THF (2 mL). At the completion of the reduction, the reaction was quenched with an aqueous solution of 10% Na SO . The precipitate was removed by filtration, and the organic filtrate was washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation. Purification by flash chromatography on silica gel with a gradient elution of 5-30% MeOH/CH2Cl2 to yield 6 as a white solid (0.065 g). 1H NMR δ (DMSO, 400 MHz), 11.22 (s, 1 H), 7.79 (s, 1 H), 7.44 (d, J = 8.5 Hz, 1 H), 7.29 (d, J = 8.5 Hz, 1 H), 5.35 (s, 1 H), 3.56 (m, 4 H), 3.31 (m, 3 H), 3.02 (m, 1 H), 2.74 (m, 1 H), 2.62 (m, 1 H), 2.43 (m, 3 H), 1.95 (m, 1 H), 1.86 (m, 1 H), 1.75 (m, 1 H), 1.60 (m, 2 H), 1.25 - 1.5 (m, 10 H). MS (ES): 451 [M+H].
Example 4
Figure imgf000044_0002
7 A mixture of ester 5 (0.80 g, 1.7 mmol) and LiOH-H2O (0.80 g, 19 mmol) in dioxane (10 mL) and water (5 mL) was heated at refluxing for 7 h. The reaction mixture was cooled. On acidification with HOAc (to pH ca. 5) white precipitate was formed. The solid was collected by filtration, rinsed with water and finally with ether to yield the compound 7 as white solid (0.50 g). 1H NMR δ (DMSO, 500 MHz), 11.21 (s, 1 H), 7.79 (s, 1 H), 7.44 (d, J = 8.5 Hz, 1 H), 7.29 (d, J = 8.5 Hz, 1 H), 3.71 (m, 2 H), 3.31 (m, 2 H), 2.97 (m, 1 H), 2.75 (m, 1 H), 2.62 (m, 1 H), 2.44 (m, 4 H), 1.6 - 2.05 (m, 8 H), 1.2 - 1.5 (m, 8 H). MS (ES): 465 [M+H].
Example 5
Figure imgf000045_0001
8
To a mixture of acid 7 (1.86 g, 4 mmol) in CH2C12 (60 mL) with 4 drops of DMF was added oxalyl chloride (17 mL, 2M in CH2C1 , 34 mmol). After stirring at r.t for 1 h, the mixture was concentrated and pumped under high vacuum to obtain a solid. To this solid was added sat. solution of NH4OH in CH2C12 (60 mL). The mixture was stirred overnight and directly loaded onto a chromatographic column with a gradient elution of 10-20% MeOH/CH2Cl2 with increasing percentage (0-10%) of NB OH added to yield compound 8 as a white solid (1.674 g). 1H NMR δ (DMSO, 400 MHz), 11.28 (s, 1H), 7.80 (s, 1H), 7.45 (d, J - 8.5 Hz, 1H), 7.29 (d, J = 8.5 Hz, 1H), 7.00 (s, br, 2H), 3.67 (m, 2H), 3.38 (m, 5H), 2.6 - 3.0 (m, 3H), 2.43 (m, 1H), 2.20 (m, 1H), 1.99 (m, 3H), 1.6 - 1.9 (m, 4H), 1.35 - 1.5 (m, 7H). MS (ES): 464 [M+H].
Example 6
Figure imgf000045_0002
To a mixture of acid 7 (0.120 g, 0.256 mmol), DMF (2 drops) in CH2C12 (2 mL) was added (COCl)2 (1.2 mL, 2M/CH2C12, 2.4 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. This solid was resuspended in CH2C12 (2 mL), to it was added (S)(+) 2-amino-l -propanol (0.100 mL, 1.7 mmol) and NEt3 (0.140 mL, 1 mmol). After 1 h stirring at r.t, the mixture was poured into saturated solution of NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10-20% MeOH/CH2Cl2 with 0-10% NH4OH added to yield compound 9 as a white solid (0.098 g). 1H NMR δ (DMSO, 400 MHz), 11.22 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (m, 2H), 4.70 (s, IH), 3.92 (m, IH), 3.66 (m, 2H), 3.34 (m, 6H), 2.90 (m, IH), 2.75 (m, IH), 2.60 (m, IH), 2.40 (m, IH), 2.25 (m, 2H), 2.20 (m, 3H), 1.84 (m, IH), 1.72 (m, 3H), 1.41 (m, 7H), 1.07 (d, J = 6.6 Hz, 3H). MS (ES): 522 [M+H].
Example 7
Figure imgf000046_0001
10
To a mixture of acid 7 (0.075 g, 0.16 mmol), DMF (2 drops) in CH2C12 (2 mL) was added (COCl)2 (0.8 mL, 2 M/CH2C12, 1.6 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. To this solid was added CH2C12 (2 mL), (S)(+) 2-(aminomethyl)-pyπolidine (0.20 mL, 1.87 mmol) and NEt3 (0.170 mL, 1.2 mmol). The mixture was stiπed for 1 h at r.t., poured into sat. NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20-40% MeOH/CH2Cl in 0 - 10% NH4OH to yield compound 10 as a yellowish solid (0.053 g). 1H NMR δ (DMSO, 400 MHz), 11.21 (s, IH), 7.78 (s, IH), 7.58 (m, IH), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.67 (m, 2H), 3.1 - 3.5 (m, 8H), 2.75 - 2.9 (m, 4H), 2.6 (m, IH), 2.38 (m, IH), 2.22 (m, 2H), 2.01 (m, 2H), 1.6 - 1.9 (m, 8H), 1.4 (m, 7H), 1.47 (m, IH). MS (ES): 547 [M+H].
Example 8
Figure imgf000047_0001
A mixture of amide 8 (0.695 g, 1.50 mmol) and POCl3 (0.42 mL, 4.5 mmol) in anhydrous pyridine (14 mL) was heated to 120°C in a sealed vessel for 2 h. It was cooled to r.t., poured into saturated NaHCO3 solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 5-30% MeOH/CH2Cl2 to yield the corresponding nitrile as a yellowish solid (0.400 g).
The above nitrile was heated with Bu3SnN3 (0.74 mL, 2.7 mmol) in toluene (3 mL) at 120° C in a sealed vessel for 2 days. At the completion of the reaction, the reaction mixture was cooled to r.t., acidified with 1M HC1 in ether and purified by flash chromatography on silica gel with a gradient elution of 20-40% MeOH/ CH2C12 in 0-10% NH4OH to yield compound 11 as a yellowish solid (0.196 g). 1H NMR δ (DMSO, 400 MHz), 11.26 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.30 (d, J = 8.5 Hz, IH), 3.37 (m, 3H), 3.14 (m, 4H), 3.14 (m, IH), 2.77 (m, IH), 2.64 (m, 3H), 2.43 (m, 2H), 2.25 (m, 3H), 2.01 (m, 2H), 1.91 (m, IH), 1.76 (m, 2H), 1.55 (m, IH), 1.35 - 1.5 (m, 4H). MS (ES): 489 [M+H].
Example 9
Figure imgf000047_0002
12
Compound 12 was prepared following the procedures detailed in Example 4, substituting 4-(2-oxo-ethyl)-tetrahydro-pyran-4-carboxylic acid methyl ester with cyclohexane carboxylic acid methyl ester. 1H NMR δ (d6-DMSO) 12.47 (bs, IH), 11.31 (s, IH), 10.46 (bs, IH), 7.78 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.28 (dd, J = 8.5, 1.4 Hz, IH), 3.78 (d, J = 11.6 Hz, IH), 3.56 (d, J = 11.6 Hz, IH), 3.10 (td, J = 12.2, 5.0 Hz, IH), 2.94 (m, 2H), 2.81 (d, J = 14.4 Hz, 2H), 2.65 (t, J = 5.7 Hz, IH), 2.40 (dd, J = 15.7, 9.8 Hz, IH), 2.05-1.90 (m, 5H), 1.80-1.65 (m, 3H), 1.54-1.39 (m, 3H), 1.39 (d, J = 6.6 Hz, 3H), 1.38-1.22 (m, 5H). MS (ES): 463 [M+H].
Example 10
Figure imgf000048_0001
Compound 13 was synthesized in 5 steps according to the following scheme.
Scheme 4
Figure imgf000048_0002
Nal04, Os04 eOH/H20
Figure imgf000048_0003
LIOH dioxane/h^O
Figure imgf000048_0004
13
Step 1. A mixture of 4-allyl-tetrahydropyran carboxylic acid methyl ester (15.7 g, 92.4 mmol) and LiOH-H2O (29 g, 688 mmol) in THF (110 mL), MeOH (110 mL) and water (5 mL) was heated to 85° C in a sealed vessel overnight. Upon cooling to r.t., it was extracted with EtOAc, washed with water , dried with anhydrous Na2SO and concentrated by rotary evaporation to yield the corresponding carboxylic acid as a white solid (12.82 g). Step 2. To a mixture of the above acid (12.82 g, 75.4 mmol), DMF (4 drops) in
CH2C12 (300 mL) was added (COCl)2 (75.4 mL, 2 M in CH2C12, 150.8 mmol). When the gas release ceased, the mixture was placed on a rotary evaporator. The obtained acid chloride (in 150 mL of dry acetone) was added to NaN (48.75 g, 0.75 mol, in 300 mL of water) at 0° C over 30 min. After stirring 2 h at r.t., the mixture was poured into ice-water and extracted with ether. The organic layer was separated, washed with brine, dried over anhydrous Na2SO4 and concentrated by rotary evaporation. The obtained azide was dissolved in 100 mL of benzene and added slowly to 100 mL of refluxing benzene. The reflux continued for another 40 min at which time no more gas was released. Benzene was distilled off and 120 mL of MeOH was added to the mixture. The mixture was heated at refluxing for 36 h, cooled to r.t. and was directly chromatographed using 40% EtOAc/hexanes as eluent. The carbamate product was obtained as a white solid (14.15 g).
Step 3. The obtained carbamate (0.498 g, 2.5 mmol) was stiπed with NaIO4
(1.18 g, 5.5 mmol) and OsO4 (30 mg) in MeOH (5 mL) and H2O (5 mL) for 15 min. The mixture was directly loaded onto a column for chromatography with a gradient elution of 70 - 90% EtOAc/hexanes as the eluent to yield the aldehyde product as an oil (0.48 g).
Step 4. The above aldehyde (0.38 g, 1.9 mmol) was stiπed with amine 4 (0.587 g, 1.9 mmpl) and NaBH(OAc)3 (1.61 g, 7.6 mmol) in C1CH2CH2C1 (15 mL) overnight. The reaction mixture was poured into a dilute aqueous ammonia solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20-40% MeOH/CH2Cl2 with 0-10% NH4OH added to yield the coupled product as a solid (0.618 g). Step 5. A mixture of the obtained product (0.618 g, 1.25 mmol) and LiOHΗ2O
(3.0 g, 71.5 mmol) in dioxane (20 mL) and water (10 mL) was heated to 120 C in a sealed vessel for 8 h. Upon cooling to r.t., it was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20-40% MeOH/CH2Cl2 with 0- 10% NH4OH added to yield compound 13 as a yellowish solid (0.268 g). MS (ES): 436 [M+H].
Example 11
Figure imgf000050_0001
To a mixture of amine 13 (0.096 g, 0.22 mmol) and NEt3 (0.084 mL, 0.6 mmol) in CH2C12 (1.5 mL) was added trifluoromethanesulfonyl anhydride (0.067 mL, 0.40 mmol) at 0 C. After 20 min, it was poured into saturated NaHCO3 solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 5-20% MeOH/CH2Cl to yield compound 14 as a yellowish solid (0.077 g). 1H NMR δ (DMSO, 400 MHz), 11.27 (s, IH), 7.81 (s, IH), 7.46 (d, J = 8.5 Hz, IH), 7.30 (d, J = 8.5 Hz, IH), 3.5 - 4.1 (m, 10H), 2.95 (m, 2H), 2.83 (m, IH), 2.80 (m, IH), 2.69 (m, IH), 2.35 (m, 2H), 2.00 (m, 5H), 1.73 (m, 3H), 1.40 (d, J = 6.5 Hz, 3H). MS (ES): 568 [M+H].
Example 12
Figure imgf000050_0002
15
To a mixture of acid 7 (0.070 g, 0.15 mmol), DMF (2 drops) and DCM (2 mL) was added (COCl)2 (0.6 mL, 2 M/DCM, 1.2 mmol). When the gas release ceased, solvents were evaporated to obtain a solid. To this solid was added DCM (2 mL), morpholine (0.2 mL, 2.3 mmol) and TEA (0.15 mL, 1.1 mmol). The mixture was stiπed at r.t. for 1 h, poured into saturated NaHCO3 and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with 20% MeOH/DCM to yield 15 as a yellowish solid (0.080 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.69 (m, 2 H), 3.58 (m, 8H), 3.44 (m, 2 H), 3.32 (m, 2H), 3.24 (m, IH), 2.90 (m, IH), 2.74 (m, IH), 2.42 (m, IH), 2.23 (m, 2H), 2.08 (m, 2H), 1.86 (m, 4H), 1.68 (m, IH), 1.52 (m, 2H), 1.29 (m, 2H), 1.27 (d, J - 8.5 Hz, 3H). MS (ES): 534 [M+H].
Example 13
Figure imgf000051_0001
16
A mixture of acid 7 (0.306 g, 0.66 mmol), 4-(2-aminoethyl)morpholine (0.375 mL, 2.64 mmol), EDC.HC1 (0.381 g, 1.98 mmol), HOBt (0.267 g, 1.98 mmol), NMP (0.44 mL, 4 mmol), DCM (5 mL) and DMF (5 mL) was stiπed at r.t. for 3 h. The mixture was poured into saturated NaHCO3 and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM containing 1 - 3% NH4OH to yield 16 as a white solid (0.310 g). MS (ES): 577 [M+H]. Example 14
Figure imgf000052_0001
17 A mixture of acid 7 (0.102 g, 0.22 mmol), 2-aminoethyl-l- ethylpyπolidine (0.128 g, 1 mmol), EDC.HCl (0.127 g, 0.66 mmol), HOBt (0.089 g, 0.66 mmol), NMP (0.22 mL, 2 mmol), DCM (1.5 mL) and DMF (1.5 mL) was stiπed at r.t. for 8 h. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 25% MeOH/DCM containing 1 - 3% NH4OH to yield 17 as a white solid (0.095 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.78 (s, IH), 7.60 (t, J = 5.5 Hz, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J - 8.5 Hz, IH), 3.66 (m, 2H), 3.30 (m, IH), 3.21 (m, IH), 3.02 (m, IH), 2.60 - 3.0 (m, 3H), 2.73 (m, IH), 2.60 (m, IH), 2.47 (m, IH), 2.40 (m, IH), 2.20 (m, 3H), 2.10 (m, IH), 2.00 (m, 2H), 1.79 - 1.95 (m, 4H), 1.5 - 1.7 (m, 6H), 1.40 (m, 5H), 1.37 (d, J = 6.5 Hz, 3H), 1.06 (m, IH), 1.04 (t, J = 7.5 Hz, 3H). MS (ES): 575 [M+H].
Example 15
Figure imgf000052_0002
18
To a mixture of acid 7 (0.325 g, 0.7 mmol), DMF (2 drops) and DCM (8 mL) was added (COCl)2 (3.6 mL, 2 M in DCM, 7.2 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. To this solid was added DCM (8 mL), MeONH2.HCl (0.800 g, 9.6 mmol) and TEA (0.60 mL, 4.3 mmol). The mixture was stiπed at r.t. for 1 h, poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 10% NH4OH to yield 18 as an off-white solid (0.24 g). 1H NMR δ
(DMSO, 500 MHz): 11.20 (s, IH), 11.15 (s, IH), 7.79 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.68 (m, 2H), 3.63 (s, 3H), 3.35 (m, 2H), 3.23 (m, IH), 2.89 (m, IH), 2.75 (m, IH), 2.60 (m, IH), 2.40 (m, IH), 2.26 (m, 2H), 1.87 - 2.0 (m, 3H), 1.81 (m, IH), 1.69 (m, 3H), 1.43 (m, 4H), 1.38 (d, J = 6.5 Hz, 3H), 1.19 (m, IH). MS (ES): 494 [M+H].
Example 16
Figure imgf000053_0001
19 To a mixture of acid 7 (0.060 g, 0.13 mmol), DMF (1 drop) and DCM (2 mL) was added (COCl)2 (0.6 mL, 2 M in DCM, 1.2 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. To this solid was added DCM (2 mL), NH2CH2CN (0.200 g, 3.6 mmol) and TEA (0.15 mL, 1.07 mmol). After stirring at r.t. for 1 h, the mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 8% NH4OH to yield 19 as a white solid (0.028 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, IH), 8.50 (s, IH), 7.78 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 4.15 (d, J = 5.5 Hz, 2H), 3.69 (m, 2H), 3.33 (s, 3H), 3.23 (m, IH), 2.88 (m, IH), 2.75 (m, IH), 2.60 (m, IH), 2.40 (m, IH), 2.22 (m, 2H), 2.00 (m, 2H), 1.6 - 1.95 (m, 4H), 1.50 (m, 2H), 1.39 (m, 2H), 1.37 (d, J = 6.5 Hz, 3H), 1.28 (m, IH). MS (ES): 503 [M+H]. Example 17
Figure imgf000054_0001
20 A mixture of acid 7 (0.102 g, 0.22 mmol), 2-aminomethyl-2-propanol
(0.095 g, 0.88 mmol, prepared according to Rai, B.; Dekhordi, L. S.; Khodr, H.; Jin, Y.; Liu, Z.; R C. Hider (1998) J. Med. Chem. 41:3347-3359), EDC-HC1 (0.127 g, 0.66 mmol), HOBt (0.089 g, 0.66 mmol), NMP (0.11 mL, 1 mmol), DCM (2 mL) and DMF (2 mL) was stirred at r.t. overnight. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 10% NH4OH to yield 20 as a white solid (0.085 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, 1 H), 7.79 (s, 1 H), 7.54 (m, 1 H), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 4.51 (s, IH), 3.68 (m, 2H), 3.38 (m, 2H), 3.26 (m, IH), 3.13 (d, J = 6.0 Hz, IH), 2.90 (m, IH), 2.75 (m, IH), 2.60 (m, IH), 2.40 (m, IH), 2.28 (m, 2H), 2.00 (m, 2H), 1.80 (m, 5H), 1.45 (m, 4H), 1.40 (d, J = 6.5 Hz, 3H), 1.29 (m, IH), 1.09 (s, 6H). MS (ES): 536 [M+H].
Example 18
Figure imgf000054_0002
21
Synthesized in two steps: To a mixture of acid 7 (1.00 g, 2.2 mmol), DMF (3 drops) and DCM (20 mL) was added (COCl)2 (10 mL, 2 M in DCM, 20 mmol). When the gas release ceased, the mixture was placed under a rotary evaporator followed by a high vacuum pump to obtain the corresponding acyl chloride as a solid. The acyl chloride (dissolved in 10 mL of DCM and 5 mL of DMF) was added to a flask containing NH2CH2CH2NH2 (4.42 mL, 66 mmol) in DCM (15 mL). The mixture was stiπed at r.t. for 1 h, poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 10% NH4OH to yield the corresponding aminoethyl amide as a yellowish solid.
The above amide (0.035 g, 0.07 mmol) was stiπed with AcCl (0.01 mL, 0.14 mmol) and TEA (0.035 mL, 0.25 mmol) in DCM (1 mL) for 10 min. The mixture was directly loaded onto a silica gel column eluted with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 7% NH4OH to yield 21 as a yellowish solid (0.022 g). MS (ES): 549 [M+H].
Example 19
Figure imgf000055_0001
22 Synthesized in two steps: A mixture of the intermediate aminoethyl amide of Example 18 (0.67 g, 1.65 mmol), trifluoroacetic anhydride (0.292 mL, 2.1 mmol), TEA (0.42 mL, 3 mmol) and DCM (10 mL) was stiπed at r.t. for 10 min. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM mixed with 1 -2% NH4OH to yield the coπesponding trifluoromethyl acetamide as a yellowish solid (0.510 g).
The above amide (0.300 g, 0.5 mmol) was refluxed with LAH (0.050 g, 1.3 mmol) in THF (5 mL) for 1 h. At this time HPLC-MS indicated half completion of the reaction. A second portion of LAH (0.050 g, 1.3 mmol) was added. After refluxing for another 1 h, no more progress was observed. The mixture was poured into dilute ammonium hydroxide and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation. Flash chromatography on silica gel with a gradient elution of 20 - 40% MeOH/DCM mixed with 1 - 10% NH4OH afforded no separation. Finally separation was achieved by preparative HPLC to yield 22 as a white solid (0.037 g). MS (ES): 589 [M+H].
Figure imgf000056_0001
A mixture of amine 13 (0.090 g, 0.2 mmol), tetiahydro-3-furoic acid (0.047 g, 0.4 mmol), EDC.HCl (0.115 g, 0.6 mmol), HOBt (0.081 g, 0.6 mmol), TEA (0.140 mL, 1 mmol), DCM (1 mL) and DMF (1 mL) was stiπed at r.t. overnight. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 10% NH4OH to yield 23 as a yellowish solid (0.069 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, IH), 7.78 (s, IH), 7.47 (s, br, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.89 (t, J = 7.8 Hz, IH), 3.58 - 3.85 (m, 6H), 3.48 (m, 2H), 3.30 (m, IH), 3.05 (m, IH), 2.74 (m, IH), 2.62 (m, IH), 2.51 (m, IH), 2.35 (m, IH), 2.09 (m, 2H), 2.00 (m, 2H), 1.92 (m, 2H), 1.84 (m, IH), 1.4 - 1.55 (m, 3H), 1.37 (d, J = 6.5 Hz, 3H), 1.30 (m, IH). MS (ES): 534 [M+H]. Example 21
Figure imgf000057_0001
24
A mixture of amine 13 (0.065 g, 0.15 mmol), methanesulfonyl acetic acid (0.061 g, 0.45 mmol), EDC.HCl (0.086 g, 0.45 mmol), HOBt (0.061 g, 0.45 mmol), NMP (0.165 mL, 1.5 mmol), DCM (1.5 mL) and DMF (1.5 mL) was stiπed at r.t. for 24 h. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 1 - 2% NH OH to yield 24 as a white solid (0.052 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.90 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 4.13 (s, 2H), 3.63 (m, 2H), 3.54 (m, 2H), 3.32 (m, 2H), 3.11 (s, 3H), 2.92 (m, IH), 2.74 (m, IH), 2.60 (m, IH), 2.40 (m, 3H), 2.08 (m, 2H), 1.92 (m, 2H), 1.83 (m, IH), 1.69 (m, IH), 1.54 (m, 2H), 1.40 (m, 2H), 1.38 (d, J - 6.5 Hz, 3H), 1.19 (m, IH). MS (ES): 556 [M+H].
Example 22
Figure imgf000057_0002
25
A mixture of amine 13 (0.097 g, 0.224 mmol), 4,4-dioxo- tetrahydrothiopyranyl carboxylic acid (0.040 g, 0.224 mmol, prepared as following), EDC.HC1 (0.107 g, 0.56 mmol), HOBt (0.076 g, 0.56 mmol), NMP (0.275 mL, 2.5 mmol), DCM (1.5 mL) and DMF (1.5 mL) was stiπed at r.t. overnight. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20% MeOH/DCM mixed with 1 - 3% NH4OH to yield 25 as a white solid (0.074 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.78 (s, IH), 7.50 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.62 (m, 2H), 3.48 (m, 2H), 3.25 (m, IH), 3.14 (m, 4H), 2.90 (m, IH), 2.74 (m, IH), 2.58 (m, 3H), 2.2 - 2.45 (m, 3H), 2.09 (m, 6H), 1.8 - 1.98 (m, 4H), 1.65 (m, IH), 1.43 (m, 3H), 1.38 (d, J = 6.5 Hz, 3H), 1.28 (m, IH). MS (ES): 596 [M+H]. Preparation of 4,4-dioxo-tetiahydrothiopyranyl carboxylic acid: A mixture of 2,2-dimethyl-l,3-dioxane-4,6-dione (10 g, 69.4 mmol), vinyl sulfone (8.19 g, 69.4 mmol), KOH (9.72 g, 173.6 mmol) and tBuOH (140 mL) was refluxed overnight. The supernant was decanted and 120 mL of 20% aqueous H2SO4 was added to the residual solid. The obtained mixture was refluxed for another 3 h, extracted with iPrOH/CHCl3, dried with anhydrous Na SO4 and concentrated by rotary evaporation to afford the desired 4,4-dioxo-tetiahydrothiopyranyl carboxylic acid.
Example 23
Figure imgf000058_0001
26
Synthesized in two steps: A mixture of amine 13 (0.100 g, 0.23 mmol), ClSO2CH2CO2Me (0.039 g, 0.3 mmol), pyrindine (0.49 mL, 0.6 mmol) and DCM (2 mL) was stiπed at r.t. for 20 min. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by prep. HPLC to yield the coπesponding sulfonamide.
The above sulfonamide (0.020 g, 0.038 mmol) was reacted with LAH (0.20 mL, 1 M in THF, 0.2 mmol) in THF (1 mL) at r.t. for 15 min. The reaction mixture was poured into dilute ammonium hydroxide and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO , concentrated by rotary evaporation. Flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 1 - 3% NH4OH afforded compound 26 as a white solid (0.004 g). MS (ES): 544 [M+H].
Example 24
Figure imgf000059_0001
27 A mixture of amine 13 (0.239 g, 0.55 mmol), C(Me)2(OH)CF2CO2H
(0.085 g, 0.55 mmol, prepared according to Dolbier, Jr. W. R; Ocampo, R. (1995) J. Organic Chem. 60:5378 and Hallinan, E. A.; Fried, J. (1984) Tetrahedron Lett. 25:2301), EDC-HC1 (0.264 g, 1.37 mmol), HOBt (0.186 g, 1.37 mmol), NMP (0.44 mL, 4 mmol), DCM (3 mL) and DMF (3 mL) was stiπed at r.t. for 24 h. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 0 - 2% NH4OH to yield 27 as a white solid (0.098 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.79 (m, 2H), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 5.59 (s, IH), 3.63 (m, 2H), 3.52 (m, 2H), 3.25 (m, IH), 2.93 (m, IH), 2.75 (m, IH), 2.61 (m, IH), 2.40 (m, 3H), 2.25 (m, 2H), 1.94 (m, 3H), 1.81 (m, IH), 1.66 (m, IH), 1.56 (m, 2H), 1.42 (m, 2H), 1.38 (d, J = 6.5 Hz, 3H), 1.19 (m, 7H). MS (ES): 572 [M+H]. Example 25
Figure imgf000060_0001
28
Step 1. A mixture of amine 13 (0.654 g, 1.5 mmol), OHCCH2CH2CH2CO2Et (0.260 g, 2 mmol), NaBH(OAc)3 (1.27 g, 6 mmol) and
C1CH CH2C1 (10 mL) was stiπed at r.t. overnight. The mixture was poured into dilute NH OH and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 1 - 4% NH4OH to yield the coπesponding monoalkylated amine (0.35 g). The product obtained above (0.35 g, 0.63 mmol) was hydrolyzed by treating with LiOH.H O (0.50 g, 12 mmol) in dioxane (4 mL) and H2O (2 mL) at r.t. for 3 h. The mixture was acidified with HO Ac to slightly acidic, and was concentrated to dryness. The above acid (~ 0.1 mmol, contained inorganic salt) was heated with
NaOAc (0.200 g) in Ac2O (2 mL) at 105°C for 20 min. The reaction mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 0 - 3 % NH4OH to yield 28 as a white solid
(0.040 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.78 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.64 (m, 2H), 3.45 (m, 4H), 3.36 (m, IH), 2.92 (m, IH), 2.75 (m, IH), 2.61 (m, IH), 2.38 (m, 5H), 2.26 (t, J = 8.0 Hz, 2H), 1.89 (m, 6H), 1.68 (m, IH), 1.41 (m, 2H), 1.38 (d, J = 6.5 Hz, 3H), 1.26 (m, IH). MS (ES): 504 [M+H]. Example 26
Figure imgf000061_0001
29
Synthesized in two steps: A mixture of amine 13 (0.664 g, 1.52 mmol), 2,5-dimethoxy-3-tetiahydrofuran-carboxaldehyde (0.487 g, 3.0 mmol) and HOAc (8 mL) was heated to 70 C for 2 h. The mixture was cooled to r.t., basified with saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM mixed with 0 - 2% NH4OH to yield, the coπesponding formylpyrrole derivative (0.70 g).
The above aldehyde was converted to nitrile by the following reaction. a vial containing NH2OH.HCl (0.083 g, 1.2 mmol) and CH3CN (3 mL) at 0 °C was added TEA (0.168 mL, 1.2 mmol) and the aldehyde (0.470 g, 0.91 mmol). The mixture was stiπed at 0 °C for 30 min. and at r.t. for 4 h. At this time, phthalic anhydride (0.178 g, 1.2 mmol) was added and the mixture was heated to 90 °C for 1 h, poured into saturated NaHCO and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM mixed with 0 - 1% NH4OH to yield 29 as a yellowish solid (0.167 g). 1H NMR δ (DMSO, 500 MHz): 11.18 (s, IH), 7.84 (s, IH), 7.77 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 7.17 (m, IH), 6.54 (m, IH), 3.71 (m, 2H), 3.41 (m, 2H), 3.08 (m, IH), 2.73 (m, 2H), 2.55 (m, 2H), 2.38 (m, IH), 2.25 (m, 2H), 1.85 - 2.1 (m, 6H), 1.77 (m, 2H), 1.54 (m, IH), 1.36 (d, J = 6.5 Hz, 3H), 1.23 (m, 2H). MS (ES): 511 [M+H]. Example 27
Figure imgf000062_0001
30
Synthesized in three steps: A mixture of amine 13 (0.600 g, 1.38 mmol), BocNHCH2CHO (0.220 g, 0.38 mmol), NaBH(OAc)3 (1.17 g, 5.5 mmol) and
C1CH2CH2C1 (14 mL) was stiπed at r.t. overnight. The mixture was poured into dilute NH OH and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 1 - 4% NH4OH to yield the coπesponding reductive animation product (0.311 g).
The product obtained above (0.311 g, 0.537 mmol) was stiπed with HC1 (1.5 mL, 4 M in dioxane, 6 mmol) in 1.5 mL of DCM for 30 min. The mixture was poured into dilute NH4OH and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentiated by rotary evaporation to yield the de-protected product which was directed used for the next step.
The resulted de-Boc product (0.129 g, 0.27 mmol) was refluxed with carbonyldiimidazole (0.087 g, 0.54 mmol) in DCM (8 mL) for 1 h. The reaction mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM mixed with 1 - 2% NH4OH to yield 30 as a white solid (0.123 g). MS (ES): 505 [M+H]. Example 28
Figure imgf000063_0001
31
A sample of the alcohol compound from Example 3 was converted to the coπesponding aldehyde by oxidation with SO3-Py and DMSO. To the solution of this aldehyde (0.100 g, 0.2 mmol) in MeOH (1 mL) at 0 °C was added glyoxal (0.055 g, 0.95 mmol, 50% in water) followed by NH3 (0.50 mL, 1 mmol, 2 M in MeOH). The mixture was allowed to warm to r.t. and stiπed for 24 h. The mixture was poured into with saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 15 - 20% MeOH/DCM with 1 - 4% NH4OH added to yield 31 as a yellowish solid (0.027 g). 1H NMR δ (DMSO, 500 MHz): 11.60 (s, br, IH), 11.20 (s, IH), 7.77 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 6.96 (s, br, 2H), 3.70 (m, 2H), 3.32 (m, 4H), 3.19 , (m, IH), 2.88 (s, IH), 2.72 (m, IH), 2.57 (m, IH), 2.51 (m, IH), 2.38 (m, IH), 2.23 (m, 2H), 2.09 (m, IH), 1.82 (m, 3H), 1.66 (m, 2H), 1.39 (m, 2H), 1.65 (d, J = 6.5 Hz, 3H), 1.19 (m, IH). MS (ES): 487 [M+H].
Example 29
Figure imgf000063_0002
32
Synthesized in two steps: To a solution of diazomethylphosphonate (0.180 g, 1.2 mmol, Seyferth, D.; Marmor, R. S.; Hubert, P. (1971) J. Organic Chem. 36:1379) in THF (6 mL) cooled to -78 °C under a nitrogen atmosphere was added KOtBu (1.8 mL, 1.8 mmol, 1.0 M in THF) dropwise. The obtained mixture was stirred for 10 min. at the low temperature. An aldehyde (0.268 g, 0.6 mmol, dissolved in THF, same one used as for the preparation of compound 31) was added to the above mixture dropwise. The mixture was stiπed for 30 min. at -78 °C and another 30 min. at r.t. The reaction was quenched with water, and the mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with gradient elution of 5 - 20% MeOH/DCM to yield the coπesponding alkyne as a white solid (0.200 g).
The obtained alkyne (0.100 g, 0.22 mmol) was heated with TMSN3 (1 mL) in a sealed vial at 140 °C for two days. The whole mixture was directly loaded onto a column, eluted with 15 - 20% MeOH/DCM mixed with 1 - 5% NH4OH to yield 32 as a white solid (0.0048 g). MS (ES): 488 [M+H].
Figure imgf000064_0001
33 Lithium diisopropylamide (1.1 mL,2.0 M, 2.2 mmol) was added to a solution of THF (0.25 M solution) containing N-allyl-N-t-butoxycarbonyl- methanesulfonamide (0.518 g, 2.2 mmol) at -78 °C. After stirring for 50 min. at —78 °C, the solution of the aldehyde intermediate from Example 28 in dry THF (0.470 g, 1.05 mmol) was added and the mixture was stiπed at -78 °C to room temperature overnight. The reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 3% MeOH/DCM to yield 3 (R1 = Boc, R2 = allyl) as a yellowish oil (0.25 g). The resulting oil was treated with trifloroacetic acid in DCM (0.5 M) to yield 33 as yellow film. 1H NMR (400 MHz, CDC13) δ 7.93 (s, IH), 7.84 (s, IH), 7.33 (s, IH), 6.71 (d, J= 15.6 Hz, IH), 6.16 (d, J= 15.6 Hz, IH), 5.86 (m, IH), 5.30 (dd, J= 1.3, J= 3.8 Hz, IH), 5.27 (d, /= 1.2 Hz, IH), 5.22 (dd, , J= 1.1, 10.2 Hz, IH), 4.38 (s, IH), 3.91 (m, 2H), 3.69 (m, 2H), 3.58 (m, 2H), 3.30 (d, , J= 10.5 Hz, IH), 2.98 (d, J= 10.5 Hz, IH), 2.68 (m, 2H), 2.30-2.48 (m, 3H), 1.95 (m, IH), 1.86 (d, , J= 9.1 Hz, IH), 1.70-1.81 (m, 6H), 1.49-1.59 (m, 4H), 1.45 (d, J= 6.6 Hz, 3H). ESI (MH+) m/z 566.
Figure imgf000065_0001
34
A sample of compound 33 (0.067g, 0.12 mmol) was treated with tetiakis(triphenylphosphine)-plladium (0.012 g, O.Olmmol) and 1,3-dimethyl barbituric acid (0.2 g, 1.28 mmol) in DCM at 35 °C overnight, to yield 34 as a solid (0.006 g). 1H NMR (400 MHz, CDC13) δ 8.04 (s, IH), 7.92 (s, IH), 7.33 (s, 2H), 6.77 (d, J= 15.6 Hz, IH), 6.34 (d, J= 15.6 Hz, IH), 4.68 (s, IH), 3.77 (m, 2H), 3.69 (m, 2H), 3. 3.29 (d, , J= 10 Hz, IH), 2.97 (d, J= 10 Hz, IH), 2.66 (m, 2H), 2.31-2.50 (m, 3H), 1.95 (m, IH), 1.86 (d, , J= 9.1 Hz IH), 1.70-1.81 (m, 7H), 1.49-1.59 (m, 4H), 1.45 (d, J= 6.6 Hz,1 3H). ESI (MH+) m/z 526.
Figure imgf000065_0002
The mixture of aldehyde compound from Example 28 (0.1 g, 0.22 mmol), hydroxylamine hydrochloride (0.024 g, 0.33 mmol) and triethylamine (0.045 g, 0.45 mmol) in MeOH (0.2 M solution) stiπed at room temperature for 5.5 h. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentrated, redissolved in DCM, pale yellow solid came out, rinsed the solid with more DCM, dried to yield 0.04 g 35 as pale yellow solid. 1H NMR (400 MHz, DMSO) δ 11.2 (s, IH), 10.55 (s, IH), 7.80 (s, IH), 7.41 (d, J= 9.0 Hz, IH), 7.26 (d, J= 9.0 Hz, 1H), 7.23 (s, IH), 3.64 (br, 2H), 3.43 (m, 2H), 3.30 (s, 3H), 3.21 (d, J= 10.0 Hz, IH), 2.88 (d, J= 10.0 Hz, IH), 2.72 (dd, J= A Hz, J= 13 Hz, IH), 2.58 (m, IH), 2.49 (t, J= 1.75 Hz, IH), 2.39 (m, IH), 2.26 (m, IH), 1.86 (m, IH), 1.72-1.80 (m, 3H), 1.62 (t, J- 7.5 Hz, 2H), 1.49 (td, J= 3.8, Hz J- 14 Hz, 2H), 1.38 (m, IH), 1.35 (d, J= 6.6 Hz, 3H), 1.27 (m, IH). ESI (MH+) m/z A6A.
Figure imgf000066_0001
Synthesized in the same way as in Example 29 with the exception of replacing hydroxylamine hydrochloride with methoxyamine hydrochloride. 1H NMR (400 MHz, DMSO) δ 11.12 (s, IH), 7.77 (s, IH), 7.42 (d, J= 8.3 Hz, IH), 7.35 (s, IH), 7.27 (d, J= 8.3 Hz, IH), 3.74 (s, 2H), 3.65 (d, J= 11.4 Hz, 2H), 3.45 (t, J= 11.4 Hz, 2H), 3.31 (s, 3H), 3.23 (d, J= 10.6 Hz, IH), 2.89 (d, J= 8.8 Hz, IH), 2.72 (dd, J= 4 Hz, J- 13 Hz, 1 H), 2.59 (m, IH), 2.49 (br, IH), 2.39 (m, IH), 2.28 (m, 2H), 1.80 (m, IH), 1.72-1.80 (m, 3H), 1.66 (br, 3H), 1.50 (br, IH), 1.40 (br, IH), 1.36 (d, J= 6.6 Hz, 3H), 1.26 (m, IH). ESI (MH+) m/z 478.
Figure imgf000066_0002
The mixture of aldehyde intermediate from Example 28 (0.10 g, 0.22 mmol), 2-amino-ethanethiol hydrochloride (0.062 g, 0.54 mmol) and sodium methoxide (0.078 g, 1.4 mmol) in MeOH (0.2 M solution) was stiπed at room temperature overnight. The reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine, dried, concentrated and purified by flash chromatography on silica gel eluted with 10:1:0.1 DCM-MeOH-NH4OH to yield 37 as yellow solid (0.06 g). 1H NMR (400 MHz, CDC13) δ 8.09 (s, IH), 7.83 (s, IH), 7.33 (s, 2H), 4.65 (d, , J= 6.1 Hz, IH), 3.79 (m, 2H), 3.70 (dd, J= 2.76 Hz, J= 9.7 Hz, IH), 3.53-3.66 (m, 4H), 3.48 (s, 3H), 3.29 (d, J= 9.5 Hz, IH), 3.16 (d, J= 9.5 Hz, IH), 2.95-2.98 (m, 2H), 2.82 (m, IH), 2.67 (m, 4H), 2.35-2.6 (br, 2H), 2.20 (m, 2H), 2.09 (m, IH), 1.86 (m, 2H), 1.60-1.80 (m, 2H), 1.2-1.55(m, 3H). ESI (MH+) m/z 508.
Figure imgf000067_0001
Synthesized in two steps: A mixture of amide 8 (0.150 g, 0.32 mmol) and (MeO)2CHN(Me)2 was heated to 120 °C for 15 min. After cooled to r.t., the whole was directly loaded onto a column eluted with 15 - 30% MeOH/DCM mixed with 1 - 10% NH4OH to yield the coπesponding acylamidine as a yellow solid (0.120 g).
The above acylamidine was treated with NH2NH2Η O (0.030 mL, 0.6 mmol) in AcOH (lmL) at 90 °C for 10 min. After cooling to r.t., the whole was directly loaded onto a column eluted with 10 - 30% MeOH/DCM mixed with 1 - 4% NH4OH to yield triazole 38 as a white solid (0.050 g). 1H NMR δ (DMSO, 500 MHz): 11.18 (s, IH), 7.77 (s, IH), 7.43 (d, j = 8.5 Hz, IH), 7.28 (d, j = 8.5 Hz, IH), 3.72 (m, 2H), 3.28 (m, 3H), 3.12 (m, IH), 2.79 (m, 1), 2.71 (m, IH), 2.55 (m, 2H), 2.37 (m, IH), 2.23 (m, 2H), 2.04 (m, 2H), 1.77 (m, 6H), 1.53 (m, IH), 1.33 (m, 5H), 1.22 (m, IH). MS (ES): 488 [M+H].
Example 36
Figure imgf000067_0002
39 The mixture of amide 8 (0.695 g, 1.50 mmol) and POCl3 (0.42 mL, 4.5 mmol) in anhydrous pyridine (14 mL) was heated to 120 °C in a sealed vessel for 2 h. The mixture was cooled to r.t., poured into saturated NaHCO3 solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 5 - 30% MeOH/DCM to yield the corresponding nitrile as a yellowish solid (0.400 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 9.79 (s, 1 H), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 3.87 (m, 2H), 3.50 (m, 2H), 3.31 (m, 2H), 2.96 (m, IH), 2.75 (m, IH), 2.61 (m, IH), 2.54 (m, IH), 2.51 (m, IH), 1.96 (m, IH), 1.86 (m, 5H), 1.72 (m, IH), 1.65 (m, 2H), 1.42 (m, 2H), 1.40 (d, J = 6.5 Hz, 3H), 1.32 (m, IH). MS (ES): 446 [M+H].
Example 37
Figure imgf000068_0001
40
Synthesized in two steps: A mixture of amide 8 (0.630 g, 1.36 mmol) and LAH (4.76 mL, 1 M in THF, 4.76 mmol) was heated to 70 °C for 1 h. After cooling to r.t., the mixture was poured into dilute NH4OH and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 - 50% MeOH/DCM mixed with 0 - 10% NH4OH to yield the coπesponding amine as a solid (0.500 g).
The above amine (0.060 g, 0.133 mmol) was reacted with (CF3SO2)2O (0.067 mL, 0.4 mmol), TEA (0.084 mL, 0.6 mmol) in DCM (2 mL) at 0°C for 10 min. The mixture was poured into water, basified with saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution with 0 - 10% MeOH/EtOAc to yield 40 as a yellowish solid (0.045 g). 1H NMR δ (DMSO, 500 MHz): 11.26 (s, IH), 7.81 (s, IH), 7.46 (d, J = 8.5 Hz, IH), 7.30 (d, J = 8.5 Hz, IH), 3.67 (m, 1 H), 3.56 (m, 4H), 3.35 (m, IH), 2.97 (m, 4H), 2.82 (m, 2H), 2.66 (m, 2H), 2.31 (m, 2H), 1.99 (m, IH), 1.80 (m, 4H), 1.68 (m, IH), 1.4 - 1.5 (m, 4H), 1.39 (d, J - 6.5 Hz, 3H). MS (ES): 582 [M+H].
Figure imgf000069_0001
A mixture of ester 5 (0.070 g, 0.15 mmol) and MeLi (1.0 mL, 1.6 M in ether, 1.6 mmol) in THF (1.5 mL) was stiπed at r.t. for 30 min. The mixture was poured into with saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel eluted with 20% MeOH/DCM to yield 41 as a white solid (0.012 g). 1H NMR δ (DMSO, 500 MHz): 11.21 (s, IH), 7.78 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 5.95 (s, IH), 3.69 (m, 2H), 3.47 (m, 2H), 3.32 (m, 2H), 2.99 (m, IH), 2.76 (m, IH), 2.63 (m, IH), 2.52 (m, IH), 2.42 (m, 2H), 1.85 (m, 4H), 1.67 (m, 2H), 1.2 - 1.55 (m, 5H), 1.38 (d, J = 6.5 Hz, 3H), 1.08 (m, 6H). MS (ES): 479 [M+H].
Figure imgf000069_0002
Synthesized in three steps: To a solution of tetrahydro-4H-pyran-4-one (5.00 g, 50 mmol) in THF (80 mL) was added allylmagnesium bromide (60 mL, 1 M/ ether, 60 mmol). After stirring at r.t. for 30 min, the reaction was quenched with aqueous NH4C1 and extracted with ether. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 50 - 60% EtOAc/hexanes to yield the alcohol adduct as a clear oil (2.51 g).
The obtained alkenyl alcohol (1.00 g, 7 mmol) was stiπed with NaIO4 (3.30 g, 15.4 mmol) and OsO4 (40 mg) in MeOH (15 mL) and H2O (15 mL) for 15 min. The entire mixture was directly loaded onto a column. Elution with EtOAc yielded the coπesponding aldehyde as a brownish oil (0.70 g).
The above aldehyde (0.250 g, 1.7 mmol) was stiπed with amine 4 (0.309 g, 1 mmol) and NaBH(OAc)3 (0.856 g, 4 mmol) in C1CH2CH2C1 (7 mL) for 1.5 h. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentrated by rotary evaporation and purified by flash chromatography on silica -gel with a gradient elution of 10 - 40% MeOH/DCM with 0 - 10% NH4OH added to yield 42 as a brownish solid (0.145 g). 1H NMR δ (DMSO, 500 MHz): 11.22 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 5.11 (s, IH), 3.62 (m, 5H), 3.36 (m, IH), 3.03 (m, IH), 2.76 (m, IH), 2.62 (m, IH), 2.51 (m, IH), 2.42 (m, IH), 1.87 (m, IH), 1.65 (m, 2H), 1.25 - 1.6 (m, 9H), 1.39 (d, J = 6.5 Hz, 3H). MS (ES): 437 [M+H].
Example 40
Figure imgf000070_0001
43
The mixture of acid 7 (1 g, 2.15 mmol), N-Boc-cystine methyl ester (1.06 g, 4.52 mmol), bis(2-oxo-3-oxazolidinyl)phosphinic chloride (1.096 g, 4.30 mmol) and triethylamine (0.456 g, 4.52 mmol) in DCM (0.4 M solution) was stiπed at room temperature overnight. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentiated and purified by flash chromatography on silica gel eluted with 20:1 :0.1 DCM:MeOH:NH4OH to yield a yellow solid (0.6 g).
The resulting solid was treated with 20% trifluoroacetic acid in DCM. At the completion of the deprotection, excess reagent and solvent were removed by evaporation. The residue was refluxed in benzene (20 mL) and DCE (5 mL, to aid the solubility) overnight. The reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 5% MeOH/DCM to yield 43 as a yellow solid (0.16 g). 1H NMR (400 MHz, CDC13) δ 8.1 (br, IH), 7.90 (s, IH), 7.32 (s, 2H), 5.24 (t, J- 8.0 Hz, IH), 3.80 (m, 6H), 3.53-3.66 (m, 5H), 3.48 (s, 3H), 2.66-2.74 (m, 2H), 2.52 (br, 2H), 1.80-2.10 (m, 6H), 1.73 (m, 3H), 1.45 (d, J= 6.6 Hz, 3H), 0.95 (s, IH). ESI (MH ) m/z 564.
Figure imgf000071_0001
44
A sample of compound from Example 40 (0.2 g, 0.35 mmol) was treated with activated manganese dioxide (0.15 g, 1.75 mmol) in benzene refluxing overnight, to yield 44 as yellow solid (0.045 g). 1H NMR (400 MHz, CDC13) δ 8.17 (s, IH), 7.91 (s, IH), 7.75 (s, IH), 7.32 (s, 2H), 5.24 (t, J= 8.0 Hz, IH), 3.93 (s, 3H), 3.84 (m, 2H), 3.58(m, 2H), 3.20 (br, IH), 2.82 (br, IH), 2.65 (m, 2H), 2.43 (m, IH), 2.34 (m, 2H), 1.90-2.10 (m, 5 H), 1.80 (m, 2H), 1.57 (br, 4H), 1.45 (d, J= 6.6 Hz, 3H). ESI (MH+) m/z 562.
Figure imgf000071_0002
The mixture of amine 4 (0.107 g, 0.35 mmol), [4-(thiazole-2-carbonyl)- tetrahydro-pyran-4-yl]-acetaldehyde (0.069 g, 0.29 mmol, prepared similarly as described previously), and sodium triacetoxborohydride (0.245 g, 1.15 mmol) in DCE (0.25 M solution) stiπed at room temperature for 2 h. The reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 5% MeOH/DCM to yield 45 as yellow solid (0.09 g). 1H NMR (400 MHz, CDC13) δ 7.9 (d, , J= 2.2 Hz, 2H), 7.80 (s, IH), 7.46 (d, J= 2.2 Hz, IH), 7.30 (s, 2H), 3.82 (m, 2H), 3.61 (t, J= 9.8 Hz, IH), 3.46 (t, , J= 9.8 Hz, IH), 3.04 (d, , J= 9.8 Hz, IH), 2.46-2.68 (m, 6H), 2.18-2.33 (m, 4H), 1.87 (m, 2H), 1.74 (m, IH), 1.45-1.63 (m, 3H), 1.30 (d, , J= 6.6 Hz, 3H), 1.02 (m, IH), 0.56 (br, IH). ESI (MH+) m/z 532.
Figure imgf000072_0001
A sample of ketone from Example 42 (0.039g, 0.073 mmol) was treated with sodium borohydride (0.02 g, 0.5 mmol) in THF for 30 min. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 5%-10% MeOH/DCM to yield 46 as yellow solid (0.005 g). 1H NMR (400 MHz, CDC13) δ 7.95 (s, IH), 7.80 (s, IH), 7.79 (dd, , J = 3.3 Hz, J= 4.6 Hz IH), 7.38 (dd, J= 3.3 Hz J= 6.3 Hz, IH), 7.35 (s, IH), 4.91 (d, J= 4.4 Hz, IH), 3.85 (m, 2H), 3.75 (q, J= 11.0 Hz, 2H), 3.41 (m, IH), 3.46 (t, , J= 9.8 Hz, IH), 3.19 (dd, J= 4.7 Hz, J= 7.4 Hz 2H), 3.0 (m, IH), 2.85 (m, IH), 2.73 (m, IH), 2.61 (dd, J= 9.0 Hz, J= 14.9 Hz, 2H), 2.05-2.50 (m, 4H), 1.91 (m, 3H), 1.67 (m, 2H), 1.60 (m, 2H), 1.44 (dd, J= 6.7 Hz J= 9.4 Hz 3H). ESI (MH1") m/z 534.
Example 44
Figure imgf000072_0002
Step 1. LAH (0.64 g, 16.7 mL) was added to a dry THF solution (70 mL) containing the ester compound from Example 1 (4.0 g, 8.4 mmol) at room temperature. The solution was heated at reflux for 2 h. After heating, water (0.6 mL) was added followed by a 1 N solution of NaOH (0.6 mL), and a final addition of water (1.2 mL). The resulting solid was filtered washed with copious amount of dichloromethane. The filtrate was concentrated and used in the next step without purification: ESI (MH+) m/z 451.
Step 2. The resulting alcohol compound from above (0.6 g, 1.33 mmol) was dissolved in DMSO/Et N (2.5:1, 0.2 M) and was treated with SO3-pyridine complex (0.85 g, 5.33 mmol) and at room temperature. After stirring for 2 h, the mixture was poured into water (60 mL) and extracted with dichloromethane (3 x 100 mL). The organic layers were washed with brine, dried over Na SO3, and concentrated to give the aldehyde intermediate. ESI (MH*) m/z 449.
Step 3. Sodium hydride (0.96, 40 mmol) was added to a dry DMF (0.2 M) solution containing tiiethyl phosphonoacetate (4 mL, 20.0 mmol) at room temperature. After stirring for 10 min., the aldehyde intermediate from above (4.51 g, 10.1 mmol) was added and the mixture was stiπed at room temperature overnight. Excess DMF was removed under vacuum and the remaimng residue was taken up in a 10% MeOH/DCM solution, washed with water, dried with Na2SO3 and concentrated. A portion of this material was purified by using preparative HPLC (C 18 column, 10%- 90% acetonitrile/water gradient). 1H NMR (400 MHz, MeOD) δ 7.79 (s, IH), 7.39 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 6.87 (d, J= 16 Hz, IH), 6.00 (d, J= 16 Hz, IH), 4.22 (q, J= 7 Hz, 2H), 3.89 (d, J= 10 Hz, IH), 3.77-3.81 (m, 3H), 3.67 (d, J= 15 Hz, IH), 3.57 (t, J= 10 Hz, 3H), 3.01-3.18 (m, 4H), 2.78-2.91 (m, 4H), 2.52 (m, IH), 2.20 (d, J- 12 Hz, IH), 2.00 (qn, J= 6 Hz, 2H), 1.61-1.85 (m, 9H), 1.49 (d, J= 6 Hz, 3H),1.31 (t, J = 7 Hz, 3H). ESI (MH+) m/z 519.
Example 45
Figure imgf000073_0001
48 DIBAL (2.1 eq) was added to a dry THF solution (0.2 M) containing the ester from Example 44 (1 equiv.) at 0 °C. After stirring for 4 h. the reaction was quenched at 0 °C with methanol and warmed to room temperature. The solution was concentrated under reduced pressure and the remaining residue was purified on silica eluting with 0-20% methanol/dichloromethane gradient: ESI (Mϊ ) m/z 475.
Sodium triacetoxyborohydride (3 equiv.) was added to a dichloromethane solution (0.2 M) containing aldehyde intermediate from above (1 equiv.) and pyrroline (2 equiv.) at room temperature. After stirring overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient). 1HNMR (400 MHz, MeOD,) δ 7.78 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.93 (d, J = 16 Hz, IH), 5.71 (td, J= 7 , J= 16 Hz, IH), 3.89 (m, 3H), 3.80 (m, 2H), 3.56-3.69 (m, 5H), 3.12-3.19 (m, 4 H), 3.01 (t, J= 14 Hz, IH), 2.75-2.92 (m, 3H), 2.52 (dd, J= 12, J= 14Hz, IH), 2.10-2.22 (m, 3H), 2.04 (m, 2H), 1.95 (m, 2H), 1.64-1.84 (m, 7H), 1.49 (d, J= 7 Hz, 3H); ESI (MH*) m/z 530.
Example 46
Figure imgf000074_0001
49 This compound was prepared in the same way as described in Example
45. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.95 (d, J= 16 Hz, IH), 5.74 (td, J= 1, J= 16 Hz, IH), 3.90 (m, 3H), 3.66-3.80 (m, 7H), 3.40-3.60 (m, 6H), 3.10-3.26 (m, 2H), 2.99 (t, J= 14 Hz, 1 H), 2.75-2.91 (m, 3H), 2.50 (dd, J= 12, J= 14 IH), 2.17 (d, J= 12 Hz, IH), 1.96 (m, 2H), 1.62-1.82 (m, 7H). 1.48 (d, J= 1 Hz, 3H). ESI (Mϊt) m/z 594.
Figure imgf000075_0001
50
This compound was prepared in the same way as described in Example 45. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.30 (d, J= 8 Hz, IH), 5.87 (d, J= 16 Hz, IH), 5.66 (td, J= 7 Hz, J= 16 Hz, IH), 3.93 (m, 3H), 3.60-3.80 (m, 7 H), 3.10-3.21 (m, 2H), 3.00 (t, J= 14 Hz, IH), 2.75-2.93 (m, 3H), 2.54 (dd, J= 12, J= 14Hz, IH), 2.22 (d, J= 12 Hz, IH), 1.80-1.96 (m, 3H), 1.64- 1.79 (m, 7H). 1.59 (d, J= 7 Hz, 3H), 1.50 (d, J= 1 Hz, 3H). ESI (MH+) m/z 547.
Figure imgf000075_0002
51
This compound was prepared in the same way as described in Example 45. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.78 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.87 (d, J= 16 Hz, IH), 5.69 (td, J= 7 , J= 16 Hz, IH), 3.90 (d, J= 10 Hz, IH), 3.59-3.80 (m, 7H), 3.10-3.21 (m, 2H), 3.00 (m, 3H), 2.79-2.92 (m, 3H), 2.53 (dd, J= 12, J= 14Hz, 1H), 2.19 (d, J= 12 Hz, IH), 1.89-1.96 (m, 3H), 1.64-1.79 (m, 6H). 1.49 (d, J= 7 Hz, 3H), 1.33 (s, 6H). ESI (MH+) m/z 548.
Figure imgf000076_0001
52 This compound was prepared in the same way as described in Example 45. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.78 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.91 (d, J= 16 Hz, IH), 5.69 (td, J- 7 Hz, J= 16 Hz, IH), 4.03 (q, J= 9 Hz, 2H), 3.84-3.91 (m, 3H), 3.78 (m, 2H), 3.58-3.70 (m, 3H), 3.10-3.21 (m, 2H), 2.99 (t, J= 14 Hz, IH), 2.77-2.91 ( , 3H), 2.53 (dd, J= 12, J= 14Hz, 1 H), 2.19 (d, J= 12 Hz, IH), 1.78-1.97 (m, 3H), 1.57-1.78 (m, 6H). 1.48 (d, J= 7 Hz, 3H). ESI (MH+) m/z 558.
Example 50
Figure imgf000076_0002
53 A sample of the aldehyde intermediate obtained in Step 1 in Example 44 was treated methyl Grinard reagent (2.5 equiv.) in dry THF (0.2 M) at room temperature. After stirring overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient). (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.62 (ddd, J= 2, j = 6 , J= 16 Hz, IH), 5.51 (d, J= 16 Hz, IH), 4.33 (qn, J= 6 Hz, IH), 3.89 (d, J= 12 Hz, IH) 3.77 (m, 2H), 3.59-3.69 (m, 3H), 3.10-3.21 (m, 2H), 3.00 (t, J= 14 Hz, IH), 2.77-2.92 (m, 3H), 2.52 (dd, J= 12, J= 14 Hz, IH), 2.20 (d, J= 12 Hz, IH), 1.83-1.90 (m, 3 H), 1.59-1.74 (m, 6H). 1.50 (d, J= 7 Hz, 3H), 1.30 (d, J- 7 Hz, 3H). ESI (MH+) m/z 491.
Figure imgf000077_0001
54 btained from treatment of aldehyde intermediate from Step 1 in Example 44 with an isopropyl Grinard. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.43-5.60 (m, 2H), 3.88 (m, 2H), 3.77 (m, 2H), 3.59-3.69 (m, 3H), 3.10-3.21 (m, 2H), 3.01 (t, J= 14 Hz, IH), 2.80- 2.92 (m, 3H), 2.52 (dd, J= 12, J= 14 Hz, IH), 2.20 (d, J= 12 Hz, IH), 1.63-1.93 (m, 10H), 1.49 (d, J= 7 Hz, 3H), 0.97 (d, J= 6 Hz, 3H), 0.94 (d, J= 6 Hz, 3H). ESI (MH+) m/z 519.
Figure imgf000077_0002
55 Obtained from treatment of aldehyde intermediate from Step 1 in
Example 44 with a t-butyl Grinard. (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 5.65 (dd, J= 1, J= 16 Hz, IH), 5.51 (d, J= 16 Hz, IH), 3.85 (m, IH), 3.78 (m, 3H), 3.59-3.69 (m, 3H), 3.10-3.21 (m, 2H), 3.02 (t, J= 14 Hz, IH), 2.80-2.93 (m, 3H), 2.53 (dd, J= 12, J= 14 Hz, IH), 2.19 (d, J= 12 Hz, IH), 1.80-1.90 (m, 3H), 1.62-1.77 (m. 6H), 1.50 (d, J= 7 Hz, 3H), 0.96 (s, 9 H). ESI (Mϊ ) m/z 533.
Figure imgf000078_0001
56
Lithium hydroxide (30 mg, 1.3 mmol) was added to a THF/water solution (1:1, 0.2 M) containing ester intermediate from Example 44 (0.54 g, 1.0 mmol) and heated at reflux for 3 h. After cooling to room temperature, the solution was titrated with a 3 N HC1 solution to neutral pH and concentrated to dryness using reduced pressure. This material was used in the next step without purification. ESI (MH+) m/z 491. General synthesis for analogs amide formation. . HBTU (3 equiv.) was added to a dichloromethane solution (0.2 M) containing, triethylamine (3 equiv.), acid intermediate from above(l equiv.), and the respective amine (2 equiv.) at room temperature. After stirring overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient).
R = Et, (TFA Salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 6.62 (d, J= 16 Hz, IH), 6.03 (d, J= 16 Hz, IH), 3.89 (d, J= 10 Hz, IH), 3.79(m, 2H), 3.50-3.73 (m, 4H), 3.13-3.21 (m, 2H), 3.02 (m, IH), 2.80-3.0 (m, 3H), 2.53 (dd, J= 12, j= 14 Hz, IH), 2.20 (d, J= 12 Hz, IH), 1.90- 2.0 (m, 3H), 1.60-1.87 (m. 7H), 1.50 (m, 4H), 1.57 (t, J= 1 Hz, 3H). ESI (MH+) m/z 518.5.
Example 54
Figure imgf000078_0002
57 (TFA salt). 1H NMR (400 MHz, MeOD,) δ 7.79 (s, 1 H), 7.40 (d, J= 8
Hz, IH), 7.29 (d, J= 8 Hz, IH), 6.63 (d, J= 16 Hz, IH), 6.02 (d, J= 16 Hz, IH), 4.06 (qn, J= 1 Hz, IH), 3.84 (d, J= 10 Hz, IH), 3.70 (m, 2H), 3.56-3.69 (m, 3H), 3.10-3.19 (m, 2H), 3.02 (t, J= 14 Hz, IH), 2.80-2.93 (m, 3H), 2.52 (dd, J= 12, J- 14 Hz, IH), 2.19 (d, J= 12 Hz, IH), 1.62-1.99 (m. 9H), 1.50 (d, J= 1 Hz, 3H), 1.19 (d, J= 7 Hz, 6H). ESI (MET*) m/z 532.5.
Figure imgf000079_0001
58
(TFA salt) 1H NMR (400 MHz, MeOD,) δ 7.76 (s, IH), 7.35 (d, J= 8 Hz, IH), 7.24 (d, J= 8 Hz, IH), 6.70 (d, J= 16 Hz, IH), 6.07 (d, J= 16 Hz, IH), 3.76 (m, 2H), 3.58 (t, J= 9 Hz, 2H), 3.40 (s, 2H), 3.31 (m, 1 H), 3.02 (d, J= 10 Hz, IH), 2.75 (dd, J= 3 Hz, J= 16 Hz, IH), 2.64 (qn, J= 1 Hz, IH), 2.40 (m, 3H), 2.04 (t, J= 10 Hz, IH), 1.69-1.93 (m, 9H), 1.63 (m, 6H), 1.49 (m, 2H), 1.43 (d, J= 7 Hz, 3H), 1.34 (m, IH). ESI (MH+) m/z 588.
Figure imgf000079_0002
Tosylmethyl isocyanide (22 mg, 0.1 mmol) was added to a methanol solution (5 mL) containing potassium carbonate (19 mg, 1.2 mmol) and aldehyde intermediate from Step 2 of Example 44 (50 mg, 0.1 mmol). The solution was heated at reflux for 24 h. After cooling, the product was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient): 1H NMR (400 MHz, MeOD,) δ 8.23 (s, IH), 7.75 (s, IH), 7.39 (d, J= 8 Hz, IH), 7.25 (d, J= 8 Hz, IH), 7.11 (s, IH), 3.81 (d, J= 12 Hz, 3H), 3.59 (d, J= 12 Hz, IH), 3.45 (t, J= 11 Hz, 2H), 2.72-3.06 (m, 6H), 2.47(dd, J= 12, J= 14 Hz, IH), 2.13 (m, 4H), 1.77-1.88 (m, 3H), 1.55-1.67 (m, 2H), 1.44 (d, J- 7 Hz, 3H). ESI (MH ) m/z 488.
Example 57
Figure imgf000080_0001
60
To a mixture of acid 12 (0.150 g, 0.32 mmol), DMF (2 drops) and DCM (4 mL) was added (COCl)2 (1.0 mL, 2 M in DCM, 7.2 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. To this solid was added DCM (4 mL) and MeNH2 (8 mL, 2 M/THF, 16 mmol). The mixture was stiπed for 1 h at r.t., poured into brine and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel eluted with a gradient elution of 20 - 40% MeOH/DCM mixed with 0 - 7% NH4OH to yield 60 as a yellowish solid (0.095 g). MS (ES): 476 [M+H].
Example 58
Figure imgf000080_0002
61 A mixture of acid 12 (0.926 g, 2 mmol), glycinamide HC1 (0.442 g, 4 mmol), EDC.HC1 (0.960 g, 5 mmol), HOBt (0.676 g, 5 mmol), NMP (2.0 mL, 18 mmol), DCM (10 mL) and DMF (10 mL) was stirred at r.t. for 3 h. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 - 40% MeOH/DCM mixed with 1 - 8% NH4OH to yield 61 as a brownish solid (0.650 g). 1H NMR δ (DMSO, 500 MHz): 11.19 (s, IH), 7.89 (t, J = 7.5 Hz, IH), 7.78 (s, IH), 7.43 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 7.20 (s, IH), 6.97 (s, IH), 3.66 (m, 4H), 3.39 (m, 2H), 3.24 (m, IH), 3.29 (m, IH), 2.74 (m, IH), 2.60 (m, IH), 2.40 (m, IH), 2.28 (m, 2H), 2.12 (m, IH), 2.03 (m, 2H), 1.89 (m, IH), 1.80 (m, IH), 1.71 (m, 2H), 1.63 (m, IH), 1.42 (m, 5H), 1.38 (d, J = 6.5 Hz, 3H), 1.27 (m, IH). MS (ES): 519 [M+H].
Example 59
Figure imgf000081_0001
62
The coπesponding amine compound was prepared following the same procedures as depicted for Example 15, with the exception of substituting 4-allyl- tetrahydropyran carboxylic acid methyl ester with 1 -allyl- 1-cyclohexyl carboxylic acid methyl ester.
A mixture of this amine (0.235 g, 0.54 mmol), EtSO2Cl (0.139 g, 1.08 mmol), TEA (0.109 g, 1.08 mmol) in DCM (5 mL) was stiπed at r.t. for 45 min. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 % MeOH/DCM mixed with 0 - 1% NH4OH to yield 62 as a solid (0.037 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 7.16 (s, IH), 3.33 (m, 2H), 3.00 (m, 3H), 2.74 (m, IH), 2.63 (m, IH), 2.37 - 2.55 (m, 4H), 1.95 (m, IH), 1.84 (m, 6H), 1.72 (m, IH), 1.55 (m, 2H), 1.40 (m, 6H), 1.39 (d, J = 6.5 Hz, 3H), 1.25 (t, J = 7.2 Hz, 3 H). MS (ES): 526 [M+H]. Example 60
Figure imgf000082_0001
63
A mixture of amine intermediate from Example 59 (0.056 g, 0.13 mmol), CF3CH2SO2Cl (0.036 g, 0.15 mmol), TEA (0.042 mL, 0.3 mmol) in DCM (1 mL) was stirred at r.t. for 15 min. The mixture was poured into saturated NaHCO3 and extracted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 20 % MeOH/DCM mixed with 0 - 1% NH4OH to yield 63 as a solid (0.030 g). 1H NMR δ (DMSO, 500 MHz): 11.20 (s, 1 H), 8.19 (s, IH), 7.79 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.28 (d, J = 8.5 Hz, IH), 4.31 (m, 2H), 3.28 (m, 2H), 2.97 (m, IH), 2.75 (m, IH), 2.63 (m, IH), 2.48 (m, 2H), 2.37 (m, IH), 1.95 (m, IH), 1.84 (m, 5H), 1.74 (m, IH), 1.5 - 1.68 (m, 5H), 1.2 - 1.5 (m, 6H), 1.38 (d, J = 6.5 Hz, 3H). MS (ES): 580 [M+H].
Example 61
Figure imgf000082_0002
64 A mixture of amine intermediate from Example 59 (0.097 g, 0.224 mmol), 4,4-dioxo-tetiahydrothiopyranyl carboxylic acid (0.040 g, 0.224 mmol), EDC.HC1 (0.107 g, 0.56 mmol), HOBt (0.076 g, 0.56 mmol), NMP (0.275 mL, 2.5 mmol), DCM (1.5 mL) and DMF (1.5 mL) was stiπed at r.t. overnight. The mixture was poured into saturated NaHCO and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentrated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM mixed with 0 - 3% NH4OH to yield 64 as a white solid (0.023 g). 1H NMR δ (DMSO, 500 MHz): 11.25 (s, IH), 7.79 (s, IH), 7.45 (d, J = 8.5 Hz, IH), 7.29 (d, J - 8.5 Hz, IH), 3.13 (m, 5H), 2.78 (m, 2H), 2.64 (m, IH), 2.55 (m, IH), 2.43 (m, 2H), 2.08 (m, 10H), 1.2 - 1.55 (m, 16H). MS (ES): 594 [M+H].
Figure imgf000083_0001
65
Synthesized according to the same sequence as used for the synthesis of compound in Example 4. 1H NMR δ (DMSO, 500 MHz): 11.20 (s, IH), 7.78 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 3.58 (s, IH), 3.30 (m, 2H), 2.97 (m, IH), 2.75 (m, IH), 2.61 (m, IH), 2.39 (m, 3H), 2.02 (m, 3H), 1.80 (m, 4H), 1.59 (m, 4H), 1.45 (m, 3H), 1.38 (d, J = 6.5 Hz, 3H), 1.32 (m, IH). MS (ES): 449 [M+H].
Example 63
Figure imgf000083_0002
66 To a mixture of acid 65 (0.070 g, 0.15 mmol), DMF (1 drop) and DCM
(2 mL) was added (COCl)2 (0.5 mL, 2 M in DCM, 1 mmol). When the gas release ceased, the mixture was placed under high vacuum to obtain a solid. To this solid was added DCM (2 mL), MeNH2(2 mL, 2 M in THF, 2 mmol). The mixture was stiπed at r.t. for 1 h, poured into saturated NaHCO3 and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO , concentrated by rotary evaporation and purified by flash chromatography on silica gel eluted with a gradient elution of 5 - 30% MeOH/DCM mixed with 0 - 5% NH4OH to yield 66 as a white solid (0.060 g). 1H NMR δ (DMSO, 500 MHz): 11.21 (s, IH), 7.79 (s, IH), 7.59 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 2.76 (m, IH), 2.55 (m, IH), 2.60 (d, J = 4.0 Hz, 3H), 2.41 (m, 2H), 2.19 (m, IH), 2.01 (m, 2H), 1.81 (m, 4H), 1.33 - 1.58 (m, 10H), 1.39 (d, J = 6.5 Hz, 3H). MS (ES): 462 [M+H].
Example 64
Figure imgf000084_0001
67
A mixture of acid 65 (0.100 g, 0.22 mmol), 2-aminomethyl-2-propanol (0.120 g, 0.67 mmol), EDC.HCl (0.127 g, 0.66 mmol), HOBt (0.089 g, 0.66 mmol), NMP (0.25 mL, 2.3 mmol), DCM (2 mL) and DMF (2 mL) was stiπed at r.t. overnight. The mixture was poured into saturated NaHCO3 and extiacted with EtOAc. The organic layer was separated, washed with brine, dried with anhydrous Na2SO4, concentiated by rotary evaporation and purified by flash chromatography on silica gel with a gradient elution of 10 - 20% MeOH/DCM mixed with 1 - 2% NH4OH to yield 67 as a white solid (0.100 g). MS (ES): 520 [M+H].
Figure imgf000084_0002
68
Synthesized according to the same sequence as was used for the synthesis of compound 12 (Example 9). 1H NMR δ (DMSO, 500 MHz): 11.21 (s, IH), 7.78 (s, IH), 7.44 (d, J = 8.5 Hz, IH), 7.29 (d, J = 8.5 Hz, IH), 3.31 (m, 2H), 2.99 (m, IH), 2.75 (m, IH), 2.61 (m, IH), 2.37 (m, 6H), 1.8 - 2.05 (m, 8H), 1.25 - 1.50 (m, 6H). MS (ES): 435 [M+H].
Figure imgf000085_0001
69
Synthesized according to the same sequence as was used for the synthesis of example 2 1H NMR (400 MHz, CD3OD) δ 7.78 (s, 1 H), 7.39 (d, J= 8.0 Hz, IH), 7.27 (d, J= 8.0 Hz, IH), 3.75 (m, IH), 3.40 (m, IH), 3.14 (m, 2H), 3.00 (m, 2H), 2.60(m, IH), 2.40 - 2.50 (m, 4H), 1.50 - 2.00 (m, 4H) 1.44 (d, J- 6.4 Hz, 3H), 1.22 (s, 6H). ESI (MH+) m/z 423.
Figure imgf000085_0002
Compound 69 from Example 66 (1.03 g, 2.43 mmol) in 5 mL DCM was treated with oxalyl chloride (2.1 mL, 24.3 mmol) and two drops of dry DMF. After 30 min., the reaction was brought to dryness under low pressure. DCM (15 mL) was added and the flask was put into an ice bath. Much excess ammonia in dry DCM was added slowly to react with the previously formed carbonyl chloride. In around 20 min., the reaction showed completion by LC-MS. Water (30 mL) was added and the aqueous layer was extiacted with DCM (3x20 mL). The organic layers were combined, dried and concentiated. Purification by flash chromatography on silica gel with 5-10% MeOH/DCM afforded 70 (800 mg, 1.90 mmol) as light brown film. 1H NMR (400 MHz, CD3OD) δ 7.78 (s, IH), 7.39 (d, J= 8.0 Hz, IH), 7.26 (d, J= 8.0 Hz, IH), 3.77 (m, IH), 3.41 (m, IH), 2.98 (m, 2H), 2.55(m, IH), 2.10 - 2.50 (m, 4H),1.45-2.00 (m, 4H) 1.41 (d, , J= 6.4 Hz, 3H), 1.21 (s, 6H). ESI (MH+) m/z 422. Example 68
Figure imgf000086_0001
69 71
The mixture of the acid 69 (Example 66) (50 mg, 0.118 mmol), EDC (68 mg, 0.354 mmol), HOBT (16 mg, 0.118 mmol), NMM (0.039 mL, 0.354 mmol) and excess ethylamine (-10 equiv.) in 2 mL DCM was stirred at room temperature overnight. The solvent was removed under vacuum and 1 mL DMF and 0.2 mL water were added. The solution was injected directly to HPLC (reverse phase) to render 30 mg (0.066 mmol) of 71 as yellow film. 1H NMR (400 MHz, CD3OD) δ 7.77 (s, IH), 7.37 (d, J- 8.4 Hz, IH), 7.26 (d, J= 8.4 Hz, IH), 3.30 (m, 2H), 3.21 (q, J= 7.2 Hz, IH), 3.10 - 3.20 (m, 4H), 2.55(m, IH), 2.15 - 2.50 (m, 4H), 1.50-2.00 (m, 4.44 (d, J= 6.4 Hz, 3H), 1.22 (s, 6H), 1.13 (t, J= 7.2 Hz, 3H). ESI (MH+) m/z 450.
Example 69
Figure imgf000086_0002
69 72
Compound 72 was prepared following same procedures as described for Example 67. 1H NMR (400 MHz, CD3OD) δ 7.79 (s, IH), 7.39 (d, J= 8.4 Hz, IH), 7.28 (d, J= 8.4 Hz, IH), 7.07 (d, J= 8.4 Hz, 2H), 6.75 (d, J= 8.4 Hz, 2H), 3.82 (m, IH), 3.61 (m, IH), 3.20 (m, 2H), 2.65 - 3.00 (m, 6H), 2.50 (m, IH), 2.15 (m, 3H), 1.60 -1.90 (m, 3H) 1.45 (d, J= 6.4 Hz, 3H). ESI (MH+) m/z 443.
Example 70
Figure imgf000086_0003
69 73 The same procedure was followed as for compound 71 (Example 68). 1H NMR (400 MHz, CD3OD) δ 7.78 (s, 1 H), 7.39 (d, J= 8.4 Hz, IH), 7.29 (d, J= 8.4 Hz, IH), 3.90 (m, IH), 3.70 (m, IH), 3.5 (m, 2H), 3.30 (m, 3H), 2.95 (s, 6H), 2.50 - 2.90 (m, 5H), 2.50 (m, IH), 2.00 - 2.20 (m, 4H), 1.60 -1.90 (m, 2H) 1.49 (d, J= 6.8 Hz, 3H), 1.27 (s, 6H). ESI (MH+) m/z 493.
Example 71
Figure imgf000087_0001
69 74
The same procedure was followed as for compound 71 (Example 68). 1H NMR (400 MHz, CD3OD) δ 7.77 (s, 1 H), 7.38 (d, J= 8.4 Hz, IH), 7.27 (d, J= 8.4 Hz, IH), 7.20 - 7.40 (m, 5H), 3.90 (m, IH), 3.70 (br, 2H), 3.30 - 3.65 (m, 8H), 3.20 (m, 2H), 2.50 - 2.90 (m, 5H), 2.50 (m, IH), 2.00 - 2.20 (m, 3H), 1.60 -1.90 (m, 3 H) 1.49 (d, J= 6.8 Hz, 3H), 1.27 (s, 6H). ESI (MH+) m/z 582.
Example 72
Figure imgf000087_0002
69 75
The same procedure was followed as for compound 71 (Example 68). 1H NMR (400 MHz, CD3OD) δ 7.77 (s, 1 H), 7.38 (d, J= 8.4 Hz, IH), 7.27 (d, J= 8.4 Hz, IH), 7.20 - 7.40 (m, 5H), 3.88 (m, 1 H), 3.69 (br, 2H), 3.30 - 3.65 (m, 5 H), 3.21 (m, 2H), 2.50 - 2.90 (m, 5H), 2.50 (m, IH), 2.00 - 2.20 (m, 4H), 1.60 -1.90 (m, 6H) 1.48 (d, J= 6.8 Hz, 3H), 1.26 (s, 6H). ESI (MH+) m/z 596. Example 73
Figure imgf000088_0001
76 77
Compound 76 was prepared by reducing compound 69 and oxidizing the coπesponding alcohol with SO3-pyridine in DMSO/TEA as described in Example 44. To the solution of compound 76 (25 mg, 0.061 mmol) in 2 mL DCM was added NaBH(OAc)3 (26.0 mg, 0.122 mmol) followed by the addition of amine (11 mg, 0.122 mmol). The reaction mixture was kept stirring for overnight. The solvent was removed and 1 mL DMF was added to dissolve the mixture. After filtration, the solution was injected directly to reverse HPLC to render pure yellow film 77 (18 mg, 0.038 mmol) as the product. 1H NMR (400 MHz, CD3OD) δ 7.78 (s, 1 H), 7.40 (d, J= 8.4 Hz, IH), 7.30 (d, J= 8.4 Hz, 1 H), 3.93 (m, 1 H), 3.68 (m, 1 H), 3.60 (s, 2 H), 3.30 - 3.40 (m, 4 H), 2.75 - 3.10 (m, 4 H), 2.50 (m, 1 H), 2.00 - 2.20 (m, 3 H),1.60 -1.90 (m, 3 H) 1.49 (d, J= 6.A Hz, 3 H), 1.38 (s, 6 H), 1.14 (s, 6 H). ESI (MH+) m/z 480.
Example 74
Figure imgf000088_0002
78 79
Compound 78 was obtained in several steps following a sequence similar to that of Example 10. At 0 °C, to the solution of compound 78 (6 mg, 0.015 mmol) and triethylamine (6 mg, 0.06 mmol), was added methansulfonic anhydride (7.8 mg, 0.045 mmol). The reaction was kept stirring for overnight. The solvent was removed under vacuum and the resulted residue was subjected to reverse HPLC to render pure yellow solid 79 (2.0 mg, 0.004 mmol). 1H NMR (400 MHz, CD3OD) δ 7.78 (s, 1 H), 7.40 (d, J= 8.4 Hz, 1 H), 7.30 (d, J= 8.4 Hz, 1 H), 3.50 - 3.70 (m, 2 H), 3.00 - 3.30 (m, 3 H), 2.70 (s, 3 H), 2.60 - 3.00 (m, 4 H), 1.60 - 2.20 (m, 6 H), 1.47 (d, J= 6.4 Hz, 3 H), 1.20 (s, 6 H). ESI (MH+) m/z 472.
Example 75
Figure imgf000089_0001
80
R=Me, 83
R=Ms, 84
A mixture of amine 4 (0.9 g, 2.92 mmol), sodium triacetoxborohydride (2.5 g, 11.7 mmol) and aldehyde 80 (l.Og, 3.5 mmol) in DCE (0.25 M solution) was stiπed at room temperature for 3 h. The reaction mixture was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 3% MeOH/DCM to yield 81 as yellow solid (0.73 g). Compound
81 was treated with trifloroacetic acid in DCM for 0.5 h, concentrated and purified by flash chromatography on silica gel eluted with 20:1 :0.1 DCM:MeOH:NH4OH to yield
82 as pale yellow solid (0.49 g). 1H NMR (400 MHz, DMSO) δ 11.2 (s, 1 H), 8.03 (s, 1 H), 7.41 (d, , J= 8.4 Hz, 1 H), 7.25 (d, J- 8.4 Hz, 1 H), 3.64 (s, 3 H), 3.2 (dd, J= 2.4
Hz, J= 9.2 Hz, l H), 2.86 (m, 2 H), 2.84 (dd, J= 2.4 Hz J= 9.2 Hz, 1 H), 2.50-2.75 (m, 4 H), 2.10-2.45 (m, 4 H), 2.0 (d, J= 12 Hz ,2 H), 1.55-1.90 (m, 5 H), 1.47 (m, 2 H), 1.35-1.4 (m, 2H), 1.35 (d, , J= 6.6 Hz, 3 H), 1.25 (m, 1 H). ESI (MH+) m/z 478.
Compound 82 (O.lg, 0.21 mmol) was treated with formaldehyde (0.01 g, 0.3 mmol) and sodium triacetoxborohydride (0.178 g, 0.84 mmol) in DCE for 3 h. at room temperature. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentiated and purified by flash chromatography on silica gel eluted with 20: 1 :0.1 DCM:MeOH:NH4OH to yield 83 as yellow solid (0.01 g). !H NMR (400 MHz, CDC13) δ 7.93 (s, 1 H), 7.84 (s,l H), 7.33 (s, 2 H), 3.71 (s, 3 H), 3.4 (d, J= 6.4 Hz, 1 H), 2.96 (d, J= 6.4 Hz,l H), 2.67 (m, 4 H), 2.47 (dd, J= 8.0 Hz J= 12 Hz, 1 H), 2.10- 2.40 (m, 7 H), 1.45-1.95 (m, 11 H), 1.35 (d, , J= 6.6 Hz, 3 H). ESI (MH+) m/z 492.
Compound 82 (0.05g, 0.105 mmol) was treated with mesyl chloride (0.012 g, 0.105 mmol) and triethylamine (0.013 g, 0.013 mmol) in DCM for 2 h. at 0 °C. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 20:1:0.1 DCM:MeOH:NH4OH to yield 84 as yellow solid (0.055 g). 1H NMR (400 MHz, CDC13) δ 8.10 (s, 1 H), 7.86 (s,l H), 7.32 (s, 2 H), 3.74 (s, 3 H), 3.63 (d, J= 9.5 Hz, 1 H), 2.93 (d, J= 9.5 Hz, 1 H), 2.60-2.80 (m, 7 H), 2.20-2.50 (m, 5 H), 1.96 (t, J= 10.8 Hz, 1 H), 1.8 (m, 4 H), 1.73 (t, J= 7.8, 1 H), 1.60 (t, J- 12 Hz, 2 H), 1.30-1.50 (m, 3H), 1.35 (d, J= 6.6 Hz, 3 H). ESI (MH+) m/z 556.
Example 76
Figure imgf000090_0001
85
Comound 83 (0.02g, 0.04 mmol) was treated with lithium aluminumhydride (0.02 g, 0.042mmol) in THF for 2 h. at room temperature. The reaction was treated with saturated sodium bicarbonate solution and extiacted with ethyl acetate, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 20:1:0.1 DCM:MeOH:NH4OH to yield 85 as yellow solid (0.055 g). 1H NMR (400 MHz, CDC13) δ 8.1 (s, 1 H), 7.80 (s,l H), 7.33 (s, 2 H), 3.41 (s, 3 H), 3.15 (d, J= 10.5 Hz, 1 H), 2.35-2.65 (m, 7 H), 2.67 (m, 4 H), 2.05 (m, 2 H), 1.5-1.9 (m, 12 H), 1.40 (d, , J= 6.6 Hz, 3 H). ESI (MH+) m/z 464.
Figure imgf000091_0001
86
Compound 83 (4g, 8.15 mmol) was treated with lithium hydroxide (0.391 g, 16.3 mmol) in THF, MeOH and water reflux overnight. The reaction was treated with 1 N HC1 solution and extracted with isopropyl alcohol and chloroform, the organic layer was washed with brine and dried, concentrated and purified by flash chromatography on silica gel eluted with 10:1:0.1 DCM:MeOH:HOAc to yield 2 g pink powder (acid). The acid (0.052g, 0.11 mmol) was reacted with 2-methoxyethylamine (0.0098 g, 0.13 mmol), under the condition of EDC (0.063 g, 0.33 mmol), HOBt (0.0147g, 0.11 mmol), and NMM (0.033 g, 0.33 mmol) in DMF (1 mL) at room temperature overnight. The reaction was treated with saturated sodium bicarbonate solution and extracted with ethyl acetate, the organic layer was washed with brine and dried, concentiated and purified by flash chromatography on silica gel eluted with 20: 1 :0.1 DCM:MeOH:NH4OH to yield 86 as brown oil (0.003 g). 1H NMR (400 MHz, CDC13) δ 8.05 (s, IH), 7.83 (s, IH), 7.34 (s, 2H), 3.48 (s, 3H), 3.35 (s, 3H), 3.05 (d, J= 11.2 Hz, IH), 2.88 (m, 2H), 2.60-2.75 (m, 3H), 2.35-2.55 (m, 9H), 2.05 (m, 2H), 1.48- 1.60 (m, 5H), 1.22 (s, 6H), 0.85 (m, 2H). ESI (MH+) m/z A6A.
Example 78
Figure imgf000092_0001
90 91
Intermediate 88. EDC (1.12 g, 5.9 mmol) was added to a dichloromethane solution (0.2 M) containing, triethylamine (1 mL.), amine 4 (1.2 g, 3.9 mmol), and the carboxylic acid 87 (0.97 g, 3.9 mmol) at room temperature. After stirring overnight the mixture was washed with water, dried over Na2SO4, and concentrated to give intermediate 88. This material was used in the next step without purification. ESI (MH*) m/z 539.
Analog 89 (TFA Salt). LAH (200 mg) was added to a dry THF solution (10 mL) containing 88 (2.0 g, 3.9 mmol) at room temperature. The solution was heated at reflux for 2 h. After heating, water (0.2 mL) was added followed by a 1 N solution of NaOH (0.2 mL), and a final addition of water (0.4 mL). The resulting solid was filtered washed with copious amounts of dichloromethane. A portion of this material was purified by using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient). 1H NMR (400 MHz, MeOD) δ 7.79 (s, IH), 7.36-7.42 (m, 5H), 7.25-7.32 (m, 2H), 4.14 (d, J= 13 Hz, IH), 3.90 (t, J= 11 Hz, IH), 3.67 (d, J= 11 Hz, IH) 3.32- 3.52 (m, 3H), 2.81-3.14 (m, 8H), 2.53 (m, 2H), 2.19-2.31 (m, 3H), 1.66-1.89 (m, 3H), 1.50 (m, 3H). ESI (MH+) m/z 511. General synthesis for analogs 90 and 91. Sodium triacetoxyborohydride (3 equiv.) was added to a dichloromethane solution (0.2 M) containing amine 89 (1 equiv.) and the respective ketone or aldehyde (3 equiv.) at room temperature. After stiπing overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C18 column, 10%-90% acetonitrile/water gradient).
Analog 90 (TFA Salt). 1H NMR (400 MHz, MeOD) δ 7.79 (s, IH), 7.36-7.42 (m, 5H), 7.25-7.32 (m, 2H), 4.14 (d, J= 13 Hz, IH), 3.86 (t, J= 11 Hz, IH), 3.62 (d, J= 11 Hz, IH) 3.32-3.52 (m, 3H), 2.81-3.14 (m, 13H), 2.51 (dd, J= 12, J= 14 Hz, 2H), 2.20 (m, 3H), 1.68-1.87 (m, 3H), 1.50 (m, 3H); ESI (MH+) m/z 525.
Analog 29 (TFA Salt). 1H NMR (400 MHz, MeOD) δ 7.79 (s, IH), 7.36-7.42 (m, 5H), 7.25-7.32 (m, 2H), 4.14 (d, J= 13 Hz, IH), 3.86 (t, J= 11 Hz, IH), 3.62 (d, J- 11 Hz, IH) 3.32-3.52 (m, 5H), 2.81-3.14 (m, 8H), 2.51 (m, 3H), 2.20 (m, 2H), 1.68-1.87 (m, 4H), 1.50 (m, 3H), 1.42 (m, 6H). ESI (MH+) m/z 553.
Example 79
Figure imgf000093_0001
91
92
NaBH(OAc)3
BnNHc
Figure imgf000093_0002
93
Analog 91 (TFA Salt). 1H NMR (400 MHz, MeOD) δ 7.79 (s, IH), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 4.25 (q, J= 7 Hz, 2H), 3.88 (d, J= 12 Hz, IH), 3.67 (d, J= 12 Hz, IH), 3.57 (m, IH), 3.10-3.21 (m, 2H), 3.02 (t, J= 14 Hz, IH), 2.80-2.93 (m, 3H), 2.53 (dd, J= 12, J= 14 Hz, IH), 2.19-2.25 (m, 3H), 1.99 (m, 2H), 1.86 (m, 3H), 1.62-1.70 (m. 2H), 1.49 (d, J= 7 Hz, 3H), 1.24-1.38 (m, 7H); ESI (MH+) m/z 507. Intermediate 92. SO3 pyridine complex (42 g, 2.7 mmol) was added to a
DMSO/Et3N (2.5:1, 0.2 M) solution containing compound 91 (0.34 g, 0.65 mmol) at room temperature. After stirring for 2 h, the mixture was poured into water (25 mL) and extracted with dichloromethane (3 x 75 mL). The organic layers were washed with brine, dried over Na2SO3, and concentrated to give intermediate 92. Compound 31 was used in the next step without purification: ESI (MH+) m/z 505.
Analog 93 (TFA Salt). Sodium triacetoxyborohydride (3 equiv.) was added to a dichloromethane solution (0.1 M) containing benzylamine (2 equiv.) and the ketone 92 (1 equiv.) at room temperature. After stirring overnight the solvent was removed using evaporation, and the remaining residue was purified using preparative HPLC (C 18 column, 10%-90% acetonitrile/water gradient). !H NMR (400 MHz, MeOD,) δ 7.79 (s, IH), 7.44-7.53 (m, 5H), 7.40 (d, J= 8 Hz, IH), 7.29 (d, J= 8 Hz, IH), 4.25 (m, 4H), 3.88 (t, J= 15 Hz, IH). 3.75 (d, J= 14 Hz, IH), 3.60 (d, J- 14 Hz, IH), 2.75-3.26 (m, 7H), 2.55 (m, IH), 2.41 (m, IH), 2.03-2.26 (m, 5H), 1.5-1.9 (m, 7H), 1.40-1.50 (m, 4H), 1.31 (m, 3H). ESI (MH+) m/z 596.5.
Example 80
The MCHR modulatory activity of the compounds of the invention can be assessed using the in vitro and in vivo assay methods described above. Exemplary in vitro methods include fluorometric imaging plate reader
(FLIPR) functional assays (see, e.g., G Protein-Coupled Receptors (1999) pp. 105-108 (T. Haga, G. Bernstein, eds.) CRC Press; Lembo et al. (1999) Nature Cell Biol. 1:267- 271; Saito et al. (1999) Nature 400:265-269; Wood et al (2000) Eur. J. Pharmacol. 396:1-8 and Miller et al. (1999) J. Biomol Screen. 4:249-258) and radioligand binding assays (see, e.g., Receptor Binding Techniques (1999) pp. 37-47 (M. Keen, ed.) Humana Press; Buckley et al. (1989) Mol. Pharmacol. 35:469-476; Mihara et al. (1994) J. Pharmacol. Exp. Tlier. 268:1122-1128; Newman et al. (2000) Eur. J. Pharmacol. 397:255-262 and Audinot et al. (2001) Br. J. Pharmacol. 133:371-378). Exemplary compounds demonstrated MCHR1 modulatory activity.
All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A compound having the formula (I) :
Figure imgf000096_0001
wherein
Figure imgf000096_0002
represents a single or fused aryl or heteroaryl ring; Q is -N(R)- or -N(R)-(C1-C3)alkylene-;
Figure imgf000096_0003
L1 is a bond, (Cι-C )alkylene, (C1-C4)alkylenoxy and (d- C4)alkylenamino; L is a bond, ( -C^alkylene, (C2-C4)alkenylene, (C2-C4)alkynylene, ( -C- alkylenoxy or (CrC^alkylenamino; R" is hydrogen or (C1-C8)alkyl; each R1 is independently selected from the group consisting of halogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(CrC4)alkyl, -OR5, -SR5, fluoro(CrC4)alkoxy, aryl, aryl(CrC4)alkyl, -NO2, -NR5R6, -C(O)R5, -CO2R5, - C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, - S(O)mR5, -CN and -N(R6)S(O)mR5; R2 and R3 are independently selected from the group consisting of hydrogen, halogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, - OR8, -SR8, fluoro(C1-C4)alkoxy, aryl, aryl(C C4)alkyl, -NO2, -NR8R9, =O, -C(O)R8, - CO2R8, -C(O)NR8R9, -N(R9)C(O)R8, -N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9 -S(O)mR8, -CN and -N(R9)S(O)mR8; R4 is selected from the group consisting of hydrogen, -OR1 λ, -C(O)R11, - CO2R! -C(O)NRπR12, -CN, (CrC4)alkyl and aryl; X and Y are independently selected from the group consisting of (C\- C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, -CO2R13 and -C(O)NR13R14; optionally, X and Y may be combined to form a 3-, A-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms independently selected from the group consisting of N, O and S; Z is selected from the group consisting of -OR15, -NR15R16, -NR15R18, -C(O)R15, -CO2R15, -R18, -C(O)NR15R16, -C(O)NR15R18, -SO2NR15R16, -SO2NR15R18, -NR16SO2R15, -N(R15)N(R16)SO2R17, -C(O)N(R16)OR15, hydroxy d- C8)alkyl, fluoro(Cι-C4)alkyl, heteroaryl, -C(=NOR15)NR16R17, -C(R16)=NOR15, -NR16(OR15), -C(O)NR17C(O)NR15R16, -NR17C(O)NR16C(O)R15 and -NR17C(O)NR15R16; R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16 and R17 are independently selected from the group consisting of hydrogen, (Cj-C8)alkyl, (C2- C8)alkenyl, (C2-C8)alkynyl, cyclo(C3-C6)alkyl, fluoro(Cι-C4)alkyl, hetero(C!-C4)alkyl, cyclohetero(C3-C6)alkyl, aryl and aryK -G alkyl; R is a 5- or 6-membered ring containing from 0 to 4 heteroatoms selected from the group consisting of N, O and S (e.g. tetrazole); optionally, when two R groups selected from the group consisting of R5, R6, R8, R9, R11, R12, R13, R14, R15, R16 and R17 are attached to the same nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from the group consisting of N, O and S; the subscript m is 1 or 2; and the subscript n is 0, 1 or 2.
2. The compound of Claim 1 wherein
Figure imgf000097_0001
represents a benzene ring.
3. The compound of Claim 1 wherein Q is -N(R)-.
4. The compound of Claim 1 wherein R3 is hydrogen or =O.
5. The compound of Claim 1 wherein
Figure imgf000097_0002
represents a benzene ring, R" is hydrogen and R is hydrogen.
6. A compound having the formula (II):
Figure imgf000098_0001
II or a pharmaceutically acceptable salt, hydrate, solvate or prodrug thereof, wherein L1 is a bond, (Cι-C4)alkylene, ( -C^alkylenoxy or ( - C4)alkylenamino; L is a bond, (Cι-C4)alkylene, (C2-C4)alkenylene, (C2-C4)alkynylene, (Cι-C4)alkylenoxy or
Figure imgf000098_0002
R" is hydrogen or (Cι-C8)alkyl; each R1 is independently selected from the group consisting of halogen, (Ci-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, -OR5, -SR5, fluoro(C1-C4)alkoxy, aryl, aryl(Cι-C4)al yl, -NO2, -NR5R6, -C(O)R5, -CO R5, - C(O)NR5R6, -N(R6)C(O)R5, -N(R6)CO2R5, -N(R7)C(O)NR5R6, -S(O)mNR5R6, - S(O)mR5, -CN and -N(R6)S(O)mR5; R2 is selected from the group consisting of hydrogen, halogen, (Ci- C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, -OR8, -SR8, fluoro(Cr C4)alkoxy, aryl, aryl(CrC4)alkyl, -NO2, -NR8R9, =O, -C(O)R8, -CO2R8, -C(O)NR8R9, N(R9)C(O)R8, -N(R9)CO2R8, -N(R10)C(O)NR8R9, -S(O)mNR8R9, -S(O)mR8, -CN and - N(R9)S(O)raR8; R4 is selected from the group consisting of hydrogen, -OR11, -C(O)Rn, CO2R! J , -C(O)NR1 !R12, -CN, (CrC4)alkyl and aryl; X and Y are independently selected from the group consisting of (Q- C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, -CO2R13 and -C(O)NR13R14; optionally, X and Y may be combined to form a 3-, 4-, 5-, 6- or 7- membered ring containing from 0 to 2 heteroatoms selected from the group consisting of N, O and S; Z is selected from the group consisting of -OR15, -NR15R16, -CO2R15, -R18, -C(O)NR15R16, -C(O)NR15R18, -SO2NR15R16, -SO2NR15R18, -NR16SO2R15, -N(R15)N(R16)SO2R17, -C(O)N(R16)OR15, fluoro(C1-C4)alkyl, heteroaryl, -C(-NOR15)NR16R17, -C(R16>=NOR15, -NR16(OR15), -C(O)NR17C(O)NR15R16, 84 -NR17C(O)NR16C(O)R15 and -NR17C(O)NR15R16;
85 R5, R6, R7, R8, R9, R10, Rπ, R12, R13, R14, R15, R16 and R17 are
86 independently selected from the group consisting of hydrogen, (d-C8)alkyl, (C2-
87 C8)alkenyl, (C2-C8)alkynyl, fluoro(C1-C4)alkyl, hetero(C C4)alkyl, aryl and aryl(d-
88 C4)alkyl;
89 R18 is a 5- or 6-membered ring containing from 1 to 3 heteroatoms
90 selected from the group consisting of N, O and S;
91 optionally, when two R groups selected from the group consisting of R5,
92 R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16, R17 and R18 are attached to the same
93 nitrogen atom, the R groups may be combined to form a 3-, 4-, 5-, 6- or 7-membered
94 ring containing the nitrogen atom and from 0 to 2 additional heteroatoms selected from
95 the group consisting of N, O and S;
96 the subscript m is 1 or 2; and
97 the subscript n is 0, 1 or 2.
1 7. The compound of Claim 6, wherein R4 is hydrogen.
1 8. The compound of Claim 6, wherein R" is hydrogen.
1 9. The compound of Claim 8, wherein R2 is (d-C4)alkyl or aryl.
1 10. The compound of Claim 9, wherein R1 is independently selected
2 from the group consisting of halogen, (d-C4)alkyl, fluoro(C1-C4)alkyl, -OR5,
3 fluoro(C1-C4)alkoxy, -CO2R5, -S(O)mNR5R6, -S(O)mR5 and -CN.
1 11. The compound of Claim 10, wherein R1 is halogen or fluoro(Cι-
2 C4)alkyl.
1 12. The compound of Claim 10, wherein n is 0 or 1.
1 13. The compound of Claim 12, wherein L1 is (d-C4)alkylene.
1 14. The compound of Claim 13, having the formula (III):
Figure imgf000100_0001
III wherein the subscript p is an integer of from 1 to 4.
15. The compound of Claim 13, wherein p is 1, 2 or 3.
16. The compound of Claim 15, wherein L2 is a bond.
17. The compound of Claim 16, wherein Z is -CO2R15 or -CO2NR15R16.
18. The compound of Claim 15, wherein X and Y are combined to form a 3-, A-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from the group consisting of O, N and S.
19. The compound of Claim 18, wherein X and Y are combined to form a 5- or 6-membered ring containing from 0 to 2 heteroatoms selected from the group consisting of O, N and S.
20. The compound of Claim 19, wherein X and Y are combined to form a 5- or 6-membered ring containing 0 heteroatoms, 1 nitiogen atom or 1 oxygen atom.
21. The compound of Claim 6, having the formula (IV):
Figure imgf000100_0002
IV wherein the subscript p is an integer of from 1 to 4.
22. The compound of Claim 21 , wherein p is 1 , 2 or 3.
23. The compound of Claim 22, wherein p is 2.
24. The compound of Claim 23, wherein Y is -CO2H.
25. The compound of Claim 23, wherein X and Y are combined to form a 3-, A-, 5-, 6- or 7-membered ring containing from 0 to 2 heteroatoms selected from the group consisting of O, N and S.
26. The compound of Claim 23, wherein X and Y are combined to form a 5- or 6-membered ring containing from 0 to 2 heteroatoms selected from the group consisting of O, N and S.
27. The compound of Claim 23, wherein X and Y are combined to form a 5- or 6-membered ring containing 0 heteroatoms, 1 mtrogen atom or 1 oxygen atom.
28. The compound of Claim 23, wherein X and Y are combined to form a 5- or 6-membered ring containing 0 heteroatoms, 1 nitiogen atom or 1 oxygen atom and Y is -CO2H.
29. The compound of Claim 23, wherein R2 is methyl.
30. The compound of Claim 23, wherein R1 is CF3.
31. The compound of Claim 30, wherein R1 is 9-trifluoromethyl.
32. The compound of Claim 23, wherein R1 is CF3 and R2 is methyl.
33. The compound of Claim 23, wherein R1 is CF3, R2 is methyl and Y is -CO2H.
34. The compound of Claim 33, wherein said compound is selected from the group consisting of the group consisting of:
Figure imgf000102_0001
35. A pharmaceutical composition comprising a pharmaceutically acceptable carrier or excipient and a compound of any one of Claims 1-34.
36. Use of a compound of Claim 1 or 6 for the manufacture of a medicament for treating a condition or disorder is selected from the group consisting of obesity, an eating disorder, an anxiety disorder and a mood disorder.
37. The use of Claim 36, wherein said condition or disorder is selected from the group consisting of obesity, anorexia nervosa, anxiety, panic disorder and obsessive-compulsive disorder and depression.
38. The use of Claim 36, wherein said medicament is for administration in combination with an anti-obesity agent, an antidepressant or an anxiolytic agent.
39. The use of Claim 36, wherein said medicament is for adminsitiation orally.
40. The use of Claim 36, wherein said medicament is for adminsitiation parenterally.
41. The use of Claim 36, wherein said compound modulates MCHR.
42. Use of a compound of Claim 1 or 6 for the manufacture of a medicament for modifying eating behavior.
43. The use of Claim 42, wherein said medicament is for decreasing food intake.
44. The use of Claim 42, wherein medicament is for increasing food intake.
45. Use of a compound of Claim 1 or 6 for the manufacture of a medicament for treating a condition or disorder mediated by MCHR.
46. The use of Claim 45, wherein said condition or disorder is selected from the group consisting of obesity, an eating disorder, an anxiety disorder and a mood disorder.
47. The use of Claim 46, wherein said eating disorder is anorexia nervosa.
48. The use of Claim 46, wherein said anxiety disorder is selected from the group consisting of anxiety, panic disorder and obsessive-compulsive disorder.
49. The use of Claim 46, wherein said mood disorder is depression.
50. A method for modulating MCHR, comprising contacting a cell with a compound of Claim 1 or 6.
51. The method of Claim 50, wherein said compound is an MCHR antagonist.
52. The method of Claim 50, wherein said compound is an MCHR agonist.
PCT/US2003/035543 2002-11-06 2003-11-06 Fused heterocyclic compounds WO2004043958A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
CA002505372A CA2505372A1 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
EP03779483A EP1562943A1 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
NZ540328A NZ540328A (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
BR0316070-0A BR0316070A (en) 2002-11-06 2003-11-06 Compound, pharmaceutical composition, use of a compound and method for modulating mchr
AU2003285160A AU2003285160B2 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
MXPA05004739A MXPA05004739A (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds.
EA200500770A EA009421B1 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
JP2004551872A JP4547266B2 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds
IS7833A IS7833A (en) 2002-11-06 2005-05-02 Combined heterocyclic compounds
NO20052655A NO20052655L (en) 2002-11-06 2005-06-02 Merged heterocyclic compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42445602P 2002-11-06 2002-11-06
US60/424,456 2002-11-06

Publications (1)

Publication Number Publication Date
WO2004043958A1 true WO2004043958A1 (en) 2004-05-27

Family

ID=32312811

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/035543 WO2004043958A1 (en) 2002-11-06 2003-11-06 Fused heterocyclic compounds

Country Status (16)

Country Link
US (2) US7253179B2 (en)
EP (1) EP1562943A1 (en)
JP (1) JP4547266B2 (en)
KR (1) KR20050103184A (en)
CN (2) CN101134757A (en)
AU (1) AU2003285160B2 (en)
BR (1) BR0316070A (en)
CA (1) CA2505372A1 (en)
EA (1) EA009421B1 (en)
IS (1) IS7833A (en)
MX (1) MXPA05004739A (en)
NO (1) NO20052655L (en)
NZ (1) NZ540328A (en)
PL (1) PL376659A1 (en)
WO (1) WO2004043958A1 (en)
ZA (1) ZA200503654B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7125885B2 (en) 2001-05-04 2006-10-24 Amgen Inc. Fused heterocyclic compounds
US7253179B2 (en) 2002-11-06 2007-08-07 Amgen Inc. Fused heterocyclic compounds
US7727998B2 (en) 2003-02-10 2010-06-01 Banyu Pharmaceutical Co., Ltd. Melanin-concentrating hormone receptor antagonists containing piperidine derivatives as the active ingredient
WO2012000608A3 (en) * 2010-06-30 2012-03-08 Gluesenkamp Karl-Heinz Novel beta-aminoaldehyde derivatives, process for their preparation and their chemical use as reactive intermediates
WO2013062966A3 (en) * 2011-10-24 2013-08-22 Abbvie Inc. Methanol derivatives as trpv3 modulators
US9012651B2 (en) 2011-03-24 2015-04-21 Abbvie Inc. TRPV3 modulators
US9156788B2 (en) 2010-08-10 2015-10-13 Abbvie Inc. TRPV3 modulators
CN106588812A (en) * 2016-12-07 2017-04-26 烟台大学 Method for preparing 2,4-disubstituted thiazoline compound from acyl chloride
EP3911631A4 (en) * 2019-01-14 2022-09-28 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2023242810A1 (en) 2022-06-17 2023-12-21 Richter Gedeon Nyrt. Mchr1 antagonists for the treatment of prader-willi syndrome

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070135425A1 (en) * 2001-05-04 2007-06-14 Amgen Inc. Fused heterocyclic compounds

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002004433A2 (en) * 2000-07-06 2002-01-17 Neurogen Corporation Melanin concentrating hormone receptor ligands
WO2002051809A1 (en) * 2000-12-22 2002-07-04 Schering Corporation Piperidine mch antagonists and their use in the treatment of obesity
WO2002083134A1 (en) * 2001-04-12 2002-10-24 Pharmacopeia, Inc. Aryl and biaryl piperidines used as mch antagonists
WO2002089729A2 (en) * 2001-05-04 2002-11-14 Tularik Inc. Fused heterocyclic compounds
WO2003060475A2 (en) * 2002-01-10 2003-07-24 Neurogen Corporation Melanin concentrating hormone receptor ligands: substituted 2-(4-benzyl-piperazin-1-ylmethyl)- and 2-(4-benzyl-diazepan-1-ylmethyl)-1h-benzoimidazole analogues
US20030199549A1 (en) * 2001-11-26 2003-10-23 Schering Corporation Novel piperidine based MCH antagonists for treatment of obesity and CNS disorders

Family Cites Families (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US23085A (en) * 1859-03-01 chandler
US199549A (en) * 1878-01-22 Improvement in screw-propellers
US176694A (en) * 1876-04-25 Improvement in screw-cutting die-stocks
JPS5825980B2 (en) 1976-02-12 1983-05-31 ヤマサ醤油株式会社 Cyclic nucleotide quantification method
JPS63258474A (en) 1986-08-15 1988-10-25 セドナ・ファ−マセウチカルズ・ビ−・ブイ Preparation of hydroxyl group derivative of compound containing carbazole, dibenzofuran or dibenzothiophene group
US5049655A (en) 1989-03-22 1991-09-17 The Salk Institute For Biological Studies Melanin-concentrating hormones
JP2906654B2 (en) 1989-11-28 1999-06-21 東レ株式会社 Immunosuppressant and method for producing the same
US5401629A (en) 1990-08-07 1995-03-28 The Salk Institute Biotechnology/Industrial Associates, Inc. Assay methods and compositions useful for measuring the transduction of an intracellular signal
JP3160941B2 (en) 1991-06-17 2001-04-25 東レ株式会社 Carbazole derivatives and immunosuppressants
AU662007B2 (en) 1991-07-05 1995-08-17 Smithkline Beecham Farmaceutici S.P.A. Hydroisoquinoline derivatives
ATE239732T1 (en) 1992-09-29 2003-05-15 Toray Industries INDOLE DERIVATIVE, METHOD FOR THE PRODUCTION THEREOF AND ITS MEDICAL APPLICATION
US5272146A (en) 1992-10-02 1993-12-21 The United States Of America As Represented By The United States Department Of Health And Human Services 1,2-dihydroellipticines with activity against CNS specific cancer cell lines
US5457208A (en) 1993-06-21 1995-10-10 Regents Of The University Of Minnesota Kappa opioid receptor antagonists
US5464841A (en) 1993-11-08 1995-11-07 Univ Minnesota Use of delta opioid receptor antagonists to treat immunoregulatory disorders
US5578725A (en) 1995-01-30 1996-11-26 Regents Of The University Of Minnesota Delta opioid receptor antagonists
US5849708A (en) 1995-06-06 1998-12-15 Joslin Diabetes Center, Inc. Promotion of eating behavior
EP0848060A3 (en) 1996-12-11 2000-02-02 Smithkline Beecham Corporation Novel human 11CB splice variant
US6156762A (en) 1997-01-16 2000-12-05 Toray Industries, Inc. Indolomorphinan derivative and agent for curing and preventing cerebral disorder
US6555339B1 (en) 1997-04-14 2003-04-29 Arena Pharmaceuticals, Inc. Non-endogenous, constitutively activated human protein-coupled receptors
JP2001525178A (en) 1997-12-03 2001-12-11 スミスクライン ビーチャム コーポレーション Method for searching for agonist and antagonist for human 11cb splice variant
EP1085869A4 (en) 1998-06-11 2001-10-04 Merck & Co Inc Spiropiperidine derivatives as melanocortin receptor agonists
CA2342833A1 (en) 1998-09-17 2000-03-23 Incyte Pharmaceuticals, Inc. Human gpcr proteins
GB9822158D0 (en) 1998-10-09 1998-12-02 Nycomed Imaging As Compositions
CA2356412A1 (en) 1998-12-28 2000-07-13 Takeda Chemical Industries, Ltd. Screening method
US6221613B1 (en) 1998-12-31 2001-04-24 Synaptic Pharmaceutical Corporation DNA encoding a human melanin concentrating hormone receptor (MCH1) and uses thereof
WO2000049170A1 (en) 1999-02-18 2000-08-24 Smithkline Beecham Corporation MURINE 11cby RECEPTOR
AU2573900A (en) 1999-02-19 2000-09-04 Takeda Chemical Industries Ltd. Novel g protein-coupled receptor protein and dna thereof
EP1183539A1 (en) 1999-05-19 2002-03-06 AstraZeneca AB Assays for agonists, agonists and inverse agonists of melanin concentrating hormone (mch) binding to the somatostatin-like receptor (slc-1)
WO2000075166A1 (en) 1999-06-08 2000-12-14 The Regents Of The University Of California Melanin concentrating hormone receptor
WO2001005947A1 (en) 1999-07-14 2001-01-25 Merck & Co., Inc. Melanin-concentrating hormone receptor
CA2378403A1 (en) 1999-07-26 2001-02-01 Genentech, Inc. Novel polynucleotides and method for the use thereof
WO2001007606A1 (en) 1999-07-27 2001-02-01 Smithkline Beecham Plc Axor21, a g-protein coupled receptor
CA2386474A1 (en) 1999-09-20 2001-03-29 Takeda Chemical Industries, Ltd. Melanin concentrating hormone antagonist
EP1219294A4 (en) 1999-09-20 2005-01-26 Melanin concentrating hormone antagonists
US6593108B1 (en) 1999-11-16 2003-07-15 Merck & Co., Inc. Nucleic acid molecule encoding a melanin-concentrating hormone receptor 2 polypeptide
JP2001226269A (en) 2000-02-18 2001-08-21 Takeda Chem Ind Ltd Melanin-concentrating hormone antagonist
WO2001068706A1 (en) 2000-03-15 2001-09-20 Merck & Co., Inc. Melanin concentrating hormone receptor chimeric and fusion proteins
JP2005229804A (en) 2000-03-24 2005-09-02 Astellas Pharma Inc New melanin concentrating hormone receptor
CA2408913A1 (en) 2000-05-16 2001-11-22 Takeda Chemical Industries, Ltd. Melanin-concentrating hormone antagonist
JP2004502423A (en) 2000-07-05 2004-01-29 シナプティック・ファーマスーティカル・コーポレーション DNA encoding human melanin-concentrating hormone receptor (MCH1) and use thereof
JP2004504303A (en) 2000-07-05 2004-02-12 シナプティック・ファーマスーティカル・コーポレーション Selective melanin-concentrating hormone-1 (MCH1) receptor antagonists and uses thereof
DE60143525D1 (en) 2000-07-05 2011-01-05 Takeda Pharmaceutical PROCESS FOR SCREENING MHC RECEPTOR ANTAGONISTS OR AGONISTS
PL362076A1 (en) 2000-10-20 2004-10-18 Pfizer Products Inc. Alpha-aryl ethanolamines and their use as beta-3 adrenergic receptor agonists
US20030022891A1 (en) 2000-12-01 2003-01-30 Anandan Palani MCH antagonists and their use in the treatment of obesity
JP4557492B2 (en) 2001-03-21 2010-10-06 ファーマコペイア ドラッグ ディスカバリー, インコーポレイテッド Aryl and biaryl compounds having MCH modulating activity
US6900329B2 (en) 2001-03-21 2005-05-31 Schering Corporation MCH antagonists and their use in the treatment of obesity
US6809104B2 (en) 2001-05-04 2004-10-26 Tularik Inc. Fused heterocyclic compounds
CA2448080A1 (en) 2001-05-22 2002-11-28 Neurogen Corporation Melanin concentrating hormone receptor ligands: substituted 1-benzyl-4-aryl piperazine analogues
AU2003285160B2 (en) * 2002-11-06 2009-06-04 Amgen Inc. Fused heterocyclic compounds

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002004433A2 (en) * 2000-07-06 2002-01-17 Neurogen Corporation Melanin concentrating hormone receptor ligands
WO2002051809A1 (en) * 2000-12-22 2002-07-04 Schering Corporation Piperidine mch antagonists and their use in the treatment of obesity
WO2002083134A1 (en) * 2001-04-12 2002-10-24 Pharmacopeia, Inc. Aryl and biaryl piperidines used as mch antagonists
WO2002089729A2 (en) * 2001-05-04 2002-11-14 Tularik Inc. Fused heterocyclic compounds
US20030199549A1 (en) * 2001-11-26 2003-10-23 Schering Corporation Novel piperidine based MCH antagonists for treatment of obesity and CNS disorders
WO2003060475A2 (en) * 2002-01-10 2003-07-24 Neurogen Corporation Melanin concentrating hormone receptor ligands: substituted 2-(4-benzyl-piperazin-1-ylmethyl)- and 2-(4-benzyl-diazepan-1-ylmethyl)-1h-benzoimidazole analogues

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7125885B2 (en) 2001-05-04 2006-10-24 Amgen Inc. Fused heterocyclic compounds
US7253179B2 (en) 2002-11-06 2007-08-07 Amgen Inc. Fused heterocyclic compounds
US7727998B2 (en) 2003-02-10 2010-06-01 Banyu Pharmaceutical Co., Ltd. Melanin-concentrating hormone receptor antagonists containing piperidine derivatives as the active ingredient
WO2012000608A3 (en) * 2010-06-30 2012-03-08 Gluesenkamp Karl-Heinz Novel beta-aminoaldehyde derivatives, process for their preparation and their chemical use as reactive intermediates
US9156788B2 (en) 2010-08-10 2015-10-13 Abbvie Inc. TRPV3 modulators
US9012651B2 (en) 2011-03-24 2015-04-21 Abbvie Inc. TRPV3 modulators
WO2013062966A3 (en) * 2011-10-24 2013-08-22 Abbvie Inc. Methanol derivatives as trpv3 modulators
US8772499B2 (en) 2011-10-24 2014-07-08 Abbvie Inc. TRPV3 modulators
US8772500B2 (en) 2011-10-24 2014-07-08 Abbvie Inc. TRPV3 modulators
CN106588812A (en) * 2016-12-07 2017-04-26 烟台大学 Method for preparing 2,4-disubstituted thiazoline compound from acyl chloride
EP3911631A4 (en) * 2019-01-14 2022-09-28 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2023242810A1 (en) 2022-06-17 2023-12-21 Richter Gedeon Nyrt. Mchr1 antagonists for the treatment of prader-willi syndrome

Also Published As

Publication number Publication date
MXPA05004739A (en) 2005-08-02
CN1735613A (en) 2006-02-15
JP4547266B2 (en) 2010-09-22
EA200500770A1 (en) 2005-12-29
IS7833A (en) 2005-05-02
CN1330651C (en) 2007-08-08
CN101134757A (en) 2008-03-05
JP2006508130A (en) 2006-03-09
AU2003285160B2 (en) 2009-06-04
ZA200503654B (en) 2006-09-27
KR20050103184A (en) 2005-10-27
AU2003285160A1 (en) 2004-06-03
EA009421B1 (en) 2007-12-28
NZ540328A (en) 2008-02-29
NO20052655L (en) 2005-07-26
US20080058365A1 (en) 2008-03-06
US20040147538A1 (en) 2004-07-29
BR0316070A (en) 2005-09-27
US7253179B2 (en) 2007-08-07
PL376659A1 (en) 2006-01-09
CA2505372A1 (en) 2004-05-27
EP1562943A1 (en) 2005-08-17
NO20052655D0 (en) 2005-06-02

Similar Documents

Publication Publication Date Title
US20080058365A1 (en) Fused heterocyclic compounds
US7045527B2 (en) Piperidine derivatives
EP1392298B1 (en) Fused heterocyclic compounds
US6624162B2 (en) Imidazopyridine compounds as 5-HT4 receptor modulators
BRPI0809567A2 (en) COMPOUND, PHARMACEUTICAL COMPOSITION, METHOD FOR MODULATING THE ACTIVITY OF A P2X7 RECEIVER, TO TREAT A CONDITION RESPONSIBLE FOR P2X7 RECEPTOR MODULATION IN A PATIENT, TO INHIBIT DEATH OF CELLS OF THE RETENTIAL OR RENINAL GENTEN, AND Absence of P2X7 RECEIVER IN A SAMPLE, PACKAGED PHARMACEUTICAL PREPARATION, AND USE OF A COMPOUND
AU2007249925A1 (en) Substituted azaspiro derivatives
US20100317679A1 (en) Substituted aryl-fused spirocyclic amines
EP1620435A1 (en) IMADAZOPYRIDINE COMPOUNDS HAVING 5-HT sb 4 /sb RECEPTOR AGONISTIC ACTIVITY AND 5-HT sb 3 /sb RECEPTOR ANTAGONISTIC ACTIVITY
CA2611296A1 (en) Pyrazolopyridine derivatives as inhibitors of beta-adrenergic receptor kinase 1
US6809104B2 (en) Fused heterocyclic compounds
KR20240013134A (en) Pyridine-sulfonamide derivatives as sigma ligands
EP3652157B1 (en) 1,3-diaza-spiro-[3.4]-octane derivatives
US20070135425A1 (en) Fused heterocyclic compounds
BR112020018767A2 (en) protein secretion inhibitors based on triazacyclododecansulfonamide (tcd)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/004739

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 168448

Country of ref document: IL

Ref document number: 1-2005-500859

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2005/03654

Country of ref document: ZA

Ref document number: 2505372

Country of ref document: CA

Ref document number: 376659

Country of ref document: PL

Ref document number: 200503654

Country of ref document: ZA

Ref document number: 1020057008174

Country of ref document: KR

Ref document number: 2004551872

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 540328

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 200500770

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2003779483

Country of ref document: EP

Ref document number: 2003285160

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 20038A82225

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2003779483

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0316070

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 1020057008174

Country of ref document: KR